Preclinical MRI - Methods and Protocols (PDFDrive)
Preclinical MRI - Methods and Protocols (PDFDrive)
Preclinical MRI - Methods and Protocols (PDFDrive)
Preclinical
MRI
Methods and Protocols
METHODS IN MOLECULAR BIOLOGY
Series Editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, AL10 9AB, UK
Edited by
Pilar López-Larrubia
Instituto de Investigaciones Biomédicas “Alberto Sols”, CSIC/UAM, Madrid, Spain
Editors
Marı́a Luisa Garcı́a-Martı́n Pilar López-Larrubia
BIONAND, Andalusian Centre Instituto de Investigaciones Biomédicas “Alberto Sols”
for Nanomedicine and Biotechnology CSIC/UAM
Junta de Andalucı́a Madrid, Spain
Universidad de Málaga
Málaga, Spain
Networking Research Center on Bioengineering
Biomaterials and Nanomedicine, CIBER-BBN
Málaga, Spain
Magnetic resonance imaging (MRI), the most versatile of all in vivo imaging modalities, was
born in 1973, when the Nobel Prize awardees, Paul C. Lauterbur and Peter Mansfield, at
the State University of New York and the University of Nottingham, published their
pioneering works on the use of magnetic field gradients to spatially localize the NMR signal.
Lauterbur obtained images of two water filled tubes using magnetic field gradients and
backprojection. He named this new imaging technique “zeugmatography,” derived from
the Greek word zeugma, meaning “that which is used for joining,” in reference to the joint
action of magnetic field gradients and radiofrequency to generate the image. In the same
year, Mansfield published his work demonstrating how a linear field gradient, along with the
Fourier transform, could be used to localize the NMR signal from different layers within a
solid sample, which is the basis of the slice selection used nowadays. Later, in 1977,
Mansfield and Maudsley obtained the first image of a part of the human body, a finger.
Since then, MRI has experienced a tremendous evolution thanks to the joint effort of
scientists from many different fields. Today, MRI is undoubtedly the leading technique in
diagnostic imaging. It has attracted a great deal of interest because of its unique combination
of qualities. MRI uses non-ionizing radiation, which is harmless to human tissue; offers very
high image quality, providing excellent anatomical detail; and additionally is also capable of
providing functional and metabolic information. On the negative side, it was conventionally
argued that MRI suffered from low sensitivity compared to other imaging modalities.
However, a new generation of contrast agents based on nanotechnology is making it
possible to overcome this limitation and bring MRI into the molecular imaging category.
Consequently, interest in MRI continues to grow and gain new adepts from different
fields who see MRI as a very powerful tool capable of answering many of their scientific
questions. Thus, in addition to the unquestionable growth of MRI use in clinical diagnosis,
its applications in basic and translational research have also increased enormously in recent
years, resulting in the creation of a multitude of preclinical imaging units worldwide.
This book was conceived with the idea of providing an update on a wide variety of
preclinical MRI methods and protocols to help technicians and researchers interested in this
technology to perform studies that have already been implemented by recognized experts in
the field.
The book is organized in seven parts:
Part I covers the basics of MRI physics, relaxation, image contrast, and main acquisition
sequences.
Part II describes updated methodology and protocols for diffusion, perfusion, and
functional imaging.
Part III is dedicated to in vivo spectroscopy, covering both proton and heteronuclear
spectroscopy, as well as spectroscopic imaging.
Part IV is intended to include some less common advanced techniques that we thought
might be of high interest to the readers of this book.
Parts V and VI illustrate some applications of the methods described above.
Part VII includes theoretical chapters aimed at providing relevant information on
anesthesia and contrast agents.
v
vi Preface
Finally, we would like to thank the collaboration of all the excellent experts who have
generously contributed with their chapters to the elaboration of this book.
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . xi
vii
viii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 459
Contributors
xi
xii Contributors
MRI Basics
Chapter 1
Abstract
Magnetic resonance imaging (MRI) is an imaging technique derived from radiofrequency (RF) signals of
proton that are magnetized by a strong magnetic field. These protons typically originate from water, fat, or
metabolites. The application of RF pulses is used to excite the magnetization, whereas pulsed magnetic field
gradients are used to provide spatial localization. This chapter describes the fundamental principles giving
rise to MR images. Furthermore, the connection between relaxation and image contrast is discussed.
1 Introduction
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0_1, © Springer Science+Business Media, LLC 2018
3
4 Gary V. Martinez
2.1 The Mechanics The acronym MRI is used for imaging and MRS refers to the
of MR Image spectroscopy of in vivo systems (which will be used here instead
Acquisition of NMR). MR images are made from H2O molecules in the body.
Due to their abundance, they compensate for low sensitivity.
Because the initial magnetic resonance experiments were per-
formed to probe the spectroscopic principles involved in spin phys-
ics, only a magnetic field and RF electronics were needed.
Historically, MR was developed without applied gradients, result-
ing in a non-localized spectrum. A critical distinction is that captur-
ing an MR image additionally requires the application of temporal
magnetic field gradients along three axes.
Because of the historical development, and because MR spec-
troscopy is an active and ongoing area of research, we will begin
with the explanation of an MR spectrum and proceed to the more
complicated MRI scan. In short, they both give spectroscopic
(frequency) information, but the image results from the signals
emanating from distinct and separate physical locations, whereas a
spectrum comes from the entire object. Wherever 1H density is
present, the localized signals can be displayed in terms of distances,
or coordinates on a Cartesian grid, hence forming an image. Fur-
thermore, localized images can be obtained providing spectro-
scopic (MRS) noninvasive insight into the interior of an object.
For example, common applications include images in the interior of
the knee, the brain, and in the diagnosis and therapy of cancer.
In addition to these, a hybrid type of acquisition can be per-
formed, giving rise to spectroscopic imaging (MRSI). In MRSI, it is
typically run with two spatial dimensions and 1 spectroscopic
dimension, although 3D spatial is possible. The primary examples
of these are shift imaging (CSI) [7], and echo planar spectroscopic
imaging (EPSI) [8, 9]. This chapter will not go in depth into
MRSI, but rather focus on the commonality of the basic principles
of spectroscopy and imaging.
2.1.1 Larmor Frequency When placed in a magnetic field, spin ½ MR active nuclei experi-
and RF Excitation ence a separation into two energy levels. The separation in energy
between the two levels results in a frequency difference. In MRI,
the peak center frequency is where the 1H in water resonates and is
given by:
ω0 ¼ γ H B 0 ð1Þ
Fig. 1 Fourier transform of single doubly exponential decaying signals: (a) with no sinusoidal modulation: the
on-resonance condition. (b) Fourier transform of (a). (c) Signal with two components, both shifted and hence
modulated creating an interference pattern. (d) Fourier transform of (c), showing a spectrum that is far more
simplistic than the superposition time-series signal in (a)
2.1.2 Fourier Transform The MR signal is a periodic function of time, which is quite com-
plicated and is not readily understandable by the human eye. How-
ever, there exists a transformation of these time-series data to a
8 Gary V. Martinez
2.1.3 MR Spectrum After FT, an MRS spectrum is achieved in the absence of magnetic
field gradients that encode after application of an RF pulse or
during acquisition of signal. It displays the spectroscopic character-
istics of a water molecule, such as the breadth of the peak, which is
known as the linewidth. The linewidth of a peak has an inverse
relationship with the rate of decay in the acquisition data. The FID
and spectrum are related through Fourier transformation, as shown
in Fig. 1a, b. However, due to heterogeneity in an image or
differences in the chemical shift, protons may resonate at different
frequencies, resulting in a superposed FID that is modulated by
different frequencies, which Fourier transforms into a spectrum of
broadened and/or multiple peaks. There are many applications to
understanding spectroscopic peak locations. For example, there is a
wide array of characteristic metabolite frequencies. Also, water in
tissue may experience different magnetic susceptibilities, which
could inform on the content and proximity of structural interfaces,
such as those due to bone, air, or regions of high iron content.
It is of interest to systematically introduce local magnetic field
changes so that signal from a particular position can be
MRI Physics 9
Fig. 2 Effect on resonance frequency due to a magnetic field gradient along the
z direction: there is a linear dependence of frequency and spatial position with
magnetic field
2.1.4 Magnetic Field A magnetic field gradient is defined as the derivative of the mag-
Gradient netic field as a function of spatial coordinate (Gz ¼ dBz/dx). A linear
gradient is transiently applied in a particular direction in the labo-
ratory frame (Fig. 2). If one considers a tube of pure water, it is
quite homogenous and produces a single MR peak, and if well
shimmed, results in narrow breadth (linewidth). However, in the
presence of a magnetic field gradient, a distribution of frequencies
results.
2.1.5 MRI: The Effect All of the water protons resonate at the Larmor frequency, yet the
of Magnetic Field Gradients frequency changes when turning on a magnetic field gradient. This
change is spatially dependent. If one assumes that a gradient is
applied in the z direction, then the resonance condition is given by:
ωðz Þ ¼ γ H þ γ H z G z ð5Þ
2.1.6 Slice Selection Once again considering an object with uniform 1H density, the
application of a gradient, along the z-direction, Gz, generates a
range of resonance frequencies across the object in that direction.
Linear change in the magnetic field as a function of position gives
rise to linear change in frequency. If one had a way to simulta-
neously excite multiple frequencies at once, then one could satisfy
the resonance condition (Eq. 1) for each of those frequencies, and
this would result in multiple locations being excited. This is where
the significance of the shape of an RF pulse comes into play.
There is indeed a relationship between the temporal domain
and the spectral domain. The shorter the RF pulse is, the broader
its excitation profile is, and the wider the range of frequencies that
are excited. The excitation pulse shape will dictate the shape of the
band of spins that is excited. In order to end up with a slab or slice
that is approximately a rectangular cuboid, RF pulses must have a
shape in the time domain. For example, a low flip angle sinc shaped
RF pulse has an approximately rectangular spectral profile, and
hence, excites a rectangular shaped band of spins in the object
that is being imaged (Fig. 3).
The strength of a signal is dictated in part by how many spins
were excited at that particular location, and other factors such as
slice thickness and pixel resolution (a signal equation and para-
meters used for acquisition), and these are influenced by the details
of how the RF pulse and the gradient were applied. At a given
gradient strength, the RF pulse excitation profile describes how
thick the slice is and the frequency of the pulse determines the
spatial location of the slice (Eq. 5) and Fig. 3). In particular, the
thickness of the slice can be determined from:
Δν ¼ γ H G z Δz ð6Þ
which shows that the pulse width Δν relates to the slice thickness Δz,
based on the gradient strength and gyromagnetic ratio. In a multi-
slice experiment, additional bands of spins, typically referred to as
slices, can be excited by changing the frequency of the pulse, which
serves to alter the location in the direction of Gz (Fig. 3). Once the
spins in a slice are excited, it is of interest to encode the other
directions.
Fig. 3 Schematic depiction of: (a) Spectroscopy and (b and c) 1D imaging (profile). In the spectroscopic case, a
slice-selective RF pulse excite a band of spins but is not spatially encoded after the initial RF pulse. In (b), the
spins are frequency encoded as indicated by the frequency encoding gradient being on during acquisition of
data. (c) A phase encode gradient precedes, and is turned off before, the acquisition of data. For the purposes
of comparison, the same direction is encoded in (c) as in (b). The differences between the spectroscopy and
imaging scenarios highlight the effects of encoding by a gradient and its ability to spatially localize signal
2.1.8 Phase-Encoding A phase-encoding gradient (Fig. 3c) imparts a distance and gradi-
ent dependent phase shift, where sampling is not performed simul-
taneously, which distinguishes it from frequency encoding. The end
result is the same as in a frequency-encoding gradient, but in the
phase-encoding case, there is a temporary frequency change while
gradient is turned on. After it is shut off, one could almost imagine
that the frequency information is lost; yet, there is memory of the
phase that is accumulated during the time that the gradient was on,
and this is retained by the spins in a position dependent manner.
In many pulse sequences, a phase-encode gradient is typically
applied orthogonal to a read gradient and is able to indirectly
encode over the course of the entire acquisition of multiple repeti-
tion time (TR) periods, rather than in a single shot (echo planar
imaging excepted).
The matrix size and gradient strength determine the in-plane
resolution of the image. In phase-encoding, the number of phase-
encode steps is the number of samples in this direction, and as such
dictates the resolution. Because phase-encoding occurs over the
course of the indirect dimension, it can be affected by changes
that may occur on this time-scale. Such changes include motion
and/or flow. Another limitation of the phase-encode gradient is
that sampling occurs without any hardware filtering and is suscep-
tible to aliasing artifacts. In such cases, one must either have a
sufficiently large field of view, or combine it with saturating
bands, to eliminate or reduce the amount of wrap around, which
can corrupt the image.
2.3 Basic Sequences The tools for MRI, described above, form the foundation for more
advanced sequences. They are an integral part of both gradient
echo or spin echo sequences. As such, most of the advanced
sequences in MRI are some variant of these. We will therefore
discuss these sequences to demonstrate the basic principles of
how to create an MR image. After a discussion of relaxation in
following sections, we shall describe the basic acquisition para-
meters that can be adjusted to determine relaxation and contrast
characteristics of the image, and impact how they can be applied to
provide physiological insight.
2.3.1 Gradient Echo A gradient echo (GE) sequence is shown in Fig. 4. Considering first
the RF pulse, it is composed of a single pulse, a phase encode gradi-
ent, and then acquisition of signal during the frequency encoding
gradient. If one is interested in the spectral characteristics, we
would acquire data across time. However, our immediate concern
is the formation of an image, which means that we hope to sample
k-space long before the FID has completed. We thus apply an RF
pulse in the presence of a slice-select gradient, and execute a phase-
encoding gradient, and a frequency-encoding gradient as part of a
single unit, a repetition. Within each frequency-encoding gradient,
there are data points acquired while the gradient is on (NFE). This
TR period gets repeated multiple times (NPE), where the phase-
encoding gradient gets varied each time. After scanning is com-
plete, raw k-space data contain a matrix of dimension NFE NPE,
which, together with the field of view (FOV), defines the in-plane
resolution of the scan. The through plane resolution is defined by
the slice thickness (described above).
It should be noted that a GE sequence cannot refocus the
effects of magnetic field inhomogeneity.
14 Gary V. Martinez
Fig. 4 A gradient echo sequence. The RF pulse is applied during the first slice
selection gradient, and is immediately followed by a negative refocusing lobe of
half the slice select area. A single element of the phase-encode gradient is
shown. Throughout the entire acquisition of a scan, the phase-encode gradient
iterates to different values that range from positive to negative. The read gradient
is preceded by a dephasing lobe such that both sides of the echo may be
captured during the read. During the read gradient, the signal is
simultaneously being sampled. For each RF excitation, the echo time is the
time from the middle of the RF pulse to the center of k-space, which is the
maximum of the signal. The repetition time (TR) is the duration for the entire unit.
Note that the gradients are depicted as rectangles, but in reality are closer to
trapezoids
2.3.2 Spin Echo The spin echo (SE) was first described by Hahn [10]. A spin echo
imaging sequence is shown in Fig. 5. The main difference between
the SE is that it can refocus the effects of magnetic field inhomoge-
neity. Unlike the GE, the spin echo sequence decays according to
T2, which is often much longer than T2*, meaning that it can be
used to increase the TE to large times (40–90 ms), which might
otherwise not be feasible. In fact, differences in T2 values at much
longer TE values can be interrogated with T2-weighted imaging,
which provides rich contrast. A weakness of it is that it is not
sensitive to short echo times as the time required to execute an
additional RF pulse and gradient makes the minimum TE signifi-
cantly longer than that for a GE sequence.
Fig. 5 A spin echo sequence. The overall design is similar to the gradient echo
pulse sequence; however, in brief there are some important distinctions. There
are two RF pulse that make up a spin echo, which is an RF phenomenon. The
first RF pulse is a 90 pulse followed by a delay (TE/2) and then a 180 pulse.
Acquisition of data is timed such that the center of k-space, a gradient induced
phenomenon, coincides with the spin echo maximum at TE. The time to repeat
this entire unit is the repetition time, TR
3.1.1 Longitudinal Magnetization is aligned with the main static magnetic field B0, and
Relaxation by convention is set to point along the z-axis. After excitation on
resonance with an RF pulse, the return to longitudinal thermal
equilibrium is well described by a relaxation time constant T1, in
Eq. (6).
d 1
M z ¼ ðM z M 0 Þ ð6Þ
dt T1
where Mz is the magnetization in the z direction and M0 is its
equilibrium value. Assuming a 90 pulse, the solution to this is
given by Eq. (7).
M z ðt Þ ¼ M 0 1 e t=T 1 ð7Þ
3.1.2 Transverse After excitation by a 90 pulse, relaxation in the transverse plane is
Relaxation governed by the equations:
d M xy
M xy ðt Þ ¼ ð10Þ
dt T2
This results in transverse signal given by:
M xy ¼ M 0 e t=T 2 ð11Þ
3.2 Acquisition The acquisition parameters are critical for the appearance of the MR
Parameters image, and the kind of contrast information that can be extracted
from it. To adequately set acquisition parameters in a useful fashion,
it helps to be aware of the relaxation behavior of the body parts that
are being imaged, and how these relate to the pulse sequence that is
being used. There are some basic principles required to design an
imaging experiment, and here we summarize a few.
In Figs. 4 and 5, the pulse sequence timing diagrams indicate
the highly relevant acquisition parameters TR and TE. These have
profound influence on the contrast that is achieved. TR dictates
how frequently the sequence is executed and has profound influ-
ence on how the T1 of each region affects the image. In fact, varying
the value of TR is how one achieves either a T1-weighted image, or
a proton density (PD) image. Assuming a small value of TE, a small
TR results in a T1-weighted image, and large TR yields a proton
density. When a T1-weighting sequence is acquired, those protons
with a long T1 become saturated and are weak, whereas those with a
short T1 recover much more quickly and appear bright. This results
in variations in intensity across the image based on the value of T1.
For example, after a CA has been administered, those regions that
have high concentrations will have a short T1, and hence will appear
bright.
In contrast, when a TR is chosen to be much longer than the
T1-value of the longest T1 region (typically 5T1), then the result is a
proton density image. Although the signal is strong, the distinction
between T1 is lost, and these images typically display poor contrast.
In addition, the large TR results in a scan time that is relatively long.
On the other hand, choosing a large value of TE attenuates the
signal of regions that have short T2. Therefore, with knowledge of
MRI Physics 19
References
1. Ardenkjær-Larsen JH, Fridlund B, Gram A, 6. Lauterbur PC (1973) Image formation by
Hansson G, Hansson L, Lerche MH, induced local interactions. Examples employ-
Servin R, Thaning M, Golman K (2003) ing nuclear magnetic resonance. Nature
Increase in signal-to-noise ratio of >10,000 242:190–191
times in liquid-state NMR. Proc Natl Acad Sci 7. Pykett I, Rosen B (1983) Nuclear magnetic
100(18):10158–10163 resonance: in vivo proton chemical shift imag-
2. Golman K, Lerche M, Pehrson R, Ardenkjaer- ing. Work in progress. Radiology 149
Larsen JH (2006) Metabolic imaging by hyper- (1):197–201
polarized 13C magnetic resonance imaging for 8. Mansfield P (1984) Spatial mapping of the
in vivo tumor diagnosis. Cancer Res 66 chemical shift in NMR. Magn Reson Med 1
(22):10855–10860 (3):370–386
3. Rabi II, Zacharias JR, Millman S, Kusch P 9. Posse S, DeCarli C, Le Bihan D (1994) Three-
(1938) A new method of measuring nuclear dimensional echo-planar MR spectroscopic
magnetic moment. Phys Rev 53(4):318 imaging at short echo times in the human
4. Bloch F (1946) Nuclear induction. Phys Rev brain. Radiology 192(3):733–738
70(7–8):460 10. Hahn EL (1950) Spin echoes. Phys Rev 80
5. Damadian R (1971) Tumor detection by (4):580
nuclear magnetic resonance. Science 171
(3976):1151–1153
Chapter 2
Abstract
Magnetic resonance images are obtained by a combination of different radiofrequency pulses and gradient
waveforms applied to the subject inside a magnetic field. There are multiple pulse sequences used in clinical
and preclinical studies adjusted to whatever physician or researches want to analyze, from basic anatomic
images to accurate diagnostic techniques as diffusion, perfusion, or functional imaging. In this chapter, we
present the most used radiofrequency pulse combinations of the two groups of sequences available in
magnetic resonance imaging: spin-echo and gradient-echo sequences.
Key words Magnetic resonance imaging, Pulse sequences, Spin echo, Gradient echo
1 Introduction
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0_2, © Springer Science+Business Media, LLC 2018
21
22 Daniel Calle and Teresa Navarro
Fig. 1 Scheme of a basic pulse sequence diagram. First trace corresponds to the
radiofrequency pulse train (α) and the lower ones to the slice selection gradient
(second), the variable phase encoding gradient (third), the readout gradient of the
echo (forth) and the echo signal (fifth trace). TE is the time between the excitation
pulse (α) and the echo signal
measured in mm (or cm), matrix size (Mtx), is the phase steps plus
the points of the echo signal and corresponds with the number of
pixels of the image; number of averages (Nex), etc. The final
duration of a spin echo (SE) or gradient echo (GE) sequence, in
absence of any acceleration factor, corresponds with the following
equation:
Acquisition time ¼ T R∙Phase Steps ∙N ex ð1Þ
where E is the electric field and σ and ρ are the electric conductivity
and density of the biological tissue. The magnetic field strength and
the frequency of the RF pulses influence directly in the SAR
increase. Clinical magnetic fields entails low SAR, but it is necessary
to do a good pulse sequence design if short TR or high flip angles
are used because of possible temperature increases which can even-
tually produce burns to the patients. In general, commercial MRI
scanners calculate the SAR for each sequence design preventing the
critical values, which depends of region of the body to be studied,
and are defined by international committees (FDA or IEC) which
established a maximum SAR value of 4 W/kg for the whole body.
Depending on the RF sequence parameters, the image
obtained can be weighted in T1 (longitudinal relaxation time), T2
(transverse relaxation time), proton density (PD), or T2*. T1 is
defined as the time that the longitudinal magnetization takes to
recover the original value. A sequence with short TR and short TE
results in a T1-weighted image. T2 represents the time that the
transversal magnetization takes to disappear. A T2-weighted image
is obtained with long TR and TE. A proton density image repre-
sents the water content (water hydrogens) and is obtained with
long TR and short TE. T2* is related to T2. It represents the loss of
phase coherence due to magnetic inhomogeneities and produces a
faster decay of T2. T2*-weighted images can be obtained using
Gradient Echo sequences. Tissues have different magnetic relaxa-
tion properties, so, depending on the image sequence, they will
have different intensities. As an example, in a T1-weighted image,
liquid (cerebrospinal fluid, edemas, etc.) is darker than other tissues
because it has not restrictions to recover the longitudinal magneti-
zation, it has a long T1, while in a T2-weighted image liquid is
brighter than other tissues, it has a long T2 (Fig. 2).
24 Daniel Calle and Teresa Navarro
Fig. 2 T1- and T2-weighted images of mice brain. Note brighter zones (long T2) in the T2-weighted image
corresponding to cerebrospinal fluid (red arrows). The same area is dark in the T1-weighted image because its
T1 value is long
1.1 Contrast Agents The magnetic relaxation properties of a tissue (T1, T2, T2*, proton
density) can be altered through the use of pharmacochemical com-
pounds known as contrast agents (CAs). MR contrast agent acts
reducing the relaxation times of tissue water (T1 or T2). Two types
of MRI contrast agents have been developed in based on the effect
on the signal intensity. They can be positives (signal hyperintensity
or T1 enhancement) or negatives (signal hypointensity or T2
enhancement), also called positive and negative contrast agents,
respectively. CAs for MRI, involve mainly Gd(III) chelates, able
to enhance water relaxation in those tissues where they accumulate
[1]. Gd(III) is used because it has seven unpaired, slow relaxing
electrons, and depicts the largest magnetic moment among the rare
earth series. The ligands most frequently used are linear chelates
derived from diethylenetriaminepentaacetic acid (DTPA) or cyclic
chelates derived from the tetraaza macrocycles or cyclen derivatives
(DOTA). In all these cases, the ligand provides eight binding sites
anchoring the Gd(III), leaving free one the nine chelating sites of
the metal, for water contact. The contact between water molecules
in the tissue with the Gd(III), and the fast exchange of this water
molecule with the bulk solution, reduces very significantly the
relaxation times of the tissue, resulting in clearly enhanced image
intensity in those regions containing the chelate [1]. It is very useful
to distinguish between tissues with similar relaxation times like
intracranial tumors and cerebral parenchyma. Gadolinium chelates
do not cross the intact blood–brain barrier (BBB), but when a
tumor is growing in the brain, usually a disruption of the BBB
take place allowing the extravasation of the CA that causes a signal
increase in the MR image becoming easily to distinguish tumor
from edema or healthy tissue (Fig. 3).
The use of other lanthanides as Dy(III) may transform the same
chelates in T2 enhancing probes due to the inherent T2 relaxing
properties of this element [2].
Basic MRI Sequences 25
Fig. 3 T1-weighted images of a glioblastoma rat model before (left) and after (right) the intravenous
administration of a contrast agent (gadolinium chelate). It can be appreciated the areas where the blood
brain barrier is disrupted in the tumor and the contrast agent is captured reducing the T1 value and therefore
increasing the intensity of the damaged brain region
1.2 MRI Sequences There are many pulse sequence classifications in MRI. In this
chapter, we are going to talk about the two main groups of MRI
pulse sequences depending on how the echo signal is acquired: spin
echo and gradient echo sequences. Considering both of them,
there are numerous variations that include (or not) modifications
to accelerate the image acquisition and reduce the acquisition time.
26 Daniel Calle and Teresa Navarro
Table 1
Equivalent MR manufacturer’s abbreviations with the corresponding type of sequence
The spin echo pulse sequence is one of the most commonly used in
MRI and historically it was the first sequence to be used in this
imaging technique. It was described in 1950 by Erwin Hahn [10],
so a spin echo is sometimes also referred as a Hahn echo. A spin
echo consists in an echo formed after application of two pulses, an
excitation pulse and a refocusing pulse, separated by suitable time
delays. This sequence, like the Carr Purcell Meiboom Gill sequence
that will be described later in this section, was first designed for
NMR in the 1950s and adapted for MRI later in the 1970s.
In the pulse sequence diagram (Fig. 4), a 90 (π/2) excitation
RF pulse is applied followed by one (or more) 180 (π) refocusing
RF pulse to generate signal echoes when the spins are again in
phase. The 90 pulse rotates the nuclear magnetization down into
Basic MRI Sequences 27
Fig. 4 Scheme of a spin echo (SE) sequence. Top trace corresponds to the
radiofrequency pulse train (π/2, π) and the lower ones correspond to the slice
selection gradient, the variable phase encoding gradient, the readout gradient of
the echo and the echo signal. TE is the time between the π/2 pulse and the echo
the transversal plane (XY plane). Then, the transverse nuclear mag-
netization begins to dephase due to the T2* dephasing (some spins
slow down and others speed up due to the field inhomogeneities).
A 180 pulse is then applied to rephase the nuclear magnetization
(partially) and, after a time delay equal to TE/2, produces a signal
called echo (the 180 pulse is exactly located in the half way
between the echo and the 90 pulse). Both pulses are applied in
conjunction with the slice selection gradient. The phase encoding
gradient is applied between the 90 and 180 pulses. Finally, the
frequency encoding gradient is applied during the readout of the
echo at time TE and the first raw of the K-space is filling. In order to
generate an image we have to repeat this process changing the
strength in the phase encoding gradient to fill another line in the
K-space. Normally, this process starts with large, negative phase
encoding gradient amplitudes going through zero phase encoding
gradient amplitude and finally to a large, positive phase encoding
gradient amplitude.
Other pulses besides a 90–180 combination can produce a
spin echo. Hahn employed a 90 –90 RF pulses in his original
description of spin echo. The 90–180 pulses are most commonly
used because with this combination it is obtained the higher echo
signal.
A spin echo pulse sequence produces an echo whose formation
can be compared with a horse race. First all horses are lined up at
28 Daniel Calle and Teresa Navarro
the starting lines. Once the race starts (90 pulse), the faster horses
separate from the slower horses (dephasing). After a while (in the
spin-echo sequence, TE/2, and corresponding to the 180 pulse)
the horses are transposed (refocuses), and now the faster horses are
behind the slower ones. Nevertheless, because the last horses are
the faster than the first ones, at the end of the race all horses reach
the finishing line at the same time (echo signal).
The main disadvantage of conventional spin echo sequence is
the relatively long acquisition time, especially when long TR are
required. The Carr Purcell Meiboom Gill (CPMG) sequence, first
designed by Carr and Purcell in 1954 [11] and completed by
Meiboom and Gill in 1958 [12], is a variety of spin echo pulse
sequence which is particularly useful for measuring T2. It consists of
a 90 RF pulse followed by a train of evenly spaced 180 RF pulses
applied alternately along the XY plane to acquire several echo
signals. Each echo is encoded with a phase encoding gradient.
This pack of echoes is known as an echo train, and the total number
of 180 RF pulses and echoes is called an echo train length. This is a
fast SE pulse sequence and the acquisition time is greatly reduced
with respect to a conventional spin echo sequence.
Important parameters in spin echo sequences are the TR and
the TE, which can be varied to control the contrast and, therefore,
to obtain T1-weighted, T2-weighted, or proton density images
(Fig. 5). Typical values for a 7 T equipment are:
– A short TR and short TE will give a T1-weighted image (Fig. 5,
T1 SE, TE ¼ 12 ms, TR ¼ 500 ms), a TR < 700 ms increases the
effect of T1 on image contrast and a TE < 20 ms minimizes T2
contrast. In T1-weighted images short T1 tissues appears bright
and long T1 tissues appears dark (hyposignal).
– A long TR and a short TE will give a proton density image
(Fig. 5, PD SE, TE ¼ 12 ms, TR ¼ 2000 ms). The contrast
Fig. 5 T1, T2 and proton density (PD) weighted axial images of a rat brain using spin echo sequences (MSME
and RARE) in a 7 T spectrometer. The TE/TR can be varied to control contrast in spin echo imaging
Basic MRI Sequences 29
2.1 Inversion This sequence is a variant of a spin echo sequence with the advan-
Recovery Sequence tage to null or suppress the signal from any tissue based on its
longitudinal relaxation value. It can be chosen a specific time
(inversion time, TI) value to null the signal from fat, white matter,
gray matter, cerebral spinal fluid, etc. This value it is known as the
“null point” or “zero crossing point.”
First a 180 pulse is applied, which inverts 180 the longitudi-
nal magnetization vector to the –Z axis. After the 180 pulse, the
spinning nuclei begin to relax and the net magnetization vector
passes to the transverse plane (the null point) at a time TI. At this
moment, a combination of 90 and 180 pulses are applied to
obtain an image without the signal of a specific tissue. The time
between the 180 and the 90 pulse is known as time to inversion
(TI). Figure 6 shows the inversion recovery pulse sequence.
2.1.1 STIR (Short TI The clinical evaluation, the differential diagnosis and the preclinical
Inversion Recovery) research, can be complicated by the amount of fat in the tissue. To
avoid this problem, it is essential to suppress the signal from adipose
tissue. The use of STIR sequences entails the null of the MRI signal
from fat in a T1-weighted image, and they are based on the differ-
ence in T1 between water and lipids hydrogens. Not only the fat
signal can be suppressed on a STIR sequence, other tissues with a
short T1 also will be nulled, such as proteinaceous fluid or
hemorrhage.
Basic MRI Sequences 31
2.1.2 FLAIR (Fluid In clinical environment, the use of spin echo pulse sequences in
Attenuated Inversion some anatomy regions such as spinal cord or periventricular lesions
Recovery) in which the bright pathologic areas are close to the equally bright
areas is problematic due to the high signal from cerebrospinal fluid
(CSF), which makes difficult to delineate the cord outline and
reduce the image quality due to the motion of CSF. Similar pro-
blems can take place in preclinical research. In this line, FLAIR
sequences can be used to null the fluid signal and the artifacts can
be reduced in the images to get a more accurate study.
FLAIR sequences use a long inversion time to null or reduce
the signals from fluids (like CSF, edema, urine. . .) and a long echo
time to produce a very heavy T2-weighted image. The problem is
that this can result in a very long acquisition time. Nowadays, the
combination of the FLAIR and the FSE avoid using this protocol
for routine imaging acquisition. In clinic, FLAIR pulse sequence is
very useful to detect lesions in the brain not easily distinguishable
within the subarachnoid space and brain parenchyma.
2.2 FAST (TURBO) In 1986 Henning et al. [13] developed the RARE sequence (Rapid
Spin Echo Acquisition with Relaxation Enhancement) to allow a more rapid
data acquisition with the same resolution than an image acquired
with a conventional spin echo. Nowadays it’s the most commonly
used sequence in MRI. The terms fast spin echo (FSE) or turbo
spin echo (TSE) mean the same, it depends on the manufacturer.
These are conventional spin echo sequences that use a series of
180 refocusing pulses after just one 90 excitation pulse to gener-
ate a train of echoes. The number of echoes acquired during the TR
is known as the echo train length or turbo factor (generally from
2 to 32 for routine imaging). Each echo has a different phase
encoding value (in a conventional multi-echo sequence all echoes
are collected with the same phase encoding) and this allows to
acquire multiple lines of K-space within a given repetition time.
The main advantage of this sequence is the speed. K-space is
filled faster and scan time is decreased. This technique allows
obtaining heavy T2-weighted imaging and a contrast to noise
ratio good due to the long TR (around twofold than in a conven-
tional spin echo). In this sequence, the acquisition time is equal to
the repetition time multiplied by the number of encoding steps in
the phase direction and divided by the turbo factor/echo train
length.
32 Daniel Calle and Teresa Navarro
Fig. 7 Gradient echo pulse (GE) sequence diagram. In this sequence, the RF pulse
angle is variable depending on the type of image weighting to be obtained. Here,
the echo signal is acquired using a dephasing readout gradient
Fig. 8 T1, T2*, and proton density (PD) weighted axial images of a rat brain using gradient echo sequences
(FLASH and GEFC) in a 7 T spectrometer. Varying the flip angle and TE/TR different MRI weighting can be
obtained
Table 2
Typical values of Flip Angle, TE, and TR for T1, T2*, and proton density weighting using a GE sequence
4 Preclinical Applications
MRI is the best choice for the study of diseases located in the
structures of the nervous and musculoskeletal systems because
provides excellent contrast between soft tissues and has a high
spatial resolution. There are specific protocols to obtain images
Basic MRI Sequences 35
References
16. Henninger B, Kremser C, Rauch S, Eder R, 19. Qin Y, Zhu W, Zhan C, Zhao L, Wang J,
Judmaier W, Zoller H, Michaely H, Schocke Tian Q, Wang W (2011) Investigation on pos-
M (2013) Evaluation of liver fat in the presence itive correlation of increased brain iron deposi-
of iron with MRI using T2* correction: a tion with cognitive impairment in Alzheimer
clinical approach. Eur Radiol 23 disease by using quantitative MR R2’ mapping.
(6):1643–1649. https://doi.org/10.1007/ J Huazhong Univ Sci Technol Med Sci 31
s00330-012-2745-2 (4):578–585. https://doi.org/10.1007/
17. Kolnagou A, Natsiopoulos K, Kleanthous M, s11596-011-0493-1
Ioannou A, Kontoghiorghes GJ (2013) Liver 20. Mihai G, He X, Zhang X, McCarthy B, Tran T,
iron and serum ferritin levels are misleading for Pennell M, Blank J, Simonetti OP, Jackson RD,
estimating cardiac, pancreatic, splenic and total Raman SV (2011) Design and rationale for the
body iron load in thalassemia patients: factors study of changes in iron and atherosclerosis risk
influencing the heterogenic distribution of in Perimenopause. J Clin Exp Cardiol 2:152.
excess storage iron in organs as identified by https://doi.org/10.4172/2155-9880.
MRI T2*. Toxicol Mech Methods 23 1000152
(1):48–56. https://doi.org/10.3109/ 21. Greenberg SM, Vernooij MW, Cordonnier C,
15376516.2012.727198 Viswanathan A, Salman RA-S, Warach S, Lau-
18. Barzin M, Kowsarian M, Akhlaghpoor S, ner LJ, Van Buchem MA, Breteler MM, Group
Jalalian R, Taremi M (2012) Correlation of MS (2009) Cerebral microbleeds: a guide to
cardiac MRI T2* with echocardiography in detection and interpretation. Lancet Neurol 8
thalassemia major. Eur Rev Med Pharmacol (2):165–174
Sci 16(2):254–260
Part II
Abstract
The use of magnetic resonance imaging (MRI) for studying the cerebral perfusion mechanisms is well
proved and contrasted in the clinical and research setups. This methodology is a promising tool in assessing
numerous brain diseases like intracranial tumors, neurodegeneration processes, mental disorders, injuries
and so on. In the preclinical environment, perfusion MRI offers a powerful resource for characterizing
pathological models and specially identifying biomarkers to monitor the illness and validate the efficacy of
therapeutical approaches. This chapter presents the theoretical bases and experimental protocols of dynamic
susceptibility contrast MRI acquisitions for developing perfusion MRI studies in small animals.
Key words Preclinical MRI, Brain perfusion, Dynamic susceptibility contrast, Bolus tracking, Cere-
bral blood flow, Cerebral blood volume, Mean transit time, Animal model
1 Introduction
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0_3, © Springer Science+Business Media, LLC 2018
41
42 Pilar López-Larrubia
1.2 “Bolus Tracking” The use of exogenous contrast agents allows obtaining unique
Perfusion MRI information of tissular perfusion and so, improving the edge of
regions with a deficient blood flow in diseased conditions. Com-
pounds that include gadolinium (Gd), dysprosium (Dy), or iron
(Fe) provoke a regional decrease of the signal intensity in T2- and
T2*-weighted images. This phenomenon can be exploited to indi-
rectly valuate the perfusion profiles in tissues and calculate hemo-
dynamic parameters to be used as biomarkers of diseases. The MRI
methodology to do that is generally called bolus tracking technique
[9], or hemodynamic-weighted magnetic resonance imaging, and
takes part of the dynamic susceptibility contrast techniques
(DSC) [10].
To map perfusion entails the ability of visualizing alterations in
the MRI signal during the transit of a bolus of CA—quickly
injected—with ultra-fast image acquisition sensitive to T2*. The
method relies in the ability of detecting magnetic susceptibility
changes in the microvasculature employing very rapid imaging to
capture the circulation of a bolus of CA rapidly injected. In the
brain, the first-pass extraction of the agent is zero when the blood–-
brain barrier (BBB) is intact, and the intravascular compartmentali-
zation of the contrast creates strong, microscopic susceptibility
gradients. These microscopic gradients cause dephasing of spins
as the CA diffuse among them, and in terms of signal loss, all
water molecules in the proximity of the vessels will be affected
Dynamic Susceptibility Contrast MRI 43
Fig. 1 Representative scheme of a bolus tracking perfusion MRI study. A pixel-by-pixel representation of the
signal intensity with time, assesses the passage of the contrast agent through the cerebral microvasculature.
Basing on the relationship between T2* (or relaxation R2*) and the CA concentration (Ct), the data can be
converted into a concentration-time curve that fitted to a gamma function allows the measurement of
hemodynamic parameters like CBV (that corresponds to the area under the curve) at any pixel or region of
interest
2 Materials
R
τ ΔR∗2 ðτ Þdτ
MTT ¼ R ∗ ð3Þ
ΔR2 ðτÞdτ
CBV
CBF ¼ ð4Þ
MTT
Later on, sections of interest are manually selected in the maps
and data statistically analyzed, with image analysis free software like
Image J and data statistically analyzed with adequate programs (see
Subheading 3.4).
Table 1
Preclinical MRI system and associated equipment/devices for developing the protocols described in
this chapter
3 Methods
3.1 Vessel For these studies, a vessel of the animal has to be cannulated for a
Cannulation quick administration of the contrast agent during acquisition.
Although an artery could be the best choice because of the faster
reaching of the brain, especially carotid arteries, in practice, due to
the higher difficulty of the procedure, a tail vein is usually the
selected one (Fig. 3, up). Besides, the cannulation of a tail vessel
is less invasive for the animals allowing the surveillance of them after
the study. Intravenous cannulation of a rat lateral tail vein can be a
challenging procedure, even more in a mouse [17]. The method
entails:
1. Place the animal in an anesthetic box induction with isoflurane
(or any other inhalational anesthesia) 3–4% in oxygen (or any
other gas carrier) at a flow of 1 L/min and then place it in an
appropriate surface with a mask supplying a lower amount of
anesthesia (1–1.5%).
Fig. 3 Cannulation of a tail vein in a rat (or mouse). Up: Schematic picture of a transversal section of the tail
showing the vessels to cannulate; down: cannulation of the dorsal venin in a rat with a catheter
Dynamic Susceptibility Contrast MRI 49
3.2 Animal Before starting MRI studies, make sure to observe all the healthy
Positioning and safety rules in a high magnetic field room:
in the Scanner
1. Not approximate to the magnet with any metal, either in the
animal or in the operator.
2. Check that the oxygen level sensors are operative.
3. Verify that vaporizer and oxygen bottle are full enough and the
halogenated gas filter is working properly.
After that:
1. Place the animal in the appropriate bed/holder—rat or mouse
adapted—usually in the prone position, over a heated blanket
(or any other system to maintain the body temperature at
around 37 C). The bed has to have an anesthesia inlet and a
1 L/min flow of 1–1.5% isoflurane/oxygen maintained during
the whole study. Make sure that anesthesia is flowing without
impediments. A thermometer sensor has to be inserted in the
rectum, to control the body temperature, and breathing sen-
sors allocated under the animal chest to monitor the animal
status and modulate the plane of anesthesia. Respiration rate
should be between 40 and 60 rpm (respirations per minute) to
minimize motion artifacts in images (see Note 2). The animal
head has to be immobilized during acquisition because
50 Pilar López-Larrubia
3.3 MRI Acquisition 1. Identify the animal and the study in the operating software of
Protocols the system. Select the protocols to be acquired starting with a
rapid acquisition to check the correct positioning of the brain
in the magnet. Prior the first scan, an automatic or manual
adjustment of some parameters has to be achieved:
(a) Tuning and matching the coil—when it is possible—to
adjust its frequency of resonance and impedance accord-
ing to the animal.
(b) Shimming the magnet to improve its homogeneity.
(c) Determining the RF power for the 90 excitation pulse.
(d) Adjusting the basic frequency of the system.
(e) Calculating the optimal receiver gain.
2. Acquire structural images to image the brain anatomy and use
them as reference. Normally a T2W sequence is employed for
obtaining anatomical information. If a T2 study is destined to
cover the whole brain, do not forget to acquire a new one with
the same geometry—number of slices, slice thickness, and
separation—as the DSC acquisition. These T2W images will
be used to superimpose the parametric perfusion maps and
better identify the cerebral regions. (Acquisition parameters
are shown in Table 2.)
3. DSC studies are performed using ultra-fast gradient-echo
methods (EPI-T2*). These sequences that employ a single-
shot echo-planar acquisition module to get the necessary tem-
poral resolution, are highly sensitive to magnetic field inhomo-
geneities. So, a very good shimming to improve the magnetic
field homogeneities is a prerequisite for successful. There are
several automatic methods to achieve that:
(a) Optimizing the shimming of the whole sample covered by
the coil—it is a global approach—by varying the first and
second shim coil values and making adjustment according
to the signal response.
Dynamic Susceptibility Contrast MRI 51
Table 2
Normal acquisition parameters used in the sequences employed to acquire a perfusion DSC MRI
experiment
3.4 Post-Processing 1. Data can be processed and analyzed with a dedicated tool
and Data Analysis incorporated to the software of the MRI spectrometer. Data
are also read with different analysis software like Matlab (The
MathWorks Inc., Natick, MA, 2000) or IDL (Iterative Data
Language, Research System, Boulder, CO, USA) and managed
either with commercial or homemade packages. Images can be
managed as raw-data or DICOM format (Digital Imaging and
Communication in Medicine) that is a standard for handling,
storing, printing, and transmitting information in medical
imaging (http://dicom.nema.org).
2. Images are computed by fitting the signal to the adequate
equations (Eqs. 2–4) in a pixel-by-pixel bases. Depending on
the software, acquisition parameters like number of slices,
Dynamic Susceptibility Contrast MRI 53
Fig. 4 Screen shot of a DSC study processing with a homemade application (MyMapAnalyzer) developed in
Matlab
54 Pilar López-Larrubia
3.5 CBF, CBV, To illustrate the protocol, some examples of parametric images
and MTT Maps obtained from perfusion DSC studies in our lab are shown in Fig. 5.
4 Notes
Fig. 5 CBF, CBV, and MTT parametric images. Maps were obtained from perfusion MRI studies of a healthy
animal (upper images) and a rat with a C6 glioma in the right hemisphere (lower images). In both cases the
bolus tracking technique was employed 250 μL of a 0.3 M gadolinium chelate (Gd-DTPA) solution rapidly
injected 10 s after starting acquisition. MR images were computed with homemade software (MyMapAnalyzer)
Acknowledgements
References
1. Calamante F, Thomas DL, Pell GS, Wiersma J, 5. Stewart G (1894) Researches on the circulation
Turner R (1999) Measuring cerebral blood time in organs and on the influences which
flow using magnetic resonance imaging techni- affect it: I–III. J Physiol 15:1–27
ques. J Cereb Blood Flow Metab 19 6. Kim SG (2012) Perfusion MR imaging: evolu-
(7):701–735. https://doi.org/10.1097/ tion from initial development to functional
00004647-199907000-00001 studies. NeuroImage 62(2):672–675.
2. Le Bihan D (1995) Diffusion, perfusion and https://doi.org/10.1016/j.neuroimage.
functional magnetic resonance imaging. J Mal 2012.01.015
Vasc 20(3):203–214 7. MacDonald ME, Frayne R (2015) Cerebrovas-
3. Le Bihan D (1992) Theoretical principles of cular MRI: a review of state-of-the-art
perfusion imaging. Application to magnetic approaches, methods and techniques. NMR
resonance imaging. Investig Radiol 27(Suppl Biomed 28(7):767–791. https://doi.org/10.
2):S6–11 1002/nbm.3322
4. Jahng GH, Li KL, Ostergaard L, Calamante F 8. Buxton RB (2012) Dynamic models of BOLD
(2014) Perfusion magnetic resonance imaging: contrast. NeuroImage 62(2):953–961.
a comprehensive update on principles and tech- https://doi.org/10.1016/j.neuroimage.
niques. Korean J Radiol 15(5):554–577. 2012.01.012
https://doi.org/10.3348/kjr.2014.15.5.554 9. Ostergaard L (2005) Principles of cerebral per-
fusion imaging by bolus tracking. J Magn
Dynamic Susceptibility Contrast MRI 57
Reson Imaging 22(6):710–717. https://doi. 17. Shimizu S (2004) Routes of administration. In:
org/10.1002/jmri.20460 Hans J Hedrich, GBaPP (ed) The laboratory
10. Koshimoto Y, Yamada H, Kimura H, mouse. Academic Press, London, p 14
Maeda M, Tsuchida C, Kawamura Y, Ishii Y 18. Gruetter R (1993) Automatic, localized in vivo
(1999) Quantitative analysis of cerebral micro- adjustment of all first- and second-order shim
vascular hemodynamics with T2-weighted coils. Magn Reson Med 29(6):804–811
dynamic MR imaging. J Magn Reson Imaging 19. Kanayama S, Kuhara S, Satoh K (1996) In vivo
9(3):462–467 rapid magnetic field measurement and shim-
11. Barbier EL, Lamalle L, Decorps M (2001) ming using single scan differential phase
Methodology of brain perfusion imaging. J mapping. Magn Reson Med 36(4):637–642
Magn Reson Imaging 13(4):496–520 20. Perez-Carro R, Cauli O, Lopez-Larrubia P
12. Conturo TE, Akbudak E, Kotys MS, Chen (2014) Multiparametric magnetic resonance
ML, Chun SJ, Hsu RM, Sweeney CC, Mark- in the assessment of the gender differences in
ham J (2005) Arterial input functions for a high-grade glioma rat model. EJNMMI Res 4
dynamic susceptibility contrast MRI: require- (1):44. https://doi.org/10.1186/s13550-
ments and signal options. J Magn Reson Imag- 014-0044-4
ing 22(6):697–703. https://doi.org/10. 21. Garcia-Palmero I, Lopez-Larrubia P, Cerdan S,
1002/jmri.20457 Villalobo A (2013) Nuclear magnetic reso-
13. Borges AR, Lopez-Larrubia P, Marques JB, nance imaging of tumour growth and neovas-
Cerdan SG (2012) MR imaging features of culature performance in vivo reveals Grb7 as a
high-grade gliomas in murine models: how novel antiangiogenic target. NMR Biomed 26
they compare with human disease, reflect (9):1059–1069. https://doi.org/10.1002/
tumor biology, and play a role in preclinical nbm.2918
trials. AJNR Am J Neuroradiol 33(1):24–36. 22. Yankam Njiwa J, Ratering D, Baltes C, Rudin
https://doi.org/10.3174/ajnr.A2959 M (2010) Increasing temporal resolution of
14. López-Larrubia P, Cañadillas-Cárdenas E, DSC perfusion MRI using the analytic image
Metelo AM, Arias N, Martı́nez-Maestro M, concept. MAGMA 23(4):251–261. https://
Salguero LA, Cerdán S (2011) Magnetic reso- doi.org/10.1007/s10334-010-0224-9
nance imaging of gliomas. In: Chen CC 23. Yin J, Sun H, Yang J, Guo Q (2015) Auto-
(ed) Advances in the biology, imaging and mated detection of the arterial input function
therapies for glioblastoma. InTech, Rijeka, pp using normalized cut clustering to determine
225–250 cerebral perfusion by dynamic susceptibility
15. Liu W, Wang B, Wolfowitz R, Yeh PH, Nathan contrast-magnetic resonance imaging. J Magn
DE, Graner J, Tang H, Pan H, Harper J, Reson Imaging 41(4):1071–1078. https://
Pham D, Oakes TR, French LM, Riedy G doi.org/10.1002/jmri.24642
(2013) Perfusion deficits in patients with mild 24. Duhamel G, Schlaug G, Alsop DC (2006)
traumatic brain injury characterized by Measurement of arterial input functions for
dynamic susceptibility contrast MRI. NMR dynamic susceptibility contrast magnetic reso-
Biomed 26(6):651–663. https://doi.org/10. nance imaging using echoplanar images: com-
1002/nbm.2910 parison of physical simulations with in vivo
16. Duong TQ (2013) Magnetic resonance imag- results. Magn Reson Med 55(3):514–523.
ing of perfusion-diffusion mismatch in rodent https://doi.org/10.1002/mrm.20802
and non-human primate stroke models. Neurol
Res 35(5):465–469. https://doi.org/10.
1179/1743132813Y.0000000211
Chapter 4
Abstract
Magnetic resonance imaging has been utilized as a quantitative and noninvasive method to image blood
flow. Arterial spin labeling (ASL) is an MRI technique that images blood flow using arterial blood water as
an endogenous tracer. Herein we describe the use of ASL to measure cerebral blood flow completely
noninvasively in rodents, including methods, analysis, and important considerations when utilizing this
technique.
Key words Arterial spin labeling, Blood flow, Perfusion, Cerebral blood flow, Magnetic resonance
imaging, Rodent
1 Introduction
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0_4, © Springer Science+Business Media, LLC 2018
59
60 Eric R. Muir
2 Materials
2.2 MRI 1. Preclinical 7 T MRI scanner (Bruker, Billerica, MA, USA) (see
Note 3).
2. Magnetic gradients, 150 G/cm with 6 cm inner diameter
gradient insert for mice, 40 G/cm with 12 cm inner diameter
for rats (see Note 3).
3. Animal holder equipped with ear and tooth bars.
4. Radiofrequency (RF) transmitter/receiver coil for brain imag-
ing (see Note 4).
5. RF transmitter coil for arterial spin labeling at the chest or neck
position for ASL (see Note 5).
6. Switch box to actively detune RF coils for ASL (see Note 6).
3 Methods
3.1 Animal 1. Place the animal in an anesthesia induction chamber with 4–5%
Preparation isoflurane delivered to the box in air or oxygen at 1.5–2 L/min
until the animal loses its righting reflex. Different anesthetics
can be used if preferred, but be aware this will affect blood flow
(see Note 2).
2. Remove the animal from the induction chamber, place a nose
cone over its nose, switch isoflurane delivery to the nose cone,
and reduce the isoflurane to 2%. Check paw reflexes by pinch-
ing the paws and looking for reflex withdrawal to ensure ade-
quate anesthesia.
3. Put eye ointment on both eyes of the animal to prevent the
cornea from drying out while under anesthesia.
4. Place the animal into a head holder with ear bars and tooth bar
to immobilize the skull and minimize animal motion through-
out scanning procedures.
62 Eric R. Muir
3.2 MRI 1. The animal should be positioned so that the labeling coil is at
the neck for rats or at the upper chest for mice, to allow labeling
of blood flowing to the brain (see Note 10).
2. Position the imaging RF coil as close to the center of the brain
as possible (see Note 11).
3. Move the animal into the MRI scanner.
4. If the RF coils allow for tuning and matching, then adjust the
tune and match of the imaging and labeling RF coils by adjust-
ing the 1H resonance frequency (300 MHz at 7 T) and
impedance.
5. Use a positioning scan to center the brain in the MRI scanner
in all three directions. Use a large field of view (FOV) if needed
if the brain is initially far away from the center.
6. Run automatic scanner adjustments to adjust frequency, shim,
and calibrate RF pulses.
7. Perform a pilot scan using a 2D gradient echo fast low angle
shot (FLASH) sequence (20 s). Then run a midsagittal rapid
acquisition with relaxation enhancement (RARE) or fast spin
echo scan (30 s).
8. For the following steps, plan three to nine coronal slices to
cover the region of interest in the brain using the pilot scans
from step 7 as reference. To consistently position the imaging
slices in the anterior-posterior direction between animals, use
the midsagittal scan as a reference to center one of the imaging
slices on the anterior commissure (e.g., the third-most anterior
slice out of seven slices total as in Fig. 1).
9. Perform localized volume shimming if needed to improve
magnetic field homogeneity and image quality (see Note 12).
Arterial Spin Labeling 63
Fig. 1 Example midsagittal RARE image of a rat at 7 T. The black arrow indicates
the anterior commissure. The centers of seven slices spaced 1.5 mm apart are
shown, with the third-most anterior slice positioned on the anterior commissure.
MRI parameters are: data matrix ¼ 128 128, field of view ¼ 25.6 25.6 mm,
spectral width ¼ 50 kHz, effective echo time ¼ 60 ms, repetition time ¼ 2.0 s,
and echo train length ¼ 8
Fig. 2 Example CBF maps (in mL/g/min) from continuous ASL in a (a) rat and (b) mouse. Some brain structures
should be able to be distinguished in the CBF maps, such as the corpus callosum with low blood flow
step 10, but without the ASL preparation and with a long TR
of 10 s.
12. Optionally, an anatomical reference image can be acquired
using a T2-weighted RARE sequence with parameters such as
data matrix ¼ 128 128, FOV ¼ 12.8 12.8 mm for mice or
25.6 25.6 mm for rats, slice thickness ¼ 1.0 mm for mice or
1.5 mm for rats, spectral width ¼ 50 kHz, echo train length ¼ 8,
effective TE ¼ 50 ms, TR ¼ 2.0 s, and 8 averages (4.3 min).
3.3 Animal Recovery 1. At the conclusion of imaging, remove the animal from the
scanner and holder. Discontinue anesthesia after removing
physiological monitoring probes. Place the animal on a heating
pad on low until it is fully awake and ambulatory (see Note 16).
2. Once the animal is awake and ambulatory it can then be
returned to its home cage for recovery.
3. The animal should be monitored throughout the day for signs
of distress.
3.4 Image Analysis 1. MRI image processing and analysis can be done using custom
written codes or various freely available MRI analysis tools.
Blood flow calculation from ASL data can be done using soft-
ware available online such as ASLtbx (University of Pennsylva-
nia) [28] or using custom written codes implementing the
equation in step 2. If needed, image co-registration can be
performed using software such as Statistical Parametric
Mapping (SPM, Wellcome Trust Centre for Neuroimaging,
University College London). If animals are properly secured
in ear and tooth bars and adequate anesthesia is maintained,
motion should not occur. Image display can be done with
software such as Mango (Research Imaging Institute, Univer-
sity of Texas Health Science Center at San Antonio). Several
other free software programs are also available to process and
display MRI images.
Arterial Spin Labeling 65
4 Notes
11. For surface coils, avoid pressing the coil too hard on the
animal’s head as it would increase loading. Having the coil or
its circuitry too near to the head can also cause artifacts at the
cortical surface.
12. High-order localized shimming on the brain can improve
image quality and reduce distortion in EPI images. However,
for ASL, we have found that this can cause poor shim quality
outside of the brain leading to poor labeling efficiency at the
neck, so first order global shimming may be preferable.
13. Calibration of labeling RF power and gradients can be done by
acquiring multiple short ASL scans with arrayed RF labeling
power or gradient amplitude to find the optimal power or
gradient that provide the maximum difference between
non-label and label images [35]. However, too high a labeling
power can directly saturate the brain, which will be seen as very
bright areas in the CBF maps in the posterior brain. With the
long RF pulse used for labeling in CASL, high RF power also
poses a risk of heating and burns. Since these scans would
lengthen imaging times and the optimal values are usually
consistent, this is usually done initially on a small number of
subjects to determine optimal settings to use for future studies.
14. Accurate calculation of blood flow assumes that all the labeled
blood has been delivered to the tissue, so a delay is usually
given after the end of the labeling period to allow time for the
labeled blood to flow from the labeling region to the tissue.
The post labeling delay should thus be longer than this arterial
transit time, but with too long a PLD the labeled blood water
signal may be reduced too much as it decays with T1. The
arterial transit time to the rat gray matter is around
200–400 ms, dependent upon brain region [21, 22]. Some
diseases, such as stroke, can significantly prolong the transmit
time which can affect CBF quantification. The arterial transit
time can be measured in addition to blood flow by acquiring a
series of ASL scans with multiple PLDs or by other methods, at
the cost of longer imaging times [21, 22, 36].
15. The non-labeled images of the ASL scan are sometimes used
instead of a separately acquired proton density image for blood
flow calculation. This will introduce some inaccuracy in the
calculated blood flow as the TR used for ASL scans is usually
short enough to have some T1 weighting. However, using the
non-labeled images avoids the acquisition of an extra proton
density scan and avoids errors if the intensities of the ASL and
proton density data are scaled differently by the MRI scanner
and not accounted for. The non-labeled images can be scaled
by 1/(1exp(TR/T1)), where T1 is the tissue T1, to com-
pensate if TR is not several times longer than T1 [2]. If
68 Eric R. Muir
References
1. Williams DS, Detre JA, Leigh JS, Koretsky AP 10. Muir ER, De La Garza B, Duong TQ (2013)
(1992) Magnetic resonance imaging of perfu- Blood flow and anatomical MRI in a mouse
sion using spin inversion of arterial water. Proc model of retinitis pigmentosa. Magn Reson
Natl Acad Sci U S A 89:212–216 Med 69(1):221–228. https://doi.org/10.
2. Alsop DC, Detre JA, Golay X, Gunther M, 1002/mrm.24232
Hendrikse J, Hernandez-Garcia L, Lu H, Mac- 11. Calamante F, Thomas DL, Pell GS, Wiersma J,
Intosh BJ, Parkes LM, Smits M, van Osch MJ, Turner R (1999) Measuring cerebral blood
Wang DJ, Wong EC, Zaharchuk G (2015) flow using magnetic resonance imaging techni-
Recommended implementation of arterial ques. J Cereb Blood Flow Metab 19:701–735
spin-labeled perfusion MRI for clinical applica- 12. Barbier EL, Lamalle L, Decorps M (2001)
tions: a consensus of the ISMRM perfusion Methodology of brain perfusion imaging. J
study group and the European consortium for Magn Reson Imaging 13:496–520
ASL in dementia. Magn Reson Med 73 13. Wong EC (2014) An introduction to ASL
(1):102–116. https://doi.org/10.1002/ labeling techniques. J Magn Reson Imaging
mrm.25197 40(1):1–10. https://doi.org/10.1002/jmri.
3. Brown GG, Clark C, Liu TT (2007) Measure- 24565
ment of cerebral perfusion with arterial spin 14. Dai W, Garcia D, de Bazelaire C, Alsop DC
labeling: part 2. Applications. J Int Neuropsy- (2008) Continuous flow-driven inversion for
chol Soc 13:526–538 arterial spin labeling using pulsed radio fre-
4. Wolf RL, Detre JA (2007) Clinical neuroimag- quency and gradient fields. Magn Reson Med
ing using arterial spin-labeled perfusion MRI. 60(6):1488–1497. https://doi.org/10.1002/
Neurotherapeutics 4:346–359 mrm.21790
5. Telischak NA, Detre JA, Zaharchuk G (2015) 15. Wong EC, Buxton RB, Frank LR (1998)
Arterial spin labeling MRI: clinical applications Quantitative imaging of perfusion using a sin-
in the brain. J Magn Reson Imaging 41 gle subtraction (QUIPSS and QUIPSSII).
(5):1165–1180. https://doi.org/10.1002/ Magn Reson Med 39:702–708
jmri.24751 16. Kim S-G (1995) Quantification of relative cere-
6. Shen Q, Fisher M, Sotak CH, Duong TQ bral blood flow change by flow-sensitive alter-
(2004) Effects of reperfusion on ADC and nating inversion recovery (FAIR) technique:
CBF pixel-by-pixel dynamics in stroke: charac- application to functional mapping. Magn
terizing tissue fates using quantitative diffusion Reson Med 34:293–301
and perfusion imaging. J Cereb Blood Flow 17. Alsop DC, Detre JA (1998) Multisection cere-
Metab 24:280–290 bral blood flow MR imaging with continuous
7. Silva AC, Zhang W, Williams DS, Koretsky AP arterial spin labeling. Radiology 208
(1995) Multi-slice MRI of rat brain perfusion (2):410–416. https://doi.org/10.1148/radi
during amphetamine stimulation using arterial ology.208.2.9680569
spin labeling. Magn Reson Med 33:209–214 18. Alsop D, Detre J (1996) Reduced transit-time
8. Sicard K, Shen Q, Brevard ME, Sullivan R, sensitivity in noninvasive magnetic resonance
Ferris CF, King JA, Duong TQ (2003) imaging of human cerebral blood flow. J
Regional cerebral blood flow and BOLD Cereb Blood Flow Metab 16:1236–1249
responses in conscious and anesthetized rats 19. Muir ER, Shen Q, Duong TQ (2008) Cerebral
under basal and hypercapnic conditions: impli- blood flow MRI in mice using the cardiac spin-
cations for functional MRI studies. J Cereb labeling technique. Magn Reson Med
Blood Flow Metab 23(4):472–481 60:744–748
9. Lei H, Grinberg O, Nwaigwe CI, Hou HG, 20. Shen Q, Duong TQ (2011) Background sup-
Williams H, Swartz HM, Dunn JF (2001) The pression in arterial spin labeling MRI with a
effects of ketamine-xylazine anesthesia on cere- separate neck labeling coil. NMR Biomed
bral blood flow and oxygenation observed 24:1111–1118
using nuclear magnetic resonance perfusion
imaging and electron paramagnetic resonance 21. Thomas DL, Lythgoe MF, van der Weerd L,
oximetry. Brain Res 913:174–179 Ordidge RJ, Gadian DG (2006) Regional vari-
ation of cerebral blood flow and arterial transit
70 Eric R. Muir
time in the normal and hypoperfused rat brain coefficient for water? J Cereb Blood Flow
measured using continuous arterial spin label- Metab 5:65–69
ing MRI. J Cereb Blood Flow Metab 26 31. Muir ER, Cardenas D, Huang S, Roby J, Li G,
(2):274–282. https://doi.org/10.1038/sj. Duong TQ (2014) MRI under hyperbaric air
jcbfm.9600185 and oxygen: effects on local magnetic field and
22. Meng Y, Wang P, Kim SG (2012) Simulta- relaxation times. Magn Reson Med 72
neous measurement of cerebral blood flow (4):1176–1181. https://doi.org/10.1002/
and transit time with turbo dynamic arterial mrm.25027
spin labeling (Turbo-DASL): application to 32. Dobre MC, Ugurbil K, Marjanska M (2007)
functional studies. Magn Reson Med 68 Determination of blood longitudinal relaxation
(3):762–771. https://doi.org/10.1002/ time (T1) at high magnetic field strengths.
mrm.23294 Magn Reson Imaging 25(5):733–735.
23. Debacker CS, Daoust A, Kohler S, Voiron J, https://doi.org/10.1016/j.mri.2006.10.020
Warnking JM, Barbier EL (2016) Impact of 33. Wegener S, Wu WC, Perthen JE, Wong EC
tissue T1 on perfusion measurement with arte- (2007) Quantification of rodent cerebral
rial spin labeling. Magn Reson Med. https:// blood flow (CBF) in normal and high flow
doi.org/10.1002/mrm.26255 states using pulsed arterial spin labeling mag-
24. Rajendran R, Lew SK, Yong CX, Tan J, Wang netic resonance imaging. J Magn Reson Imag-
DJ, Chuang KH (2013) Quantitative mouse ing 26(4):855–862. https://doi.org/10.
renal perfusion using arterial spin labeling. 1002/jmri.21045
NMR Biomed 26(10):1225–1232. https:// 34. Doty FD, Entzminger G, Kulkarni J,
doi.org/10.1002/nbm.2939 Pamarthy K, Staab JP (2007) Radio frequency
25. Campbell-Washburn AE, Price AN, Wells JA, coil technology for small-animal MRI. NMR
Thomas DL, Ordidge RJ, Lythgoe MF (2013) Biomed 20(3):304–325. https://doi.org/10.
Cardiac arterial spin labeling using segmented 1002/nbm.1149
ECG-gated Look-Locker FAIR: variability and 35. Maccotta L, Detre JA, Alsop DC (1997) The
repeatability in preclinical studies. Magn Reson efficiency of adiabatic inversion for perfusion
Med 69(1):238–247. https://doi.org/10. imaging by arterial spin labeling. NMR Biomed
1002/mrm.24243 10(4–5):216–221
26. Muir ER, Duong TQ (2011) MRI of retinal 36. Wells JA, Lythgoe MF, Gadian DG, Ordidge
and choroidal blood flow with laminar resolu- RJ, Thomas DL (2010) In vivo Hadamard
tion. NMR Biomed 24:216–223 encoded continuous arterial spin labeling
27. Li G, De La Garza B, Shih YY, Muir ER, (H-CASL). Magn Reson Med 63
Duong TQ (2012) Layer-specific blood-flow (4):1111–1118. https://doi.org/10.1002/
MRI of retinitis pigmentosa in RCS rats. Exp mrm.22266
Eye Res 101:90–96. https://doi.org/10. 37. Maleki N, Dai W, Alsop DC (2012) Optimiza-
1016/j.exer.2012.06.006 tion of background suppression for arterial spin
28. Wang Z, Aguirre GK, Rao H, Wang J, labeling perfusion imaging. MAGMA 25
Fernandez-Seara MA, Childress AR, Detre JA (2):127–133. https://doi.org/10.1007/
(2008) Empirical optimization of ASL data s10334-011-0286-3
analysis using an ASL data processing toolbox: 38. Steger TR, White RA, Jackson EF (2005)
ASLtbx. Magn Reson Imaging 26(2):261–269 Input parameter sensitivity analysis and com-
29. Buxton RB, Frank LR, Wong EC, Siewert B, parison of quantification model for continuous
Warach S, Edelman RR (1998) A general arterial spin labeling. Magn Reson Med
kinetic model for quantitative perfusion imag- 53:895–903
ing with arterial spin labeling. Magn Reson 39. Talagala SL, Ye FQ, Ledden PJ, Chesnick S
Med 40(3):383–396 (2004) Whole-brain 3D perfusion MRI at 3.0
30. Herscovitch P, Raichle ME (1985) What is the T using CASL with a separate labeling coil.
correct value for the brain-blood partition Magn Reson Med 52:131–140
Chapter 5
Abstract
Dynamic contrast-enhanced MRI in pre-clinical imaging allows the in-vivo monitoring of vascular,
physiological properties in normal and diseased tissue. There is considerable variation in the methods
employed owing to the different questions that can be asked and answered about the physiologic alterations
as well as morphologic changes in tissue. Here we review the typical decisions in the design and execution of
a dynamic contrast-enhanced MRI study in mice although the findings can easily be transferred to other
species. Emphasis is placed on highlighting the many pitfalls that wait for the unaware pre-clinical MRI
practitioner and that go often unmentioned in the abundant literature dealing with dynamic contrast-
enhanced MRI in animal models.
Key words Pharmacodynamics, Gadolinium chelate, Blood flow, Vessel permeability, Ktrans, IAUC60
1 Introduction
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0_5, © Springer Science+Business Media, LLC 2018
71
72 Jennifer Moroz and Stefan A. Reinsberg
1.3 Arterial Input The arterial input function (AIF) describes the concentration of a
Function contrast agent in the blood. Ideally, the AIF should be sampled in a
vessel feeding the tissue of interest and concurrently with the DCE
experiment (see [13] for an elegant example). Measuring the AIF in
small animals is difficult due to their small spatial dimensions and
limited number of vessels of a sufficient size [14]. Common sites for
74 Jennifer Moroz and Stefan A. Reinsberg
AIF measurements include the heart [15], the aorta/vena cava, the
iliac artery [16], and the tail veins [17]. For increased accuracy in
the model parameters, the AIF should be sampled with a time
resolution at, or greater than, that of the DCE images [18].
There are four main methods to obtain the AIF:
1. Using a population-averaged AIF from the literature [19–22],
2. Measuring an AIF in control population using your planned
procedure,
3. Measuring the AIF from a small injection prior to the actual
DCE-MRI experiment, also known as dual-bolus
method [23, 24], and
4. Employing a reference-region approach by imaging known
tissue (commonly muscle) in addition to the site of interest
during the DCE-MRI experiment [25] (see Note 4.1).
There is much debate on which method produces the most
accurate measurement of the true curve. Naturally, methods 2, 3,
and 4 are preferred in cases where the variation between individuals
is large or where the injection and imaging protocols deviate from
those published in the literature [14, 15]. For the purposes of this
chapter, measuring an AIF separately in a control experiment
(method 2) will be outlined in greater detail in Subheading 3.
Those interested in the other methods are referred to the notes.
2 Materials
2.1 Analysis Almost none of the vendor-software packaged with the MRI system
Software consoles allows data analysis of DCE-MRI. Consequently, a great
number of alternative solutions have emerged with no clear leader
in sight. A further problem is the large inter-software variability for
parameter maps derived from the same data but different software
solutions as recently reviewed by Beuzit et al. [26]. A large number
of closed-source, proprietary solutions also exist and require further
on-site quality assurance to ascertain the suitability for the task. At
our institution, we use a custom-built software solution relying on
an array of open-source Python packages mostly from the scipy
ecosystem [27]; in particular, pandas [28], and nibabel [29].
Tools such as the Osirix (Pixmeo SARL and Osirix Foundation)
plugin DCE_Tool by K.H. Sung, or Brandon Whitcher’s and
Volker Schmid’s DCEMRI package for the R programming envi-
ronment streamline some of the data processing steps described in
the Methods below. However, they do not remove the need to
critically check the parameter maps obtained and, unfortunately,
sometimes make it harder to inspect the intermediate results in the
calculation and optimization.
76 Jennifer Moroz and Stefan A. Reinsberg
3 Methods
3.1 Preparation The contrast agents should be administered with a line that avoids
of the Contrast the premature injection of contrast agents. Since the blood pressure
Injection of the animal fluctuates from the time the intra-venous access is
established to the time the DCE-MRI experiments is performed, it
is possible for blood to be pushed into the connected line (increase
in blood pressure) or intra-venous fluid to flow into the animal.
Figure 1 shows the assembly of the line. A heparin lock of 25 μL
volume (segment A in the figure) will act as a buffer between the
animal and the actual contrast-agent volume.
1. Dilute the stock contrast agent with saline. Determine the
volume of contrast agent to be injected based on a dose of
0.1 mmol/kg but not exceeding 0.3 mmol/kg body
weight [5].
2. Determine the length of PE20 (segment B in the figure) tubing
needed for this volume. The length of tubing is calculated from
l ¼ VCA/(π ∗ r2), where r ¼ 0. 19 mm for PE20.
3. Fill the line with the contrast agent and set aside.
4. Connect a PE20 line of a length sufficient to hold a volume of
25 μL pre-bolus heparinized saline with the contrast-agent
PE20 line. Blunt stents may be used to create dead-volume
free connections between these tubes.
5. A further tube connecting the PE20 assembly with the power
injector (segment C in the figure) is filled with (heparin-free)
saline for the post-injection flush.
3.2 Catheterization 1. Prior to anesthesia and after sufficient warming of the animal
of the Mouse Tail using a heat lamp, a catheter is placed in the mouse tail vain. A
and Anesthesia proper restraint such that the experimenter has ready access to a
motionless tail is required such as our custom-made perspex
box with a V-shaped slit in one of the walls (see Fig. 2). The
catheter should be glued or taped in place.
Fig. 1 Setup for the injection line with heparin lock, bolus line, and saline flush
Preclinical MRI 77
Fig. 2 Mouse tail catheterized (step 2) and anesthesia induced. At this stage care must be taken to maintain
body temperature. Mouse can now be transferred to the RF coil and animal holder (left in the photo). The
catheter can be cut and connected to the bolus line
6. Remove the syringe from the catheter line, and connect the line
with contrast agent to it. This connection can be done with
stents, a butterfly needle or a regular needle tip.
7. Connect the saline flush to the end of the contrast-agent line.
Naturally, it is important to avoid the inclusion of air bubbles in
any of the liquid that will be injected into the animal.
3.4 Data Acquisition DCE-MRI experiments acquire images before, during, and after
injection of a contrast agent. Most studies estimate the concentra-
tion of contrast agent in the tissue through the change in the T1
relaxation time constant, and using Eq. 1.
3.4.2 DCE Image A typical DCE-MRI consists of pre-injection scans, the injection,
Acquisition and a long time course evaluation post-injection. It may be benefi-
cial to acquire a second T1 map after the full scan for a comparison.
A large number of MR sequences have been used to acquire
DCE-MRI data. A very common technique is a 3D FLASH
sequence (also known as spoiled gradient echo sequence) since it
allows for shorter repetition times, TR, by using small flip angles.
Parameters usually are TR ¼ 4. . .6 ms, TE ¼ 1. . .3 ms, flip angle, θ
¼ 10∘. . .15∘. A final determination on the ideal repetition time/flip
angle determination has to be made through the optimization of
signal and contrast to noise ratio for an expected baseline and
change of T1. The required spatial coverage, field of view, and
spatial resolution determine the final temporal resolution. A viable
alternative for certain geometries is the use of a multi-slice 2D
FLASH DCEMRI sequence. For those the repetition time, TR, is
lengthened to about 100 ms while the flip angle is increased to θ ¼
25∘. . .40∘ to continue acquiring signal in a signal-to-noise per unit
time optimized fashion.
While parallel imaging is very useful in many human DCE-MRI
studies to achieve acceleration factors of 2–3, this has not been
routinely done in pre-clinical DCE-MRI scans. When parallel imag-
ing has been applied in pre-clinical imaging it is often used over
surface coil due to need to extend homogeneous coverage of
patient anatomy rather than the acquisition speed up. An area of
active research is the use of compressed sensing, which can achieve
remarkable speed-up [31].
1. Acquire survey scan to plan positioning of DCE-MRI slices.
2. Acquire T1 baseline preferably at exactly the same location and
resolution as the subsequent DCE-MRI experiment. For a
variable-flip angle FLASH sequence, this is straightforward
due to the only difference arising from the flip angle and lack
of repeat acquisition.
80 Jennifer Moroz and Stefan A. Reinsberg
3.5 Arterial Input The AIF should be sampled for each individual and for each experi-
Function ment. However this may not be possible if a high temporal resolu-
tion is desired for the DCE images. One method is to estimate the
AIF from a population averaged curve using your planned proce-
dure, but in a separate experiment.
1. Identify a large vessel in the near vicinity of the tissue of
interest. Common sites are the heart, aorta/vena cava, iliac
artery, or the tail veins.
2. Set up the animal as detailed above, but position the animal
such that the site of the AIF measurement is centered above
the coil.
3. Run the desired scans for the AIF measurement. The total scan
time should be the same as the DCE experiment.
4. Repeat for several animals.
5. The AIF may be determined from the signal magnitude [32] or
phase [33– 36] in the images. Image phase has the advantages
that it has a directly linear relationship over a wide range of
concentrations, has greater SNR, it is less sensitive to flow, and
it is less sensitive to changes in the scan parameters.
6. The change in T1 can be used to determine the change in signal
for a magnitude based AIF.
7. To get the concentration from the image phase, a calibration
factor is required for your scanner. This may be attained from a
separate experiment using a large range of contrast agent con-
centrations and fitting a linear regression to the data. The phase
may need to be unwrapped first due to the 2π dynamic range. If
using the signal phase, a reference phantom should be placed
within the imaging plane to track phase drift [37].
8. Average the individual measurements for all animals.
Preclinical MRI 81
3.6 Data Processing Since the data processing requires a parameter fit of an data time
course, it is important to ensure that subsequent images are
registered with each other.
1. At a minimum, a visual inspection is required to check that all
slices line up as time advances. Slices with motion present
should be discarded. More robustly, it is possible to recover
data that might otherwise be lost to motion artifacts, by regis-
tering the DCE images to a suitable baseline image.
2. When signal intensities from separate scans are computationally
combined, the receiver gain used to acquire these different
scans has to be fixed. Many scanners will automatically adjust
the receiver gain and thereby introduce an unknown overall
intensity scaling factor. While scanner-reconstructed data
sometimes also stores information to undo the effect of a
variable receiver gain, it is advisable to fix the gain if possible.
3. Baseline T1 map is calculated and registered to the DCE-MRI
time course. The calculation of the baseline T1 may be aug-
mented with the calculated flip-angle map.
4. Using Eq. 5, the baseline T1 map, and the averaged signal prior
to contrast injection, the change in T1 for every pixel in the
tissue of interest can be calculated. The lower panel in Fig. 3
shows such two example time courses.
5. Using Eq. 1, one can calculate the concentration of the con-
trast agent for every time in the DCE-MRI time course. This
data forms the basis for semi-quantitative parameters and the
subsequent fit. Note 4.4 discusses the options of fitting signal
time courses vs. concentration time courses.
6. Simpler but less computationally involved, semi-quantitative
parameters such as area-under-the curve (AUC) may be calcu-
lated. There is now widespread consensus that parameters such
as AUC have limitations when it comes to translation and inter-
group comparisons.
7. Data quality, temporal resolution, and signal-to-noise will dic-
tate the choice of model and the number of free parameters
that can be supported by the data. Rukat et al. studied the
effects of these factors and provide a methodology for deciding
on a suitable model [38].
8. A suitable arterial input function, ca(t) is chosen.
9. Using the Tofts model as an example, the observed concentra-
tion data can be fit using Eq. 2. As a result, parameters
Ktrans and ve can be determined. See example in Fig. 3. For
different models (such as extended Tofts or 2CXM) other/
more parameters will result from this step. See Note 4.5 for
details on an additional parameter originating from the uncer-
tainty of the bolus arrival in the tissue.
82 Jennifer Moroz and Stefan A. Reinsberg
Ktrans ve R2
0.00 0.02 0.04 0.06 0.08 0.10 0.0 0.1 0.2 0.3 0.4 0.5 0.6 0.0 0.2 0.4 0.6 0.8 1.0
concentration time course and fit T1 − weighted image concentration time course and fit
0.25 0.25
0.20 0.20
0.10 0.10
0.05 0.05
0.00 0.00
0 100 200 300 0 100 200 300
Fig. 3 Parameter map and typical concentration time course and Tofts model fit. Top row shows the parameter
maps for Ktrans and ve. Top right shows R2 as a measure of fit quality. While reservations exist for the use of R2
for assessing different models in non-linear regression, it is useful to judge fit quality for one model applied to
different time curves. The lower row shows two different for a quickly enhancing pixel (left) and a slower
enhancing one (right)
4 Notes
4.1 Reference DCE-MRI models are based on the indicator-dilution theory, and
Region as Alternative therefore require knowledge of the concentration-time curve (the
to Direct AIF AIF) in the blood plasma. It has been suggested that the AIF should
Acquisition be measured with a temporal resolution exceeding 5 s/image for
accurate results [39]. However, this can be difficult to achieve. To
relax the time restraint, Yankeelov et al. proposed a technique that
replaces the AIF with a calibration against another tissue with known
contrast kinetics; similar to what is used in PET imaging [25].
The technique is derived in [25], providing a final model of:
K T rans , RR K T rans , T OI
C T OI ðT Þ ¼ R C RR ðT Þ þ R
ve , RR v e , T OI
ZT
K trans , T OI
C RR ðtÞ exp ∗ðT tÞdt
ve , T OI
0
Preclinical MRI 83
where CTOI and CRR are the contrast agent concentrations in the
tissue of interest and the reference region tissue, KTrans,TOI and
KTrans,RR are the volume transfer coefficients, ve, TOI and ve, RR are
the fractional volume of the extravascular-extracellular space, and
R ¼ KTrans,TOI/KTrans,RR. This technique assumes that the AIF is
similar for both the reference tissue and the tissue of interest [39].
Procedure for model fitting with the Reference Region
model [25].
1. Acquire a T1o map over the tissue of interest. A variable flip
angle spoiled gradient echo may be used with a number of
different excitation angles. Fit the data to Eq. 5 to solve for T1.
2. Perform the DCE experiment as proposed. Allow for several
pre-bolus images to be acquired.
3. Co-register all images against the first pre-contrast image. A
mutual information-based rigid registration algorithm is
suggested.
4. Estimate the T1 time courses for each voxel in both the refer-
ence region tissue and the tissue of interest from the change in
signal intensity of the time course data.
5. Input both T1 curves into the proposed model described above
into a curve-fitting routine.
dc p
vp ðtÞ ¼ F p c a ðtÞ F p c p ðtÞ þ PSc e ðtÞ PSc p ðtÞ: ð7Þ
dt
The response function in a two-compartment model is a
double-exponential with four free parameters (two amplitudes
and two decay constants).
τ 1 Fp
A 1=2 ¼ F p and B 1=2 ¼ ð9Þ
τþ τ ðv p þ ve Þτ
where
ve
e¼ and τ
vp þ ve
rffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffi!
E Ee þ e 1 4Eeð1 EÞð1 eÞ
¼ 1 : ð10Þ
2E E Ee þ e
4.3 Predicting Since a considerable cost in time and effort is incurred with
Injection Success DCE-MRI experiments it is desirable to have an early indication
While Scanning of a successful bonus injected. At our institutions we have devel-
oped an early alert should there not have been a successful injec-
tion—this early alert can be run as soon as the contrast is injected.
Using signal from the entire field of view and reconstructing a
“global” time course of signal change. Figure 4 shows an example
of a successful injection and unsuccessful injection. The latter
allows the immediate discontinuation of the experiment thereby
saving precious scan time. This diagnostic early reconstruction
requires a tolerance to partially written data files.
4.4 Fitting The methods described in this chapter advocate the calculation of
Concentration or the concentration time curves from the DCE-MRI data and the
Signal-Time Curves subsequent fitting of this curved C(t) by a model of choice. While
this is a more obvious approach, an alternative method involves the
calculation of the expected signal time curve from a concentration
time curve C(t) in terms of the still undetermined parameters of the
model. This will form a model of the expected signal time curve, S
(t), and can be fit by the data acquired.
In principle, these two methods are equivalent. However, the
experimenter needs to take care to adjust the weighting of data
points as dictated by the transformation from C(t) to S(t). Further-
more since this transformation is not linear, the cost function used
in the minimization will result in slightly different optimization
results that are usually inconsequential in practice.
Preclinical MRI 85
2650
2800
2600
2600
2550
signal intensity / a.u.
2000 2400
1800 2350
1600 2300
1400 2250
0 100 200 300 0 50 100 150 200 250
image frame number image frame number
Fig. 4 Early image showing a successful injection (left) and failed injection (right). Note the vastly different
scale in image intensity between the two. At the time of injection, motion-induced change in intensity is
sometimes visible but clear enhancement is lacking
4.5 Bolus In fitting the pharmacokinetic models such as by the Tofts equa-
Arrival Time tion (Eq. 2), there is an implicit assumption of the convolution
starting from time t ¼ 0 extending into the feature. In reality this
time zero is the bolus arrival time and needs to be determined
separately. A simple method is to set the bolus arrival time to the
time when the injection was initiated. A more sophisticated
approach is to leave this bolus arrival time flexible. It can then be
fit as part of the model optimization or it can be determined directly
from the data using control chart decision criteria [42]. In either
case, the correct choice of bolus arrival time has a strong influence
on pharmacokinetic parameters.
Acknowledgements
This work was made possible through support from the Discovery
Program of NSERC and CIHR.
References
1. O’Connor JPB, Aboagye EO, Adams JE, et al 3. Keara Boss M, Muradyan N, Thrall DE (2011)
(2016) Imaging biomarker roadmap for cancer DCE-MRI: a review and applications in veteri-
studies. Nat Rev Clin Oncol 14:169–186. doi: nary oncology. Vet Comp Oncol 11(2):87–100
10.1038/nrclinonc.2016.162 4. Zhou Z, Lu ZR (2013) Gadolinium-based
2. Weinmann HJ, Brasch RC, Press WR, et al contrast agents for magnetic resonance cancer
(1984) Characteristics of gadolinium-DTPA imaging. Wiley Interdiscip Rev Nanomed
complex: a potential NMR contrast agent. Am Nanobiotechnol 5(1):1–18. doi: 10.1002/
J Roentgenol 142:619–624 wnan.1198
86 Jennifer Moroz and Stefan A. Reinsberg
5. Yan GP, Robinson L, Hogg P (2007) Magnetic function can be achieved by phase imaging.
resonance imaging contrast agents: overview Magn Reson Med 71(2):544–550. doi: 10.
and perspectives. Radiol 13:e5–e19. doi: 10. 1002/mrm.24682
1016/j.radi.2006.07.005 17. Pathak AP, Artemov D, Bhujwalla ZM (2004)
6. Carr DH, Brown J, Bydder GM, et al (1984) Novel system for determining contrast agent
Intravenous chelated gadolinium as a contrast concentration in mouse blood in vivo. Magn
agent in NMR imaging of cerebral tumours. Reson Med 51:612–615. doi: 10.1002/mrm.
Lancet 323(8375):484–486. doi: 10.1016/ 10727
S0140-6736(84)92852-6 18. Henderson E, Rutt BK, Lee TY (1998) Tem-
7. Shen Y, Goerner FL, Snyder C, et al (2015) T1 poral sampling requirements for the tracer
relaxivities of gadolinium-based magnetic reso- kinetics modeling of breast disease. Magn
nance contrast agents in human whole blood at Reson Imaging 16(9):1057–1073. doi: 10.
1.5, 3, and 7 T. Invest Radiol 50(5):330–338. 1016/S0730-725X(98)00130-1
doi: 10.1097/RLI.0000000000000132 19. Lyng H, Dahle GA, Kaalhus O, et al (1998)
8. Tofts P (2010) T1-weighted DCE imaging Measurement of perfusion rate in human mel-
concepts: modelling, acquisition and analysis. anoma xenografts by contrast-enhanced mag-
MAGNETOM Flash 3:30–39 netic resonance imaging. Magn Reson Med
9. Tofts PS, Kermode AG (1991) Measurement 40:89–98
of the blood-brain barrier permeability and 20. Parker GJM, Roberts C, Macdonald A, et al
leakage space using dynamic MR imaging. (2006) Experimentally-derived functional
1. Fundamental concepts. Magn Reson Med form for a population-averaged high-temporal-
17(2):357–367 resolution arterial input function for dynamic
10. Larsson HBW, Stubgaard M, Frederiksen JL, contrast-enhanced MRI. Magn Reson Med 56
et al (1990) Quantitation of blood-brain bar- (5):993–1000. doi: 10.1002/mrm.21066
rier defect by magnetic resonance imaging and 21. Sanz-Requena R, Prats-Montalban J, Marti-
gadolinium-DTPA in patients with multiple Bonmati L, et al (2015) Automatic individual
sclerosis and brain tumors. Magn Reson Med arterial input functions calculated from PCA
16(1):117–131. doi: 10.1002/mrm. outperform manual and population-averaged
1910160111 approaches for the pharmacokinetic modeling
11. Brix G, Semmler W, Port R, et al (1991) Phar- of DCE-MR images. J Magn Reson Imaging
macokinetic parameters in CNS Gd-DTPA 42(2):477–487. doi: 10.1002/jmri.24805
enhanced MR imaging. J Comput Assist 22. Onxley JD, Yoo DS, Muradyan N, et al (2014)
Tomogr 15(4):621–628. doi: 10.1097/ Comprehensive population-averaged arterial
00004728-199107000-00018 input function for dynamic contrast-enhanced
12. Sourbron SP, Buckley DL (2011) On the scope vmagnetic resonance imaging of head and neck
and interpretation of the Tofts models for cancer. Int J Radiat Oncol 89(3):658–665. doi:
DCE-MRI. Magn Reson Imaging 66 10.1016/j.ijrobp.2014.03.006
(3):735–745. doi: 10.1002/mrm.22861 23. Kershaw LE, Cheng MHL (2011) A general
13. McIntyre DJO, Ludwig C, Pasan A, et al dual-bolus approach for quantitative
(2004) A method for interleaved acquisition DCE-MRI. Magn Reson Imaging 29
of a vascular input function for dynamic (2):160–166. doi: 10.1016/j.mri.2010.08.
contrast-enhanced MRI in experimental rat 009
tumours. NMR Biomed 17(3):132–143. doi: 24. Kim TH, Pack NA, Chen L, et al (2010) Quan-
10.1002/nbm.868 tification of myocardial perfusion using CMR
14. Pickup S, Zhou R, Glickson J (2003) MRI with a radial data acquisition: comparison with
estimation of the arterial input function in a dual-bolus method. J Cardiovasc Magn
mice1. Acad Radiol 10:963–968. doi: 10. Reson 12(1):45–52. doi: 10.1186/1532-
1016/S1076-6332(03)00291-5 429X-12-45
15. Ragan DK, Lai SY, Bankson JA (2011) Fast, 25. Yankeelov TE, Luci JJ, Lepage M, et al (2005)
reproducible measurement of the vascular Quantitative pharmacokinetic analysis of
input function in mice using constrained DCE-MRI data without an arterial input func-
reconstruction and cardiac sampling. NMR tion: a reference region model. Magn Reson
Biomed 24:373–384. doi: 10.1002/nbm. Imaging 23(4):519–529. doi: 10.1016/j.mri.
1601 2005.02.013
16. Fruytier AC, Magat J, Colliez F, et al (2014) 26. Beuzit L, Eliat PA, Brun V, et al (2015)
Dynamic contrast-enhanced MRI in mice at Dynamic contrast-enhanced MRI: study of
high field: estimation of the arterial input inter-software accuracy and reproducibility
Preclinical MRI 87
using simulated and clinical data. J Magn for quantification of dynamic contrast-
Reson Imaging 43(6):1288–300. doi: 10. enhanced (DCE) MRI and comparison with
1002/jmri.25101 DCE-CT. Magn Reson Med 66
27. van der Walt S, Colbert SC, Varoquaux G (5):1267–1274. doi: 10.1002/mrm.22905
(2011) The NumPy array: a structure for effi- 36. Garpebring A, Wirestam R, Yu J, et al (2011)
cient numerical computation. Comput Sci Eng Phase-based arterial input functions in humans
13(2):22–30. doi: 10.1109/MCSE.2011.37 applied to dynamic contrast-enhanced MRI:
28. McKinney W (2010) Data structures for statis- potential usefulness and limitations. Magn
tical computing in Python. In: van der Walt S, Reson Mater Phy 24(4):233–245. doi: 10.
Millman J (eds) Proceedings of the 9th Python 1007/s10334-011-0257-8
in science conference, pp 51–56 37. Akbudak E, Norberg RE, Conturo TE (1997)
29. Brett M, Hanke M, Cipollini B, et al (2016) Contrast-agent phase effects: an experimental
nibabel: 2.1.0. doi: 10.5281/zenodo.60808 system for analysis of susceptibility, concentra-
30. Look DC, Locker DR (1970) Time saving in tion, and bolus input function kinetics. Magn
measurement of NMR and EPR relaxation Reson Med 38:990–1002
times. Rev Sci Instrum 41(2):250–251. doi: 38. Rukat T, Walker-Samuel S, Reinsberg SA
10.1063/1.1684482 (2015) Dynamic contrast-enhanced MRI in
31. Han S, Paulsen JL, Zhu G, et al (2012) Tem- mice: an investigation of model parameter
poral/spatial resolution improvement of uncertainties. Magn Reson Med 73
in vivo DCE-MRI with compressed sensing- (5):1979–1987. doi: 10.1002/mrm.25319
optimized FLASH. Magn Reson Imaging 30 39. Cárdenas-Rodrı́guez J, Howison CM, Pagel
(6):741–752. doi: 10.1016/j.mri.2012.02. MD (2013) A linear algorithm of the reference
001 region model for DCE-MRI is robust and
32. McGrath DM, Bradley DP, Tessier JL, et al relaxes requirements for temporal resolution.
(2009) Comparison of model-based arterial Magn Reson Imaging 31(4):497–507. doi:
input functions for dynamic contrast-enhanced 10.1016/j.mri.2012.10.008
MRI in tumor bearing rats. Magn Reson Med 40. Brix G, Kiessling F, Lucht R, et al (2004)
61:1173–1184. doi: 10.1002/mrm.21959 Microcirculation and microvasculature in
33. Moroz J, Wong CL, Yung AC, et al (2014) breast tumors: pharmacokinetic analysis of
Rapid measurement of arterial input function dynamic MR image series. Magn Reson Med
in mouse tail from projection phases. Magn 52(2):420–429. doi: 10.1002/mrm.20161
Reson Med 71:238–245. doi: 10.1002/mrm. 41. Sourbron SP, Buckley DL (2013) Classic mod-
24660 els for dynamic contrast-enhanced MRI. NMR
34. Bleeker EJW, van Buchem MA, Webb AG, et al Biomed 26(8):1004–1027. doi: 10.1002/
(2010) Phase-based arterial input function nbm.2940
measurements for dynamic susceptibility con- 42. McPhee KC, Baker JEH, Saatchi K, et al
trast MRI. Magn Reson Med 64:358–368. doi: (2012) Delayed bolus arrival time with high
10.1002/mrm.22420 molecular weight contrast agent indicates sig-
35. Korporaal JG, van den Berg CAT, van Osch nificant necrosis. In: Proceedings of the inter-
MJP, et al (2011) Phase-based arterial input national society for magnetic resonance in
function measurements in the femoral arteries medicine, Melbourne, p. 1528
Chapter 6
Abstract
Magnetic resonance imaging (MRI) is a technique based on the contents and relaxation features of water in
tissues. In basic MRI sequences, diffusion phenomenon of water molecules is not taken into account
although it has a notable influence in the relaxation times, and therefore in the signal intensity of images. In
fact, MRI techniques that take advantage of water diffusion have experienced a huge development in last
years. Diffusion-weighted imaging (DWI) has spectacularly evolved reaching nowadays a great impact both
in clinical and preclinical imaging—especially in the neuroimaging field—and in basic research. We present
here a protocol to perform DWI studies in a high-field preclinical setup.
Key words Magnetic resonance imaging, Preclinical MRI, Diffusion-weighted imaging, Echo-planar
imaging, Apparent diffusion coefficient
1 Introduction
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0_6, © Springer Science+Business Media, LLC 2018
89
90 Irene Guadilla et al.
Fig. 2 Representative DWI workflow. Several images are acquired increasing b value from one to the next. A
pixel-by-pixel adjustment of the logarithm of the MRI signal (related to the basal signal) versus b values,
provides the ADC values from the slope of the line. A voxel placed in cerebrospinal fluid (CSF)—where proton
can freely move across—diffuse faster originating a higher slope in the linear adjustment than those placed in
gray matter (GM)—where water molecules find restrictions to their translational movement
Diffusion MRI 93
2 Materials
2.2 Other Equipment, It is necessary to use a monitoring system that continuously reports
Devices, and Materials data of body temperature (rectal sensor) and respiratory rate (using
a pneumatic pillow placed in the animal abdomen) to follow the
2.2.1 Small Animal
physiological evolution of the anesthetized animal inside the mag-
Monitoring and Gating
net and control the level of anesthesia.
System
2.2.2 Anesthesia Device The inhalation anesthesia equipment used for small animals con-
tains an anesthetic vaporizer (dependent on the anesthetic), a flow-
meter (dependent on the gas carrier), a poly(methylmethacrylate)
(PMMA) chamber, circulating tubes, and a filter.
2.2.3 Thermal System Anesthetized animals have to be kept warmed by using a water
heated blanket (or any other appropriate and MR compatible
tool) to maintain the body temperature at 37 C during scanning.
94 Irene Guadilla et al.
2.2.4 Animal Holders Suitable animal beds of PMMA (or any other MR compatible
material) are used to place the rat or mouse inside the magnet,
provided with cap nose to supply anesthesia and adequate options
to immobilize the animal head (usually fixing tooth and ears with
bars). The holder will be slid to localize the region of interest in the
magnet isocenter.
3 Methods
3.1 Pre-scan Animals are anesthetized in a PMMA chamber using a mix of pure
oxygen and isoflurane (3–4%) and then placed on the animal holder
to be introduced inside the magnet. The anesthesia is maintained
during the whole acquisition and controlled based on the animal
respiration (see Note 1). The body temperature is maintained at
around 37 C—using the thermal blanket—and monitored with a
rectal thermometer. The animal head has to be immobilized during
acquisition because movements will cause artifacts and originate
processing errors in the calculated parametric images. To that,
immobilize it with ears and tooth bars and/or fix tape over the
head. Put eye ointment on the eyes of the animal to avoid damaging
the cornea.
3.2 Scan A fast-low angle shot magnetic resonance imaging (FLASH MRI)
sequence is used to obtain quickly an image of the three spatial
3.2.1 Localization
orientations (axial, coronal, and sagittal) of the animal and fix the
and Anatomical Imaging
correct position inside the magnet (see Note 2).
Diffusion MRI 95
Table 1
Preclinical MRI system and associated equipment/devices for developing the protocols described in
this chapter
3.2.2 Diffusion Imaging There are many options to acquire DWI, but acquisitions based on
the Stejskal-Tanner sequence are probably the most commonly
used. In these studies, as many ADC maps will be generated as
different directions are selected by the operator to apply the diffu-
sion gradients.
We present in this manuscript three different methods opti-
mized in for obtaining diffusion images depending on how the
k-space is filled, that it, the collection of the MR data.
96 Irene Guadilla et al.
Fig. 3 MRI study of a mouse brain in axial orientation. T2-weighted (A) and diffusion-weighted images of the
same slice (B). The acquisition parameters of the DWI sequence are showed in Table 2.
Table 2
Acquisition parameters of a diffusion-weighted MRI study of a mouse brain in a Bruker Biospect® 7 T
MRI system
3.3 Post-scan Diffusion studies are computed in a pixel-by-pixel base using the
Analysis appropriate software. The corresponding ADC parametric images
are generated in each direction, and also the mean ADC map in all
assessed directions are generated by fitting the MRI signal to the
Eq. 4. In our case, we employ the homemade application
98 Irene Guadilla et al.
Fig. 4 Diffusion MRI study of a mouse in axial orientation. ADC parametric images in the head-foot (HF) (A),
left–right (L–R) (B), anterior–posterior (A–P) (C) directions and mean values of ADC considering the three
directions (D). The acquisition parameters of the DWI sequence are showed in Table 2.
4 Notes
Acknowledgments
References
€
1. Einstein A (1905) Uber die von der moleku- 2. Einstein A (1956) Investigations on the theory
larkinetischen Theorie der W€arme geforderte of the Brownian movement. Dover Publica-
Bewegung von in ruhenden Fl€ ussigkeiten sus- tions, NY
pendierten Teilchen. Ann Phys-Berlin 322 3. Le Bihan D (1995) Diffusion, perfusion and
(8):549–560 functional magnetic resonance imaging. J Mal
Vasc 20(3):203–214
Diffusion MRI 101
Abstract
Diffusion Tensor Imaging is an MRI technique that allows in vivo noninvasive measurement of the
translational motion of water, providing information about its anisotropy (or lack of it) in different tissues.
DTI has been commonly used to quantitatively measure the integrity of tissues which may be compromised
by neurological disease, such as white matter tracks of the brain, which normally impart significant
anisotropy to water motion in healthy brains. However, this anisotropic effect is diminished when axonal
or neuronal damage is present. This chapter describes a standard protocol for DTI data acquisition in
preclinical studies.
Key words Diffusion tensor imaging, Fractional anisotropy, Apparent diffusion coefficient, Axial
diffusivity, Radial diffusivity, White matter, Fiber tracking
1 Introduction
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0_7, © Springer Science+Business Media, LLC 2018
103
104 Silvia Lope-Piedrafita
of equal time and amplitude, rewinds this phase shift, in such a way
that for static spins, where no net translational movement has taken
place, the phase of all the spins is brought back together after the
second gradient. However, if molecules diffuse during the time
delay between the two gradients, the refocusing is incomplete and
a phase dispersion remains after the second gradient. Accordingly,
signal intensity diminishes compared to that of static molecules.
Experimentally, the equation for signal attenuation, S, is com-
monly described as [5]
S ¼ S 0 e bD ð1Þ
where
2 δ
b ¼ ðγ δ G D Þ Δ ð2Þ
3
Fig. 1 Estimation of the diffusion coefficient (ADC map, top right) by the exponential fitting of data acquired at
three diffusion weightings (b ¼ 200, 600, and 1000 s/mm2) and a non-diffusion one (b0 image, top left)
Fig. 2 The diffusion tensor visualized as an ellipsoid in the case of isotropic diffusion (left) and anisotropic
diffusion (right). The diffusion ellipsoid represents the three-dimension mean squared displacement of mobile
hydrogen within the voxel. When the diffusion has no preferred direction, λ1 ¼ λ2 ¼ λ3, it is said that diffusion
is isotropic and is represented as a sphere. When diffusion is anisotropic, the diffusion ellipsoid is oriented
such that its long axis, λ1, is parallel to the direction of greatest diffusion, ε1, known as the primary principal
diffusion direction. Two additional vectors represent the rate of diffusion perpendicular to the primary principal
diffusion direction with axis lengths proportional to λ2 and λ3, respectively
λ2 þ λ3
λ⊥ ¼ ð8Þ
2
Changes in λ⊥ and λ|| may potentially be used to differentiate
myelin loss versus axonal injury. Both circumstances induce a
decrease in FA; however, a loss of myelin integrity is correlated
with an increase in λ⊥, whereas axonal damage is associated to a
decrease in λ|| [12, 13].
In addition, the physical direction of the preferred motion of
water in the laboratory frame can also be elucidated directly from
the eigenvectors which can be displayed in directional-encoded-
color (DEC) maps. Moreover, the diffusion tensor is used for fiber
orientation tracking by connecting voxels based on the primary
principal diffusivity direction [14]. However, the traditional
single-tensor DTI model described here has been shown to be
inadequate in many regions of the brain that contain crossing fibers,
leading to false connections. More advanced models, which are
beyond the scope of this chapter, have been introduced to over-
come this problem including multitensor models, high angular
resolution diffusion imaging (HARDI), Q-ball imaging, or diffu-
sion spectrum imaging (DSI) [15].
The quantitative anisotropic parameters calculated from DTI
data sets are sensitive to the geometry and integrity of brain tissue.
For example, in dense white matter tracks, especially highly myelin-
ated ones, water diffusion is restricted in directions perpendicular
to the axon’s long axis, leading to high values of anisotropy.
Accordingly, DTI has been widely used to study a number of
white matter diseases and developmental pathologies [12, 13, 16,
17]. In addition, there is an active field of DTI research in the area
of cancer where the usefulness of anisotropy parameters for tumor
classification and for detecting tumor invasion is being investigated
[18, 19].
2 Materials
3 Methods
3.1 Pre-scanning The aim is to have everything ready so that, once the animal is
Preparation anesthetized, we are ready to run the experiment, thus minimizing
the time that the animal is under anesthesia.
1. It is very important to remove all loose metals from the opera-
tor and the animal (metal ear tags) prior to entering the
scanner room.
2. Insert and center volume coil inside the magnet.
3. Turn on heating system (water bath or heating module fan).
DTI 109
3.2 Pilot Scan Load a pilot scan (in ParaVision named “tripilot”) in the MRI
acquisition software. Complete automatic adjustments which
enable to boost SNR: global shimming, setting the reference fre-
quency, and calibration of the reference frequency (see Note 4).
Adjust receiver gain and run the tripilot scan. As a result, three
110 Silvia Lope-Piedrafita
3.3 DTI Scan 1. Load a diffusion protocol from the MRI software library. The
most commonly used sequence for DTI studies is the bipolar
pulse field gradient diffusion sequence in its spin-echo version
and with segmented echo planar imaging (EPI) acquisition
scheme, DTI_EPI (see Note 5).
2. Select slice geometry through the region of interest. Number
of slices, and orientation (see Note 6).
3. Choose spatial resolution, field of view (FOV), matrix size, and
slice thickness. 2D or 3D options are available, although usu-
ally 2D is performed in small animal studies since 3D requires
much longer acquisition times (see Note 7). Making the voxel
isotropic is important for fiber tracking.
4. Insert number and values for the b-value. A standard DTI data
set include acquisition of one image at low b-value (b ¼ 0
usually) and several diffusion-weighted images at only 1 high
b-value (~1000 s/mm2) (see Note 8).
5. Choose diffusion time Δ and the duration of the diffusion
gradients δ. They need to be selected trying to minimize TE
while maintaining the designated high b-value.
6. Enter number of sensitive diffusion directions, and number of
b0 images. For a proper estimation of the diffusion tensor,
5–10 measurements should be made at high b-values for
every measurement made at b0 [21, 22]. On the other hand,
a minimum SNR of 20 is recommended for the b0 image
[23]. Also, diffusion directions should be well distributed in
space (see Note 9).
7. Fat suppression should be used since aids in reducing Nyquist
ghost artifacts (see Note 10).
8. Activate respiration gating to reduce motion artifacts if neces-
sary (see Note 11).
9. Run the scan.
Sample DTI parameters for rat brain are presented in Table 1.
3.4 DTI Data 1. Diffusion tensor calculation and derived parameters, described
Processing in the introduction, can be obtained in a pixel-by-pixel basis by
using the scanner manufacture software (in our case ParaVision
5.1). It outputs maps for each component of the diffusion
tensor, each eigenvalue, each direction of the three eigenvec-
tors, the tensor trace which corresponds to the ADC value, and
the fractional anisotropy (see Fig. 3). Alternatively, free software
DTI 111
Table 1
Sample parameters for DTI acquisition in a rat brain study at 7T
4 Notes
Fig. 3 Rat brain b0 image (bottom left) and DTI parametric maps: longitudinal diffusivity (λ||, top left), axial
diffusivity (λ⊥, top middle), apparent diffusion coefficient (ADC, top right), fractional anisotropy (FA, bottom
middle), and directional encoding color (DEC, bottom right). The colors in the DEC map refer to the direction of
the primary principal diffusion direction and are coded as follows: red ¼ right-left, green ¼ dorsal-ventral, and
blue ¼ rostro-caudal. The DTI data set was acquired using the sample parameters presented in Table 1. High
ADC is observed in the ventricles and nonsignificant differences in ADC are seen between white and gray
matter. However, white matter has higher values of λ|| and FA compared to gray matter
anesthesia, thus when the animal is first located in the bed its
temperature is usually a few degrees below the target tempera-
ture (37 C). Vary water temperature in such a fashion as to
reach and keep the animal temperature constant throughout
the entire scanning time. Also try to scan at the same tempera-
ture all the animals within the same study for proper compari-
son. Automated, temperature feedback warm air systems are
usually connected to the monitoring system and work well to
compensate heating power, if body temperature changes occur.
Diffusion scans tend to increase body temperature, thus we
may anticipate these changes by lowering the bath temperature
a little bit in advance.
3. It is very important to monitor respiration rate very closely,
since different strains, and even different individuals within the
same strain, respond differently to anesthesia. Vary anesthesia
DTI 113
Acknowledgements
References
1. Stejskal EO, Tanner JE (1965) Spin diffusion revealed through MRI as increased radial (but
measurements: spin echoes in the presence of a unchanged axial) diffusion of water. Neuro-
time-dependent field gradient. J Chem Phys Image 17:1429–1436
42:288–292 13. Song SK, Yoshino J, Le TQ, Lin SJ, Sun SW,
2. Le Bihan D, Breton E (1985) Imagerie de Cross AH, Armstrong RC (2005) Demyelin-
diffusion in vivo par resonance magnetique ation increases radial diffusivity in corpus callo-
nucleaire. CR Acad Sci Paris 301:1109–1112 sum of mouse brain. NeuroImage 26:132–140
3. Merboldt KD, Hanicke W, Frahm J (1985) 14. Conturo TE, Lori NF, Cull TS, Akbudak E,
Self-diffusion NMR imaging using stimulated Snyder AZ, Shimony JS, McKinstry RC,
echoes. J Magn Reson Imaging 64:479–486 Burton H, Raichle ME (1999) Tracking neu-
4. Taylor DG, Bushell MC (1985) The spatial ronal fiber pathways in the living human brain.
mapping of translational diffusion coefficients Proc Natl Acad Sci 96:10422–10427
by the NMR imaging technique. Phys Med 15. Tournier JD, Mori S, Leemans A (2011) Dif-
Biol 30:345–349 fusion tensor imaging and beyond. Magn
5. Le Bihan D, Mangin JF, Poupon C, Clark CA, Reson Med 65:1532–1556
Pappata S, Molko N, Chabriat H (2001) Dif- 16. Lope-Piedrafita S, Totenhagen JW, Hicks CM,
fusion tensor imaging: concepts and applica- Erickson RP, Trouard TP (2008) MRI detects
tions. J Magn Reson Imaging 13:534–546 therapeutic effects in weanling Niemann-Pick
6. Moseley ME, Kucharczyk J, Mintorovitch J, type C mice. J Neurosci Res 86:2802–2807
Cohen Y, Kurhanewicz J, Derugin N, 17. Breu M, Zhang J, Porambo M, Pletnikov MV,
Asgari H, Norman D (1990) Diffusion- Goeral K, Kakara M, Johnston MV, Fatemi A
weighted MR imaging of acute stroke: correla- (2016) Diffusion tensor imaging abnormalities
tion with T2-weighted and magnetic in the cerebral white matter correlate with
susceptibility-enhanced MR imaging in cats. sex-dependent neurobehavioral deficits in
Am J Neuroradiol 11:423–429 adult mice with neonatal ischemia. Dev Neu-
7. Chenevert TL, McKeever PE, Ross BD (1997) rosci 38:83–95
Monitoring early response of experimental 18. Lope-Piedrafita S, Garcia-Martin ML, Galons
brain tumors to therapy using diffusion mag- JP, Gillies RJ, Trouard TP (2008) Longitudinal
netic resonance imaging. Clin Cancer Res diffusion tensor imaging in a rat brain glioma
3:1457–1466 model. NMR Biomed 21:799–808
8. Dmytriw AA, Sawlani V, Shankar J (2017) 19. Gimenez U, Perles-Barbacaru AT, Millet A,
Diffusion-weighted imaging of the brain: Appaix F, El-Atifi M, Pernet-Gallay K, van der
beyond stroke. Can Assoc Radiol J 68 Sanden B, Berger F, Lahrech H (2016) Micro-
(2):131–146. https://doi.org/10.1016/j. scopic DTI accurately identifies early glioma
carj.2016.10.001 cell migration: correlation with multimodal
9. Basser PJ, Mattiello J, Le Bihan D (1995) MR imaging in a new glioma stem cell model.
diffusion tensor spectroscopy and imaging. NMR Biomed 29:1553–1562
Biophys J 66:259–267 20. Jiang H, van Zijl PC, Kim J, Pearlson GD,
10. Skare S, Hedehus M, Moseley ME, Li TQ Mori S (2006) DtiStudio: resource program
(2000) Condition number as a measure of for diffusion tensor computation and fiber
noise performance of diffusion tensor data bundle tracking. Comput Methods Prog
acquisition schemes with MRI. J Magn Reson Biomed 81:106–116
147:340–352 21. Jones DK, Horsfield MA, Simmons A (1999)
11. Mattiello J, Basser PJ, Le Bihan D (1997) The Optimal strategies for measuring diffusion in
b matrix in diffusion tensor echo-planar imag- anisotropic systems by magnetic resonance
ing. Magn Reson Med 37:292–300 imaging. Magn Reson Med 42:515–525
12. Song SK, Sun SW, Ramsbottom MJ, Chang C, 22. Alexander DC, Barker GJ (2005) Optimal
Russell J, Cross AH (2002) Dysmyelination imaging parameters for fiber-orientation
116 Silvia Lope-Piedrafita
estimation in diffusion MRI. NeuroImage 29. Pu LL, Trouard TP, Ryan L, Huang C, Altbach
27:357–367 MI, Bilgin A (2011) Model-based compressive
23. Mukherjee P, Chung SW, Berman JI, Hess CP, diffusion tensor imaging. 8th IEEE Int Symp
Henry RG (2008) Diffusion tensor MR imag- Biomed Imaging 1:254–257
ing and fiber tractography: technical considera- 30. Shi X, Ma X, Wu W, Huang F, Yuan C, Guo H
tions. Am J Neuroradiol 29:843–852 (2015) Parallel imaging and compressed sens-
24. Liu B, Zhu T, Zhong J (2015) Comparison of ing combined framework for accelerating high-
quality control software tools for diffusion ten- resolution diffusion tensor imaging using inter-
sor imaging. Magn Reson Imaging image correlation. Magn Reson Med
33:276–285 73:1775–1785
25. Sarlls JE, Pierpaoli C (2009) In vivo diffusion 31. Gr€u tter R (1993) Automatic, localized in Vivo
tensor imaging of the human optic chiasm at adjustment of all firstand second-order shim
sub-millimeter resolution. NeuroImage coils. Magn Reson Med 29:804–811
47:1244–1251 32. Kanayama S, Kuhara S, Satoh K (1996) In vivo
26. Totenhagen JW, Lope-Piedrafita S, Borbon IA, rapid magnetic field measurement and shim-
Yoshimaru ES, Erickson RP, Trouard TP ming using single scan differential phase
(2012) In vivo assessment of neurodegenera- mapping. Magn Reson Med 36:637–642
tion in Niemann-Pick type C mice by quantita- 33. Hasan KM, Parker DL, Alexander AL (2001)
tive T2 mapping and diffusion tensor imaging. Comparison of gradient encoding schemes for
J Magn Reson Imaging 35:528–536 diffusion-tensor MRI. J Magn Reson Imaging
27. Pipe JG, Farthing VG, Forbes KP (2002) Mul- 13:769–780
tishot diffusion-weighted FSE using PROPEL- 34. Jones DK (2004) The effect of gradient sam-
LER MRI. Magn Reson Med 47:42–52 pling schemes on measures derived from diffu-
28. Bammer R, Holdsworth SJ, Veldhuis WB, sion tensor MRI: a Monte Carlo study. Magn
Skare ST (2009) New methods in diffusion- Reson Med 51:807–815
weighted and diffusion tensor imaging. Magn
Reson Imaging Clin N Am 17:175–204
Chapter 8
Abstract
Since its discovery in the early 90s, BOLD signal-based functional Magnetic Resonance Imaging (fMRI)
has become a fundamental technique for the study of brain activity in basic and clinical research. Functional
MRI signals provide an indirect but robust and quantitative readout of brain activity through the tight
coupling between cerebral blood flow and neuronal activation, the so-called neurovascular coupling.
Combined with experimental techniques only available in animal models, such as intracerebral micro-
stimulation, optogenetics or pharmacogenetics, provides a powerful framework to investigate the impact
of specific circuit manipulations on overall brain dynamics. The purpose of this chapter is to provide a
comprehensive protocol to measure brain activity using fMRI with intracerebral electric micro-stimulation
in murine models. Preclinical research (especially in rodents) opens the door to very sophisticated and
informative experiments, but at the same time imposes important constrains (i.e., anesthetics, translatabil-
ity), some of which will be addressed here.
1 Introduction
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0_8, © Springer Science+Business Media, LLC 2018
117
118 Laura Pérez-Cervera et al.
2 Materials
2.3 Functional MRI The additional materials listed here are required to produce electri-
cal brain stimulation in the rat while it remains in the magnet and
acquire and process imaging data.
2.3.1 MRI Magnet We use a horizontal 7 Tesla scanner with a 30 cm diameter bore
(Biospec 70/30v, Bruker Medical, Ettlingen, Germany). The sys-
tem has a 675 mT/m actively shielded gradient coil (Bruker, BGA
12-S) of 11.4 cm inner diameter. Data is acquired and
pre-processed with a Hewlett-Packard console running Paravision
5.1 software (Bruker Medical GmbH, Ettlingen, Germany)
operating on a Linux platform.
2.3.2 MRI Phase It is not indispensable, but it is highly convenient. The expected
Array Coil signal changes are in the range of 1–3% of the total signal intensity,
thus it will critically contribute to achieve the highest possible
signal-to-noise ratio (SNR). We employ a1H rat brain receive-only
phase array coil with integrated combiner and preamplifier, no
tune/no match, in combination with the actively detuned
transmit-only resonator (BrukerBioSpin MRI GmbH, Germany).
2.3.4 Heating System It is crucial to keep the animal’s temperature in the physiological
range, and to maintain it stable (37 0.5 C) in order to preserve
vascular reactivity in response to neuronal activation. We use a water
blanket connected to a water bath (Thermo Scientific SAHARA
Heated Bath Circulators S5P) controlled by a temperature regu-
latory system (Thermo Scientific STANDARD Series Thermostats
SC150) (see Note 2). In this system, precise control of the animal’s
temperature requires the constant attention of dedicated personnel
to manually vary the temperature in the heating bath. In order to
automate this process, a home-made and inexpensive closed loop
regulation system has been developed to adjust water bath temper-
ature to keep a constant body temperature in the animal (see details
in Note 3).
2.3.5 Other Devices 1. Pulse generator and current source (STG2004, Multichannel
and Small Equipments Systems, Reutlingen, Germany) for electric micro-stimulation.
2. Digital Oscilloscope to check electrode functionality.
3. Eye ointment (even if it is an acute procedure).
4. MRI-compatible stereotaxic device with ear- and bite-bars.
5. Agarose (0.5%) in saline. It is prepared and introduced in a
10 cc syringe. It can be storage in the fridge before its use (see
Note 4).
6. MRI sequences: gradient Echo (GE)-Echo Planar Imaging
(EPI) sequence providing adequate temporal and spatial reso-
lution (see Notes 5 and 6); and T2-weighted anatomical
images, like a Rapid Acquisition Relaxation Enhanced
(RARE) sequence (see Note 7).
2.3.6 Image Analysis There are some commercially available software tools for fMRI
Software analysis. In our case, fMRI data are analyzed offline using our
own software developed in MatLab, which included Statistical
Parametric Mapping package (SPM8, http://fil.ion.ucl.ac.uk/
spm), Analysis of Functional NeuroImages (AFNI, http://afni.
nimh.nih.gov/afni), and FSL Software (FMRIB http://fsl.fmrib.
ox.ac.uk/fsl).
3 Methods
All animal work should be carried out only upon review and
approval of the methods by your institution’s Animal Care and
Use Committee [11, 12]. For those new to MRI and small animal
surgery, prior to initiating any studies, training and advice should
be sought from experts in the field.
Due to the presence of strong magnetic fields, surgery is per-
formed in an area separated from the magnet room. Thus, it will be
Electric-Stimulation fMRI 123
fMRI
Stim.
ON OFF
Dummy scans (not stored)
Fig. 1 Scheme representing the image acquisition (up) and the stimulus (down) during the time course of an
fMRI experiment. Green squares represent the time for dummy scans at the beginning of the acquisition
3.4 Intracerebral Most of the BOLD based fMRI experiments are acquired using EPI
Electrode Implant images, which are very sensitive to T2* changes. Practically
for fMRI speaking, a number of factors can confabulate causing a deteriora-
tion of the quality of the functional images. When working with
surgically manipulated animals, especially in acute preparations,
extreme care has to be taken to minimize bleeding. After the
surgery and before cementing the implant to the skull (see below)
thorough cleaning of the exposed cranium is mandatory.
In order to improve the SNR, the tip of the electrode is bent
(using a burner and some forceps) to form a 90 angle, so it could
go inside the brain leaving the main body of the pipette outside
parallel to the head of the rat, minimizing the implant’s height and
allowing a closer proximity between the MRI array coil and the
head of the animal.
The method described here is based on standard procedures
used in electrophysiological experiments with rats adapted to the
MRI requirements. Similar protocols are used for mice.
1. Weigh the animal.
2. Dissolve urethane in sterile 0.9% saline. Warm it to room
temperature before injection.
126 Laura Pérez-Cervera et al.
3.5 fMRI In order to prevent temperature drop, preheat the magnet’s heat-
ing system before the animal arrives to the facility.
1. Place the animal on the MRI bed.
2. Check that the correct level of anesthesia has been maintained.
3. Insert the rectal temperature probe using lubricating jelly and
tape it in place.
4. Fix the animal’s head in an MRI-compatible stereotaxic device.
5. Place the physiological monitoring device (MRI-compatible
sensor with foot clip) or the breathing piezoelectric sensor.
128 Laura Pérez-Cervera et al.
6. Cover the exposed skull and the implant with agarose, with
special emphasis in filling all the possible empty spaces between
the head of the animal and the coil (see Note 14).
7. Connect the electrode to the current source.
8. Using the oscilloscope, cross-check the impedance of the elec-
trode that should match the value obtained during its fabrica-
tion, discarding a possible breakdown in the process of
implantation or during the accommodation of the animal in
the MRI setup.
9. Fix the coil in the MRI bed over the head of the animal, as close
as possible to the skull (see Note 15).
10. Place the animal inside the RF coil aligning the approximate
center of the brain with the magnet isocenter.
11. Acquire T2-weighted anatomical images in the three orthogo-
nal planes.
12. Even when the animal positioning is accurate, there can be
small inter-animal differences when defining an exact position.
In order to do grouped analyses, it is interesting to minimize
this variability. Thus, we recommend using anatomical land-
marks to position EPI slices always in the same orientation. A
possible strategy is:
(a) Take the plane that cut the base of cerebellum and the
anterior commissure (see Fig. 2a).
(b) Take the midline plane that separates the brain in left and
right hemispheres (Fig. 2b, c).
(c) Use the above anatomical planes to define the angle and
positioning of the slices for functional imaging. In our
case, 15 slices are positioned perpendicular to the planes
with the sixth more anterior slice containing the anterior
commissure (Fig. 2d).
13. Use a shimming procedure to adjust field homogeneity in the
brain. In our case, we use the MapShim macro implemented by
Bruker.
14. Adjust the EPI images according to the landmarks mentioned
in step 12 (see Note 16) and use saturation slices around the
brain. Acquire a set of EPI images without stimulation to check
proper image acquisition (no folding, ghosts, etc.)
15. Acquire an anatomical image with the same geometry than the
EPI images but higher (at least double) in plane spatial resolu-
tion. It will help to identify anatomical landmarks and
co-registration of brain templates for grouped analysis.
16. Start data acquisition.
Electric-Stimulation fMRI 129
Fig. 2 Position of selected slices using anatomical landmarks. Sagittal image (a) is used to calculate the angle
of the plane (pink) defined by the anterior commissure and the base of the cerebellum. Horizontal (b) and
coronal (c) images are used to calculate the plane (pink) that goes through the midline. Finally, EPI planes for
functional imaging (d) are positioned perpendicular to the anatomical planes calculated previously. To assure
a similar anteroposterior location of the EPI images across animals, an anatomical landmark is also used, in
our case the sixth most anterior slices is located on the anterior commissure (d, yellow slice)
4 Notes
Corr.
5 mm
0
2
1
0
-1
-2
0 20 40 60 80 100 120 140 160 180 200 220 240 260 280 300
Time (s)
Fig. 3 Brain-wide functional connectivity of dorsal CA3. (a) fMRI BOLD map overlaid on anatomical
T2-weighted images. Color-code denotes the correlation of the BOLD signal with the stimulation protocol
convolved with a hemodynamic function (see text). The arrow shows the artifact caused by the carbon
electrode. Arrowheads show agarose. (b) Global BOLD signal time course of the significantly (P < 0.001)
activated voxels in response to ten trains of stimulation (indicated by vertical gray bars), delivered at 10 Hz.
Graph shows the mean of 3 repetitions of the same protocol
132 Laura Pérez-Cervera et al.
15. Avoid excessive pressure between the coil and electrode. Usu-
ally electrodes are fragile and break easily during the experi-
ment if there is some of pressure on them.
16. The employed FOV usually exceeds the cross section of the
subject to prevent artifacts from image folding. The slice thick-
ness is 1 mm for rats and 0.8 mm for mice, but depending on
the SNR and the expected level of activation, it could be
decreased.
Acknowledgements
References
12. European Convention for the Protection of further evidence for the lack of electrically
vertebrate animals used for experimental and induced polysynaptic propagation of activity
other scientific purposes (2006) Appendix in the neocortex. Magn Reson Imaging 29
A. Guidelines for accommodation and care of (10):1374–1381. https://doi.org/10.1016/j.
animals (Article 5 of the Convention) mri.2011.04.005
13. Hendrich KS, Kochanek PM, Melick JA, Schid- 22. Alvarez-Salvado E, Pallares V, Moreno A,
ing JK, Statler KD, Williams DS, Marion DW, Canals S (2014) Functional MRI of long-term
Ho C (2001) Cerebral perfusion during anes- potentiation: imaging network plasticity. Philos
thesia with fentanyl, isoflurane, or pentobarbi- Trans R Soc Lond Ser B Biol Sci 369
tal in normal rats studied by arterial spin- (1633):20130152. https://doi.org/10.
labeled MRI. Magn Reson Med 46 1098/rstb.2013.0152
(1):202–206 23. Canals S, Beyerlein M, Murayama Y, Logothe-
14. Schroeter A, Schlegel F, Seuwen A, tis NK (2008) Electric stimulation fMRI of the
Grandjean J, Rudin M (2014) Specificity of perforant pathway to the rat hippocampus.
stimulus-evoked fMRI responses in the Magn Reson Imaging 26(7):978–986.
mouse: the influence of systemic physiological https://doi.org/10.1016/j.mri.2008.02.018
changes associated with innocuous stimulation 24. Godino Mdel C, Romera VG, Sanchez-
under four different anesthetics. NeuroImage Tomero JA, Pacheco J, Canals S, Lerma J,
94:372–384. https://doi.org/10.1016/j. Vivancos J, Moro MA, Torres M, Lizasoain I,
neuroimage.2014.01.046 Sanchez-Prieto J (2013) Amelioration of ische-
15. Sonnay S, Just N, Duarte JM, Gruetter R mic brain damage by peritoneal dialysis. J Clin
(2015) Imaging of prolonged BOLD response Invest 123(10):4359–4363. https://doi.org/
in the somatosensory cortex of the rat. NMR 10.1172/JCI67284
Biomed 28(3):414–421. https://doi.org/10. 25. Hadar R, Vengeliene V, Barroeta Hlusicke E,
1002/nbm.3263 Canals S, Noori HR, Wieske F, Rummel J,
16. Paasonen J, Salo RA, Shatillo A, Forsberg MM, Harnack D, Heinz A, Spanagel R, Winter C
Narvainen J, Huttunen JK, Grohn O (2016) (2016) Paradoxical augmented relapse in
Comparison of seven different anesthesia pro- alcohol-dependent rats during deep-brain
tocols for nicotine pharmacologic magnetic stimulation in the nucleus accumbens. Transl
resonance imaging in rat. Eur Neuropsycho- Psychiatry 6(6):e840. https://doi.org/10.
pharmacol 26(3):518–531. https://doi.org/ 1038/tp.2016.100
10.1016/j.euroneuro.2015.12.034 26. Tehovnik EJ, Tolias AS, Sultan F, Slocum WM,
17. Maggi CA, Meli A (1986) Suitability of ure- Logothetis NK (2006) Direct and indirect acti-
thane anesthesia for physiopharmacological vation of cortical neurons by electrical micro-
investigations in various systems. Part 2: Car- stimulation. J Neurophysiol 96(2):512–521.
diovascular system. Experientia 42 https://doi.org/10.1152/jn.00126.2006
(3):292–297 27. Pallares V, Moya J, Samper-Belda FJ, Canals S,
18. Moreno A, Morris RG, Canals S (2016) Moratal D (2015) Neurosurgery planning in
Frequency-dependent gating of hippocampal- rodents using a magnetic resonance imaging
neocortical interactions. Cereb Cortex 26 assisted framework to target experimentally
(5):2105–2114. https://doi.org/10.1093/ defined networks. Comput Methods Prog
cercor/bhv033 Biomed 121(2):66–76. https://doi.org/10.
19. Pawela CP, Biswal BB, Hudetz AG, Schulte 1016/j.cmpb.2015.05.011
ML, Li R, Jones SR, Cho YR, Matloub HS, 28. Eklund A, Nichols TE, Knutsson H (2016)
Hyde JS (2009) A protocol for use of medeto- Cluster failure: why fMRI inferences for spatial
midine anesthesia in rats for extended studies extent have inflated false-positive rates. Proc
using task-induced BOLD contrast and Natl Acad Sci U S A 113(28):7900–7905.
resting-state functional connectivity. Neuro- https://doi.org/10.1073/pnas.1602413113
Image 46(4):1137–1147. https://doi.org/ 29. Poldrack RA, Mumford JA, Nichols TE (2011)
10.1016/j.neuroimage.2009.03.004 Handbook of functional MRI data analysis.
20. Shyu BC, Lin CY, Sun JJ, Sylantyev S, Chang C Cambridge University Press, New York
(2004) A method for direct thalamic stimula- 30. Ashby FG (2011) Statistical analysis of FMRI
tion in fMRI studies using a glass-coated car- Data. MIT Press, Cambridge, MA
bon fiber electrode. J Neurosci Methods 137 31. Paxinos G, Watson C (2007) The rat brain in
(1):123–131. https://doi.org/10.1016/j. stereotaxic coordinates. Academic Press, Else-
jneumeth.2004.02.015 vier, New York
21. Sultan F, Augath M, Murayama Y, Tolias AS,
Logothetis N (2011) esfMRI of the upper STS:
Chapter 9
Abstract
Functional diffusion magnetic resonance imaging (fDMRI) is a noninvasive technique that allows elucidat-
ing physiological and anatomical changes at a microscopic scale by detection of water molecular displace-
ments in tissue structures. These displacements likely reflect microstructural changes associated with
neuronal or glial cells activation. In this chapter, we will describe the physical and biological concepts of
fDMRI and how images of brain activation can be acquired in a preclinical setup.
Key words Magnetic resonance imaging, Functional imaging, Diffusion imaging, Diffusion biexpo-
nential parameters
1 Introduction
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0_9, © Springer Science+Business Media, LLC 2018
135
136 Rita Maria Rocha Oliveira et al.
S ðb Þ
¼ SDP:e b:Dslow þ FDP:e b:Dfast ð2Þ
S ð0Þ
behaviors [14–16] that have been published but from our point of
view that one is the most correct approach to interpret molecular
water motion as a sensitive marker to study changes associated with
physiological and pathological states.
Some authors have demonstrated changes in water coefficients
diffusion during neural activation. In their investigations, they
quantified slow and fast diffusion phases and they observed micro-
structural properties changes that are probably linked to volume
variations and cell swelling. Such cell size variations were inter-
preted as a reflection from the neuronal soma, dendritic areas, or
also glial cells [17–19]. These results strongly suggest that diffusion
MRI can be used as a powerful technique to monitor neural activa-
tion. Based on this, the biexponential DWI analysis brings forward
a new approach to produce brain images directly associated with
neural activation.
2 Materials
2.2 Other Equipment, 1. Monitoring System: Small animal monitoring and gating sys-
Devices, and Materials tem with a rectal sensor and a pneumatic pillow placed in the
animal abdomen that continuously reports data of body tem-
perature and respiratory rate. This allows to monitor the state
of the anesthetized animal inside the magnet and controlling
the level of anesthesia.
2. Anesthesia device: It is formed by an anesthetic vaporizer, a
flowmeter, a poly(methylmethacrylate) (PMMA) chamber, a
nose cap, gases circulating tubes, and a filter. The small rodent
inhalation anesthesia equipment allows to induced and control
the anesthetized state of the animal.
3. Thermal blanket: Anesthetized animals are placed on a water
heated blanket to maintain the body temperature at approx.
37 C during scanning (any other heating setup can be used).
Functional Diffusion MRI 139
3 Methods
3.2 Scan Acquire a position image of the animal in the three spatial orienta-
tions (axial, coronal, and sagittal) by using a fast-low angle shot
3.2.1 Localization/
(FLASH) sequence (these sequences are called Tripilot or Location
Positioning Images
in Bruker systems). This allows to fix the correct position inside the
magnet (see Note 2).
140 Rita Maria Rocha Oliveira et al.
Table 1
Preclinical MRI system and associated equipment/devices for developing the functional DWI
protocols described in this chapter
3.2.3 Functional For obtaining functional diffusion images, DWI sequences avoid-
Diffusion Imaging ing low b-values have to be used. We present here three different
methods optimized in a Bruker Biospect® MRI system for acquir-
ing fDWI depending on the collection of the MR data (k-space
filling):
1. Standard acquisition. It is based on a SE sequence in which
each line of the k-space (echo signal) is filled at each phase
Functional Diffusion MRI 141
encoding step (Fig. 1). This method entails a very good signal-
to-noise ratio (SNR) but suffers of a long acquisition time,
increasing the possibility of animal movements that cause arti-
facts (see Note 3). The operator has to select the number of
spatial directions to apply the diffusion gradients.
2. Single-shot echo-planar imaging (ss-EPI). The whole k-space is
filled after the first 90 –180 pulse set in the SE sequence, and
each image acquisition takes the TR value. EPI acquisition is
much faster than standard one, nevertheless suffers from the
presence of artifacts (EPI ghost) and has low SNR. These
disadvantages make mandatory to achieve a very good shim-
ming of the magnetic field to reduce the artifacts associated to
the echo-planar readout (see above). Also, the adjustment of
the basic frequency need to be made previously the acquisition
of each diffusion EPI sequence.
3. Segmented EPI. In this case, K-space is filled in different shots
or segments, that is, several lines are collected after each
90 –180 pulse combination. This sequence has a better SNR
(see Note 4) but it is slower than the ss-EPI as many times as
segments selected. In this case, it is also crucial to get a good
shimming.
Before starting EPI DWI acquisition, it is necessary to shim
adequately the system to get the higher homogeneity of the mag-
netic field. In Bruker systems there are several automatic options to
achieve that:
1. Adjusting the first and second shim coil values to optimize the
shimming of the whole sample covered based on the signal
response after each modification.
2. Using the FASTMAP module implemented in Bruker scanners
[22]. This efficient tool allows to shimming in a localized cubic
region which is virtually divided in smaller sticks. The main
disadvantage of this method is that not always the volume of
interest can be covered by a cubic space.
3. Employing the MAPSHIM protocol that optimizes the mag-
netic field homogeneity in a rectangular region of arbitrary size
and orientation [23] after the acquisition of a 3D magnetic
field distribution.
Once the shim is improved, the DWI sequence is acquired after
selecting the b-values that will control the diffusion weighting in
the study (see Note 5). Figure 2 shows representative T2 and DW
images obtained in a diffusion study. Table 2 shows the parameters
of a common diffusion sequence (see Note 6).
3.3 Post-scan The values of fast and slow diffusion coefficients (FDC and SDC,
Analysis respectively) and fast and slow diffusion phases (FDP and SDP) can
142 Rita Maria Rocha Oliveira et al.
Fig. 2 MRI study of a rat brain in axial orientation. T2-weighted (a) and diffusion-weighted (b) images of the
same slice. The acquisition parameters of the DWI sequence are showed in Table 2
Table 2
Acquisition parameters of a diffusion-weighted MRI study of a mouse brain in a Bruker Biospect® 7T
MRI system
Fig. 3 Functional diffusion MRI study carried out in a rat model of paclitaxel induced-peripheral neuropathy.
Parametric images shown correspond to the application of the diffusion gradient in the left-right (L–R)
direction, and acquired in axial orientation. (a) Diffusion-weighted image, (b) fast diffusion coefficient (FDC)
map, (c) slow diffusion coefficient (SDC) map, (d) slow diffusion phase (SDP) map. The acquisition parameters
of the DWI sequence are showed in Table 2
3.4 Applications Functional diffusion MRI is a recent approach for assessing brain
functionality also in animal models and human being (see Note 7).
In our group we have previously used this methodology for analyz-
ing hypothalamic activity of healthy subjects under an appetite
paradigm, also in the mouse and human brain [17], and with
mice genetically deficient in leptin or neuropeptide Y [24]. Besides,
we characterized the hypothalamic metabolic compartmentation
during appetite regulation in mice [25], and carried out fDWI
studies in glioma bearing rats and an animal model of chemothera-
peutic induced-peripheral neuropathy (unpublished results).
This tool can be employed for studying specific activation path-
ways in the brain (using or not dedicated paradigm) and for
144 Rita Maria Rocha Oliveira et al.
4 Notes
Acknowledgments
References
1. Padhani AR, Liu G, Koh DM, Chenevert TL, imaging as a cancer biomarker: consensus and
Thoeny HC, Takahara T, Dzik-Jurasz A, Ross recommendations. Neoplasia 11(2):102–125
BD, Van Cauteren M, Collins D, Hammoud 2. Le Bihan D (2007) The ‘wet mind’: water and
DA, Rustin GJ, Taouli B, Choyke PL (2009) functional neuroimaging. Phys Med Biol 52
Diffusion-weighted magnetic resonance
Functional Diffusion MRI 147
In Vivo Spectroscopy
Chapter 10
Abstract
In vivo Magnetic Resonance Spectroscopy (MRS) allows the non-invasive detection and quantification of a
number of metabolites from localized volumes within a living organism. MRS localization techniques can
be divided into two main groups, single voxel and multi-voxel. Single voxel techniques provide the
metabolic profile from a specific small volume, whereas multi-voxel techniques are used to obtain the
spatial distribution of metabolites throughout a large volume subdivided into small contiguous voxels. This
chapter describes standard protocols for the acquisition and processing of in vivo single voxel1H MRS data
from the rodent brain.
1 Introduction
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0_10, © Springer Science+Business Media, LLC 2018
151
152 M. Carmen Muñoz-Hernández and Marı́a Luisa Garcı́a-Martı́n
Table 1
NMR properties of isotopes commonly used in biological studies
follow-up [8, 9]. From the biological point of view, MRS can be
classified into three different modalities: in vitro (in-solution),
in vivo, and ex vivo (high resolution magic angle spinning,
HR-MAS). The aim of all of them when applied to biomedicine is
the characterization of the metabolic profile of living organisms.
Whereas in vitro and ex vivo spectroscopy are high resolution
techniques based on the same principles as the classical spectros-
copy used in chemistry labs, in vivo spectroscopy combines imaging
and spectroscopy methodology to obtain spectra from specific
volumes within a living organism.
Most in vivo spectroscopy studies are devoted to1H spectros-
copy, but any other nucleus with magnetic moment that is present
in biomolecules, such as phosphorus or carbon, could, in principle,
be observed.1H MRS has the advantage of its higher sensitivity
(Table 1), although it also has the drawback of the large water
signal, which has to be suppressed to allow for the observation of
tissue metabolites.31P and13C MRS, on the other hand, do not
need water suppression to be performed, but they have the disad-
vantage of their much lower sensitivity and, in the case of13C, the
low natural abundance and, therefore, the need for enriched
substrates.
Finally, in vivo spectroscopy techniques can be divided into two
main modalities, single- and multi-voxel, which allow for the detec-
tion of the average metabolic profile within a small single volume or
the spatial distribution of metabolites within a larger volume, sub-
divided into smaller ones.
In this chapter we will focus on the in vivo1H MRS studies of
the rodent brain at 9.4 T using single voxel techniques. Multi-voxel
and heteronuclear MRS will be discussed in other chapters.
1
H MRS 153
1.1 Single-Voxel1H There are several acquisition schemes that allow spectra to be
Spectroscopy obtained from a defined region within the tissue under study. In
Sequences the case of1H spectroscopy, there are basically two different meth-
ods, as described below:
1.1.1 Point RESolved This technique employs one 90 excitation pulse together with a
Spectroscopy (PRESS) [10] field gradient to select a slice in one spatial direction, and two 180
refocusing pulses, also combined with field gradients, in the other
two spatial directions, so that the intersection of the three selected
slices defines a cubic voxel. This technique is also known as double
spin echo (Fig. 1A). The first 180 pulse is applied at time τ1 after
the 90 pulse, giving rise to the formation of a spin echo at time 2τ1,
and the second 180 pulse refocuses this echo at time 2τ2. There-
fore, the final spin echo is formed at time 2τ1 þ 2τ2, which is the
total echo time (TE) of the sequence.
Fig. 1 Pulse sequences for in vivo single voxel1H NMR spectroscopy. (a) Point RESolved Spectroscopy
(PRESS), (b) STimulated Echo Acquisition Mode (STEAM), (c) voxel localization by the intersection of three
orthogonal slices
154 M. Carmen Muñoz-Hernández and Marı́a Luisa Garcı́a-Martı́n
1.1.2 STimulated Echo The STEAM sequence uses three 90 pulses combined with three
Acquisition Mode (STEAM) orthogonal field gradients to select a cubic voxel (Fig. 1b), similarly
[11] to the PRESS sequence. To understand how the signal is generated
from this selected volume, let us focus just on the effect of the three
rf pulses, leaving the gradients aside. If the two first pulses are
separated by a time TE/2 and the last two by a time TM, four
spin echoes will be generated as follows: SE12 (formed by the
combination of pulses 1 and 2), SE13 (combination of pulses
1 and 3), SE23 (combination of pulses 2 and 3), and SE123
(formed by the combination of the three pulses). SE12 will be
formed at time TE after the first pulse, SE13 will be formed at
time TEþ2TM, SE23 at time TE/2þ2TM, and S123 at time 2TM.
Moreover, besides the spin echoes, other type of echoes, the
so-called stimulated echoes (STE), will appear at time TEþTM. If
we now take into consideration the field gradients, needed for
spatial localization, together with the three rf pulses, we can
observe that the first signal that appears after the three pulses is
precisely the STE, and therefore this is the signal used in the
STEAM sequence to acquire the spectrum. Finally, the spin-echo
signals are eliminated using crusher gradients during TM to avoid
undesired contribution of these signals to the spectrum.
The main advantage of STEAM as compared to PRESS is that
the former allows for very short echo times; however, it has the
disadvantage that the stimulated echo amplitude corresponds to
about half of that obtained with the spin echo in the PRESS
sequence. This is due to the fact that the 90 pulse only takes half
of the total magnetization to the longitudinal axis, while the other
half is de-phased by the crusher gradient. Furthermore, STEAM is
more sensitive to motion and diffusion artifacts. Consequently,
PRESS will be the method of choice unless very short echo times
are needed. Indeed, PRESS is by far the most commonly used
sequence in clinical practice as well as in preclinical research.
1.2 Brain The number of metabolites that can be detected in vivo is limited by
Metabolites Detectable the intrinsic low sensitivity of magnetic resonance and by the
In Vivo reduced spectral resolution as compared to liquid and HR-MAS
NMR. As this chapter is focused on brain spectroscopy, a list of the
main cerebral metabolites typically measured in vivo, and their
metabolic meaning, is described below:
1.2.1 N-acetyl aspartic This metabolite shows a very prominent signal at 2.01 ppm,
acid (NAA) corresponding to its acetyl group. Is considered a neuronal marker
[12] and its decrease is associated with neuronal loss, which can be
due to multiple causes, such as tumors [13–15], stroke [16], mul-
tiple sclerosis [17], etc.
1.2.2 Total Choline (tCho) A prominent peak appears at 3.20 ppm which we refer to as total
choline (tCho) and contains contributions from the trimethyl
1
H MRS 155
1.2.3 Creatine (Cr) The methyl group of creatine appears as a singlet at 3.03 ppm. This
singlet actually contains signals from two metabolites that cannot
be resolved in vivo, creatine and phosphocreatine, both involved in
energy metabolism. Creatine is synthesized in the liver and trans-
ported to the brain, therefore the diseases affecting the liver chron-
ically can lead to a decrease in brain creatine [18]. Moreover,
creatine levels can be altered in other conditions, such as brain
tumors, where decreased creatine levels have been reported as a
feature of high grade glioma [19] or as a favorable prognostic factor
in low grade gliomas [20], among others.
1.2.4 Lactate The methyl group of lactate appears at 1.31 ppm, but it can barely
be detected in the brain under normal conditions because of its low
concentration. However, lactate levels increase and can be easily
detected in various pathological conditions, such as cancer [21, 22]
or ischemia [23–25]. Sometimes lactate can be masked by over-
lapping with a large signal from mobile lipids. A variety of editing
methods have been developed to differentiate both signals. The
simplest is based on the use of an echo time corresponding to 1/J
(144 ms), where J is the homonuclear coupling constant. The scalar
coupling gives rise to a phase evolution of the methyl doublet,
which depends on the echo time. At 144 ms this phase evolution
is 180 , which results in an inverted doublet in the1H NMR
spectrum [26].
1.2.5 myo-Inositol (myo- myo-Ins is much more abundant in glial cells as compared to neu-
Ins) rons and therefore its increase is associated with an increase in the
glial component. At short echo times a large myo-Ins signal can be
observed at 3.56 ppm. At long echo time, however, since it is a
strongly coupled system, its signal disappears. The myo-Ins signal
increases significantly in low grade glial tumors, where cell dediffer-
entiation is still low, and decreases in most undifferentiated tumors
(high grade) [27].
1.2.6 Glutamate (Glu) These two molecules are essential in brain metabolism, with Glu
and Glutamine (Gln) being the predominant neurotransmitter. They are both closely
related through the glutamate-glutamine cycle [28]. The methine
156 M. Carmen Muñoz-Hernández and Marı́a Luisa Garcı́a-Martı́n
proton of both Glu and Gln can be detected at short echo times
around 3.7 ppm, and the resonances from the four protons of the
two methylene groups are closely grouped between 2.1 and
2.4 ppm. At low magnetic fields there is a large overlap between
both metabolites and they are generally measured together, being
designated as Glx. At higher magnetic fields, like those used in
preclinical studies, they can sometimes be resolved, depending on
the spectral quality, but in general it is difficult to obtain reliable
measurements of both metabolites separately. Consequently, sev-
eral editing techniques have been developed in recent years to allow
both metabolites to be measured independently even at clinical
magnetic fields [29]. Glu and Gln can be altered in a wide range
of pathological conditions, such as depression [30], cancer
[31–33], hepatic encephalopathy [34, 35], multiple sclerosis
[17], etc.
2 Materials
2.2 Other Equipment 1. Vaporizer and induction chamber to anesthetize the animals
outside the MR spectrometer.
2. Small Animal Monitoring and Gating system (Small Animal
Instruments Inc. Stony Brook, NY, USA), provided with respi-
ration and temperature sensors.
3. Animal physiology monitoring software (PC-SAM 32 v8.02,
Small Animal Instrument Inc.).
1
H MRS 157
2.3 Data Analysis 1. Manufacturer software for spectroscopic data processing Top-
Spin 3.0 (Bruker, Ettlingen, Germany).
2. Third party software for quantitative analysis (as an example in
this chapter we describe the use of LCModel [36, 37], but there
are several options available).
3 Methods
3.1 Animal Handling 1. Turn on the gas flow (oxygen or air at a flow rate of 1–1.5 mL/
min) and anesthesia (4–5% isoflurane), directed to the induc-
tion chamber, and place the rat inside.
2. Once the animal is asleep, redirect the anesthesia to the animal
holder already placed on the scanner, reduce the isoflurane to
2–2.5%, and quickly move the animal to the animal cradle,
which should have a temperature control system (water or
air) to maintain temperature between 36.5 and 37.5 C during
the experimental procedure.
3. Immobilize the animal’s head using ear bars and tooth bars to
minimize motion effects during acquisition.
4. Place the breathing sensor under the lower part of the abdo-
men, so that interferences from the heartbeat are avoided, then
place the rectal temperature probe and check the animal moni-
toring unit. Anesthesia should be adjusted throughout the
experiment to maintain a breathing rate between 50 and
80 breaths per minute.
5. At the end of the experiment, remove the animal from the
animal holder and place it on a warm location until it is awake
(2–3 min). Then return the animal to its cage.
3.2 MRS 1. Position the surface RF coil as close to the skull as possible and
also properly centered respect to the brain (see Note 1).
2. Move the animal into the scanner so that the center of the
surface coil gets to the center of the volume coil, which should
have been previously position at the isocenter of the magnet.
3. Adjust the tune and match of the volume coil (see Note 2).
4. Load a positioning protocol and acquire the image with a field
of view (FOV) of 40–50 mm. Use these images to re-position
the animal inside the scanner if necessary (see Note 3).
5. Acquire axial, sagittal, and coronal T2-weighted images using
the RARE (rapid acquisition with relaxation enhancement)
sequence with the following or approximate parameters:
FOV ¼ 32 mm, matrix size ¼ 256 256, slice thick-
ness ¼ 1 mm, number of contiguous slices ¼ 10–15, repetition
158 M. Carmen Muñoz-Hernández and Marı́a Luisa Garcı́a-Martı́n
Fig. 2 Voxel positioning and shim volume selection. The voxel (4 4 4 mm3) from which the1H NMR
spectrum will be acquired is outlined in yellow, and the volume for local shim adjustment
(5.5 5.5 5.5 mm3) in green
time (TR) ¼ 2.5 s, echo time (TE) ¼ 33 ms, rare factor ¼ 4 (see
Note 4).
6. Perform a local shim adjustment. In our case this will be done
using the “2_Localized_Shim” protocol and the “Map_Shim”
subroutine, as follows:
(a) Load the protocol.
(b) Define the voxel size (4 4 4 mm3 in the example
reported herein) and the position on the T2-weighted
images (Fig. 2, see Note 5).
(c) Define the shim volume. This can be done by adding a
margin to the voxel volume (Fig. 2, see Note 6).
(d) Acquire a B0 map (see Note 7).
(e) Run the localized shim scan (see Notes 8 and 9).
7. Load the localized spectroscopy protocol, PRESS or STEAM,
and copy the voxel position from the previous experiment.
8. Define acquisition parameters, which in the current example
using PRESS are the following: sweep width ¼ 10 ppm, num-
ber of points ¼ 2048, TR ¼ 3 s, TE ¼ 35 or 144 ms, number of
averages ¼ 128.
9. Adjust the bandwidth of the RF pulses to minimize the chemi-
cal shift displacement error (see Note 10). This step is funda-
mental to avoid lipid contamination from the skull (Fig. 3).
10. Optimize water suppression for by fine-tuning the power level
of the selective pulses. In Paravision 6.0 follow the next steps:
(a) In the “Water Sup” sub-card of the “Setup” card deselect
“Calc Sup Pulse Ampl.”
(b) Start the acquisition in setup mode and manually adjust
the power level of the suppression pulses (“Vp Pulse
1
H MRS 159
Fig. 3 RF bandwidth and chemical shift displacement error. (a) The RF bandwidth has been optimized to
minimize the chemical shift displacement error and thus avoid lipid signal contamination (bandwidths of 90
and 180 are 5400 and 3600 Hz, and the corresponding chemical shift displacements are 1.04 mm and
1.57 mm); (b) The default RF bandwidths have been used, which gives rise to a chemical shift displacement of
2.24 mm in x and y directions, resulting in substantial lipid signal contamination
Fig. 4 In vivo1H NMR spectra acquired with PRESS. (a) Echo time ¼ 35 ms; (b) Echo time ¼ 144 ms. NNA: N-
acetyl aspartic acid, Glx: glutamate þ glutamine, tCr: total Creatine (phosphocreatine þ creatine), tCho: total
Choline (choline þ phosphocholine þ glycerophosphocholine), myo-Ins: myo-Inositol
3.3 Spectra Basic manual analysis can be performed using the manufacturer
Processing software (TopSpin in Bruker) or using third party software, such
as MestReNova (Mestrelab Research, Galicia, Spain). Although the
3.3.1 Basic Processing
step-by-step processing of NMR spectra is out of the scope of this
chapter, some basic steps with TopSpin software are described
below:
1. Transfer the spectra from Paravision to TopSpin (right click on
the acquired scan and transfer to TopSpin).
2. In the command line write “lb ¼ 3” and “si ¼ 4096.” Then
write “efp,” which executes exponential multiplication, Fourier
transform, and phase adjustment.
3. If phase is not properly adjusted, re-adjust it manually.
These steps should provide spectra similar to those in Fig. 4.
3.3.2 Metabolite For quantitative analysis, specific software able to provide auto-
Quantification matic processing with high reproducibility is advised, otherwise,
data comparison and hence robust statistical analysis could be
hampered. Among the most widely used are LCModel [37] and
jMRUI [38]. In this chapter we have chosen LCModel as an
example. The main steps of the analysis are described below:
1. Open FID data in LCModel (see Note 15).
2. Activate “Do water-scaling,” but do not activate “Do eddy-
current correction” (see Note 16).
3. Choose the basis-set corresponding to your acquisition con-
ditions (field strength, vendor, sequence, echo time. . .) (see
Note 17).
4. In advance settings, edit the control parameters and make sure
you add the modifications needed for your specific data type/
source. With Bruker data using Paravision 6.0, a correction
factor that accounts for the difference in receiver gain between
the suppressed and unsuppressed spectra must be applied (see
Note 18).
5. Click on “Run LCModel” and select the unsuppressed water
FID when asked. The quantified spectrum will pop up as
shown in Fig. 5 (see Note 19).
4 Notes
Fig. 5 LCModel analysis of an in vivo1H NMR spectrum acquired with PRESS. The black trace corresponds to
the experimental spectrum and the red trace to the fitted spectrum. The table shows, from left to right, the
calculated concentrations in mM (Conc.), the estimated standard deviations expressed in percent of the
estimated metabolite concentrations (% SD), the concentration ratios relative to total-creatine (/Cr þ PCr), and
the abbreviation of metabolites (Metabolite). Highlighted in blue are the metabolites with concentration
estimates of acceptable reliability (SD < 15%). Ala: alanine, Asp: aspartic acid, Cr: creatine, PCr: phospho-
creatine, GABA: gamma-aminobutyric acid, Glc: glucose, Gln: glutamine, Glu: glutamate, GPC: glyceropho-
sphocholine, PCho: phosphocholine, GSH: glutathione, Ins: myo-inositol, Lac: lactate, NAA: N-acetyl aspartic
acid, NAAG: N-acetylaspartylglutamic acid, Scyllo: scyllo-inositol, Tau: taurine, Gly: glycine. Sums of meta-
bolites are also provided when strong overlap occurs and therefore the uncertainty of the estimates in greatly
reduced by computing the sum instead of the individual metabolites
Acknowledgements
The MRI system used in this work has been funded by the Spanish
Ministry of Science and Innovation (National Plan for Scientific
Research, Development and Technological Innovation 2008-
2011) and the European Regional Development Fund
(PCT-420000-2010-3).
References
1. Bloch F (1946) Nuclear induction. Phys Rev 4. Ernst RR, Anderson WA (1966) Applications
70:460–474 of Fourier transform spectroscopy to magnetic
2. Gutowsky HS, McCall DW (1951) Nuclear resonance. Rev Sci Instrum 37:93–102
magnetic resonance fin structure in liquids. 5. Moon RB, Richards JH (1973) Determination
Phys Rev 82:748–749 of intracellular pH by 31P magnetic resonance.
3. Proctor WG, FC Y (1950) The dependence of J Biol Chem 248(20):7276–7278
a nuclear magnetic resonance frequency. Phys 6. Hoult DI, Busby SJ, Gadian DG, Radda GK,
Rev 77:717 Richards RE, Seeley PJ (1974) Observation of
166 M. Carmen Muñoz-Hernández and Marı́a Luisa Garcı́a-Martı́n
tissue metabolites using 31P nuclear magnetic 19. Yerli H, Agildere AM, Ozen O, Geyik E,
resonance. Nature 252(5481):285–287 Atalay B, Elhan AH (2007) Evaluation of cere-
7. Garcia-Martin ML, Garcia-Espinosa MA, Cer- bral glioma grade by using normal side creatine
dan S (1998) Biochemistry detectable by MRS. as an internal reference in multi-voxel 1H-MR
In: S. Cerdán AHaFT (ed) Methodology, spec- spectroscopy. Diagn Interv Radiol 13(1):3–9
troscopy and clinical MRI, Springer, Milan, pp 20. Hattingen E, Raab P, Franz K, Lanfermann H,
7–13 Setzer M, Gerlach R, Zanella FE, Pilatus U
8. Gillies RJ, Morse DL (2005) In vivo magnetic (2008) Prognostic value of choline and crea-
resonance spectroscopy in cancer. Annu Rev tine in WHO grade II gliomas. Neuroradiology
Biomed Eng 7:287–326 50(9):759–767. https://doi.org/10.1007/
9. Castillo M, Kwock L, Mukherji SK (1996) s00234-008-0409-3
Clinical applications of proton MR spectros- 21. Herholz K, Heindel W, Luyten PR, denHol-
copy. Am J Neuroradiol 17(1):1–15 lander JA, Pietrzyk U, Voges J, Kugel H,
10. Bottomley PA (1987) Spatial localization in Friedmann G, Heiss WD (1992) In vivo imag-
NMR spectroscopy in vivo. Ann N Y Acad Sci ing of glucose consumption and lactate con-
508:333–348 centration in human gliomas. Ann Neurol 31
(3):319–327. https://doi.org/10.1002/ana.
11. Frahm J, Merboldt KD, Hanicke W (1987) 410310315
Localized proton spectroscopy using stimu-
lated echoes. J Magn Reson 72:502–508 22. Law M, Yang S, Wang H, Babb JS, Johnson G,
Cha S, Knopp EA, Zagzag D (2003) Glioma
12. Simmons ML, Frondoza CG, Coyle JT (1991) grading: sensitivity, specificity, and predictive
Immunocytochemical localization of N-acetyl- values of perfusion MR imaging and proton
aspartate with monoclonal antibodies. Neuro- MR spectroscopic imaging compared with con-
science 45(1):37–45 ventional MR imaging. Am J Neuroradiol 24
13. Negendank WG, Sauter R, Brown TR, Evel- (10):1989–1998
hoch JL, Falini A, Gotsis ED, Heerschap A, 23. Higuchi T, Fernandez EJ, Maudsley AA,
Kamada K, Lee BC, Mengeot MM, Moser E, Shimizu H, Weiner MW, Weinstein PR
Padavic-Shaller KA, Sanders JA, Spraggins TA, (1996) Mapping of lactate and N-acetyl-L-
Stillman AE, Terwey B, Vogl TJ, Wicklow K, aspartate predicts infarction during acute focal
Zimmerman RA (1996) Proton magnetic res- ischemia: in vivo 1H magnetic resonance spec-
onance spectroscopy in patients with glial troscopy in rats. Neurosurgery 38
tumors: a multicenter study. J Neurosurg 84 (1):121–129. discussion 129–130
(3):449–458. https://doi.org/10.3171/jns.
1996.84.3.0449 24. Graham GD, Blamire AM, Howseman AM,
Rothman DL, Fayad PB, Brass LM, Petroff
14. Poptani H, Gupta RK, Roy R, Pandey R, Jain OA, Shulman RG, Prichard JW (1992) Proton
VK, Chhabra DK (1995) Characterization of magnetic resonance spectroscopy of cerebral
intracranial mass lesions with in vivo proton lactate and other metabolites in stroke patients.
MR spectroscopy. Am J Neuroradiol 16 Stroke 23(3):333–340
(8):1593–1603
25. Woo CW, Lee BS, Kim ST, Kim KS (2010)
15. Brandao LA, Castillo M (2013) Adult brain Correlation between lactate and neuronal cell
tumors: clinical applications of magnetic reso- damage in the rat brain after focal ischemia: An
nance spectroscopy. Neuroimaging Clin N Am in vivo 1H magnetic resonance spectroscopic
23(3):527–555. https://doi.org/10.1016/j. (1H-MRS) study. Acta Radiol 51(3):344–350.
nic.2013.03.002 https://doi.org/10.3109/
16. Barker PB, Gillard JH, van Zijl PC, Soher BJ, 02841850903515395
Hanley DF, Agildere AM, Oppenheimer SM, 26. Kelley DA, Wald LL, Star-Lack JM (1999)
Bryan RN (1994) Acute stroke: evaluation Lactate detection at 3T: compensating J cou-
with serial proton MR spectroscopic imaging. pling effects with BASING. J Magn Reson
Radiology 192(3):723–732. https://doi.org/ Imaging 9(5):732–737
10.1148/radiology.192.3.8058940
27. Castillo M, Smith JK, Kwock L (2000) Corre-
17. Sajja BR, Wolinsky JS, Narayana PA (2009) lation of myo-inositol levels and grading of
Proton magnetic resonance spectroscopy in cerebral astrocytomas. AJNR Am J Neurora-
multiple sclerosis. Neuroimaging Clin N Am diol 21(9):1645–1649
19(1):45–58. https://doi.org/10.1016/j.nic.
2008.08.002 28. Sonnewald U, Schousboe A (2016) Introduc-
tion to the glutamate-glutamine cycle. Adv
18. Ross B, Michaelis T (1994) Clinical applica- Neurobiol 13:1–7. https://doi.org/10.1007/
tions of magnetic resonance spectroscopy. 978-3-319-45096-4_1
Magn Reson Q 10(4):191–247
1
H MRS 167
29. Ramadan S, Lin A, Stanwell P (2013) Gluta- 35. Cordoba J, Sanpedro F, Alonso J, Rovira A
mate and glutamine: a review of in vivo MRS in (2002) 1H magnetic resonance in the study
the human brain. NMR Biomed 26 of hepatic encephalopathy in humans. Metab
(12):1630–1646. https://doi.org/10.1002/ Brain Dis 17(4):415–429
nbm.3045 36. Provencher SW (1993) Estimation of metabo-
30. Yildiz-Yesiloglu A, Ankerst DP (2006) Review lite concentrations from localized in vivo pro-
of 1H magnetic resonance spectroscopy find- ton NMR spectra. Magn Reson Med 30
ings in major depressive disorder: a meta- (6):672–679
analysis. Psychiatry Res 147(1):1–25. https:// 37. Provencher SW (2001) Automatic quantitation
doi.org/10.1016/j.pscychresns.2005.12.004 of localized in vivo 1H spectra with LCModel.
31. Hazany S, Hesselink JR, Healy JF, Imbesi SG NMR Biomed 14(4):260–264
(2007) Utilization of glutamate/creatine ratios 38. Naressi A, Couturier C, Castang I, de Beer R,
for proton spectroscopic diagnosis of menin- Graveron-Demilly D (2001) Java-based graph-
giomas. Neuroradiology 49(2):121–127. ical user interface for MRUI, a software pack-
https://doi.org/10.1007/s00234-006-0167- age for quantitation of in vivo/medical
z magnetic resonance spectroscopy signals.
32. Li Y, Park I, Nelson SJ (2015) Imaging tumor Comput Biol Med 31(4):269–286
metabolism using in vivo magnetic resonance 39. Gruetter R (1993) Automatic, localized in vivo
spectroscopy. Cancer J 21(2):123–128. adjustment of all first- and second-order shim
https://doi.org/10.1097/PPO. coils. Magn Reson Med 29(6):804–811
0000000000000097 40. Garcia-Martin ML, Adrados M, Ortega MP,
33. Rijpkema M, Schuuring J, van der Meulen Y, Fernandez Gonzalez I, Lopez-Larrubia P,
van der Graaf M, Bernsen H, Boerman R, van Viano J, Garcia-Segura JM (2011) Quantita-
der Kogel A, Heerschap A (2003) Characteri- tive (1) H MR spectroscopic imaging of the
zation of oligodendrogliomas using short echo prostate gland using LCModel and a dedicated
time 1H MR spectroscopic imaging. NMR basis-set: correlation with histologic findings.
Biomed 16(1):12–18. https://doi.org/10. Magn Reson Med 65(2):329–339. https://
1002/nbm.807 doi.org/10.1002/mrm.22631
34. Chavarria L, Alonso J, Garcia-Martinez R, 41. Barker PB, Soher BJ, Blackband SJ, Chatham
Simon-Talero M, Ventura-Cots M, JC, Mathews VP, Bryan RN (1993) Quantita-
Ramirez C, Torrens M, Vargas V, Rovira A, tion of proton NMR spectra of the human
Cordoba J (2013) Brain magnetic resonance brain using tissue water as an internal concen-
spectroscopy in episodic hepatic encephalopa- tration reference. NMR Biomed 6(1):89–94
thy. J Cereb Blood Flow Metab 33
(2):272–277. https://doi.org/10.1038/
jcbfm.2012.173
Chapter 11
Abstract
Magnetic Resonance Spectroscopy is a technique that has the capability of measuring metabolites in vivo
and, in appropriate conditions, to infer its metabolic rates. The success of MRS depends a lot on its
sensitivity, which limits the usage of X-nuclei MRS. However, technological developments and refinements
in methods have made in vivo heteronuclear MRS possible in humans and in small animals. This chapter
provides detailed descriptions of the main procedures needed to perform successful in vivo heteronuclear
MRS experiments, with a particular focus on experimental setup in 13C MRS experiments in rodents.
13
Key words Magnetic Resonance Spectroscopy, X-nuclei, Surface coil, Infusion, Tracer, C labeled
glucose, Mouse
1 Introduction
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0_11, © Springer Science+Business Media, LLC 2018
169
170 Blanca Lizarbe et al.
Fig. 1 1H localized broadband 13C NMR spectroscopy of the rat brain in vivo at 9.4 T. (a) the original DEPT
sequence; (b) modified sequence. The 13C part of DEPT is replaced by a segmented adiabatic 0 BIR-4 pulse.
Dephasing gradients (1 ms duration, 46 mT/m) are added between the RF pulses in DEPT to eliminate
unwanted, offset-dependent coherences. The flip angle of last 1H pulse is set to a nominal 45 to detect
signals simultaneously from the CH, CH2, and CH3 groups
Fig. 2 (a) Sequence diagram of SPECIAL-BISEP (TE ¼ 2.8 ms). The editing is achieved by applying an AFP in
the 13C channel on alternate scans; (b) in vitro validation of the editing scheme performed at 14.1 T on a
solution of 67% enriched [2-13C]sodium acetate (no 13C decoupling during the acquisition is applied). When
the 13C AFP is turned off, the inversion BISEP pulse acts as a 0 BIR-4 pulse (top), while when AFP is turned on,
13
C-coupled 1H resonances are inverted (middle). In the difference spectrum (bottom), the uncoupled
resonances (1H-[2-12C]) are minimized and only the 13C-coupled 1H resonances (1H-[2-13C]) are detected
1.2.1 Design of Specific RF coils are frequency-dependent, and a coil that is to be used
RF Coils during heteronuclear NMR studies should consist of several
loops, each one corresponding to the different nuclei to be inves-
tigated. Moreover, the RF coil should be designed in a way that the
respective RF fields of the different loops are electrically isolated
[18]. A more detailed description of the characteristics of the RF
coils used in direct and indirect non-proton MRS is given in the
materials section.
2 Materials
13
3. C labeled substrates. Glucose is the most common substrate
that allows investigating both oxidative and glycolytic metabo-
lism; however, other substrates have been used to target specific
metabolic pathways. Notably, acetate can be used to measure
specifically astrocytic oxidative metabolism [21], whereas lac-
tate can be used for investigating neuronal oxidative metabo-
lism [22]. Besides the differences in cost, the different 13C-
labeled glucose substrates can be used for different purpose. It
is thus primordial to choose the appropriate substrate accord-
ing to the technique (mainly direct and indirect 13C MRS) and
the need for an increased signal relative to cost. After glycolysis,
carbon positions 1 and 6 of glucose will label the methyl group
of pyruvate, leading primarily to the labeling of glutamate C4
following the pyruvate dehydrogenase pathway and glutamate
C2 for the pyruvate carboxylase pathway [23]. [1, 6-13C]
glucose is thus twice as efficient as [1-13C] glucose labeling,
and thus detecting, TCA cycle aminoacids. The use of [1,
6-13C] glucose is particularly important for 13C direct detec-
tion, in order to avoid splitting of peaks due to coupling
between adjacent 13C carbons. Because of this coupling,
[U-13C] glucose should best used in 1H-[13C] MRS, where
decoupling in the carbon channel allows to eliminate reso-
nances from heteronuclear coupling. [2-13C] glucose can be
used to investigate the anaplerotic metabolism of glial cells as it
will label specifically glutamine positions C2 and C3 due to the
presence of pyruvate carboxylase [24].
4. Infusion pump. Infusion can be done with a pump suitable for
infusion from any syringe and operating in both infusion and
withdrawal mode. The syringe containing the labeled glucose
solution is connected to a polyethylene line that has to be long
enough to cover the distance between the catheter and the
pump, i.e., the distance between the animal in the magnet
and the operator in the monitoring room. Adding a transparent
tube connector at the end of the line can help for controlling
the adjunction of the 99% enriched 13C glucose bolus solution.
5. Physiology monitoring. Temperature of the animal can be con-
trolled with a water circulating system covering the animal and
controllable in the preparation room. Respiration and temper-
ature can be monitored with a module located near the animal
in the magnet bore and a control/gating module connected to
a computer located near the operator console module. Glucose
can be measured with a single drop of blood using any com-
mercial system. Small volume sampling is critical for small
animals with small blood volumes such as mice or for repeated
measurements.
176 Blanca Lizarbe et al.
3 Methods
9:9∗ 100
)X ¼ ¼ 70:71 ½mL ð2Þ
20∗ 0:7
3.2 Animal Setup Before putting the animal inside the scanner, the following steps
should be followed:
1. Checking general status of the animal. Before starting an exper-
iment, the health of the animal needs to be checked and his
status reported. For 13C glucose infusion experiments, the
animal’s body weight and blood glucose levels should be
measured (see Note 1). Importantly, these values will be
178 Blanca Lizarbe et al.
Fig. 3 Animal immobilization in holders and supporting instrumentation. Left: The mouse head is fixed through
ear bars (in white) which are secured by small screwdrivers (in red). A small piece of acrylic glass covers the
animal’s mouth to avoid loss of anesthesia and prevent potential movements. An extra bar connects the
anesthetic tubing with the animal’s mouth. On the edge of this bar a small hole (in red) is used to place the
animal’s teeth. Blue water tubing is placed on top of the animal to maintain body temperature. The
thermometer cable can be seen coming out from the mouse body. The blue tubing on the right lateral of
the holder belongs to the respiration sensor, which is placed underneath of the animal’s belly. Right: The rat
head is fixed also through ear bars. A surface coil (double 1H in quadrature and single 31P) is place centered in
the animals’ head
3.3 MRS: Preparation Once the animal is in the magnet, the MRS preparation before
and Acquisition acquisition consists in: (1) positioning the holder in such a way
that the area to be investigated is in the center of the magnet;
(2) defining the voxel of interest (VOI), which normally involves
acquisition of high resolution anatomical images, and (3) adjusting
the first and second order gradient shims in the voxel [26], a
process that leads to an optimized resolution of the 1H spectra.
Note that for defining the VOI with a good anatomical reference it
might be convenient to perform T2-weighted acquisitions. Once
the VOI localization and shimming processes are performed, the
type of the study and the particular conditions of the experiment
will determine the type of sequences employed and the calibrations
performed. A description of the potential techniques to be used and
their respective adjustments is provided in the next subsections.
3.3.1 Localization To perform single voxel MRS, approaches that limit the excitation
Techniques in specific areas are used. Usually, localization is performed on the
1
H magnetization for both direct and indirect detection methods,
but localization on the 13C is frequently possible [18]. Generally,
the specific conditions of the study will determine which localiza-
tion method is more suitable. Single voxel MRS can be accom-
plished by intersecting three different RF pulses, like in Point
Resolved Spectroscopy (PRESS) [27] and STimulated Echo Acqui-
sition (STEAM) [28] methodologies. Both techniques are based on
the spin echo (SE) sequence, with PRESS using a 90 –180 –180
scheme and STEAM employing three 90 pulses. In PRESS, the
90 pulse shifts the magnetization into the transverse plane, the
180 refocuses it only from one column and the last pulse refocuses
magnetization in the third dimension. Since the second pulse in
STEAM flips only half of the magnetization along z, only half of the
In Vivo Heteronuclear MRS 181
3.3.2 Suppression Suppression of the water signal can be achieved using CHEmical
of the Water Signal Shift Selective pulses (CHESS) [32]. Values have to be calibrated
during each experimental session to obtain a minimum contribu-
tion of the water signal to the spectra. This can be achieved by using
a sequence with variable-power RF pulses with optimized relaxa-
tion delays, like (VAPOR) [33], and selecting the power that mini-
mizes the longitudinal magnetization of water.
3.3.3 Calibrations in 13C In general, due to the low sensitivity of the 13C signal, calibrations
Channel cannot be done in vivo and should be performed previously in vitro
in a phantom containing a known concentration of 13C labeled
molecules. Interestingly, using external bodies during calibration
and during the in vivo studies, such as formic acid spheres, is very
convenient (see Subheading 2).
182 Blanca Lizarbe et al.
3.3.4 Decoupling Heteronuclear coupling between the X-nucleus and the 1H is rela-
Schemes tively large, and decoupling schemes [18] need to be implemented
to avoid splitting of the signal in several lines. 1H (or 13C) decou-
pling is usually done by applying a RF field (B2) at the Larmor
frequency of 1H (or 13C) [13], which can result in increased RF
power deposition and cause excessive heating in the tissue. In this
sense, it is useful to optimize decoupling power to the minimum.
The value should correspond to the minimum power that elimi-
nates most if not all effects of heteronuclear coupling signal.
3.3.5 13
C Editing Block For 1H-[13C] methods, Adiabatic Full Passage (AFP) pulses in the
13
C channel should be also calibrated previously in a phantom
containing a known concentration of 13C labeled molecules. Opti-
mized power should invert the total 1H-[13C] signal. Figure 2b
shows an example of a non-inverted, not-decoupled spectra of
labeled acetate (top), and inverted not-decoupled spectra of labeled
acetate (middle) after the application of the AFP pulse in the 13C
channel. The use of an external reference allows detecting potential
variations on the inversion performance of AFP pulses (see Sub-
heading 2).
3.4 Tracer Infusion Infusing a tracer inevitably increases the content of the substance in
particular. Because of the non-toxicity of the tracers used and the
intrinsic low sensitivity of heteronuclear MRS, the label is normally
administered in high quantities, with enrichments ranging between
50 and 100% [34]. Typically, the targeted final IE at steady state is
around 70%, and the infusion process is divided in two parts:
(1) administration of a bolus of labeled substrate (99% enriched)
during a short period of time at an exponential rate and (2) infusion
at a constant rate of 70% enriched solution that keeps the IE at
steady state during the rest of the experiment. Several factors such
as the volume of the glucose bolus infused, rates and time constant
of its administration need to be calculated beforehand. The next
sections explain in detail these calculations.
3.4.1 Glucose Plasma The amount of 13C glucose in blood at the steady state of IE
Levels (Glcfinal) (Glcfinal) should be equal to the sum of 13C glucose in blood at
initial basal levels (Glcbasal), enriched with natural abundance of
1.1%, and the administration of the 99% enriched bolus. This
relationship is expressed in Eqs. (4) and (5).
1:1 99
Glcfinal ∗ IR ¼ Glcbasal ∗ þ ðGlcfinal Glcbasal Þ∗ ð4Þ
100 100
With IE ¼ 70%
1:1 99
Glcfinal ¼ Glcbasal ∗ ð5Þ
70 99
In Vivo Heteronuclear MRS 183
1:1 99 1 1
if ffi ) Glcfinal ¼ Glcbasal ð6Þ
70 99 ð1 70Þ ð1 IR Þ
With IE ¼ 70%
3.4.2 Glucose Bolus Taking into account that the percentage of extracellular fluid in
Volume rodents is around 20% of total body weight [35], and using Eq. 7 to
calculate the difference between basal and final glucose concentra-
tion, we can estimate that the total glucose bolus administrated in
g/kg of animal is 0.0047 * Glcbasal, as expressed in Eq. 8.
Total glc bolus½g=kg ¼ 0:2∗ ðGlcfinal Glcbasal Þ
¼ 0:0047∗ Glcbasal ð8Þ
3.4.3 Rate The glucose bolus is habitually administered during 5 min follow-
of Administration ing an exponential decay. To calculate the time constant of the
of the Bolus decay (k), we can use the system of equations expressed in
Eqs. 10 and 11.
V ðt ¼ 0Þ ¼ V 0 ¼ 0:45 Total glc bolus ð10Þ
3.4.4 Infusion Rate After the administration of the glucose bolus, 70% enriched [1,
6-13C] glucose is infused at a rate equivalent to the eGDR,
33.2 mg/kg/min. If we are using a glucose solution of 200 g/L,
this infusion rate can be also expressed as 9.96 mL/kg/h. Ideally,
to keep the IE at a steady state, this value might need to be adjusted
based depending on concomitantly measured plasma glucose con-
centrations; if plasma glucose levels decrease infusion rate should be
increased by the same percentage, and if plasma glucose levels
increase the speed of administration of the glucose solution should
be decreased. In rats, these concomitant measurements can be
easily done by analyzing the sample extracts of the femoral artery,
which are normally performed every 15–30 min. In mice, however,
blood extractions are not performed routinely and alternative ways
of warranting IE stability are needed. In this respect, there are
basically two ways of checking this constancy of IE. The first one
consists in taking out the holder from center of the magnet and
removing a small drop of blood from the tip of the tail, with the
counterpart of having to reposition the animal in the exact same
place afterwards. Finally, values can also be checked using the brain
glucose and/or lactate FE calculations from MRS measurements.
4 Notes
2. Tail vein vs. femoral vein. Tail vein cannulations are more fragile
than cannulations in femoral vein, but they are mandatory to
perform longitudinal studies with measurements at different
time points. The protocol for cauterization is similar than for
femoral vein, but its smaller size and localization makes it easier
for the needle to accidentally come out. This means that animal
manipulation has to be done with special care. Also, the smaller
size of the vein makes tail vein cannulation more prone to be
closed and blocked (partially or completely) the entrance of
infusion. To avoid this, two precautions should be taken: keep
the tail at warm temperatures while animal preparation and
check its circulation by infusing of small amounts of PBS during
animal preparation. In fact, once the animal is in the scanner it is
always convenient to start infusion only with PBS to keep the
line opened. Finally, it is important to take special care of the tail
at the end of each MRS session. Sometimes small injuries caused
by cannulations can end up with damages in the tail that prevent
its usage for the following time points.
3. Careful positioning of the surface coil. Being poor sensitivity one
of the main issues in heteronuclear MRS, the position of the
coil should be optimized to increase SNR in the area of interest.
With this aim, it is convenient to perform pilot studies to
determine which position enhances mostly the signal in this
area. Besides, it is recommended to implement periodically
additional tests in phantoms that make sure performance of
the coil is not affected.
References
1. Rothman DL, De Feyter HM, de Graaf RA, 5. Doddrell DM, Pegg DT, Bendall MR (1982)
Mason GF, Behar KL (2011) 13C MRS studies Distortionless enhancement of NMR signals by
of neuroenergetics and neurotransmitter polarization transfer. J Magn Reson 48
cycling in humans. NMR Biomed 24 (2):323–327. https://doi.org/10.1016/
(8):943–957. https://doi.org/10.1002/nbm. 0022-2364(82)90286-4
1772 6. Lagemaat MW, van de Bank BL, Sati P, Li SZ,
2. Zhu XH, Du F, Zhang N, Zhang Y, Lei H, Maas MC, Scheenen TWJ (2016) Repeatability
Zhang X, Qiao H, Ugurbil K, Chen W (2009) of P-31 MRSI in the human brain at 7T with
Advanced in vivo heteronuclear MRS and without the nuclear Overhauser effect.
approaches for studying brain bioenergetics NMR Biomed 29(3):256–263. https://doi.
driven by mitochondria. Methods Mol Biol org/10.1002/nbm.3455
489:317–357. https://doi.org/10.1007/ 7. Henry PG, Tkac I, Gruetter R (2003)
978-1-59745-543-5_15 1H-localized broadband 13C NMR spectros-
3. de Graaf RA (2007) Basic principles. In: In vivo copy of the rat brain in vivo at 9.4 T. Magn
NMR spectroscopy. Wiley, pp 1–42. doi: Reson Med 50(4):684–692. https://doi.org/
https://doi.org/10.1002/9780470512968. 10.1002/mrm.10601
ch1 8. Schroeder MA, Atherton HJ, Ball DR, Cole
4. de Graaf RA, Rothman DL, Behar KL (2011) MA, Heather LC, Griffin JL, Clarke K, Radda
State of the art direct 13C and indirect GK, Tyler DJ (2009) Real-time assessment of
1H-[13C] NMR spectroscopy in vivo. A prac- Krebs cycle metabolism using hyperpolarized
tical guide. NMR Biomed 24(8):958–972. 13C magnetic resonance spectroscopy. FASEB
https://doi.org/10.1002/nbm.1761 J 23(8):2529–2538. https://doi.org/10.
1096/fj.09-129171
186 Blanca Lizarbe et al.
new technique for spatially selective 34. Gruetter R (2002) In vivo 13C NMR studies
NMR-spectroscopy. J Magn Reson 66 of compartmentalized cerebral carbohydrate
(2):283–294. https://doi.org/10.1016/ metabolism. Neurochem Int 41(2-3):143–154
0022-2364(86)90031-4 35. Chapman ME, Hu L, Plato CF, Kohan DE
31. Mlynarik V, Gambarota G, Frenkel H, Gruetter (2010) Bioimpedance spectroscopy for the
R (2006) Localized short-echo-time proton estimation of body fluid volumes in mice. Am
MR spectroscopy with full signal-intensity J Physiol Ren Physiol 299(1):F280–F283.
acquisition. Magn Reson Med 56 https://doi.org/10.1152/ajprenal.00113.
(5):965–970. https://doi.org/10.1002/ 2010
mrm.21043 36. Jucker BM, Schaeffer TR, Haimbach RE,
32. Haase A, Frahm J, Hanicke W, Matthaei D McIntosh TS, Chun D, Mayer M, Ohlstein
(1985) 1H NMR chemical shift selective DH, Davis HM, Smith SA, Cobitz AR, Sarkar
(CHESS) imaging. Phys Med Biol 30 SK (2002) Normalization of skeletal muscle
(4):341–344 glycogen synthesis and glycolysis in
33. Tkac I, Gruetter R (2005) Methodology of H rosiglitazone-treated Zucker fatty rats: an
NMR spectroscopy of the human brain at very in vivo nuclear magnetic resonance study. Dia-
high magnetic fields. Appl Magn Reson 29 betes 51(7):2066–2073
(1):139–157. https://doi.org/10.1007/
BF03166960
Chapter 12
1
H Spectroscopic Imaging of the Rodent Brain
Rui V. Simões, Emma Muñoz-Moreno, Raúl Tudela, and Guadalupe Soria
Abstract
Proton MR spectroscopic imaging (MRSI) can provide a variety of “molecular images” from animal models
of human disease, which are useful for different research purposes. This chapter describes a protocol for
in vivo acquisition and analysis of MRSI data from the rodent brain.
Key words Magnetic Resonance Spectroscopic Imaging (MRSI), Chemical shift imaging (CSI), Field
of view (FOV), Volume of interest (VOI), Point resolved spectroscopy (PRESS), Acquisition-weight-
ing, Spectral analysis
1 Introduction
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0_12, © Springer Science+Business Media, LLC 2018
189
190 Rui V. Simões et al.
Fig. 1 MRSI of the neonatal rabbit brain. Data acquired at two days post-delivery in the transversal plane, with
Point Resolved Spectroscopy (PRESS) localization and acquisition-weighted k-space sampling (Hanning filter),
as detailed previously [17]. Top left, reference T2-w image (RARE), acquired over a 24 24 mm field of view
(FOV) and 1 mm slice thickness, showing the volume of interest for MRSI (purple box, 9 9 mm—enlarged on
the right, showing the final spectral matrix within) and one specific voxel position (black box, showed enlarged
at the bottom). Additional parameters for MRSI: matrix (initial size, MTXi ¼ 8 8, with 9 μL nominal voxel
size; Fourier interpolated to final size, MTXf ¼ 32 32, with 0.6 μL voxel display size); repetition time (TR),
2500 ms; echo time (TE), 14 ms; 512 accumulation (12 in the center of k-space); water-suppression (VAPOR
module, 300 MHz); 4006.4 Hz sweep-width and 2k points; 21.5 min total acquisition time. Major peak
assignments: alanine (Ala), creatine and phosphocreatine (CrT pool), glutamine (Gln) and glutamate (Glu; Glx
pool), glycerophosphocholine and phosphocholine (ChoT pool), glycine (Gly); glutathione (GSH), lactate (Lac),
myo-inositol (Ino), N-acetylaspartate (NAA) and N-acetylaspartylglutamate (NAAT pool), phosphoetanolamine
(PE); taurine (Tau), and macromolecules (MM). Experiment performed on a Bruker BioSpec 7T running with
Paravision 5.1 and a single-channel quadrature RF coil for the rat brain. Matrix spectra processed and
displayed with Paravision 5.1 softwate (CSIvisualizer, CSIdash, and TopSpin). Enlarged single spectrum
displayed with jMRUI 5.2 software, after zero order phase adjustment and line-broadening with a 4 Hz
apodization filter
2 Materials
2.3 Animals 1. Rodent (we have tested this protocol with rats between
250 and 400 g; but is also valid for mice as long as the FOV
and VOI sizes are suitably adjusted).
2. Vaporizer and induction chamber, to anesthetize the animals
outside the MR spectrometer (isoflurane in O2/N2O gas
mixture).
3. Cradle to position the animal inside the MR spectrometer,
which includes: restraining system (stereotactic fixing with
three fixation points, for the ear cavities and teeth); mainte-
nance of anesthesia (isoflurane gas, delivered from vaporizer);
heating system (recirculating water blanket); sensors for respi-
ration and temperature monitoring (chest/abdominal pillow-
probe and rectal probe, respectively).
4. Monitoring unit to control animal physiology (control/gating
module, Small Animal Instruments Inc. Stony Brook, NY,
USA).
3 Methods
3.1 Animal Handling 1. Turn on the gas flow (30% O2, 70% N2O, 800 mL/min) and
anesthesia (3–4% isoflurane) in the induction chamber and put
the rat inside.
2. Once the animal is asleep (1–2 min), quickly move it to the MR
cradle, where anesthetic gas is already being delivered (30% O2,
70% N2O, 800 mL/min, 1–2.5% isoflurane) and the heating
system running (recirculating heated water system); position
the animal head, using the head fixation system (see Note 1).
3. Position the sensors for breathing and rectal temperature and
check the animal monitoring unit. The anesthesia and heating
system should be adjusted to control the breathing and rectal
temperature throughout the experiment, ideally between
60–80 BPM and 36.5–37.5 C, respectively.
4. At the end of the experiment, the animal should be removed
from the cradle and left to recover in a warm environment until
full recovery (2–3 min), and then returned to its cage.
3.2 Initial Reference 1. Once the animal is ready, put the 4-channel RF surface coil over
Images the head and position the cradle inside the magnet bore.
2. Syntonize the probe (Wobble) and acquire the scout localizers
in the three orthogonal planes, adjusting the animal position if
Spectroscopic Imaging 193
Fig. 2 Preparation for global shim adjustment. Automatic Map Shim adjustment is performed within an
ellipsoid adjusted to fit the brain volume, based on T2-w reference images: left, coronal view; center, sagittal
view; right, transversal view
Fig. 3 Reference T2-w image for MRSI. Left side, high-resolution reference image (HRref), with matrix size
256 256. Right side, a low-resolution image, with the same matrix size as MTXf. The inner green square
represents the VOI, and the outer square indicates the region for the local homogeneity adjustments
Fig. 4 B0 map after global and local shim adjustments. B0 map acquired in a
transversal plane, after local Map Shim adjustments. The larger square
represents the FOV for MRSI, and the inner ones the region for local
homogeneity adjustment and the VOI (smallest). The image shows that the B0
field is relatively homogeneous throughout the VOI region
3.5 Analysis 1. Once the MRSI acquisition is finished, spectra should be visu-
of MRSI Data ally inspected for quality assessment across the VOI (using
ParaVision CSIvisualizer). Thus, the spectral patterns should
be interpretable and without gross artifacts (extensively
reported in [18], e.g., outer-volume lipid contaminations due
VOI proximity to the scalp and/or suboptimal OVS adjust-
ment), and resolution (related shimming performance), SNR
(related to the number of averages), and water suppression
(related to the VAPOR pulse calibration) acceptable; if not,
the respective parameters should be revised (as indicated) and
the acquisition repeated.
2. For MRSI post-processing, third-party software is required.
Importantly, the individual raw data recorded from each chan-
nel are stored in the file “rawdata.job0,” whereas the “fid” file
is generated after reconstruction to the MTXf size defined, and
already combines the 4 channel data; if the data are analyzed
with Bruker TopSpin software, the “fid” file generated is
renamed to “ser,” keeping the same structure and size.
3. Before processing the MRSI data with available software tools,
such as jMRUI or LC Model, first make sure that the recon-
struction pipeline in ParaVision 6.0.1 and the output “acqp”
file generated are properly set to remove the voxel-to-voxel
180 flip filter and display the final matrix size (MTXf),
respectively (see Notes 4 and 5).
4. For spectral inspection and analysis of MRSI data, jMRUI 5.2 is
a suitable option; after loading the “fid” file, the voxel filling
order should be customized, as instructed in the manual
(matrix transposition required: e.g., for 32 32 MTXf size,
“0-0 1-32 2-64 (. . .) 1021-959 1022-991 1023-1023”);
finally, adjust zero order phase correction and apodization for
correct spectral display (effect of changing the MTXi size on
spectral resolution shown in Fig. 5) (see Note 5).
Fig. 5 MRSI spectra display and resolution. Two consecutive MRSI experiments (1 and 2) were acquired with
1500 ms TR, 12 ms TE, k-space sampled with a Hamming filter (10 averages in the center), 28.8 28.8 mm
FOV, 1.8 mm slice thickness, 10.8 10.8 mm VOI, and interpolated to a 32 32 MTXf size (1.5 μL display
voxel size): MRSI-1 was acquired with 16 16 MTXi size (5.8 μL nominal voxel size), 809 accumulations and
20 min acquisition time; whereas for MRSI-2 a 8 8 MTXi size was used (23.3 μL nominal voxel size), with
213 accumulations and 5 min acquisition time. (A) MRSI-2 data display with jMRUI 5.2: left-side, “mosaic
view” of the spectra selected within the VOI, after zero order phase adjustment and line-broadening with a
4 Hz apodization filter; right-side, respective HRref image showing the VOI region (12 12 matrix, in blue).
Spectra from the same voxel position (dark square) are shown enlarged for each MRSI experiment: (B) MRSI-
1; (C) MRSI-2. In smaller voxels (nominal size), SNR decreases but spectral resolution increases, as the tissue
volume becomes more homogeneous. Major peak assignments as indicated in Fig. 1, plus aspartate (Asp) and
gamma-aminobutyrate (GABA)
198 Rui V. Simões et al.
4 Notes
Fig. 7 Reference water-line MRSI. Spectral matrix corresponding to the PRESS-VOI region shown in Fig. 6,
indicating a homogeneous spatial distribution of the water peak in each voxel (water-suppression module off).
Data displayed with the ParaVision module CSI visualization tool
Acknowledgments
References
(2001) Java-based graphical user interface for classification of brain tumors in mice with per-
the MRUI quantitation package. MAGMA 12 turbation enhanced (PE)-MRSI. Integr Biol
(2–3):141–152 (Camb) 4(2):183–191
14. Pfeuffer J, Tkac I, Provencher SW, Gruetter R 17. Simões RV, Candiota AP, Julià-Sapé M, Arús C
(1999) Toward an in vivo neurochemical pro- (2013) In vivo magnetic resonance spectros-
file: quantification of 18 metabolites in short- copy imaging (MRSI) and ex vivo quantitative
echo-time (1)H NMR spectra of the rat brain. J neuropathology by high resolution magic angle
Magn Reson 141(1):104–120 spinning proton magnetic resonance spectros-
15. Ortega-Martorell S, Olier I, Juliá-Sapè M, Arús copy (HR-MAS). In: Springer CS Jr
C (2010) SpectraClassifier 1.0: a user friendly, (ed) Animal models of brain tumors, Neuro-
automated MRS-based classifier-development methods volume series, vol 77. Springer,
system. BMC Bioinformatics 11:106 New York, pp 329–365
16. Simões RV, Ortega-Martorell S, Delgado- 18. Kreis R (2004) Issues of spectral quality in
Goni T, Le Fur Y, Pumarola M, Candiota AP, clinical 1H-magnetic resonance spectroscopy
Martin J, Stoyanova R, Cozzone PJ, Julia- and a gallery of artifacts. NMR Biomed 17
Sape M, Arus C (2012) Improving the (6):361–381
Part IV
Abstract
Susceptibility Weighted Imaging (SWI) is an established part of the clinical neuroimaging toolbox and,
since its inception, has also successfully been used in various preclinical studies. Exploiting the effect of
variations of magnetic susceptibility between different tissues on the externally applied, static, homoge-
neous magnetic field, the method visualizes venous vasculature, hemorrhages and blood degradation
products, calcifications, and tissue iron deposits. The chapter describes in vivo and ex vivo protocols for
preclinical SWI in rodents.
Key words SWI, Preclinical, MRI, Phase imaging, Magnetic susceptibility, Rodents, Mice, Rat,
Gradient echo, Bold
1 Introduction
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0_13, © Springer Science+Business Media, LLC 2018
205
206 Ferdinand Schweser et al.
2 Materials
2.2 In Vivo 1. Animals: Rats or mice without any metal parts in or attached to
Experiments their bodies, such as metal ear tags (see Note 7). We have been
using primarily adult female Swiss Jim Lambert (SJL/J) mice
for Multiple Sclerosis (MS) research and adult male Wistar rats
for Traumatic Brain Injury (TBI) research, but SWI should
work in any animal that allows a stable anesthesia.
2. Radio-frequency (RF) coil configuration for rat experiments
(see Note 10): Cross-coil configuration with an anatomically
shaped four-element receive-only1H RF array coil with 2 2
coil element topology (see Note 8) and a circularly polarized
volume RF coil with 86 mm inner diameter for transmission.
208 Ferdinand Schweser et al.
Fig. 1 Illustration of in vivo SWI at 9.4 T in the lateral fluid percussion rat model of TBI. Magnitude images of
the 3D multi-echo sequence were averaged, bias-field corrected to mitigate signal inhomogeneities due to the
use of a surface receiver coil. Finally, minimum intensity projection (mIP) images were calculated over 1 mm
in coronal (top left), sagittal (top right), and horizontal (bottom) slices. The filtered phase image shows
magnetic field perturbations due to susceptibility variations in the brain
Fig. 2 Illustration of ex vivo SWI in the lateral fluid percussion rat model of TBI. Shown are mIPs of bias-field
corrected magnitude images over 50 μm. White arrows with straight tail point at TBI-related hemorrhages
(hypointense), arrows with circle-tails point at neurons in the caudate, and arrows with box-tail point at veins
3 Methods
3.1 Animal Perfusion 1. Perfuse animal at 5 ml/min with 1.5 ml per gram body weight
for the Ex Vivo of 10 mM Gadobutrol (10 μl of Gadobutrol for every 1 ml
Experiment solution) phosphate-buffered saline (PBS) followed by 1.5 ml
per gram of 10 mM Gadobutrol NBF.
2. Dissect brain.
3. Post-fix brain for 24 h in NBF with 10 mM Gadobutrol. Keep
it at 4 C.
4. After 24 h, transfer brains to PBS with 10 mM Gadobutrol.
Store brain at 4 C.
5. Transfer sample from the cold room to MRI control room in
good time before the experiment to allow warming up to room
temperature.
6. Right before the MRI experiment, transfer the brain to the
Falcon tube (see Notes 21 and 22).
7. Fill the Falcon tube with Galden and screw on the cap (see Note
23).
8. Transfer the tube into the magnet room.
9. Gently tap the tube onto the bench several times to remove/
release air bubbles from the brain’s surface and from within the
foam. Place the tube on the animal sled with its conical tip in
the nose cone. Mechanically fix the tube with the mounting
putty. Use enough putty so that the end of the tube with the
cap is slightly higher than the end of the conical tip, trapping air
bubbles in the cap.
3.2 Pre-scan 1. Connect the additional animal heating blanket to the water
Preparations for In tubing.
Vivo Experiments 2. Activate the water heating mechanism of the MRI animal cra-
dle. Set the target temperature to 45 C (see Note 24).
3. Switch on the electric heating pad under the anesthesia
chamber.
4. For mice: Prepare surgical tape to align the two SpO2 sensor
pieces (without the clamp) such as to allow for connection to
the base of the mouse tail (see Note 25).
212 Ferdinand Schweser et al.
3.3 Induction 1. Transport the animal in its cage from the animal holding room
of Anesthesia to the MRI control room and place it under the fume hood
and Positioning for In until ready for the induction. Prevent the animal from becom-
Vivo Experiment ing stressed by bright light, loud noises, strong smells, etc.
Provide drinking water to the animal.
2. Using gloves, transfer the animal to the induction chamber. Set
oxygen flow to approximately 1.0 l/min and isoflurane con-
centration to 1%. After 1 min, increase isoflurane to 4–4.5%.
3. Monitor the animal closely; gently shake induction chamber
regularly and inspect response of animal. Once the animal
reaches a depth of anesthesia that allows transporting into the
magnet room (typically indicated by ~60 breaths per minute in
mice and ~50 breaths per minute in rats) (see Note 28), which
usually happens in less than 10 min.
4. Once anesthesia is deep enough, shut off the oxygen flow and
disconnect the vaporizer from the anesthesia chamber. Con-
nect the vaporizer with the tubing running into the magnet
room and set oxygen flow to 1.2 l/min and isoflurane to
2.5–3%.
5. Using gloves, open the anesthesia chamber and quickly trans-
port the animal into the magnet room. Cover the animal with
both hands minimizing heat loss. Inside the magnet room,
SWI 213
place the animal on the cradle prone and gently push the
animal’s nose into the nose cone. Monitor physiological signs
to ensure that animal stays under stable anesthesia. If anesthesia
became lighter during the transport, wait until animal is stable
again (see Note 29).
6. Place the respiration pad under the animal’s mid-abdominal
area (rat) or the animal’s upper abdominal area (mice), or atop
of the animal and tape it onto the cradle to avoid movement
during the exam.
7. Gently pull animal into nose cone using the tooth bar. Hold the
tooth bar with one hand while with the other hand gently push
the animal’s head down into the custom-made head support.
Pull animal’s tail gently to make sure the teeth are still hooked
up in the tooth bar. Verify the head is horizontal by looking at
the position of the eyes relative to the animal cradle. Hold the
tooth bar in that position with one hand and tighten the screw
with the other hand.
8. Continue with the lateral fixation of the head. For rats, push
the side ear bars gently into the rat’s ears. For mice, press the
side support squeeze gently (see Note 30) against the head
from the sides and tighten the screws into place. Avoid rotation
of the head and ensure that the animal’s head is centered in the
left-right direction (see Notes 31 and 32).
9. Place a generous amount of eye lubricant onto eyes to avoid
drying out.
10. Gently, stretch animal’s feet and tape them to the bottom of
the sled. This stretching will ensure that the animal will not
turn/twist if, by chance, wakes up during scanning.
11. Look at the animal from the side (horizontally). Your fixation is
right if you do not see any respiration-related motion of the
hair at the top of the head.
12. For mice: Position the custom-made soft tape cushion over the
top of animal’s head. Secure it with tape.
13. For rats: Place the surface coil atop of the animal’s head. Watch
for the ears not to get caught on the sides. The center of the
coil should coincide with the center of the brain. Move the
nose cone forward or backward if needed. Tape the coil in
place, pushing down slightly.
14. Insert the temperature probe into the lubricated sleeve and
very gently slide it into the rectum using circular motions,
deep enough to ensure correct reading throughout the experi-
ment (see Note 33). Tape the rectal probe wire to the sled (not
to the animal’s tail!). Stretch gently the animal’s tail and tape to
the sled as well.
214 Ferdinand Schweser et al.
15. Install the SpO2 sensor. For mice, slide animal’s tail into the
opening of the prepared pulse oxygenation sensor setup. Make
sure sensors are as far up at the base of the tail as possible to
ensure strong reading throughout the experiment. Tape the
cables in place onto the sled’s bottom and sides. For rats, put
the rat-clap on the hind foot of the rat. Ensure proper reading
of the sensor and reposition sensors if needed. Make sure that
the foot is held in the clamp tight enough to ensure strong
reading throughout the experiment. To stabilize it even more,
tape the clamp in place to the sled’s side. Watch the monitor for
a while to make sure the signal is strong and stable.
16. If the respiration pad is placed atop of the animal, position it
now. Tape it central to animal’s back above the lung area and
the cradle, snug but not tight. Verify that the respiration wave
is stable.
17. Cover the animal’s back with the heating blanket. Tape it in
place around the animal, gentle yet snug.
18. Watch the vitals to see how stable and strong they are.
19. Mice and postmortem: Slide the animal cradle into the cryo-
probe simulator to check and resolve mechanical obstructions,
if necessary. Lift the cradle against the coil head and closely
watch respiration rate. The respiration rate should remain the
same—if it changes, reposition!
20. Slide the sled into the magnet.
21. Adjust the concentration of Isoflurane to achieve a stable res-
piration rate ideally ~70 breaths/min.
22. Record the initial body temperature reading and adjust water
bath temperature throughout the scanning to maintain
it. General body temperature range is 36.5–37.5 C, and varies
from animal to animal.
3.4 MRI Scan 1. Acquire a standard three-plane scout with default, automated
system adjustments. Confirm that the brain is positioned at the
magnet isocenter. If this is not the case, modify the location of
the animal sled. When using the cryogenic coil, also inspect the
signal inhomogeneity profile and confirm that the coil is axially
centered at the magnet’s isocenter. Adjust location of animal
bed and coils and run scout again until positions are acceptable.
In addition, for ex vivo, ensure that the specimen is rotated
such that a homogeneous intensity profile will be obtained in at
least one slice orientation.
2. In vivo experiment: Run a quick sagittal, single-slice 2D MGE
scan to check if respiration-related movements risk to deterio-
rate the SWI images (see Note 34). For mice: TE1 ¼ 3.99 ms,
ΔTE ¼ 8 ms, 5 echoes with positive readout (flyback),
SWI 215
Fig. 3 Illustration of images obtained from the 2D motion-sensing pulse sequence in a mouse. The top row
shows typical images obtained when the positioning is insufficient for SWI (increasing TE from left to right).
The white arrows point at motion-related artifacts. After improving the fixation, no artifacts are discernible on
the images of the motion-sensing sequence (bottom row)
3.6 Recovery 1. Carefully remove all tape that was used to position the blood
and Preparation oxygenation sensor, the rectal probe, feet to the cradle, head
for Next Scan soft tape cushion, and so forth.
2. Release the head from the restraint devices.
3. Turn off anesthesia.
4. Remove animal from nose cone and recover in a well-ventilated
induction box (lid open) that was kept warm with a heating pad
from below. Optionally, use infrared heating lamp from top but
ensure that animal does not overheat. Monitor recovery. Pedal
reflex should be noticeable after 1 min. Pinch animal toe or the
very tip of their tail. An immediate jerk reaction will take place
if the animal has pain reflexes.
5. Disinfect (using MB-10) all components wearing gloves, eye
protection, and lab coat.
218 Ferdinand Schweser et al.
3.7 Post-processing 1. Load the MRI data with MATLAB (see Note 20).
and Data Analysis 2. Sinc-interpolate the spatial resolution of the images by a factor
of 1.5 or higher (see Note 55).
3. Reconstruct magnitude images from the raw multichannel
k-space data with the sum-of-squares technique [48] (see
Note 56).
4. Perform a bias field correction (e.g., using N4-ITK [49]) to
reduce the signal inhomogeneity of the surface coil (see Note
57).
5. Calculate a sliding minimum intensity projection (mIP) of the
magnitude image over at least 1 mm of consecutive slices (see
Notes 57 and 58).
6. Combine the multichannel phase images and remove large-
scale field inhomogeneities (background fields) with a high-
pass filter (see Notes 60 and 62).
7. Combine the phase from different echo times into one single
phase image that may be interpreted (see Note 60).
8. Export the resulting images to your preferred data format, e.g.,
NIfTI (see Note 59).
4 Notes
Fig. 4 Illustration of the setup for mice. Panel (a) shows the 3D models of the custom side fixation bars. Panel
(b) illustrates the setup with SpO2 sensor (straight tail black arrow), respiration sensor (white arrow), and
temperature probe (circle-ended arrow). Panel (c) shows the same setup with a mouse. The arrows mark the
custom 3D printed fixation bars shown in panel (a). Panel (d) illustrates the vertical fixation using a soft tape
cushion
cradle near the nose cone and can be considered the fourth
fixation point. The animal’s head is cushioned on the sides and
on the bottom by this foam providing support below animal’s
neck without obstructing its respiration. The sides of this
U-shaped foam add even more protection when the lateral
fixation parts squeeze the animal’s head. The vertical fixation
is particularly important for smaller mice for which the stan-
dard animal sled does not provide sufficient vertical support.
12. The BOLD signal depends on cerebral metabolic oxygen con-
sumption, blood flow, and oxygenation level of the arterial
blood. Hence, the inhaled gas mixture and, in particular, the
anesthetic agent has a considerable impact on the vessel con-
trast in preclinical SWI [31]. Shen et al. [60] recommended
intraperitoneal injection of a mixture of ketamine, xylazine,
and atropine. Inhalation anesthesia allows for faster induction
and quicker recovery than injectable agents, and the dose is
easily regulated. However, Isoflurane increases the cerebral
blood flow (CBF) resulting in a reduced deoxygenation level
of venous blood and, hence, reduced SWI vein contrast. A
reduced visibility of veins is disadvantageous for venography,
but may be considered as beneficial for the detection of hemor-
rhages, iron storage, and calcium. Halothane reduces the oxy-
gen consumption without increased blood flow, but has poorer
analgesia and muscle relaxation qualities and is associated with
toxic metabolic products. The use of agents for preclinical
222 Ferdinand Schweser et al.
Fig. 5 Illustration of the ex vivo setup. The specimen is fixed within a PFPE-filled tube using soft foam. The
sagittal and horizontal perspectives illustrate the orientation of the brain in the tube. A similar set up was used
to acquire the images in Fig. 2
27. Ensure that the slices of the scout are prescribed without spatial
offsets, i.e., they are centered at the isocenter. With this slice
location, it is simple to confirm that the brain of the animal is
centered at the isocenter of the magnet.
28. A sufficient depth of anesthesia can be tested by placing a slight
pressure on the animal’s hind limb. The pedal withdrawal reflex
is absent in deeply anesthetized animals. However, never let the
animal get too deep because this can result in instability later
during the anesthesia!
29. Some strains/models (in particular smaller animals) cool off or
recover from anesthesia very quickly. When working with such
animals, the anesthesia chamber may be transported into the
magnet room and placed right next to the heated animal cradle
before opening it and transferring the animal onto the cradle.
However, this procedure exposes the operator to a higher level
of isoflurane compared to opening the chamber under the
fume hood. Furthermore, the anesthesia chamber must not
contain any ferromagnetic parts.
30. If the eyes popped up you squeezed too much!!
31. You may need to move the nose cone forward or backward to
match the ear bars with the ears’ location.
32. The fixation must not cause pain to the animal, because this
could wake the animal up or render the anesthesia unstable.
Avoid hard, strong, body-spasm-type, gasping respirations,
which indicates respiration difficulty. The respiration should
appear shallow enough that the whole body does not move
with each breath (only the abdominal area should pulse with
each breath). If you see these gasps, you’ve probably packed
the animal in too tight or isoflurane level is too high. Loosen
up the animal and/or decrease the isoflurane concentration in
small increments. Wait for the respiration to become smoother
yet stable and uniform. Do not try to save time during the
positioning. This is one of the most important steps of the
experiment!
33. Do not push if you feel it does not slide in. Rather not use the
probe instead of rupturing the rectum.
34. Motion is the most critical factor for image quality in preclini-
cal SWI, in particular when 3D sequences are employed. Later
echoes are particularly motion sensitive, which is why we rec-
ommend the use of a motion-sensing multi-echo scan with
long echo times. Unstable animals with grasping respiration
show particularly high level of artifacts that can render images
at later echo times unusable. Neither respiration nor cardiac
triggering significantly reduces the motion-related artifacts in
SWI at later echo times. The artifacts are probably related to
changes in the susceptibility-related magnetic field (phase) due
SWI 225
Fig. 6 Illustration of the temporal phase evolution (top row, from left to right), associated artifacts, and the
benefit of a magnitude-weighted combination (bottom row; from left to right more echoes used). The middle
row shows magnitude images corresponding to the phase images. The circle-ended arrow marks the field
perturbation due to a hemorrhage. The straight-ended arrows indicate phase artifacts. In the top row, the
arrow points at an intrinsic wrapping artifact due to a high susceptibility difference. Such artifacts are difficult
to eliminate using filtering and propagate into the combined phase images (bottom row), requiring an
appropriate cut-off for the echo time. The straight-ended arrow in the bottom row points at magnetic field
inhomogeneities close to the surface of the brain. Due to their spatial localization, such inhomogeneities
cannot be mitigated by field shimming and propagate into typical magnitude-phase SWI composite images,
complicating the interpretation of signal changes in the cortex. The box-ended arrow points at the improved
gray-white matter contrast and reduced noise level in the magnitude-weighted phase average image
wrapping artifacts in the brain that result from the brain tissue
susceptibility variation itself. Since these fields are not slowly
varying, such wrapping artifacts cannot be suppressed easily
(Fig. 6).
41. For QSM, relatively isotropic voxels and whole brain coverage
are beneficial. Hence, our 3D-MGE standard SWI protocol
covers the entire brain with a nearly isotropic resolution. If
smaller veins need to be detected, a 2D protocol with partial
coverage should be utilized and measurement time should be
invested into higher in-plane resolution instead. Depending on
the goal of the study, the in-slice resolution should be isotropic
to ensure that vessel visibility is independent of the orientation
of vessels. The resolution in read direction may be increased
substantially without additional measurement time by a simul-
taneous increase of the readout bandwidth (however, at the
expense of SNR). Measurement time may be decreased by
reducing the resolution in phase-encode direction.
42. We use ParaVision 6.0.1 for ex vivo MR microscopy measure-
ments because we observed inter-slice intensity variations on
magnitude images when using large matrices in PV5.1.
SWI 227
low respiration rate is needed to keep the animal still, check the
animal’s temperature and improve fixation.
54. ParaVision 5.1 acquires the specified “matrix dimension” in
read direction. If 33% echo position is specified, this effectively
results in a shift of the k-space readout window and must not
be confused with the “Partial Fourier” feature on other plat-
forms, that reduces the effectively acquired matrix size and fills
the data matrix elements that were not acquired with zeros. A
set echo position below 50% needs to be accounted for when
working with the raw data, e.g., by placing the raw data matrix
in a larger array of zeros with a centered k-space origin in that
matrix. In addition, if isotropic nominal spatial resolution is
desired, the acquisition matrix size specified in ParaVision must
be decreased accordingly.
55. In MATLAB, use the command padarray to add zeros to the
first, second, and third dimension of the k-space data. Make
sure that the parity of the matrix dimensions remains the same,
i.e., increase the matrix size from 256 to 512, but not from
256 to 515. Do not add zeros to the echo- or coil-dimensions.
Apply an 3D Fast Fourier Transform to the k-space data
(data ¼ fftshift(fft(fft(fft(ifftshift(data),[],1),[],2),[],3)); in
matlab) to obtain the image space data.
56. Square the magnitude signal in every matrix element, sum over
all coil elements and take the square root of the result
(in MATLAB: sqrt(sum(abs(data).^2,5), if the coil elements
are stored in the 5th dimension of the data matrix). If a single-
channel receive coil is used (compared to a multichannel coil),
it is sufficient to directly calculate the absolute value (abs(data))
after the inverse Fourier transform to obtain the magnitude
images.
57. This processing step may also be performed on images recon-
structed by ParaVision. The image may be found in the sub-
folder pdata/.
58. For every pixel in a certain slice jS, determine the minimum
intensity value of the equivalent voxel in the adjacent N slices
(In MATLAB: dataMIP(:,:,jS) ¼ min(data(:,:,jS-N:jS+N),[],3).
The calculation of the mIP may be performed for all slices
using a for-loop over jS. However, make sure that the mIP is
calculated over less slices at the boundaries of the imaging slab.
59. For NIfTI export from MATLAB use the Matlab package by
Jimmy Shen (https://www.mathworks.com/matlabcentral/
fileexchange/8797-tools-for-nifti-and-analyze-image).
60. Several methods can be employed to combine multichannel
phase images [72] and remove background phase contribu-
tions [73]. A simple homodyne phase combination of the
multichannel phase images may be performed by creating a
230 Ferdinand Schweser et al.
Acknowledgements
References
16. Wang D, Zhu D, Wei XE, Li YH, Li WB (2013) autoimmune encephalomyelitis mouse model
Using susceptibility-weighted images to quan- of multiple sclerosis using susceptibility MRI
tify iron deposition differences in amnestic mild and hyperoxygenation. PLoS One 10(5):
cognitive impairment and Alzheimer’s disease. e0127033. https://doi.org/10.1371/journal.
Neurol India 61(1):26–34. https://doi.org/ pone.0127033
10.4103/0028-3886.107924 26. Shen Y, Kou Z, Kreipke CW, Petrov T, Hu J,
17. Xu X, Wang Q, Zhang M (2008) Age, gender, Haacke EM (2007) In vivo measurement of
and hemispheric differences in iron deposition tissue damage, oxygen saturation changes and
in the human brain: an in vivo MRI study. blood flow changes after experimental trau-
NeuroImage 40(1):35–42. https://doi.org/ matic brain injury in rats using susceptibility
10.1016/j.neuroimage.2007.11.017 weighted imaging. Magn Reson Imaging 25
18. Yu J, Qi F, Wang N, Gao P, Dai S, Lu Y, Su Q, (2):219–227. https://doi.org/10.1016/j.
Du Y, Che F (2014) Increased iron level in mri.2006.09.018
motor cortex of amyotrophic lateral sclerosis 27. Verma SK, Kan EM, Lu J, Ng KC, Ling EA,
patients: an in vivo MR study. Amyotroph Lat- Seramani S, Kn BP, Wong YC, Tan MH, Velan
eral Scler Frontotemporal Degener 15 SS (2015) Multi-echo susceptibility-weighted
(5–6):357–361. https://doi.org/10.3109/ imaging and histology of open-field blast-
21678421.2014.906618 induced traumatic brain injury in a rat model.
19. Ogawa S, Lee TM, Nayak AS, Glynn P (1990) NMR Biomed 28(9):1069–1077. https://doi.
Oxygenation-sensitive contrast in magnetic org/10.1002/nbm.3351
resonance image of rodent brain at high mag- 28. Yang SH, Lin J, Lu F, Dai YY, Han ZH, CX F,
netic fields. Magn Reson Med 14(1):68–78 FL H, HC G (2016) Contrast-enhanced sus-
20. Krishnan AS, Lansley JA, Jager HR, Mankad K ceptibility weighted imaging with ultrasmall
(2015) New vistas in clinical practice: superparamagnetic iron oxide improves the
susceptibility-weighted imaging. Quant Imag- detection of tumor vascularity in a hepatocellu-
ing Med Surg 5(3):448–452. https://doi.org/ lar carcinoma nude mouse model. J Magn
10.3978/j.issn.2223-4292.2015.03.03 Reson Imaging 44(2):288–295. https://doi.
21. Liu S, Buch S, Chen Y, Choi HS, Dai Y, org/10.1002/jmri.25167
Habib C, Hu J, Jung JY, Luo Y, Utriainen D, 29. Kim SG, Park SH (2011) High-resolution
Wang M, Wu D, Xia S, Haacke EM (2017) venographic BOLD MRI of animal brain at
Susceptibility-weighted imaging: current status 9.4 T: implications for BOLD fMRI. Suscepti-
and future directions. NMR Biomed 30(4): bility weighted imaging in MRI: basic concepts
e3552. https://doi.org/10.1002/nbm.3552 and clinical applications. Wiley-Blackwell,
22. Bir C, Vandevord P, Shen Y, Raza W, Haacke Hoboken, pp 637–647
EM (2012) Effects of variable blast pressures 30. Ogawa S, Lee TM (1990) Magnetic resonance
on blood flow and oxygen saturation in rat imaging of blood vessels at high fields: in vivo
brain as evidenced using MRI. Magn Reson and in vitro measurements and image simula-
Imaging 30(4):527–534. https://doi.org/10. tion. Magn Reson Med 16(1):9–18
1016/j.mri.2011.12.003 31. Ogawa S, Lee TM, Kay AR, Tank DW (1990)
23. Blasiak B, Landry J, Tyson R, Sharp J, Iqbal U, Brain magnetic resonance imaging with con-
Abulrob A, Rushforth D, Matyas J, Ponjevic D, trast dependent on blood oxygenation. Proc
Sutherland GR, Wolfsberger S, Tomanek B Natl Acad Sci U S A 87(24):9868–9872
(2014) Molecular susceptibility weighted 32. Park SH, Masamoto K, Hendrich K, Kanno I,
imaging of the glioma rim in a mouse model. Kim SG (2008) Imaging brain vasculature with
J Neurosci Methods 226:132–138. https:// BOLD microscopy: MR detection limits deter-
doi.org/10.1016/j.jneumeth.2014.01.034 mined by in vivo two-photon microscopy.
24. Nathoo N, Agrawal S, Wu Y, Haylock-Jacobs S, Magn Reson Med 59(4):855–865. https://
Yong VW, Foniok T, Barnes S, Obenaus A, doi.org/10.1002/mrm.21573
Dunn JF (2013) Susceptibility-weighted imag- 33. Harrison PM, Arosio P (1996) The ferritins:
ing in the experimental autoimmune encepha- molecular properties, iron storage function and
lomyelitis model of multiple sclerosis indicates cellular regulation. Biochim Biophys Acta
elevated deoxyhemoglobin, iron deposition 1275(3):161–203
and demyelination. Mult Scler 19 34. Buch S, Liu S, Ye Y, Cheng YC, Neelavalli J,
(6):721–731. https://doi.org/10.1177/ Haacke EM (2015) Susceptibility mapping of
1352458512460602 air, bone, and calcium in the head. Magn Reson
25. Nathoo N, Rogers JA, Yong VW, Dunn JF Med 73(6):2185–2194. https://doi.org/10.
(2015) Detecting deoxyhemoglobin in spinal 1002/mrm.25350
cord vasculature of the experimental
SWI 233
35. Chen W, Zhu W, Kovanlikaya I, Kovanlikaya A, (2013) Iron deposition in the precentral grey
Liu T, Wang S, Salustri C, Wang Y (2014) matter in patients with multiple sclerosis: a
Intracranial calcifications and hemorrhages: quantitative study using susceptibility-
characterization with quantitative susceptibility weighted imaging. Eur J Radiol 82(2):
mapping. Radiology 270(2):496–505. e95–e99. https://doi.org/10.1016/j.ejrad.
https://doi.org/10.1148/radiol.13122640 2012.09.006
36. Deistung A, Schweser F, Wiestler B, Abello M, 44. Wang D, Li YY, Luo JH, Li YH (2014)
Roethke M, Sahm F, Wick W, Nagel AM, Age-related iron deposition in the basal ganglia
Heiland S, Schlemmer HP, Bendszus M, of controls and Alzheimer disease patients
Reichenbach JR, Radbruch A (2013) Quanti- quantified using susceptibility weighted imag-
tative susceptibility mapping differentiates ing. Arch Gerontol Geriatr 59(2):439–449.
between blood depositions and calcifications https://doi.org/10.1016/j.archger.2014.04.
in patients with glioblastoma. PLoS One 8(3): 002
e57924. https://doi.org/10.1371/journal. 45. Hagemeier J, Weinstock-Guttman B,
pone.0057924 Heininen-Brown M, Poloni GU,
37. Schweser F, Deistung A, Lehr BW, Reichen- Bergsland N, Schirda C, Magnano CR,
bach JR (2010) Differentiation between dia- Kennedy C, Carl E, Dwyer MG, Minagar A,
magnetic and paramagnetic cerebral lesions Zivadinov R (2013) Gray matter SWI-filtered
based on magnetic susceptibility mapping. phase and atrophy are linked to disability in
Med Phys 37(10):5165–5178. https://doi. MS. Front Biosci (Elite Ed) 5:525–532
org/10.1118/1.3481505 46. Peters AM, Brookes MJ, Hoogenraad FG,
38. Straub S, Laun FB, Emmerich J, Jobke B, Gowland PA, Francis ST, Morris PG, Bowtell
Hauswald H, Katayama S, Herfarth K, Schlem- R (2007) T2* measurements in human brain at
mer HP, Ladd ME, Ziener CH, Bonekamp D, 1.5, 3 and 7 T. Magn Reson Imaging 25
Rothke MC (2017) Potential of quantitative (6):748–753. https://doi.org/10.1016/j.
susceptibility mapping for detection of pros- mri.2007.02.014
tatic calcifications. J Magn Reson Imaging 45 47. Bourekas EC, Christoforidis GA, Abduljalil
(3):889–898. https://doi.org/10.1002/jmri. AM, Kangarlu A, Chakeres DW, Spigos DG,
25385 Robitaille PM (1999) High resolution MRI of
39. Rauscher A, Sedlacik J, Deistung A, Mentzel the deep gray nuclei at 8 Tesla. J Comput Assist
HJ, Reichenbach JR (2006) Susceptibility Tomogr 23(6):867–874
weighted imaging: data acquisition, image 48. Roemer PB, Edelstein WA, Hayes CE, Souza
reconstruction and clinical applications. Z SP, Mueller OM (1990) The NMR phased
Med Phys 16(4):240–250 array. Magn Reson Med 16(2):192–225
40. Deistung A, Rauscher A, Sedlacik J, Stadler J, 49. Tustison NJ, Avants BB, Cook PA, Zheng Y,
Witoszynskyj S, Reichenbach JR (2008) Sus- Egan A, Yushkevich PA, Gee JC (2010)
ceptibility weighted imaging at ultra high mag- N4ITK: improved N3 bias correction. IEEE
netic field strengths: theoretical considerations Trans Med Imaging 29(6):1310–1320.
and experimental results. Magn Reson Med 60 https://doi.org/10.1109/TMI.2010.
(5):1155–1168. https://doi.org/10.1002/ 2046908
mrm.21754 50. Frank LR, Crawley AP, Buxton RB (1992)
41. Duyn JH, van Gelderen P, Li TQ, de Zwart JA, Elimination of oblique flow artifacts in mag-
Koretsky AP, Fukunaga M (2007) High-field netic resonance imaging. Magn Reson Med 25
MRI of brain cortical substructure based on (2):299–307
signal phase. Proc Natl Acad Sci U S A 104 51. Denk C, Rauscher A (2010) Susceptibility
(28):11796–11801. https://doi.org/10. weighted imaging with multiple echoes. J
1073/pnas.0610821104 Magn Reson Imaging 31(1):185–191.
42. Hagemeier J, Dwyer MG, Bergsland N, https://doi.org/10.1002/jmri.21995
Schweser F, Magnano CR, Heininen-Brown M, 52. Wu B, Li W, Avram AV, Gho SM, Liu C (2012)
Ramasamy DP, Carl E, Kennedy C, Melia R, Fast and tissue-optimized mapping of magnetic
Polak P, Deistung A, Geurts JJ, Reichenbach susceptibility and T2* with multi-echo and
JR, Zivadinov R (2013) Effect of age on MRI multi-shot spirals. NeuroImage 59
phase behavior in the subcortical deep gray (1):297–305. https://doi.org/10.1016/j.
matter of healthy individuals. AJNR Am J Neu- neuroimage.2011.07.019
roradiol 34(11):2144–2151. https://doi.org/
10.3174/ajnr.A3569 53. Duyn J (2013) MR susceptibility imaging. J
Magn Reson 229:198–207. https://doi.org/
43. Rumzan R, Wang JJ, Zeng C, Chen X, Li Y, 10.1016/j.jmr.2012.11.013
Luo T, Lv F, Wang ZP, Hou H, Huang F
234 Ferdinand Schweser et al.
54. Haacke EM, Liu S, Buch S, Zheng W, Wu D, 65. van Duijn S, Nabuurs RJ, van Rooden S, Maat-
Ye Y (2015) Quantitative susceptibility Schieman ML, van Duinen SG, van Buchem
mapping: current status and future directions. MA, van der Weerd L, Natte R (2011) MRI
Magn Reson Imaging 33(1):1–25. https:// artifacts in human brain tissue after prolonged
doi.org/10.1016/j.mri.2014.09.004 formalin storage. Magn Reson Med 65
55. Liu C, Li W, Tong KA, Yeom KW, Kuzminski S (6):1750–1758. https://doi.org/10.1002/
(2015) Susceptibility-weighted imaging and mrm.22758
quantitative susceptibility mapping in the 66. Hakkarainen H, Sierra A, Mangia S,
brain. J Magn Reson Imaging 42(1):23–41. Garwood M, Michaeli S, Grohn O, Liimatai-
https://doi.org/10.1002/jmri.24768 nen T (2016) MRI relaxation in the presence of
56. Liu C, Wei H, Gong NJ, Cronin M, Dibb R, fictitious fields correlates with myelin content
Decker K (2015) Quantitative susceptibility in normal rat brain. Magn Reson Med 75
mapping: contrast mechanisms and clinical (1):161–168. https://doi.org/10.1002/
applications. Tomography 1(1):3–17. 10. mrm.25590
18383/j.tom.2015.00136 67. Meijer FJ, Goraj B, Bloem BR, Esselink RA
57. Reichenbach JR, Schweser F, Serres B, Deis- (2017) How I do it: clinical application of
tung A (2015) Quantitative susceptibility brain MRI in the diagnostic work-up of Parkin-
mapping: concepts and applications. Clin Neu- sonism. J Parkinsons Dis 7:211. https://doi.
roradiol 25(Suppl 2):225–230. https://doi. org/10.3233/JPD-150733
org/10.1007/s00062-015-0432-9 68. Birkl C, Langkammer C, Krenn H, Goessler W,
58. Wang Y, Liu T (2015) Quantitative susceptibil- Ernst C, Haybaeck J, Stollberger R, Fazekas F,
ity mapping (QSM): decoding MRI data for a Ropele S (2015) Iron mapping using the tem-
tissue magnetic biomarker. Magn Reson Med perature dependency of the magnetic suscepti-
73(1):82–101. https://doi.org/10.1002/ bility. Magn Reson Med 73(3):1282–1288.
mrm.25358 https://doi.org/10.1002/mrm.25236
59. Xu B, Liu T, Spincemaille P, Prince M, Wang Y 69. Dal-Bianco A, Grabner G, Kronnerwetter C,
(2014) Flow compensated quantitative suscep- Weber M, Hoftberger R, Berger T, Auff E,
tibility mapping for venous oxygenation imag- Leutmezer F, Trattnig S, Lassmann H,
ing. Magn Reson Med 72(2):438–445. Bagnato F, Hametner S (2017) Slow expansion
https://doi.org/10.1002/mrm.24937 of multiple sclerosis iron rim lesions: pathology
60. Shen Y, Kou Z, Haacke EM (2011) Suscepti- and 7 T magnetic resonance imaging. Acta
bility weighted imaging in rodents. Susceptibil- Neuropathol 133(1):25–42. https://doi.org/
ity weighted imaging in MRI: basic concepts 10.1007/s00401-016-1636-z
and clinical applications. Wiley-Blackwell, 70. Schweser F, Deistung A, Reichenbach JR
Hoboken, pp 649–667 (2016) Foundations of MRI phase imaging
61. Kim S, Pickup S, Hsu O, Poptani H (2009) and processing for Quantitative Susceptibility
Enhanced delineation of white matter struc- Mapping (QSM). Z Med Phys 26(1):6–34.
tures of the fixed mouse brain using https://doi.org/10.1016/j.zemedi.2015.10.
Gd-DTPA in microscopic MRI. NMR Biomed 002
22(3):303–309. https://doi.org/10.1002/ 71. Gudbjartsson H, Patz S (1995) The Rician
nbm.1324 distribution of noisy MRI data. Magn Reson
62. Aggarwal M, Mori S, Shimogori T, Med 34(6):910–914
Blackshaw S, Zhang J (2010) Three- 72. Robinson SD, Bredies K, Khabipova D,
dimensional diffusion tensor microimaging Dymerska B, Marques JP, Schweser F (2017)
for anatomical characterization of the mouse An illustrated comparison of processing meth-
brain. Magn Reson Med 64(1):249–261. ods for MR phase imaging and QSM: combin-
https://doi.org/10.1002/mrm.22426 ing array coil signals and phase unwrapping.
63. Andrews TJ, Osborne MT, Does MD (2006) NMR Biomed 30(4):e3601. https://doi.org/
Diffusion of myelin water. Magn Reson Med 10.1002/nbm.3601
56(2):381–385. https://doi.org/10.1002/ 73. Schweser F, Robinson SD, de Rochefort L,
mrm.20945 Li W, Bredies K (2017) An illustrated compari-
64. Shmueli K, van Gelderen P, Li T, Duyn J son of processing methods for phase MRI and
(2008) High resolution human brain suscepti- QSM: removal of background field contribu-
bility maps calculated from 7 Tesla MRI phase tions from sources outside the region of inter-
data. 16th Annual Meeting of the International est. NMR Biomed 30(4):e3604. https://doi.
Society for Magnetic Resonance in Medicine, org/10.1002/nbm.3604
p 642
Chapter 14
Abstract
Background-free fluorine (19F) MR imaging exhibits an excellent degree of specificity, and facilitates among
others the in vivo visualization of inflammatory processes. Merging19F MR images with morphologically
matching1H MR images enables the exact anatomic localization of the observed19F signal. Biochemically
inert nanoemulsions of perfluorocarbons, which are known to be taken up by the macrophage/monocyte
system, are widely used as contrast agents for preclinical applications. Herein, the most common protocols
are described to obtain high-resolution and artifact-free19F MR images even for compounds with com-
plex19F MR spectra. In addition, we report on the utilization of perfluorocarbons with individual spectral
identities and targeting approaches to specifically visualize thrombi by19F MRI.
Key words Fluorine MRI, Perfluorocarbons, Sterol-based post-insertion, Active targeting, Chemical
shift imaging
1 Introduction
19
F MRI is a promising noninvasive imaging technique for transla-
tional approaches, as it exhibits a great degree of specificity due to
the lack of any background signal in the body. Merging of match-
ing1H and19F datasets enables the anatomic localization of the
observed19F MR signal as “hot spot” [1–3]. The19F signal intensity
is directly proportional to the number of accumulated19F nuclear
spins which enables the convenient quantification of19F MR data.
As fluorinated contrast agents with a high payload of19F nuclei,
biochemically inert perfluorocarbon nanoemulsions (PFCs) are
promising candidates [2, 4, 5]. In particular, perfluoro-15-crown-
5 ether (PFCE; Fig. 1a) with 20 magnetically equivalent19F nuclei
per molecule is one of the most widely used PFCs for preclinical
applications. However, since PFCE displays a long biological half-
life (>250 days) [6], it is not suitable for prospective clinical
Tuba G€
uden-Silber and Sebastian Temme contributed equally to this work.
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0_14, © Springer Science+Business Media, LLC 2018
235
236 €den-Silber et al.
Tuba Gu
Fig. 1 Scheme of PFC application for in vivo tracking of biomedical processes by19F MRI using passive and
active targeting. (a) Chemical structures of PFCE, PFOB, and F-44E. (b) High-pressure homogenization with
phospholipids yields stable perfluorocarbon nanoemulsions (PFCs). The fluorophil PFC core (gray sphere) is
shielded by a lipid monolayer with the polar phosphate group (pink spheres) directed to the hydrophilic phase.
Preformed PFCs can be functionalized with targeting ligands (green spheres) through insertion of a cholesterol
anchor (red) into the lipid layer (SPIT, see text for details). After intravenous injection of PFCs (c), neat PFCs are
phagocytized by macrophages and monocytes and carried to inflammatory foci (d, left), whereas functiona-
lized PFCs can be directed to specific targets independent of phagocytosis (d, right, here thrombi)
2 Materials
2.3 Microfluidization 1. For the preparation of PFCs, an Ultra Turrax (TP 18/10;
of PFCs IKA-Werke, Staufen, Germany) is used for the primary high-
performance dispersion process, and a Microfluidizer (M110P,
Microfluidics, Newton, MA, USA) is applied for the
subsequent homogenization process at high pressure.
238 €den-Silber et al.
Tuba Gu
3 Methods
3.2 Surface Currently, different ways to modify lipid surfaces of PFC nanopar-
Functionalization ticles are in use. One of these methods is the sterol-based post-
of PFCs by Sterol- insertion technique (SPIT) which enables the modification of pre-
Based Post-insertion formed PFCs under very mild conditions using ligands coupled to
cholesterol moieties. The cholesterol moiety acts as an anchor
which spontaneously inserts into the lipid layer of the PFCs at
ambient temperature. Thus, the SPIT technique is particularly
suitable for labile targeting ligands which might be destroyed by
the harsh process of high-pressure homogenization or microfluidi-
zation [7, 8].
1. Before SPIT can be performed, a cholesterol containing com-
pound has to be prepared. The compound consists of the
cholesterol moiety (chol), a targeting ligand as well as a linker,
mostly a polyethylene glycol molecule of 1–2 kDa (PEG1000-
240 €den-Silber et al.
Tuba Gu
3.3 Disease Models Herein, we give an overview of models which were successfully
monitored by1H/19F MRI for immune cell tracking or thrombus
formation (see Subheading 4). Since the detailed description of
these models would be beyond the scope of this chapter, we refer
the interested reader to the according references. For the visualiza-
tion of inflammatory patterns or thrombus formation, mice
received in each case an intravenous bolus application of PFCs
(3 mM/kg body weight) at least 24 h prior the imaging session
to ensure adequate19F loading of circulating monocytes/macro-
phages and efficient binding to the thrombus, respectively.
1. Otitis model: Ear clipping is routinely used for animal identifi-
cation. We used ear clipping as a simple and reproducible
inflammation model as described by Jacoby et al. [6].
2. Induction of glomerulonephritis: The induction of anti-GBM
nephritis (GBM ¼ glomerular basement membrane) was per-
formed essentially as accomplished by Rosenkranz et al.
[9]. Three days after a preimmunization step, 0.25 ml of anti-
GBM antibodies were injected intravenously through the tail
vein. Control animals received 0.25 ml medium.
3. Subcutaneous inflammation model: We elaborated a simple
inflammation model injecting lipopolysaccharide (LPS) loaded
19
F MRI 241
3.4 Starting the MR 1. The animal is anesthetized with 1.5% isoflurane in a water-
Experiment saturated gas mixture of 30% oxygen in nitrogen applied at a
rate of 75 ml/min by manually restraining the animal and
placing its head in an in-house built nose cone (see Note 10).
Thereafter, it is fixed within a custom-made animal handling
system, and then, inserted into the magnet. Afterwards, the
coil is tuned and matched on the1H resonance. A pilot scan is
carried out to verify the correct positioning of the animal
within the resonator, and, if necessary, the position of the
mouse is corrected (see Note 17).
2. Before any experiment is started, it is important to adjust the
shim and basic frequency and to determine the reference atten-
uation as well as the optimal receiver gain.
3. The anatomical reference images (1H) of the region of interest
are acquired by performing either FLASH (fast low-angle shot)
or RARE (rapid acquisition with relaxation enhancement)
experiments (see other chapters in this book for detailed
description of1H MR techniques).
3.5 Adjusting 1. After the acquisition of the anatomical reference images, the
Parameters for19F MRI coil is tuned and matched to the19F resonance frequency.
2. Since the19F spin density will be quite sparse (even after injec-
tion of PFCs), most of the routinely applied automatic adjust-
ments will fail. Therefore, reference pulse gains for the distinct
242 €den-Silber et al.
Tuba Gu
3.7 Adjusting For compounds with complex19F MR spectra the spectral band-
the Spectral width used for conventional RARE imaging (cf. Subheading 3.6)
Bandwidth leads to chemical shift artifacts. However, due to the relatively large
for Multipeak MR chemical shift range of the19F nuclei (from 300 to 400 ppm)
Spectra these artifacts can be avoided by adjusting the19F RARE sequence
and aligning the effective spectral bandwidth to lower values. This
method is applicable for compounds like PFOB and F-44E with
19
F MRI 243
Fig. 2 Suitability of various PFCs for in vivo19F MR inflammation imaging in an otitis model. (a)19F MR spectra
of PFCE (top), PFOB (middle), and F-44E (bottom). PFCE generates a single resonance peak, whereas for PFOB
and F-44E multipeak MR spectra are obtained due to the presence of nonequivalent19F nuclei. Red brackets
indicate the spectral bandwidths used for the individual compound of 25 kHz for PFCE, and 10 kHz for PFOB
and F-44E, respectively. (b) In vivo visualization of inflammation after ear clipping:1H/19F MR images of the ear
acquired 24 h after clipping and intraveneous injection of PFCE (top), PFOB (middle), and F-44E (bottom),
respectively. In the1H MR images (left) the clipping areas are clearly evident. Anatomically corresponding
artifact-free19F images (middle) could be obtained for all PFCs with the spectral acquisition bandwidths given
in (a). The area of inflammation is clearly delineated by each PFC with almost comparable sensitivity.
Interestingly,19F signals were predominantly found at the proximal, highly perfused base of the ear as can
be recognized in the merged images (right)
3.8 Chemical Shift As already pointed out above, conventional gradient or spin echo
Imaging for Multipeak sequences for the imaging of PFCs with complex MR spectra
MR Spectra produce chemical shift artifacts along the readout direction. At
the same time, slice selection is impaired by displacement errors,
due to its frequency dependence. Using chemical shift imaging
(CSI) all spatial dimensions are phase encoded, wherefore chemical
shift artifacts are absent. Compared to the narrow bandwidth19F
RARE sequence (cf. see Subheading 3.7), in which only one reso-
nance frequency is used for imaging, the CSI method allows the
selection of all arising19F resonance frequencies for reconstruction
of the19F image.
1. For 2D CSI, spectra are acquired within a slice, spatially
arranged in a two-dimensional matrix. 2D CSI can be per-
formed with the following parameters: FOV 20 20 mm2,
Matrix 256 25 25 (first dimension is spectroscopic), ST
2 mm, sinc3 excitation pulse length ¼ 150 μs, flip angle ¼ 23 ,
TR ¼ 30 ms, TE ¼ 739 μs, sine-bell acquisition weighting with
a maximum of 28 averages, acquisition time ¼ 2.5 min.
2. It is also possible to examine the distribution of spectral com-
ponents within a volume of interest by performing 3D CSI
experiments using the following parameters: FOV
30 30 32 mm3, Matrix 256 33 33 17 (first
dimension is spectroscopic), sinc3 excitation pulse
length ¼ 150 μs, flip angle ¼ 23 , TR ¼ 30 ms, TE ¼ 732 μs,
averages ¼ 1, acquisition time ¼ 5.1 min.
3.9 Applications The high affinity of the macrophage/monocyte system for PFC
uptake (see Fig. 1) can be exploited to monitor the infiltration of
3.9.1 Immune Cell
PFC-loaded immune cells into inflammatory foci by19F MRI
Tracking with PFCs
in vivo [2, 6, 11, 12]. This enables the noninvasive visualization
of regions affected by inflammatory processes in several clinically
relevant disease models [2], such as viral myocarditis, ischemic
heart disease, stroke, atherosclerosis, glomerulonephritis, and
others.19F MRI is performed depending on the administered PFC
with a conventional, narrow bandwidth, or CSI sequence.
1. Conventional19F MRI using PFCE in a glomerulonephritis
model
19
F MRI 245
Fig. 3 Visualization of enhanced renal inflammation in CD73/ mice by1H/19F MRI. (a) Anatomically
corresponding sections of1H and19F MR images from individual WT and CD73/ mice acquired 10 days
after induction of nephritis showing an increased accumulation of19F signal in kidneys of the CD73-deficient
mice as compared to WT. Red arrows: cortex and medulla renalis; yellow arrows: subcutaneous fat; SC: spinal
cord; LK and RK: left and right kidney. (b) Quantification of19F signal intensity within the kidney 10 and 17 days
after immunization (n ¼ 5, *p < 0.05)
3.9.2 Active Targeting Due to the PFC uptake in macrophages and monocytes,19F MR
of Thrombi imaging with neat PFCs can be used for tracking of these cells in a
with Functionalized PFCs variety of settings. However, the functionalization of PFCs with
binding ligands enables a more specific targeting, and, therefore,
allows the visualization of structures which would not be labeled by
non-functionalized PFCs [14, 15].
19
F MRI 247
Fig. 4 Monitoring of immune cell infiltration in a subcutaneous inflammation model. (a) Anatomical location of
the LPS-doped matrigel for induction of subcutaneous inflammation. Sagittal (top) and axial slices (bottom)
show the site of injection (yellow arrows). The red, dashed rectangle corresponds to the position of the axial
slices shown on the right. (b)1H/19F MRI of mice injected with LPS-doped matrigel (1 μg/μl). F-44E was
injected 24 h after matrigel implantation, and animals were scanned by1H/19F MRI 24 h later. Yellow arrows
indicate the location of the matrigel plug.19F 3D CSI images were reconstructed with the CSI tool (cf. Fig. 6;
top:1H; middle:19F; bottom: merge). The inflamed area can be clearly located next to the matrigel
19
1. F MRI using targeted PFCs in an ex vivo as well as in vivo
thrombus model
During the early phase of thrombus formation, factor XIIIa
crosslinks α2-antiplasmin with fibrin [16]. Thus, peptides
derived from α2-antiplasmin (α2AP) can be used for specific
labeling of freshly developed thrombi. By means of SPIT, α2AP-
decorated PFCs with PFCE as core were formed (see Note 14)
and applied for active targeting and visualization of acute
thrombi by19F MRI. As negative control, a low affinity peptide
for factor XIIIa, i.e. Q3A [8, 17], was coupled to PFCs.
Figure 5a shows ex vivo human thrombi in vials incubated
with Q3A-PFCs (left vial) and α2AP-PFCs (right vial). As
expected, the application of Q3A-PFCs did not result in any
248 €den-Silber et al.
Tuba Gu
Fig. 5 Specific targeting of thrombi with α2AP-functionalized PFCs. (a)1H/19F MRI of vials containing ex vivo
formed human thrombi:1H MR reference (left),19F RARE image (middle), and merged images (right). The19F MR
signal only arises from thrombi exposed to α2AP-PFCs (right vial). Q3A-PFCs (left vial) were used as negative
control. (b) Top: In vivo visualization of thrombus formation in the murine vena cava with anatomical1H MR
reference (left),19F RARE image (middle), and merged images (right). Bottom: Enlargement of the area around
the vena cava. The formed thrombus can be vaguely recognized in the1H MR image (left), while the hot spot in
the19F MR image (middle) clearly indicates its presence within the vena cava when merged with the
anatomical reference (right). SC spinal cord, LK and RK left and right kidney, VC vena cava, AA aorta
abdominalis
4 Notes
Fig. 6 In-house developed software module for visualization and quantification of CSI datasets. (a) GUI of the
CSI analysis tool: Images loaded into the tool were acquired after injection of an emulsion mixture of PFCE and
PFOB into a healthy mouse. Top left:1H MR reference image with overlaying grid representing the in-plane
matrix. The red cross depicts the selected voxel. Top right: 2D19F CSI dataset with the individual19F MR
spectra in each voxel (FOV 2.56 2.56 cm2, matrix 64 64, zero-filling to 128 128, ST 1 mm, acquisition
time 16 min). Bottom:19F MR spectrum of the selected voxel (red cross) indicating the presence of both PFOB
and PFCE in the liver. The enlargement of this spectrum given beneath (red frame) shows the integration gates
for resonance peaks originating from PFOB. (b) Reconstruction of the19F MR image (middle) from the pixel-
wise summation of spectral intensities in the integration gates displayed above and merge (right) with the
anatomical1H MR image (left) demonstrating an equal distribution of PFOB over the entire liver
19
F MRI 251
Fig. 7 Appearance of isoflurane signals in19F MR images over time. (a) Chemical structure of isoflurane. (b)
Long-term MR investigation of a control mouse under isoflurane anesthesia (1.5%) without PFC injection.
Left:1H MR images of the mouse thorax (GE ¼ gradient echo; T1-weighted; RARE; T2-weighted) with slices
from the base of the heart (top) down to the apex (bottom) and the upper part of the liver. Right:19F MR images
(ST 2 mm, matrix 64 64) of the same locations at different time points (acquisition time 20 min) showing the
slow appearance of19F signals from isoflurane which is known to accumulate in lipid-rich areas, here in
particular in the subcutaneous fat. Of note, up to 1 h of anesthesia isoflurane-related signals are almost
negligible
Fig. 8 Setup for inhalation anesthesia. (a) The in-house-built nose cone for mice
is connected to an isoflurane vaporizer and a gas flow control unit with flow
meters (V-100) for O2 and N2. This setup enables the convenient control of
anesthesia throughout the experiment. (b) The magnification of the nose cone
shows the elastic mask, which tightly clasps around the nose of the mouse, the
inlet and the offlet system for the inhalation anesthesia to avoid accumulation of
isoflurane within the probe head
Acknowledgements
References
1. Bartusik D, Aebisher D (2014)19F applications 8. Temme S, Grapentin C, Quast C, Jacoby C,
in drug development and imaging—a review. Grandoch M, Ding Z, Owenier C, Mayenfels F,
Biomed Pharmacother 68(6):813–817. Fischer JW, Schubert R, Schrader J, Flögel U
https://doi.org/10.1016/j.biopha.2014.07. (2015) Noninvasive imaging of early venous
012 thrombosis by 19F magnetic resonance imag-
2. Flögel U, Ding Z, Hardung H, Jander S, ing with targeted perfluorocarbon nanoemul-
Reichmann G, Jacoby C, Schubert R, Schrader sions. Circulation 131(16):1405–1414.
J (2008) In vivo monitoring of inflammation https://doi.org/10.1161/
after cardiac and cerebral ischemia by fluorine CIRCULATIONAHA.114.010962
magnetic resonance imaging. Circulation 118 9. Rosenkranz AR, Mendrick DL, Cotran RS,
(2):140–148. https://doi.org/10.1161/ Mayadas TN (1999) P-selectin deficiency
CIRCULATIONAHA.107.737890 exacerbates experimental glomerulonephritis:
3. Bulte JW (2005) Hot spot MRI emerges from a protective role for endothelial P-selectin in
the background. Nat Biotechnol 23 inflammation. J Clin Invest 103(5):649–659.
(8):945–946. https://doi.org/10.1038/ https://doi.org/10.1172/JCI5183
nbt0805-945 10. Temme S, Jacoby C, Ding Z, Bönner F,
4. Zhong J, Mills PH, Hitchens TK, Ahrens Borg N, Schrader J, Flögel U (2014) Technical
(2013) Accelerated fluorine-19 MRI cell track- advance: monitoring the trafficking of neutro-
ing using compressed sensing. Magn Reson phil granulocytes and monocytes during the
Med 69(6):1683–1690. https://doi.org/10. course of tissue inflammation by noninvasive
1002/mrm.24414 19F MRI. J Leukoc Biol 95(4):689–697.
5. Krafft MP (2001) Fluorocarbons and fluori- https://doi.org/10.1189/jlb.0113032
nated amphiphiles in drug delivery and bio- 11. Flögel U, Su S, Kreideweiss I, Ding Z,
medical research. Adv Drug Deliv Rev 47 Galbarz L, Fu J, Jacoby C, Witzke O, Schrader
(2–3):209–228 J (2011) Noninvasive detection of graft rejec-
6. Jacoby C, Temme S, Mayenfels F, Benoit N, tion by in vivo (19) F MRI in the early stage.
Krafft MP, Schubert R, Schrader J, Flögel U Am J Transplant 11(2):235–244. https://doi.
(2014) Probing different perfluorocarbons for org/10.1111/j.1600-6143.2010.03372.x
in vivo inflammation imaging by 19F MRI: 12. Temme S, Bönner F, Schrader J, Flögel U
image reconstruction, biological half-lives and (2012) 19F magnetic resonance imaging of
sensitivity. NMR Biomed 27(3):261–271. endogenous macrophages in inflammation.
https://doi.org/10.1002/nbm.3059 Wiley Interdiscip Rev Nanomed Nanobiotech-
7. Molnar D, Linders J, Mayer C, Schubert R nol 4(3):329–343. https://doi.org/10.1002/
(2016) Insertion stability of poly(ethylene gly- wnan.1163
col)-cholesteryl-based lipid anchors in lipo- 13. Grenz A, Zhang H, Eckle T, Mittelbronn M,
some membranes. Eur J Pharm Biopharm Wehrmann M, Kohle C, Kloor D, Thompson
103:51–61. https://doi.org/10.1016/j.ejpb. LF, Osswald H, Eltzschig HK (2007) Protec-
2016.03.023 tive role of ecto-50 -nucleotidase (CD73) in
renal ischemia. J Am Soc Nephrol 18
19
F MRI 257
Abstract
Rodents including rats and mice are important models to study obesity, diabetes, and metabolic syndrome
in a preclinical setting. Translational and longitudinal imaging of these rodents permit investigation of
metabolic diseases and identification of imaging biomarkers suitable for clinical translation. Here we
describe the imaging protocols for achieving quantitative abdominal imaging in small animals followed
by segmentation and quantification of fat volumes.
Key words Magnetic Resonance Imaging, Abdomen, Rats, Mice, Segmentation, Visceral fat, Subcu-
taneous fat, Obesity, Quantification
1 Introduction
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0_15, © Springer Science+Business Media, LLC 2018
259
260 Bhanu Prakash KN et al.
2 Materials
2.2 Other Equipment 1. Respiration and body temperature was monitored using a phys-
iological monitoring system (ML880 16/30 power lab system,
AD Instruments, Spechbach, Germany).
2. Dedicated animal chamber for anesthetizing the animals.
3 Methods
3.1 Animal 1. Anesthesia : Place the animal in the induction chamber and
Preparation anesthetize with 2–2.5% isoflurane mixed with medical grade
oxygen and air. Monitor the vital signs including breathing and
heart rate while the animal is anesthetized. Ensure that animal is
completely anesthetized before imaging. Maintain the isoflurane
MR Imaging of Adipose Tissue 261
3.2 MR Coil Selection 1. The transmit/receive volume coils are more suitable for imag-
ing of abdomen. The dimensions of the coils can vary depend-
ing on the system and vendors. It is preferable to use a coil
which provides maximum filling factor to achieve good signal-
to-noise ratio along with superior spatial resolution.
3.3 In Vivo MR 1. Localizer: Rapid gradient echo imaging sequences can be uti-
Imaging lized to localize the abdominal area. Localizer will provide
multiple images in three orthogonal planes including axial
(transverse), sagittal, and coronal directions. Initially utilize a
larger field of view (FOV) of around 45–70 mm for rats and
relatively smaller FOV of 25–35 mm for mice. Following para-
meters can be utilized for a rat weighing around 250 g. FOV
70 mm, Repetition time (TR) 300–350 ms, Echo time (TE)
~4 ms, Flip angle (FA) 25 , matrix size 128 128, in plane
resolution 0.430 mm 0.430 mm, slice thickness
(ST) ~1 mm, slice mode- interleaved, number of averages
(NA) ~1, 15 slices in each imaging plane including axial, sagit-
tal, and coronal directions. Depending upon the animal size the
parameters can be optimized for maximum coverage and
resolution.
262 Bhanu Prakash KN et al.
Fig. 1 Localization of abdominal compartment with the help of coronal T2 TSE imaging
Fig. 2 In-phase, out-phase (top row), fat and water (bottom row) images acquired using 2point Dixon
sequence on a 7 T scanner
3.4 Post-processing In the following section we describe a general procedure for quan-
tification of subcutaneous fat (SAT) and visceral adipose tissue
(VAT). The SAT and VAT compartments are separated by the
abdominal muscular wall where the fat exterior to the abdominal
wall is referred as SAT, whereas the interior compartment to the
264 Bhanu Prakash KN et al.
Fig. 3 Block diagram representation of overall post-processing for SAT and VAT quantification used in our
study
abdominal wall is VAT (see Note 2). Figure 3 shows the different
stages of processing and quantification.
1. Collect the data from the MR scanner in DICOM format. If
DICOM format is not available then use any software that
converts the format of images to DICOM or to Analyze/
Nifti format. In the proposed framework we use the DICOM
format data.
2. Combine the individual DICOM files into a volume data,
where rows represent X, and columns are Y and slices are
represented as Z.
3. Pre-processing is an important step of segmentation. In this
stage remove information like noise, background, gantry, tub-
ing, etc. which influence the quantification. In our study we
used anisotropic diffusion filter based on Perona–Malik equa-
tions to remove the noise and enhance the edges of the abdom-
inal wall [12].
4. Bias field correction to reduce the B0 inhomogeneity was per-
formed after filtering using biased fuzzy C-means
algorithm [13].
5. Dixon imaging provides fat, water, in-phase, and out-phase
images. In the fat volume only fat is highlighted and signals
coming from water are completely suppressed. The range of
intensities in each slice might be different due to inhomogene-
ity effects (B0, RF, receive coils, etc.). Normalize the intensity
range of every slice in the volume data.
MR Imaging of Adipose Tissue 265
Fig. 4 A sample slice based representation of results at different stages of SAT and VAT segmentation
4 Notes
References
1. Brochu M, Poehlman, Ades PA (2000) Obe- Horiguchi T, Nishimoto T, Velan SS (2016)
sity, body fat distribution, and coronary artery Effect of exercise and calorie restriction on tissue
disease. J Cardpulm Rehabil 20(2):96–108 acylcarnitines, tissue desaturase indices, and fat
2. Colberg SR, Simoneau JA, Thaete FL, Kelley accumulation in diet-induced obese rats. Sci Rep
DE (1995) Skeletal muscle utilization of free 6:26445. https://doi.org/10.1038/srep26445
fatty acids in women with visceral obesity. J 7. Sadananthan SA, Prakash B, Leow MK, Khoo
Clin Invest 95(4):1846–1853. https://doi. CM, Chou H, Venkataraman K, Khoo EY, Lee
org/10.1172/JCI117864 YS, Gluckman PD, Tai ES, Velan SS (2015)
3. Evans DJ, Hoffmann RG, Kalkhoff RK, Kisse- Automated segmentation of visceral and sub-
bah AH (1984) Relationship of body fat cutaneous (deep and superficial) adipose tissues
topography to insulin sensitivity and metabolic in normal and overweight men. J Magn Reson
profiles in premenopausal women. Metab Clin Imaging 41(4):924–934. https://doi.org/10.
Exp 33(1):68–75 1002/jmri.24655
4. Vague J (1956) The degree of masculine differ- 8. Marzola P, Boschi F, Moneta F, Sbarbati A,
entiation of obesities: a factor determining pre- Zancanaro C (2016) Preclinical in vivo imaging
disposition to diabetes, atherosclerosis, gout, for fat tissue identification, quantification, and
and uric calculous disease. Am J Clin Nutr 4 functional characterization. Front Pharmacol
(1):20–34 7:336. https://doi.org/10.3389/fphar.2016.
5. Kn BP, Gopalan V, Lee SS, Velan SS (2014) 00336
Quantification of abdominal fat depots in rats 9. HH H, Kan HE (2013) Quantitative proton
and mice during obesity and weight loss inter- MR techniques for measuring fat. NMR
ventions. PLoS One 9(10):e108979. https:// Biomed 26(12):1609–1629. https://doi.org/
doi.org/10.1371/journal.pone.0108979 10.1002/nbm.3025
6. Gopalan V, Michael N, Ishino S, Lee SS, Yang 10. Schick F (1998) Simultaneous highly selective
AY, Bhanu Prakash KN, Yaligar J, Sadananthan MR water and fat imaging using a simple new
SA, Kaneko M, Zhou Z, Satomi Y, type of spectral-spatial excitation. Magn Reson
Hirayama M, Kamiguchi H, Zhu B, Med 40(2):194–202
268 Bhanu Prakash KN et al.
11. Reeder SB, McKenzie CA, Pineda AR, Yu H, of volumetric and single-slice MRI adipose
Shimakawa A, Brau AC, Hargreaves BA, Gold analysis using a novel fully automated segmen-
GE, Brittain JH (2007) Water-fat separation tation method. J Magn Reson Imaging 41
with IDEAL gradient-echo imaging. J Magn (1):233–241. https://doi.org/10.1002/jmri.
Reson Imaging 25(3):644–652. https://doi. 24526
org/10.1002/jmri.20831 19. Joshi AA, HH H, Leahy RM, Goran MI, Nayak
12. Pernona P, Malik J (1990) Scale-space and edge KS (2013) Automatic intra-subject registra-
detection using anisotropic diffusion. IEEE Trans tion-based segmentation of abdominal fat
Pattern Anal Mach Intell 12(7):629–639. from water-fat MRI. J Magn Reson Imaging
https://doi.org/10.1109/34.56205 37(2):423–430. https://doi.org/10.1002/
13. Ahmed MN, Yamany SM, Mohamed N, Farag jmri.23813
AA, Moriarty T (2002) A modified fuzzy 20. Ranefall P, Bidar AW, Hockings PD (2009)
C-means algorithm for bias field estimation Automatic segmentation of intra-abdominal
and segmentation of MRI data. IEEE Trans and subcutaneous adipose tissue in 3D whole
Med Imaging 21(3):193–199. https://doi. mouse MRI. J Magn Reson Imaging 30
org/10.1109/42.996338 (3):554–560. https://doi.org/10.1002/jmri.
14. Peli T, Malah D (1982) A study of edge detec- 21874
tion algorithms. Comput Graph Image Process 21. Shen J, Baum T, Cordes C, Ott B, Skurk T,
20(1):1–21. https://doi.org/10.1016/0146- Kooijman H, Rummeny EJ, Hauner H, Menze
664X(82)90070-3 BH, Karampinos DC (2016) Automatic seg-
15. Lankton S, Tannenbaum A (2008) Localizing mentation of abdominal organs and adipose
region-based active contours. IEEE Trans tissue compartments in water-fat MRI: applica-
Image Process 17(11):2029–2039. https:// tion to weight-loss in obesity. Eur J Radiol 85
doi.org/10.1109/TIP.2008.2004611 (9):1613–1621. https://doi.org/10.1016/j.
16. Zou KH, Warfield SK, Bharatha A, Tempany ejrad.2016.06.006
CM, Kaus MR, Haker SJ, Wells WM 3rd, 22. Tang Y, Sharma P, Nelson MD, Simerly R,
Jolesz FA, Kikinis R (2004) Statistical valida- Moats RA (2011) Automatic abdominal fat
tion of image segmentation quality based on a assessment in obese mice using a segmental
spatial overlap index. Acad Radiol 11 shape model. J Magn Reson Imaging 34
(2):178–189 (4):866–873. https://doi.org/10.1002/jmri.
17. HH H, Bornert P, Hernando D, Kellman P, 22690
Ma J, Reeder S, Sirlin C (2012) ISMRM work- 23. Wald D, Teucher B, Dinkel J, Kaaks R,
shop on fat-water separation: insights, applica- Delorme S, Boeing H, Seidensaal K, Meinzer
tions and progress in MRI. Magn Reson Med HP, Heimann T (2012) Automatic quantifica-
68(2):378–388. https://doi.org/10.1002/ tion of subcutaneous and visceral adipose tissue
mrm.24369 from whole-body magnetic resonance images
18. Addeman BT, Kutty S, Perkins TG, Soliman suitable for large cohort studies. J Magn Reson
AS, Wiens CN, McCurdy CM, Beaton MD, Imaging 36(6):1421–1434. https://doi.org/
Hegele RA, McKenzie CA (2015) Validation 10.1002/jmri.23775
Chapter 16
Abstract
Cardiac magnetic resonance (MR) imaging of mice is a valuable tool for the precise in vivo diagnosis and
prognosis of heart defects. This detailed protocol describes the method of cardiac MR imaging in mice step
by step. A series of MR images captures the contractile function of the mouse heart and post-processing of
the image data yields morphometric parameters (myocardial mass, myocardial wall thickness, ventricular
end-systolic and end-diastolic volume) as well as functional parameters (stroke volume and ejection
fraction). This protocol may also serve as a starting point for MR imaging of rats, by using larger image
dimensions (field-of-view) and MR hardware suitable for larger animals.
Key words Magnetic resonance imaging (MRI), Cardiac magnetic resonance imaging (CMR),
Mouse, Heart, Function
1 Introduction
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0_16, © Springer Science+Business Media, LLC 2018
269
270 Min-Chi Ku et al.
2 Materials
2.1 Animals Mice are housed under standard conditions according to the animal
welfare guidelines and regulations. Here we describe cardiac MRI
of C57BL/6J mice with a body weight of 20–30 g. Other strains
and ages may require some adaptations of the experimental setup
and protocol.
2.3 Magnetic An MRI system including suitable accessories for the MR acquisi-
Resonance Imaging tion such as radio frequency (RF) antennas; equipment for animal
positioning, inhalation anesthesia, warming, and monitoring
Cardiac MRI 271
3 Methods
3.3 MRI Pre-scans 1. Register a new subject and study. For the first scan load the
Protocol-1_Localizer.
2. Tune and match the volume resonator using the Wobble
function.
3. Start the 1_Localizer scan and verify on the acquired images
that the heart is close to the isocenter of the magnet and well
within the field of view. If the heart is too far away from the
center of the magnet and the RF coil (heart not visible or in
region where signal intensity starts to tail off), it may be neces-
sary correct the animal or/and Laser marker position. In that
case, repeat the tuning/matching and this pilot scan.
4. Load the Protocol-2_Multislice_Localizer. Using the 1_Loca-
lizer scan as the reference edit the geometry of the 2_Multi-
slice_Localizer such that the slices packages are centered
around the heart (Fig. 1) and run the scan.
Cardiac MRI 275
Fig. 1 Image from 1_Localizer scans displaying the mouse thorax and upper abdomen in axial, sagittal, and
coronal views. The overlaid slice contours show how to position the slice packages for 2_Multislice_Localizer
centered on the heart
Fig. 2 (a) Representative slice for axial view of the thorax provided by the 2_Multislice_Localizer. The overlaid
slice contours show how to position the slice packages for running 3_Coronal_Localizer. The slice package is
centered on the heart. (b) Representative slice for coronal view of the thorax provided by the 3_Coronal_Lo-
calizer and the overlaid yellow box shows slice contour for running the 4_Sagittal_LAX_Ig
Fig. 3 (a) Representative image of the mouse heart obtained from the 4_Sagit-
tal_LAX_Ig scan with overlaid slice contour of the 5_Coronal_LAX_Ig illustrating
how to plan the geometry. (b) Representative image obtained from 5_Coronal_-
LAX_Ig scan
(RR) of the animal just before starting the scan. If the initial
guess for nominal HR and RR is too far off then the images
may have severe artifacts. In this case the scan must be repeated
with different nominal HR and RR. After each scan the Intra-
gate method provides both HR and RR values that are derived
from the MR data (a so-called navigator) which can be found in
the reconstruction panel and can help to adjust the nominal
HR and RR to improve image quality. Set the nominal HR and
RR, then run the scan.
7. Load the Protocol-5_Coronal_LAX_Ig. Use the 4_Sagittal_-
LAX_Ig as reference image and browse through the CINE
images of the different heart phases to select the one depicting
diastole (maximum dilation of the left ventricle). Position the
slice from the 5_Coronal_LAX_Ig scans such that the slice is
perpendicular to the reference image and centered along the
long axis of the left ventricle of the heart (Fig. 3a). Set the HR
and RR, then run the scan. Check the scan result and pick the
diastolic image from the coronal view (Fig. 3b).
8. Load the Protocol-6_SAX_CINE_Ig: Use the image from
5_Coronal_LAX_Ig as reference and browse to the CINE
movie frame of diastole. Align the navigator slice (blue box in
Fig. 4a) (see Note 4) along the long axis of the left ventricle and
perpendicular to the reference image. Adjust the slice geometry
of the 6_SAX_CINE_Ig scan to be perpendicular to the heart
long axis and the reference image (yellow box in Fig. 4a); move
the slice to the middle of the heart. Set the HR and RR, then
run the scan, which serves as a test scan for the following series
of short axis (SAX) images. Check the obtained images for all
16 heart phases (Fig. 4b) and pick the diastolic phase (here is
phase 8; also see Fig. 5a).
Cardiac MRI 277
Fig. 4 (a) Representative slice for long axis (LAX) view obtained from the 5_Coronal_LAX_Ig scan. Overlaid is
the navigator slice outline (blue box) and the short axis image slice outline (yellow box) of the 6_SAX_CINE_Ig
scan. (b) 16 phases of cardiac short axis (SAX) views from a test scan of the 6_SAX_CINE_Ig scan
Fig. 5 (a) A representative slice for SAX view in diastole from 6_SAX_CINE_Ig scan with overlaid slice outline
(yellow box) of the second, improved LAX (5_Coronal_LAX_Ig scan). (b) A representative slice for improved
LAX view obtained with Protocol-5_Coronal_LAX_Ig. LV left ventricle, RV right ventricle
3.4 Heart Short Axis 1. Duplicate the Protocol-6_SAX_CINE_Ig scan. Use the
View Images improved LAX images (Fig. 5b) as the scan reference and pick
the diastolic CINE frame to fine adjust the geometry of the
278 Min-Chi Ku et al.
Fig. 6 (a) Slice positioning of the navigator slice (blue box) and 9 individual SAX CINE image slices (yellow
boxes) for 6_SAX_CINE_Ig based on the improved LAX view of the mouse heart. Adjust the slice angles
according to the heart orientation. (b, c) Stacks of 9 SAX image slices that represent the main result of the MRI
examination used for heart function assessment. (b) Mouse heart SAX images obtained with the conventional
room temperature RF coil (RT). (c) SAX images of another mouse provided by the cryogenic RF coil (CP). Please
note that the scan time by using the cryogenic RF coil is reduced to 1/4 compared to the room temperature RF
coil (see Note 2). Numbers 1–9 in yellow indicate the slice positions shown in Fig.6a
image slice and navigator slice. Shift the image slice to the
heart’s apex (slice 1 in Fig. 6a). Set the HR and RR, then run
the scan. Duplicate the scan protocol and place the second slice
1.1 mm above the slice 1. Repeat this procedure until covering
the entire heart, in particular the left ventricle (create nine slices
in this case) (Fig. 6a). These images represent the main result of
the MRI examination, which will be then used for heart func-
tion assessment.
3.5 Heart Long Axis Load the Protocol-7_LAX_CINE_Ig. Pick the diastolic slice from
View Images (2–4- a short axis view (the scan result from Protocol-6_SAX_CINE_Ig)
Chambers) and place the slice position (similar with the slice position shown in
Fig. 5a); adjust the angle of the slice to make it perpendicular to the
heart short axis. Set the HR and RR, then run the scan. Fig. 7
shows the scan result of Protocol-7. Please note that depending on
the angle of the slice orientation, different views of the heart’s long
axis might present.
3.6 End 1. Carefully remove the mouse from the mouse holder.
of the Experiment 2. Move the animal into the preparation room and place in a
supine position on a pre-warmed mat to recover from the
anesthesia.
3.7 Data Analysis 1. On the scanner console export all images in DICOM (Digital
Imaging and Communications in Medicine) format.
2. The analysis software CMR42 requires the LAX and all SAX
images to have the same DICOM frame of reference UID (user
Cardiac MRI 279
4 Notes
1. The temperature probes are rather fragile and you may want to
consider using protectively coated GaAs probes instead. In that
case be aware that a GaAs crystal based system shows a temper-
ature offset caused by the magnetic field (approx. 4.7 C at
9.4 T).
280 Min-Chi Ku et al.
Fig. 8 Illustration of image analysis in CMR42. There are shown the SAX images of 3 slices for 10 cardiac
phases, including systole (yellow box) and diastole (green box). Endo- (red circle) and epi-cardial (green circle)
borders in each slice have to be outlined manually
Fig. 9 Illustration of outlining contour for endo- and epi-cardial borders for both
LV and RV. White dashed arrows indicate papillary muscles which has to be
excluded in contour
Acknowledgement
References
1. Camacho P, Fan H, Liu Z, He JQ (2016) Small French BA, Epstein FH (2012) T(2) -weighted
mammalian animal models of heart disease. Am MRI of post-infarct myocardial edema in mice.
J Cardiovasc Dis 6(3):70–80 Magn Reson Med 67(1):201–209. https://
2. Zaragoza C, Gomez-Guerrero C, Martin- doi.org/10.1002/mrm.22975
Ventura JL, Blanco-Colio L, Lavin B, 7. Musthafa HS, Dragneva G, Lottonen L,
Mallavia B, Tarin C, Mas S, Ortiz A, Egido J Merentie M, Petrov L, Heikura T,
(2011) Animal models of cardiovascular dis- Yla-Herttuala E, Yla-Herttuala S, Grohn O,
eases. J Biomed Biotechnol 2011:497841. Liimatainen T (2013) Longitudinal rotating
https://doi.org/10.1155/2011/497841 frame relaxation time measurements in
3. Stuckey DJ, McSweeney SJ, Thin MZ, Habib J, infarcted mouse myocardium in vivo. Magn
Price AN, Fiedler LR, Gsell W, Prasad SK, Reson Med 69(5):1389–1395. https://doi.
Schneider MD (2014) T(1) mapping detects org/10.1002/mrm.24382
pharmacological retardation of diffuse cardiac 8. Vandsburger MH, Epstein FH (2011)
fibrosis in mouse pressure-overload hypertro- Emerging MRI methods in translational car-
phy. Circ Cardiovasc Imaging 7(2):240–249. diovascular research. J Cardiovasc Transl Res
https://doi.org/10.1161/CIRCIMAGING. 4(4):477–492. https://doi.org/10.1007/
113.000993 s12265-011-9275-1
4. Coelho-Filho OR, Shah RV, Mitchell R, Neilan 9. Wagenhaus B, Pohlmann A, Dieringer MA,
TG, Moreno H Jr, Simonson B, Kwong R, Els A, Waiczies H, Waiczies S, Schulz-Menger J,
Rosenzweig A, Das S, Jerosch-Herold M Niendorf T (2012) Functional and morpho-
(2013) Quantification of cardiomyocyte hyper- logical cardiac magnetic resonance imaging of
trophy by cardiac magnetic resonance: implica- mice using a cryogenic quadrature radiofre-
tions for early cardiac remodeling. Circulation quency coil. PLoS One 7(8):e42383. https://
128(11):1225–1233. https://doi.org/10. doi.org/10.1371/journal.pone.0042383
1161/CIRCULATIONAHA.112.000438 10. Niendorf T, Pohlmann A, Reimann HM,
5. Abeykoon S, Sargent M, Wansapura JP (2012) Waiczies H, Peper E, Huelnhagen T,
Quantitative myocardial perfusion in mice Seeliger E, Schreiber A, Kettritz R, Strobel K,
based on the signal intensity of flow sensitized MC K, Waiczies S (2015) Advancing cardiovas-
CMR. J Cardiovasc Magn Reson 14:73. cular, neurovascular, and renal magnetic reso-
https://doi.org/10.1186/1532-429X-14-73 nance imaging in small rodents using cryogenic
6. Beyers RJ, Smith RS, Xu Y, Piras BA, radiofrequency coil technology. Front
Salerno M, Berr SS, Meyer CH, Kramer CM,
284 Min-Chi Ku et al.
Abstract
Genetically engineered mouse models are used extensively as models of human development and develop-
mental diseases. Conventional histological approaches are static and two-dimensional, and do not provide a
full understanding of the dynamic, spatiotemporal changes in developing mouse embryos. Magnetic
resonance imaging (MRI) offers a noninvasive and longitudinal approach for three-dimensional in utero
imaging of normal and mutant mouse embryos. In this chapter, we describe MRI approaches that have been
developed for imaging the living embryonic mouse brain and vasculature. Details are provided on the
animal preparation and setup, MRI equipment, acquisition and reconstruction methods that have been
found to be most useful for in utero MRI, including examples of applications to fetal mouse neuroimaging.
Key words Diffusion MRI, Diffusion weighted gradient and spin echo (DW-GRASE), Field of
excitation (FOE), Fractional anisotropy (FA), High-field MRI, Mn-enhanced MRI (MEMRI), Phased
array coil, Three-dimensional (3D)
1 Introduction
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0_17, © Springer Science+Business Media, LLC 2018
285
286 Jiangyang Zhang et al.
2 Materials
2.1 High-Field MRI 1. Small animal MRI system: a high-field (preferably 7 Telsa or
Systems higher) MRI system is required to perform in utero MRI of
mouse embryos. The in utero MRI experiments described in
this chapter were performed on horizontal 7 Tesla (T) and
11.7 T MRI systems (Bruker Biospin, Billerica, MA, USA),
each with an integrated shim and actively shielded three-axis
gradient (B-GA9S, Bruker Biospin, Billerica, MA, USA, inner
diameter ¼ 90 mm, maximum gradient strength ¼ 740
mT/m). The 11.7 T scanner has a motor driven automatic
positioning system (Autopac, Bruker Biospin) for animal
positioning.
2. RF coils: MRI experiments (7 and 11.7 T) have been per-
formed using a 72 mm (inner diameter) quadrature volume
coil for transmit (Bruker Biospin) in combination with a
receive-only planar surface coil, using a pin-diode to decouple
the transmit and receive coils and a low-noise preamplifier on
the receive coil. At 11.7 T, a whole-body 8-channel receive-
only phased array coil was also used (Bruker Biospin, Part
No. T20035 V3) in combination with the transmit
volume coil.
3. Mouse Holders: We use either custom-made holders or the
Autopac compatible animal holders provided by the manufac-
turer of the MRI system (Bruker Biospin).
In Utero MRI 287
3 Methods
3.2 Animal Setup Anesthesia is induced in pregnant mice using 3–5% isoflurane
mixed with air (via the vaporizer), or with a 3:1 air:oxygen mixture
(see Note 2). During MRI, the amount of isoflurane should be
reduced to 1–1.5% and adjusted regularly to maintain the respira-
tory rate at 30–60 breaths per minute. A pregnant mouse may have
up to 12 embryos, each residing in their own amniotic sacs and
distributed along the two uterine horns (Fig. 1a). In order to
achieve uniform excitation while maintaining high sensitivity, it is
ideal to use a large whole body volume coil as the transmission coil
(Tx) in combination with a receive-only surface coil or a receive-
only phased array body coil (Fig. 1b). The phased array coil pro-
vides more abdominal coverage than the surface coil but the surface
coil can be positioned as close to the target embryo as possible and
provide higher sensitivity (see Note 3).
288 Jiangyang Zhang et al.
Fig. 1 Animal setup and the initial 2D multi-slice T2-weighted images. (a) The anatomy of a pregnant mouse
showing 12 embryos in the two uterine horns: (b) Placement of a pregnant mouse with a receive-only planar
surface coil (top) and a receive-only phased array coil (bottom). (c) axial T2-weighted images of mouse
embryos at embryonic day 17 (E17) in a pregnant mouse. The embryos are surrounded by the amniotic fluid,
which has strong T2 signals. With high in-plane resolution, large internal organs, e.g., the brain, liver, and
spinal cord, can be identified. (d) Three-dimensional rendering of amniotic sacs (gray) and embryonic mouse
brains (purple) in the abdomen based on the axial T2-weighted images
Fig. 2 In utero T1-weighted MEMRI and T2*-weighted vascular MRI. (a) Mn-enhancement and respiratory
gating work in combination to improve in utero MRI. Images at E14.5 with gating and without Mn (Left panel,
Gatingþ/Mn), with Mn and without gating (Middle panel, Gating/Mnþ) and with gating and Mn (Right
panel, Gatingþ/Mnþ). Scale bar is 1 mm. The images are modified with permission from [5]. (b) 3D maximum
intensity projections (MIPs) show the developing vasculature from E10.5 to E14.5. Labels: DA dorsal aorta, FV
facial vein, H heart, JV jugular vein, NV nasal vein, OA optic artery, VC vena cava, VS venous sinus. The images
are modified with permission from [9]
3.4 Two- A fast survey of the complex anatomy can be obtained using 2D
Dimensional multi-slice T2-weighted MRI, which can provide satisfactory con-
(2D) Multi-Slice trasts to distinguish individual embryos in a relatively short time
T2-Weighted MRI (~5 min) (Fig. 1c). Because of the low through-plane resolution of
of Pregnant Mice 2D MRI, both axial and sagittal images of the mouse abdomen
need to be acquired to visualize the locations and orientations of
mouse embryos and guide subsequent imaging.
290 Jiangyang Zhang et al.
3.5 Generation The next step is to select an embryo for localized imaging based on
of Tailored Radio- the 2D multi-slice T2-weighted images (see Subsection 3.4). For
Frequency (RF) Pulses localized imaging, it is necessary to define a so-called field of
for Localized Imaging excitation (FOE) that encloses the selected embryo or embryonic
of Mouse Embryos brain. To facilitate the process, we have designed a software tool to
display the T2-weighted images and define the FOE (Fig. 3). The
software is based on Matlab (Mathworks, mathworks.com) and can
run on the scanner console. Once the size and location of an FOE
are defined, the software generates a tailored 90 selective excita-
tion RF pulse based on a linear class of large tip-angle (LCLTA)
pulses [10] with spiral k-space trajectories that start and end at the
origin. Under the “incoherently refocused” condition, a 2D selec-
tive 90 RF pulse can be derived by inverse Fourier transform of the
desired excitation profile [10]. The pulse is designed to excite a
rectangular FOE in the x–y plane that covers the target region, with
a duration of 3 ms, an amplitude of 9–10 μT, and a 12-turn spiral-in
excitation k-space (maximum gradient strength ¼ 148 mT/m).
Details on the performance of the designed RF pulses can be
found in [11].
Fig. 3 Definition of the field of excitation (FOE, represented by the red box) based on the 2D multi-slice
T2-weighted images using a software tool. Using the software, users can display the T2-weighted images,
define the size and spatial offsets of an FOE, and calculate the selective excitation RF pulse to excite the FOE
with the desired parameters. The lower panel displays the amplitude and phase profiles of the generated RF
pulse
3.7 Image The 3D k-space data are first apodized with a tapered cosine win-
Reconstruction dow, zero-padded to twice the original size, and reconstructed in
Matlab. The twin-navigator echoes are Fourier transformed along
the readout direction, which are then used to correct the phases of
odd- and even-numbered echoes from each repetition and phase
errors caused by intra-scan motion [12]. The 30 direction
292 Jiangyang Zhang et al.
Fig. 4 A diagram of the 3D DW-GRASE sequence with a spatially selective excitation pulse. The diagram
shows the timing of the 2D selective excitation pulse together with the spiral gradient in the x–y plane, the
diffusion encoding gradients (represented by turquoise trapezoids), the GRASE readout module, and the twin-
navigator echoes. Each GRASE readout module acquires four gradient echoes and one spin echo, and the
readout is repeated four times to achieve an acceleration factor of 20 compared to the conventional spin echo
sequence. The images are modified with permission from [6]
4 Notes
1. For in utero MRI, we have found that MnCl2 can cause embry-
onic toxicity and death, especially for embryos staged earlier
than embryonic day (E)13.5 [5]. For embryos staged E13.5 or
In Utero MRI 293
Fig. 5 Methods for motion correction employed in in utero imaging of the embryonic mouse brain. (a) Due to
intra-scan motion, the boundary of the fourth ventricle in the embryonic mouse brain (indicated by the yellow
arrow) is not clear. Real-time navigator echoes can be used to correct intra-scan motion, and the boundary of
the 4th ventricle becomes clear. (b) Motions between scans cause images acquired sequentially to be
mis-adlinged, as indicated by the cross-hair in the images. This mis-alignment can be corrected using linear
rigid image registration. (c) The inter-scan motion can be estimated using the linear rigid image registration.
The plot shows he amount of overall brain displacement of 5 E17.5 mouse brains during 60-min scans. The
images are modified with permission from [6]
Fig. 6 In utero MRI of the embryonic mouse brain. (a) Coronal T2-weighted, FA, and directionally encoded
colormap (DEC) images of an E17.5 mouse brain. (b) Surface rendering of the ventricules in the E17.5 mouse
brain based on the T2-weighted images. (c) rendering of the gray matter structures and early white matter
tracts in the E17.5 mouse brain based on the diffusion MRI data. Abbreviations: CP cortical plate, cp cerebral
peduncle, IZ intermediate zone, fi fimbria, Hi hippocampus, LV lateral ventricle, opt optical tract, st stria
terminalis, Th thalamus, 3V and 4V the third and forth ventricles
Acknowledgements
We thank all the people, current and past, in the Zhang, Wu and
Turnbull labs who have contributed to developing the protocols
described in this chapter. This research was supported, in part, by
grants from the National Institutes of Health: R01NS038461 and
R01HL078665 (DHT); R01NS070909 and R01HD974593
(JZ); R21NS098018 (DW).
References
1. International Mouse Knockout Consortium, mouse embryonic vasculature with MRI. Magn
Collins FS, Rossant J, Wurst W (2007) A Reson Med 67(1):251–257. https://doi.org/
mouse for all reasons. Cell 128(1):9–13. 10.1002/mrm.22991
https://doi.org/10.1016/j.cell.2006.12.018 8. Nieman BJ, Szulc KU, Turnbull DH (2009)
2. Nieman BJ, Wong MD, Henkelman RM Three-dimensional, in vivo MRI with self-
(2011) Genes into geometry: imaging for gating and image coregistration in the mouse.
mouse development in 3D. Curr Opin Genet Magn Reson Med 61(5):1148–1157. https://
Dev 21(5):638–646. https://doi.org/10. doi.org/10.1002/mrm.21945
1016/j.gde.2011.08.009 9. Parasoglou P, Berrios-Otero CA, Nieman BJ,
3. Aristizabal O, Mamou J, Ketterling JA, Turn- Turnbull DH (2013) High-resolution MRI of
bull DH (2013) High-throughput, high-fre- early-stage mouse embryos. NMR Biomed 26
quency 3-D ultrasound for in utero analysis of (2):224–231. https://doi.org/10.1002/nbm.
embryonic mouse brain development. Ultra- 2843
sound Med Biol 39(12):2321–2332. https:// 10. Pauly J, Nishimura D, Macovski A (1989) A
doi.org/10.1016/j.ultrasmedbio.2013.06. linear class of large-tip-angle selective excita-
015 tion pulses. J Magn Reson 82(3):571–587.
4. Nieman BJ, Turnbull DH (2010) Ultrasound https://doi.org/10.1016/0022-2364(89)
and magnetic resonance microimaging of 90219-9
mouse development. Methods Enzymol 11. Wu D, Reisinger D, Xu J, Fatemi SA, van Zijl
476:379–400. https://doi.org/10.1016/ PC, Mori S, Zhang J (2014) Localized diffu-
S0076-6879(10)76021-3 sion magnetic resonance micro-imaging of the
5. Deans AE, Wadghiri YZ, Berrios-Otero CA, live mouse brain. NeuroImage 91:12–20.
Turnbull DH (2008) Mn enhancement and https://doi.org/10.1016/j.neuroimage.
respiratory gating for in utero MRI of the 2014.01.014
embryonic mouse central nervous system. 12. Aggarwal M, Mori S, Shimogori T,
Magn Reson Med 59(6):1320–1328. https:// Blackshaw S, Zhang J (2010) Three-
doi.org/10.1002/mrm.21609 dimensional diffusion tensor microimaging
6. Wu D, Lei J, Rosenzweig JM, Burd I, Zhang J for anatomical characterization of the mouse
(2014) In utero localized diffusion MRI of the brain. Magn Reson Med 64(1):249–261.
embryonic mouse brain microstructure and https://doi.org/10.1002/mrm.22426
injury. J Magn Reson Imaging 42:717. 13. Wu D, Xu J, McMahon MT, van Zijl PC,
https://doi.org/10.1002/jmri.24828 Mori S, Northington FJ, Zhang J (2013) In
7. Berrios-Otero CA, Nieman BJ, Parasoglou P, vivo high-resolution diffusion tensor imaging
Turnbull DH (2012) In utero phenotyping of of the mouse brain. NeuroImage 83:18–26.
296 Jiangyang Zhang et al.
https://doi.org/10.1016/j.neuroimage. https://doi.org/10.1007/978-1-61737-992-
2013.06.012 5_6
14. Mao J, Mareci TH, Andrew ER (1988) 16. Jiang H, van Zijl PC, Kim J, Pearlson GD,
Experimental-study of optimal selective Mori S (2006) DtiStudio: resource program
180-degrees radiofrequency pulses. J Magn for diffusion tensor computation and fiber
Reson 79(1):1–10. https://doi.org/10. bundle tracking. Comput Methods Prog
1016/0022-2364(88)90317-4 Biomed 81(2):106–116. https://doi.org/10.
15. Jones DK, Leemans A (2011) Diffusion tensor 1016/j.cmpb.2005.08.004
imaging. Methods Mol Biol 711:127–144.
Chapter 18
Abstract
Oxygen monitoring is a topic of exhaustive research due to its central role in many biological processes,
from energy metabolism to gene regulation. The ability to monitor in vivo the physiological distribution
and the dynamics of oxygen from subcellular to macroscopic levels is a prerequisite to better understand the
mechanisms associated with both normal and disease states (cancer, neurodegeneration, stroke, etc.). This
chapter focuses on magnetic resonance imaging (MRI) based techniques to assess oxygenation in vivo. The
first methodology uses injected fluorinated agents to provide quantitative pO2 measurements with high
precision and suitable spatial and temporal resolution for many applications. The second method exploits
changes in endogenous contrasts, i.e., deoxyhemoglobin and oxygen molecules through measurements of
T2* and T1, in response to an intervention to qualitatively evaluate hypoxia and its potential modulation.
1 Introduction
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0_18, © Springer Science+Business Media, LLC 2018
297
298 Heling Zhou et al.
been favored, because they simplify the imaging and the processing
of the pO2 maps and tend to provide higher signal-to-noise ratio
[38]. The PFC administration protocol is an important consider-
ation. In this chapter, we will describe the direct administration of
hexafluorobenzene (HFB) at the site of measurement. Neverthe-
less, alternative methods are also available based on the particular
needs of the experiment [6].
We may note that19F MRI remains relatively esoteric on human
clinical systems and thus an analogous proton MRI approach was
proposed. Hexamethyldisiloxane (HMDSO) may serve as a
novel1H NMR oximetry probe analogous to PFC [39]. It shares
some important properties with PFCs making a highly suitable
oximetry probe [40]. Its development would avoid the limitation
of special hardware and sequences needed for19F MRI allowing,
therefore, a straightforward implementation in clinical scanner.
This would make possible the combination of oximetry informa-
tion with that derived from other routinely used1H MR-based
methods, such as dynamic contrast enhancement, diffusion mea-
surements, and MR spectroscopy. We do not describe this method-
ology here, but it may be implemented using a similar protocol,
with the addition of water suppression. Interested readers can go to
[39–41].
There are alternative methods to measure oxygenation using
endogenous reporters, based on the magnetic nature of the oxygen
molecule (O2) or its main blood transporter, hemoglobin. Blood
Oxygen Level Dependent (BOLD) contrast relies on the fact that
deoxyhemoglobin (dHb) is paramagnetic, whereas its oxygenated
counterpart, oxyhemoglobin (HbO2), is diamagnetic. Briefly,
deoxygenated red blood cells generate local magnetic field gradi-
ents causing signal dephasing and thus signal loss in T2*-weighted
images [42, 43]. This approach was originally developed to investi-
gate cerebral activity (fMRI), being extended more recently to the
assessment of tumor oxygenation in response to hyperoxic gas
challenge [21, 44]. The magnitude of this effect depends on multi-
ple factors (see Note 1), preventing a simple and direct relationship
between T2* changes and pO2 [45]. These circumstances
prompted the introduction of the acronym “FLOOD” (flow and
oxygenation dependent) to refer to the BOLD experiment, as
applied to the study of oxygenation in tumors [46]. Nontheless
several reports have indicated a strong correlation between changes
in R2* (or T2*-weighted signal) and pO2 [47–50].
More recently, T1 changes in response to increased oxygen
content in the gas breathed have been demonstrated in both
tumor [50–52] and normal tissues [53] providing the so-called
Tissue Oxygen Level Dependent (TOLD) approach. This tech-
nique generally does not measure absolute pO2 directly, except in
rare circumstances such as vitreous humor or CSF [45, 54], since
many factors can influence R1. However, it has been applid to assess
300 Heling Zhou et al.
2 Materials
2.2 MRI System For pO2 mapping with PFC, the MRI system must provide dual
nuclei capability or a broad frequency response.
2.2.1 Magnetic
Resonance Imaging
System
2.2.2 MRI Coil 1. The expected changes for T1- and T2*-weighted images are
small, so it is critical to achieve excellent signal-to-noise ratio
(SNR) and field homogeneity. We used a1H selective birdcage
resonator of 38 mm and a gradient system with 360 mT/m of
maximum intensity.
2. For pO2 mapping with PFC a tunable coil to1H or19F is needed
(see Note 2). We use a homebuilt 2 or 3.5 cm single-turn
solenoid volume coil.
2.2.3 Sequences 1. Anatomical images: Used for localization of the tumor and to
acquire representative images to overly the different acquired
maps (see Note 3).
2. DOCENT: Spoiled Gradient Echo images are needed. Sug-
gested parameters: (1) T1 weighted, repetition time
MR Oximetry 301
(TR) ¼ 30 ms, echo time (TE) ¼ 5 ms, flip angle (FA) ¼ 45 ;
and (2) T2* weighted, TR ¼ 150 ms, TE ¼ 20 ms and FA ¼ 20
(see Note 4).
3. pO2 mapping: T1 map is acquired applying the FREDOM
sequence, which uses Pulse Burst Saturation Recovery
(PBSR) echo planar imaging (EPI), by arraying 14 delay
times (τ) [38]. The FREDOM parameters are: TR ¼ 50 ms,
TE ¼ 21 ms, τ ranges from ¼ 0.2 to 90 s, number of excitations
(NEX) ¼ 1–12 (depending on τ), field of view
(FOV) ¼ 40 40 mm with 32 32 acquisition matrix, slice
thickness 10 mm, giving a total acquisition time of 6½ min.
T1 map can also be performed by using a standard inversion
recovery sequence, but acquisition time will be significantly
increased, or Modified Look Locker sequence [36].
2.2.5 Heating System It is important to keep the animal temperature in the physiological
range (see Notes 5 and 6). The two common methodologies to
achieve this are blowing warm air over the subject or placing the
subject on a bed of circulating water connected to a water bath
controlled by a temperature regulatory system (see Note 7). If the
heating system only warms a small surface of the animal (i.e.,
waterbed), wrapping the animal to minimize heat loss would be
very convenient.
2.3 Software for Data Processing methods require software capable of handling MR
Post-processing images. For DOCENT analysis, DCE-like software is sufficient. In
the case of pO2 mapping, software for fitting T1 curves is needed.
There are many good free software options for such applications as
ImageJ (NIH, USA). Other choices include MATLAB (The Math-
Works Inc., Natick, MA, USA), IDL (Exelis Visual Information
Solutions, Boulder, CO, USA), or standard graphing packages such
as Sigmaplot (Systat Software, San Jose, CA, USA).
302 Heling Zhou et al.
3 Methods
3.1 Animal Handling All animal work should be carried out only upon review and
and Anesthesia approval of the methods by your institution’s Animal Care and
Use Committee [56, 57]. For those new to MRI, prior to initiating
any studies, training and advice should be sought from experts in
the field.
Anesthetized subject must be maintained in as natural as possi-
ble conditions because tissue oxygenation will be dependent on
many physiological parameters such as inspired O2, blood flow,
vascular reactivity, and heart rate [58]. Thus, proper monitoring
on the subject is crucial to obtain consistent results.
Anesthesia plays a central role in study design. There are several
anesthetics that can be used in these experiments, but it is impor-
tant to bear in mind that all of them have certain impact in the
oxygenation and in blood flow [59, 60]. The final selection will
depend on multiple factors such as the species (rat [61] vs. mouse
[62]), the duration of the experiment [63], etc. As a general rule,
injectable anesthetics provide a stable imaging condition for up to
2 h, whereas inhaled anesthesia provides stable anesthesia level over
a prolonged period [64] and can be adjusted during the course of
the experiment, but also requires additional equipment. With
injectable anesthetics, ketamine-xylazine tends to result in lower
tissue pO2 values [65], whereas propofol and remifentanil have
been reported to have limited effects on perfusion and oxygenation
[66]. Urethane has been reported to render a stronger BOLD
signal [67], but it can be used only in terminal experiments because
it is carcinogenic and compulsory euthanization must be performed
at the end of the experiment.
3.2 DOCENT 1. Place the animal in an anesthetic induction chamber with 3–5%
isoflurane in medical air (1 L/min) and wait until the animal is
sedated. If an injectable anesthetic is used, inject it and wait
until absence of withdrawal reflexes (toe and ear pinch). Addi-
tional doses can be injected depending on the specific protocol
(see Note 8).
2. Transfer the animal to the MRI bed. Check correct positioning
of the animal so the heating system can maintain physiological
temperature.
3. Check that the correct stage of anesthesia has been maintained.
If not, increase the level of anesthetic or inject additional doses.
Maintain anesthesia with isoflurane at 1.5–2%.
4. Insert the rectal temperature probe using lubricating jelly and
tape in place.
5. Place the physiological monitoring device (i.e., MRI compati-
ble sensor with foot clip) or the breathing sensor.
MR Oximetry 303
Fig. 1 Scheme of gas breathing sequence and images acquisition in each experimental condition.
304 Heling Zhou et al.
3.3 PFC pO2 Mapping 1. Inject PFC (see Note 9) directly into the tumor with a fine
sharp needle (32G). In order to minimize tissue damage, we
recommend the use of a Hamilton syringe (typically, 100 μL
capacity). To adequately sample the whole tumor’s pO2, inject
the PFC in a fan pattern in a single plane, as described in
[38]. Ideally, the PFC should be saturated with N2 (by gentle
bubbling in the bottle for a few minutes) before injection into
tissue to minimize perturbation of measured pO2 levels.
2. Transfer the animal to the MRI bed and place the physiological
monitoring device as described in steps 2–8 of the DOCENT
protocol.
3. Place the animal in a dual1H/19F coil.
4. Tune the coil to the1H frequency and perform shimming.
5. Acquire anatomical images in the three orthogonal planes.
6. Using anatomical images acquire in step 5 to plan your slice
selection. Slice positioning and geometry must be maintained
through all the subsequent experiments.
7. It is recommended to acquire an anatomical image with the
same geometry than the T119F mapping images. Whether this
is not necessary, it will provide anatomical information helping
in the identification of the areas where we are measuring pO2.
8. Tune the coil to19F frequency and perform further shimming
on the19F signal if needed.
9. Acquire T119F maps to quantify the spin-lattice relaxation rate
of the injected PFC. Obtain a series (3–5) of pO2 measure-
ments for each condition studied. Usually, we acquire five
baseline points (i.e., air breathing), five during the oxygen
challenge (i.e., breathing 100% O2 or carbogen) and five points
during return to baseline conditions or after performing any
other intervention (i.e., administration of some drug).
3.4 Data Analysis For the DOCENT experiment, data analysis can be performed on a
region of interest (ROI) or on a voxel-by-voxel basis. Briefly, signal
intensity (SI) in the baseline (air breathing) T1 and T2* images are
averaged to obtain mean baseline images. Normalized image SI
values are then calculated by following the expression:
SIi, m, n
ΔSI ð%Þi, m, n ¼ P ∗100 100 ð1Þ
5
i¼1 SIi, m, n =5
where i is the image number, m and n are the image matrix, and SI
is the voxel intensity. Similar approach can be used if a ROI analysis
is applied.
If R1 and R2* maps are acquired, R1 values for a given voxel are
obtained by fitting the signal intensities corresponding to different
TRs to a monoexponential function, using a Levenberg-Marquardt
MR Oximetry 305
where R1 is the relaxation rate for that particular voxel, R1a is the
anoxic relaxation rate, R1p is the relaxation rate due to the para-
magnetic contribution of oxygen and K is the Henry’s constant.
These parameters must be known or measured for each PFC (each
resonance), temperature and magnetic field. A detailed list of these
parameters can be found in [38]. Figure 3 shows an example of the
results obtained in a dynamic oximetry experiment performed with
HFB in a mouse bearing an orthotopic MDA-MB-231/luc human
breast tumor xenograft implanted in the left upper mammary fat
pad. Figure 4 shows an example of the pO2 maps (using HFB) and
DOCENT results obtained in a nude rat bearing a human lung
tumor line A549 implanted subcutaneously in the thigh.
4 Notes
Fig. 2 DOCENT oximetry. Changes in T1- and T2*-weighted image SI in response to transition from air to oxygen breathing in both healthy brain tissue (C,
contralateral) and high grade gliomas (T, tumor) induced in Wistar rats by stereotaxic injection of C6 cells in the right caudate nucleus. (A, B) T2*-weighted gradient
echo cross-sectional images of the tumor and normal tissue acquired while breathing (A) air (baseline) and (B) oxygen (image selected to show maximum change).
Maps are overlaid on anatomical T2-weighted images. (C) Mean BOLD (normalized SI) response in three adjacent image slices (1–3) for tumors (T SI1, T SI2, T SI3)
and contralateral hemisphere (C SI1, C SI2, C SI3), and mean value over whole area of interest (T Mean for tumor and C Mean for healthy brain tissue). (D, E) T1-
weighted gradient echo images, acquired while breathing (D) air and (E) oxygen. (F) Mean variations across areas of interest of normalized SI change versus time
(TOLD response). Heat scale bar shows percentage change.
Fig. 3 Dynamic oximetry using FREDOM. FREDOM of a SCID mouse bearing an orthotopic MDA-MB-231/luc human breast tumor xenograft implanted in the left
upper mammary fat pad. HFB (50 μL) was injected directly into the tumor in a fan pattern to ensure distribution to various locations and provide pO2 maps
representative of whole tumor. MR was performed using a 4.7 T small animal scanner. Three pO2 measurements were obtained for baseline air, and four with
oxygen breathing challenge. An indole-based vascular disrupting agent (VDA), OXi8007 was administered IP in situ (350 mg/kg) and 18 more pO2 measurements
were obtained over 2 h. In total 25 FREDOM maps were acquired over 3 h. (A) T1 curve fitting for the whole tumor at three representative points: during air
MR Oximetry
breathing, oxygen breathing, and 2 h after administration of VDA while continuing breathing oxygen. (B) Dynamic changes of the mean pO2 values in response to
oxygen breathing challenge and VDA administration. (C) pO2 maps of the tumor during air, oxygen breathing and 2 h after administration of VDA while continuing
breathing oxygen. (D) Histogram showing pO2 distributions at the three stages (OXi8007 kindly provided by Dr. Kevin Pinney, Baylor University, and a
307
Fig. 4 19F and1H MRI oximetry of subcutaneous A549 human tumor xenograft in a nude rat. FREDOM was used
to acquire pO2 maps and their corresponding histograms, while the rat breathed air (A, E) and oxygen (B, F).
TOLD effect is reflected in changes in signal intensity in T1-weighted images while the rat breathed air (C) and
oxygen (D). TOLD approach reflects tissue oxygenation while BOLD reflects blood oxygenation through mean
changes in signal intensity (G). If FREDOM and DOCENT are porfermed in the same animal, it is crucial to perform
DOCENT before PFC administration to avoid interferences in T- and T-weighted images
Acknowledgements
References
1. Lassmann H (2016) Demyelination and neu- of hypoxia. Ann Neurol 79(4):520–521.
rodegeneration in multiple sclerosis: The role https://doi.org/10.1002/ana.24632
310 Heling Zhou et al.
2. Peers C, Dallas ML, Boycott HE, Scragg JL, importance in tumor biology, noninvasive mea-
Pearson HA, Boyle JP (2009) Hypoxia and surement by imaging, and value of its measure-
neurodegeneration. Ann N Y Acad Sci ment in the management of cancer therapy. Int
1177:169–177. https://doi.org/10.1111/j. J Radiat Biol 82(10):699–757. https://doi.
1749-6632.2009.05026.x org/10.1080/09553000601002324
3. Figueroa XA, Wright JK (2016) Hyperbaric 13. Overgaard J, Horsman MR (1996) Modifica-
oxygen: B-level evidence in mild traumatic tion of hypoxia-induced radioresistance in
brain injury clinical trials. Neurology 87 tumors by the use of oxygen and sensitizers.
(13):1400–1406. https://doi.org/10.1212/ Semin Radiat Oncol 6:10–21
WNL.0000000000003146 14. Matsuo M, Matsumoto S, Mitchell JB, Krishna
4. Kones R (2011) Oxygen therapy for acute MC, Camphausen K (2014) Magnetic reso-
myocardial infarction-then and now. A century nance imaging of the tumor microenvironment
of uncertainty. Am J Med 124 in radiotherapy: perfusion, hypoxia, and
(11):1000–1005. https://doi.org/10.1016/j. metabolism. Semin Radiat Oncol 24
amjmed.2011.04.034 (3):210–217. https://doi.org/10.1016/j.
5. Rink C, Khanna S (2011) Significance of brain semradonc.2014.02.002
tissue oxygenation and the arachidonic acid 15. Nordsmark M, Bentzen SM, Rudat V,
cascade in stroke. Antioxid Redox Signal 14 Brizel D, Lartigau E, Stadler P, Becker A,
(10):1889–1903. https://doi.org/10.1089/ Adam M, Molls M, Dunst J, Terris DJ, Over-
ars.2010.3474 gaard J (2005) Prognostic value of tumor oxy-
6. Pacheco-Torres J, Lopez-Larrubia P, genation in 397 head and neck tumors after
Ballesteros P, Cerdan S (2011) Imaging primary radiation therapy. An international
tumor hypoxia by magnetic resonance meth- multi-center study. Radiother Oncol 77
ods. NMR Biomed 24(1):1–16. https://doi. (1):18–24
org/10.1002/nbm.1558 16. Vaupel P, Höckel M, Mayer A (2007) Detec-
7. Mason RP, Zhao D, Pacheco-Torres J, Cui W, tion and characterization of tumor hypoxia
Kodibagkar VD, Gulaka PK, Hao G, Thorpe P, using pO2 histography. Antioxid Redox Signal
Hahn EW, Peschke P (2010) Multimodality 9(8):1221–1236. https://doi.org/10.1089/
imaging of hypoxia in preclinical settings. Q J ars.2007.1628
Nucl Med Mol Imaging 54(3):259–280 17. Griffiths JR, Robinson SP (1999) The OxyLite:
8. Gray LH, Conger AD, Ebert M, Hornsey S, a fibre-optic oxygen sensor. Br J Radiol 72
Scott OC (1953) The concentration of oxygen (859):627–630
dissolved in tissues at the time of irradiation as a 18. Peeters SG, Zegers CM, Yaromina A, Van
factor in radiotherapy. Br J Radiol 26 Elmpt W, Dubois L, Lambin P (2015) Current
(312):638–648 preclinical and clinical applications of hypoxia
9. Overgaard J (2007) Hypoxic radiosensitiza- PET imaging using 2-nitroimidazoles. Q J
tion: adored and ignored. J Clin Oncol 25 Nucl Med Mol Imaging 59(1):39–57
(26):4066–4074. https://doi.org/10.1200/ 19. Peeters SG, Zegers CM, Lieuwes NG, van
JCO.2007.12.7878 Elmpt W, Eriksson J, van Dongen GA,
10. Tredan O, Grantab R, Dumontet C (2008) Dubois L, Lambin P (2015) A comparative
Can tumor hypoxia be turned into a chemo- study of the hypoxia PET tracers [(1)(8)F]
therapeutic advantage? Bull Cancer 95 HX4, [(1)(8)F]FAZA, and [(1)(8)F]FMISO
(5):528–534. https://doi.org/10.1684/bdc. in a preclinical tumor model. Int J Radiat
2008.0637 Oncol Biol Phys 91(2):351–359. https://doi.
11. Hay MP, Hicks KO, Wang J (2014) Hypoxia- org/10.1016/j.ijrobp.2014.09.045
directed drug strategies to target the tumor 20. Krohn KA, Link JM, Mason RP (2008) Molec-
microenvironment. Adv Exp Med Biol ular imaging of hypoxia. J Nucl Med 49(Suppl
772:111–145. https://doi.org/10.1007/ 2):129S–148S. https://doi.org/10.2967/
978-1-4614-5915-6_6 jnumed.107.045914
12. Tatum JL, Kelloff GJ, Gillies RJ, Arbeit JM, 21. Padhani AR, Krohn KA, Lewis JS, Alber M
Brown JM, Chao KS, Chapman JD, Eckelman (2007) Imaging oxygenation of human
WC, Fyles AW, Giaccia AJ, Hill RP, Koch CJ, tumours. Eur Radiol 17(4):861–872
Krishna MC, Krohn KA, Lewis JS, Mason RP, 22. Foo SS, Abbott DF, Lawrentschuk N, Scott
Melillo G, Padhani AR, Powis G, Rajendran AM (2004) Functional imaging of intratu-
JG, Reba R, Robinson SP, Semenza GL, Swartz moral hypoxia. Mol Imaging Biol 6
HM, Vaupel P, Yang D, Croft B, Hoffman J, (5):291–305
Liu G, Stone H, Sullivan D (2006) Hypoxia:
MR Oximetry 311
23. Ljungkvist AS, Bussink J, Kaanders JH, van der Blood Substit Immobil Biotechnol 22
Kogel AJ (2007) Dynamics of tumor hypoxia (4):1141–1153
measured with bioreductive hypoxic cell mar- 35. Sotak CH, Hees PS, Huang HN, Hung MH,
kers. Radiat Res 167(2):127–145 Krespan CG, Raynolds S (1993) A new per-
24. Vikram DS, Zweier JL, Kuppusamy P (2007) fluorocarbon for use in fluorine-19 magnetic
Methods for noninvasive imaging of tissue hyp- resonance imaging and spectroscopy. Magn
oxia. Antioxid Redox Signal 9(10):1745–1756 Reson Med 29(2):188–195
25. Liu H, Gu Y, Kim JG, Mason RP (2004) Near 36. Jordan BF, Cron GO, Gallez B (2009) Rapid
infrared spectroscopy and imaging of tumor monitoring of oxygenation by 19F magnetic
vascular oxygenation. Methods Enzymol resonance imaging: Simultaneous comparison
386:349–378 with fluorescence quenching. Magn Reson
26. Roussakis E, Li Z, Nichols AJ, Evans CL Med 61(3):634–638. https://doi.org/10.
(2015) Oxygen-sensing methods in biomedi- 1002/mrm.21594
cine from the macroscale to the microscale. 37. Liu S, Shah SJ, Wilmes LJ, Feiner J, Kodibag-
Angew Chem 54(29):8340–8362. https:// kar VD, Wendland MF, Mason RP, Hylton N,
doi.org/10.1002/anie.201410646 Hopf HW, Rollins MD (2011) Quantitative
27. Mason RP (2017) Oxygen breathing chal- tissue oxygen measurement in multiple organs
lenge—the simplest theranostics. Theranostics using 19F MRI in a rat model. Magn Reson
7(16):3873–3875. https://doi.org/10.7150/ Med 66(6):1722–1730. https://doi.org/10.
thno.20655 1002/mrm.22968
28. Charnley N, Donaldson S, Price P (2009) 38. Zhao D, Jiang L, Mason RP (2004) Measuring
Imaging angiogenesis. Methods Mol Biol changes in tumor oxygenation. Methods Enzy-
467:25–51 mol 386:378–418
29. Jordan BF, Runquist M, Raghunand N, 39. Kodibagkar VD, Cui W, Merritt ME, Mason
Baker A, Williams R, Kirkpatrick L, Powis G, RP (2006) Novel 1H NMR approach to quan-
Gillies RJ (2005) Dynamic contrast-enhanced titative tissue oximetry using hexamethyldisi-
and diffusion MRI show rapid and dramatic loxane. Magn Reson Med 55(4):743–748
changes in tumor microenvironment in 40. Kodibagkar VD, Wang X, Pacheco-Torres J,
response to inhibition of HIF-1alpha using Gulaka P, Mason RP (2008) Proton imaging
PX-478. Neoplasia 7(5):475–485 of siloxanes to map tissue oxygenation levels
30. Shukla HP, Mason RP, Woessner DE, Antich (PISTOL): a tool for quantitative tissue oxim-
PP (1995) A comparison of three commercial etry. NMR Biomed 21(8):899–907
perfluorocarbon emulsions as high-field 19F 41. Gulaka PK, Rastogi U, McKay MA, Wang X,
NMR probes of oxygen tension and tempera- Mason RP, Kodibagkar VD (2011)
ture. J Magn Reson B 106(2):131–141. Hexamethyldisiloxane-based nanoprobes for
https://doi.org/10.1006/jmrb.1995.1023 (1) H MRI oximetry. NMR Biomed 24
31. Ali Hamza MH, Serratrice G, Stébé M-J, Del- (10):1226–1234. https://doi.org/10.1002/
puech J-J (1981) Fluorocarbons as oxygen car- nbm.1678
riers. II. An NMR study of partially or totally 42. Greve JM (2011) The BOLD effect. Methods
fluorinated alkanes and alkenes. J Magn Reson Mol Biol 771:153–169. https://doi.org/10.
(1969) 42(2):227–241. https://doi.org/10. 1007/978-1-61779-219-9_8
1016/0022-2364(81)90214-6 43. Nield LE, Qi X, Yoo SJ, Valsangiacomo ER,
32. Thomas SR, Millard RW, Pratt RG, Shiferaw Y, Hornberger LK, Wright GA (2002)
Samaratunga RC (1994) Quantitative pO2 MRI-based blood oxygen saturation measure-
imaging in vivo with perfluorocarbon F-19 ments in infants and children with congenital
NMR: tracking oxygen from the airway heart disease. Pediatr Radiol 32(7):518–522.
through the blood to organ tissues. Artif Cells https://doi.org/10.1007/s00247-001-0652-
Blood Substit Immobil Biotechnol 22 9
(4):1029–1042 44. Ndubuizu O, La Manna JC (2007) Brain tissue
33. Mason RP, Shukla H, Antich PP (1993) In vivo oxygen concentration measurements. Antioxid
oxygen tension and temperature: simultaneous Redox Signal 9(8):1207–1219
determination using 19F NMR spectroscopy of 45. Dunn JF (2011) MR oximetry. Methods Mol
perfluorocarbon. Magn Reson Med Biol 771:227–240. https://doi.org/10.1007/
29:296–302 978-1-61779-219-9_12
34. Mason RP (1994) Non-invasive physiology: 46. Howe FA, Robinson SP, McIntyre DJ,
19F NMR of perfluorocarbons. Artif Cells Stubbs M, Griffiths JR (2001) Issues in flow
and oxygenation dependent contrast
312 Heling Zhou et al.
(FLOOD) imaging of tumours. NMR Biomed models. Cancer Res 76(4):787–795. https://
14(7–8):497–506 doi.org/10.1158/0008-5472.CAN-15-2062
47. Baudelet C, Gallez B (2005) Current issues in 56. Tennant DA, Duran RV, Gottlieb E (2010)
the utility of blood oxygen level dependent Targeting metabolic transformation for cancer
MRI for the assessment of modulations in therapy. Nat Rev Cancer 10(4):267–277.
tumor oxygenation. Curr Med Imaging Rev 1 https://doi.org/10.1038/nrc2817
(3):229–243. https://doi.org/10.2174/ 57. European Convention for the Protection of
157340505774574754 vertebrate animals used for experimental and
48. Al-Hallaq HA, River JN, Zamora M, other scientific purposes (2006) Appendix
Oikawa H, Karczmar GS (1998) Correlation A. Guidelines for accommodation and care of
of magnetic resonance and oxygen microelec- animals (Article 5 of the Convention)
trode measurements of carbogen-induced 58. Sicard KM, Duong TQ (2005) Effects of hyp-
changes in tumor oxygenation. Int J Radiat oxia, hyperoxia, and hypercapnia on baseline
Oncol Biol Phys 41(1):151–159 and stimulus-evoked BOLD, CBF, and
49. Zhao D, Jiang L, Hahn EW, Mason RP (2009) CMRO2 in spontaneously breathing animals.
Comparison of1H blood oxygen level- NeuroImage 25(3):850–858. https://doi.
dependent (BOLD) and19F MRI to investigate org/10.1016/j.neuroimage.2004.12.010
tumor oxygenation. Magn Reson Med 62 59. Masamoto K, Kanno I (2012) Anesthesia and
(2):357–364 the quantitative evaluation of neurovascular
50. Hallac RR, Zhou H, Pidikiti R, Song K, coupling. J Cereb Blood Flow Metab 32
Stojadinovic S, Zhao D, Solberg T, Peschke P, (7):1233–1247. https://doi.org/10.1038/
Mason RP (2014) Correlations of noninvasive jcbfm.2012.50
BOLD and TOLD MRI with pO2 and rele- 60. Duong TQ (2007) Cerebral blood flow and
vance to tumor radiation response. Magn BOLD fMRI responses to hypoxia in awake
Reson Med 71(5):1863–1873. https://doi. and anesthetized rats. Brain Res 1135
org/10.1002/mrm.24846 (1):186–194. https://doi.org/10.1016/j.
51. Matsumoto K, Bernardo M, Subramanian S, brainres.2006.11.097
Choyke P, Mitchell JB, Krishna MC, Lizak MJ 61. Hendrich KS, Kochanek PM, Melick JA, Schid-
(2006) MR assessment of changes of tumor in ing JK, Statler KD, Williams DS, Marion DW,
response to hyperbaric oxygen treatment. Ho C (2001) Cerebral perfusion during anes-
Magn Reson Med 56(2):240–246. https:// thesia with fentanyl, isoflurane, or pentobarbi-
doi.org/10.1002/mrm.20961 tal in normal rats studied by arterial spin-
52. Beeman SC, Shui YB, Perez-Torres CJ, Engel- labeled MRI. Magn Reson Med 46
bach JA, Ackerman JJ, Garbow JR (2016) (1):202–206
O2-sensitive MRI distinguishes brain tumor 62. Schroeter A, Schlegel F, Seuwen A,
versus radiation necrosis in murine models. Grandjean J, Rudin M (2014) Specificity of
Magn Reson Med 75(6):2442–2447. https:// stimulus-evoked fMRI responses in the
doi.org/10.1002/mrm.25821 mouse: the influence of systemic physiological
53. O’Connor JP, Naish JH, Jackson A, Waterton changes associated with innocuous stimulation
JC, Watson Y, Cheung S, Buckley DL, under four different anesthetics. NeuroImage
McGrath DM, Buonaccorsi GA, Mills SJ, 94:372–384. https://doi.org/10.1016/j.
Roberts C, Jayson GC, Parker GJ (2009) Com- neuroimage.2014.01.046
parison of normal tissue R1 and R*2 modula- 63. Sonnay S, Just N, Duarte JM, Gruetter R
tion by oxygen and carbogen. Magn Reson (2015) Imaging of prolonged BOLD response
Med 61(1):75–83 in the somatosensory cortex of the rat. NMR
54. Berkowitz BA, McDonald C, Ito Y, Tofts PS, Biomed 28(3):414–421. https://doi.org/10.
Latif Z, Gross J (2001) Measuring the human 1002/nbm.3263
retinal oxygenation response to a hyperoxic 64. Liu ZM, Schmidt KF, Sicard KM, Duong TQ
challenge using MRI: eliminating blinking arti- (2004) Imaging oxygen consumption in fore-
facts and demonstrating proof of concept. paw somatosensory stimulation in rats under
Magn Reson Med 46(2):412–416 isoflurane anesthesia. Magn Reson Med 52
55. O’Connor JP, Boult JK, Jamin Y, Babur M, (2):277–285. https://doi.org/10.1002/
Finegan KG, Williams KJ, Little RA, mrm.20148
Jackson A, Parker GJ, Reynolds AR, Waterton 65. Lei H, Grinberg O, Nwaigwe CI, Hou HG,
JC, Robinson SP (2016) Oxygen-enhanced Williams H, Swartz HM, Dunn JF (2001) The
MRI accurately identifies, quantifies, and effects of ketamine-xylazine anesthesia on cere-
maps tumor hypoxia in preclinical cancer bral blood flow and oxygenation observed
MR Oximetry 313
using nuclear magnetic resonance perfusion 69. Zhang N, Yacoub E, Zhu XH, Ugurbil K,
imaging and electron paramagnetic resonance Chen W (2009) Linearity of blood-
oximetry. Brain Res 913(2):174–179 oxygenation-level dependent signal at micro-
66. Mikkelsen ML, Ambrus R, Miles JE, Poulsen vasculature. NeuroImage 48(2):313–318.
HH, Moltke FB, Eriksen T (2016) Effect of https://doi.org/10.1016/j.neuroimage.
propofol and remifentanil on cerebral perfu- 2009.06.071
sion and oxygenation in pigs: a systematic 70. Dunn JF, Swartz HM (1997) Blood oxygena-
review. Acta Vet Scand 58(1):42. https://doi. tion. Heterogeneity of hypoxic tissues moni-
org/10.1186/s13028-016-0223-6 tored using bold MR imaging. Adv Exp Med
67. Paasonen J, Salo RA, Shatillo A, Forsberg MM, Biol 428:645–650
Narvainen J, Huttunen JK, Grohn O (2016) 71. Silvennoinen MJ, Clingman CS, Golay X,
Comparison of seven different anesthesia pro- Kauppinen RA, van Zijl PC (2003) Compari-
tocols for nicotine pharmacologic magnetic son of the dependence of blood R2 and R2* on
resonance imaging in rat. Eur Neuropsycho- oxygen saturation at 1.5 and 4.7 Tesla. Magn
pharmacol 26(3):518–531. https://doi.org/ Reson Med 49(1):47–60. https://doi.org/10.
10.1016/j.euroneuro.2015.12.034 1002/mrm.10355
68. Zhao D, Pacheco-Torres J, Hallac RR, 72. Zhou H, Hallac RR, Lopez R, Denney R, Mac-
White D, Peschke P, Cerdan S, Mason RP Donough MT, Li L, Liu L, Graves EE, Trawick
(2015) Dynamic oxygen challenge evaluated ML, Pinney KG, Mason RP (2015) Evaluation
by NMR T1 and T2*—insights into tumor of tumor ischemia in response to an indole-
oxygenation. NMR Biomed 28(8):937–947. based vascular disrupting agent using BLI and
https://doi.org/10.1002/nbm.3325 (19)F MRI. Am J Nucl Med Mol Imaging 5
(2):143–153
Chapter 19
Abstract
Molecular magnetic resonance imaging (mMRI) enables the detection of a protein of interest in vivo, in a
noninvasive manner. The general concept of mMRI is to target a contrast agent to a protein of interest, and
to perform a contrast-sensitive MRI sequence. Typically, contrast agents are made of a “contrastophore”
(the part of the construct responsible for the contrast on the images) and a targeting moiety (“pharmaco-
phore”). Recently, the development of a new family of contrastophore carrying a high payload of iron oxide
(micro-sized particles of iron oxide, MPIO) has led to a dramatic increase in the sensitivity of mMRI. Here,
we describe the production of targeted MPIO using commercially available reagents and the MRI protocols
to allow their detection in vivo.
Key words Molecular imaging, Inflammation, Vascular-cell adhesion molecule, Intercellular adhesion
molecule, Selectins, Leucocytes, USPIO, Microparticles, Endothelial cells, Endothelium, Platelets
1 Introduction
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0_19, © Springer Science+Business Media, LLC 2018
315
316 Maxime Gauberti et al.
iron oxide) labeled with a rat monoclonal antibody against VCAM-1 (MPIO-αVCAM-1) in a mouse model of sepsis (intraperitoneal LPS injection, 1 mg/kg). MPIOs
bind to activated endothelial cells (VCAM-1 positive vessels) as presented in the immunofluorescence images of the upper panels. Lower panels: mMRI allows
detection of endothelial activation (red images), whereas no binding is detectable in healthy organs (green images)
317
318 Maxime Gauberti et al.
2 Materials
3 Methods
4. After securing the mouse and its tail, locate the tail veins (one
main vein is present on each side of the tail). Under an
operating microscope, incise the skin of the tail laterally using
a scalpel. Dissect the tail vein using micro-scissors and curved
fine tips forceps.
5. Once isolated from the adjacent tissue, the superior part of the
vein should be cut using the micro-scissors allowing insertion
of a tail vein catheter in the venous lumen (SAI Infusion
technologies, Mouse Tail Vein Catheter, MTV-01) (see Note
9) [20].
6. Success of the procedure can be ascertained by two methods:
either intravenous injection of a small amount of sterile saline
(100 μL), which should not induce any swelling of the tail, or
visualization of a blood reflow inside the catheter (that is not
constantly observed despite adequate placement of the cathe-
ter) (see Note 10).
7. Once in place, the tail vein catheter should be secured on the
tail skin using medical plaster.
8. If pre-contrast imaging is required, it should be done at
this step.
9. Slowly inject 200 μL per mouse of the MPIOs solution
through the tail vein catheter. Then, rinse the catheter using
100 μL saline.
10. A delay of 5 min between injection and imaging should be
respected to allow MPIOs binding to their target and their
clearance from the blood by the reticuloendothelial system.
The mouse is then ready for post-contrast imaging.
3.3 In Vivo mMRI 1. Place the animal in the MRI holder system in order to avoid, as
much as possible, any movement of the imaged parts. Place a
pressure captor on the animal body to monitor breath rate and
anesthesia.
2. Both surface and volume coils are suitable for mMRI applica-
tions. However, surface coils allow a better signal-to-noise ratio
at the expense of a reduced field of view. We use a Bruker
Pharmascan 7 T magnet equipped with a mouse brain
surface coil.
3. If possible, maintain anesthesia throughout MRI acquisition
using 100% O2. This reduces the deoxyhemoglobin of the veins
into oxyhemoglobin. Since oxyhemoglobin is devoid of mag-
netic susceptibility effects, this allows to specifically observe the
susceptibility effects of the injected MPIOs [21, 22].
4. Brain imaging (Fig. 3): after standard shimming and localizer
scan, to visualize MPIOs, we use a coronal 3D-GEFC (gradient
echo with flow compensation) sequence with the following
Molecular MRI 323
Fig. 3 Representative mMRI of VCAM-1 in the brain in four experimental models showing the wide diversity of
labeling pattern according to the inflammatory stimulus
3.4 Visualization 1. Perfuse transcardially deeply anesthetized mice with cold hepa-
of MPIOs Using rinized saline (15 mL) followed by 150 mL of fixative (PBS
Microscopy 0.1 M. pH 7.4 containing 2% paraformaldehyde and 0.2%
picric acid or alternatively 4% paraformaldehyde without picric
acid) [23] using a peristaltic tubing pump.
2. (Facultative) Brain, kidney, or heart can be post-fixed overnight
in the same fixative solution (paraformaldehyde with or with-
out picric acid) at 4 C.
3. The collected organs should be cryoprotected using 20%
sucrose in PBS buffer during 24 h at 4 C before freezing in
Tissue-Tek (Miles Scientific, Naperville, IL, USA).
4. Using a cryomicrotome, cut the organs into 8–10 μm slices and
collect them on poly-lysine slides. If required, the slides can be
stored at 80 C until further use.
5. To allow direct white-light detection of MPIOs, washed sec-
tions can be directly coverslipped with antifade medium and
observed using a microscope. In that case, MPIOs appear as
~1 μm black and white spheres (usually a white dot surrounded
by a black ring, Fig. 4) [16].
6. To allow detection of MPIO using fluorescence microscopy,
co-incubate the sections on the slides with a fluorescently
labeled anti-pharmacophore antibody (usually, a secondary
anti-IgG antibody) for at least 90 min at ambient temperature
(see Note 11). In that case, MPIOs appear as ~1 μm fluorescent
spheres (Fig. 1) [24].
4 Notes
References
1. Laurent S, Forge D, Port M, Roch A, Robic C, iron oxide particles. Magn Reson Med 53
Vander Elst L, Muller RN (2008) Magnetic (2):329–338. https://doi.org/10.1002/
iron oxide nanoparticles: synthesis, stabiliza- mrm.20342
tion, vectorization, physicochemical character- 10. Montagne A, Gauberti M, Macrez R,
izations, and biological applications. Chem Rev Jullienne A, Briens A, Raynaud JS, Louin G,
108(6):2064–2110. https://doi.org/10. Buisson A, Haelewyn B, Docagne F, Defer G,
1021/cr068445e Vivien D, Maubert E (2012) Ultra-sensitive
2. Mahmoudi M, Sheibani S, Milani AS, molecular MRI of cerebrovascular cell activa-
Rezaee F, Gauberti M, Dinarvand R, Vali H tion enables early detection of chronic central
(2015) Crucial role of the protein corona for nervous system disorders. NeuroImage 63
the specific targeting of nanoparticles. Nano- (2):760–770. https://doi.org/10.1016/j.
medicine (Lond) 10(2):215–226. https://doi. neuroimage.2012.07.018
org/10.2217/nnm.14.69 11. Deddens LH, van Tilborg GA, van der
3. Le Behot A, Gauberti M, Martinez De Toorn A, van der Marel K, Paulis LE, van
Lizarrondo S, Montagne A, Lemarchand E, Bloois L, Storm G, Strijkers GJ, Mulder WJ,
Repesse Y, Guillou S, Denis CV, Maubert E, de Vries HE, Dijkhuizen RM (2013) MRI of
Orset C, Vivien D (2014) GpIbalpha-VWF ICAM-1 upregulation after stroke: the impor-
blockade restores vessel patency by dissolving tance of choosing the appropriate target-
platelet aggregates formed under very high specific particulate contrast agent. Mol Imag-
shear rate in mice. Blood 123 ing Biol 15(4):411–422. https://doi.org/10.
(21):3354–3363. https://doi.org/10.1182/ 1007/s11307-013-0617-z
blood-2013-12-543074 12. Blezer EL, Deddens LH, Kooij G, Drexhage J,
4. Gauberti M, Montagne A, Quenault A, Vivien van der Pol SM, Reijerkerk A, Dijkhuizen RM,
D (2014) Molecular magnetic resonance imag- de Vries HE (2015) In vivo MR imaging of
ing of brain-immune interactions. Front Cell intercellular adhesion molecule-1 expression
Neurosci 8:389. https://doi.org/10.3389/ in an animal model of multiple sclerosis. Con-
fncel.2014.00389 trast Media Mol Imaging 10(2):111–121.
5. Vivien D, Gauberti M, Montagne A, Defer G, https://doi.org/10.1002/cmmi.1602
Touze E (2011) Impact of tissue plasminogen 13. Melemenidis S, Jefferson A, Ruparelia N, Akh-
activator on the neurovascular unit: from clini- tar AM, Xie J, Allen D, Hamilton A, Larkin JR,
cal data to experimental evidence. J Cereb Perez-Balderas F, Smart SC, Muschel RJ,
Blood Flow Metab 31(11):2119–2134. Chen X, Sibson NR, Choudhury RP (2015)
https://doi.org/10.1038/jcbfm.2011.127 Molecular magnetic resonance imaging of
6. Serres S, Soto MS, Hamilton A, McAteer MA, angiogenesis in vivo using polyvalent cyclic
Carbonell WS, Robson MD, Ansorge O, RGD-iron oxide microparticle conjugates.
Khrapitchev A, Bristow C, Balathasan L, Theranostics 5(5):515–529. https://doi.org/
Weissensteiner T, Anthony DC, Choudhury 10.7150/thno.10319
RP, Muschel RJ, Sibson NR (2012) Molecular 14. Belliere J, Martinez de Lizarrondo S, Choudh-
MRI enables early and sensitive detection of ury RP, Quenault A, Le Behot A, Delage C,
brain metastases. Proc Natl Acad Sci U S A Chauveau D, Schanstra JP, Bascands JL,
109(17):6674–6679. https://doi.org/10. Vivien D, Gauberti M (2015) Unmasking
1073/pnas.1117412109 Silent Endothelial Activation in the Cardiovas-
7. Iyer AK, Khaled G, Fang J, Maeda H (2006) cular System Using Molecular Magnetic Reso-
Exploiting the enhanced permeability and nance Imaging. Theranostics 5
retention effect for tumor targeting. Drug Dis- (11):1187–1202. https://doi.org/10.7150/
cov Today 11(17–18):812–818. https://doi. thno.11835
org/10.1016/j.drudis.2006.07.005 15. Jefferson A, Ruparelia N, Choudhury RP
8. McAteer MA, Sibson NR, von Zur Muhlen C, (2013) Exogenous microparticles of iron
Schneider JE, Lowe AS, Warrick N, Channon oxide bind to activated endothelial cells but,
KM, Anthony DC, Choudhury RP (2007) In unlike monocytes, do not trigger an endothe-
vivo magnetic resonance imaging of acute brain lial response. Theranostics 3(6):428–436.
inflammation using microparticles of iron https://doi.org/10.7150/thno.5895
oxide. Nat Med 13(10):1253–1258. https:// 16. Gauberti M, Montagne A, Marcos-Contreras
doi.org/10.1038/nm1631 OA, Le Behot A, Maubert E, Vivien D (2013)
9. Shapiro EM, Skrtic S, Koretsky AP (2005) Ultra-sensitive molecular MRI of vascular cell
Sizing it up: cellular MRI using micron-sized adhesion molecule-1 reveals a dynamic
Molecular MRI 327
inflammatory penumbra after strokes. Stroke JJ, Rothwell NJ, Rubio M, Waeber C, Young
44(7):1988–1996. https://doi.org/10.1161/ AR, Touze E, Vivien D (2016) Efficacy of
strokeaha.111.000544 Alteplase in a Mouse Model of Acute Ischemic
17. Marcos-Contreras OA, Martinez de Stroke: A Retrospective Pooled Analysis.
Lizarrondo S, Bardou I, Orset C, Pruvost M, Stroke 47(5):1312–1318. https://doi.org/
Anfray A, Frigout Y, Hommet Y, Lebouvier L, 10.1161/strokeaha.116.012238
Montaner J, Vivien D, Gauberti M (2016) 21. Gaberel T, Gakuba C, Hebert M, Montagne A,
Hyperfibrinolysis increases blood-brain barrier Agin V, Rubio M, Emery E, Vivien D, Gauberti
permeability by a plasmin- and bradykinin- M (2013) Intracerebral hematomas disappear
dependent mechanism. Blood 128 on T2*-weighted images during normobaric
(20):2423–2434. https://doi.org/10.1182/ oxygen therapy. Stroke 44(12):3482–3489.
blood-2016-03-705384 https://doi.org/10.1161/strokeaha.113.
18. Quenault A, Martinez de Lizarrondo S, 002045
Etard O, Gauberti M, Orset C, Haelewyn B, 22. Gaberel T, Gakuba C, Goulay R, Martinez De
Segal HC, Rothwell PM, Vivien D, Touze E, Lizarrondo S, Hanouz JL, Emery E, Touze E,
Ali C (2017) Molecular magnetic resonance Vivien D, Gauberti M (2014) Impaired glym-
imaging discloses endothelial activation after phatic perfusion after strokes revealed by
transient ischaemic attack. Brain 140 contrast-enhanced MRI: a new target for fibri-
(Pt 1):146–157. https://doi.org/10.1093/ nolysis? Stroke 45(10):3092–3096. https://
brain/aww260 doi.org/10.1161/strokeaha.114.006617
19. Gakuba C, Gauberti M, Mazighi M, Defer G, 23. Gauberti M, Martinez de Lizarrondo S,
Hanouz JL, Vivien D (2011) Preclinical evi- Orset C, Vivien D (2014) Lack of secondary
dence toward the use of ketamine for recombi- microthrombosis after thrombin-induced
nant tissue-type plasminogen activator- stroke in mice and non-human primates. J
mediated thrombolysis under anesthesia or Thromb Haemost 12(3):409–414. https://
sedation. Stroke 42(10):2947–2949. https:// doi.org/10.1111/jth.12487
doi.org/10.1161/strokeaha.111.620468 24. Briens A, Gauberti M, Parcq J, Montaner J,
20. Orset C, Haelewyn B, Allan SM, Ansar S, Vivien D, Martinez de Lizarrondo S (2016)
Campos F, Cho TH, Durand A, El Amki M, Nano-zymography Using Laser-Scanning
Fatar M, Garcia-Yebenes I, Gauberti M, Confocal Microscopy Unmasks Proteolytic
Grudzenski S, Lizasoain I, Lo E, Macrez R, Activity of Cell-Derived Microparticles. Ther-
Margaill I, Maysami S, Meairs S, anostics 6(5):610–626. https://doi.org/10.
Nighoghossian N, Orbe J, Paramo JA, Parienti 7150/thno.13757
Part V
Abstract
Magnetic resonance spectroscopy (MRS) or spectroscopic imaging (MRSI) enables the detection of
metabolites, amino acids, and lipids, among other biomolecules, in tumors of live mouse models of cancer.
Tumor-bearing mice are anesthetized by breathing isoflurane in a magnetic resonance (MR) scanner
dedicated to small animal MR. Here we describe the overall setup and steps for measuring 1H and 31P
MRS and 1H MRSI of orthotopic breast tumor models in mice with surface coils. This protocol can be
adapted to the use of volume coils to measure 1H and 31P MRS(I) of tumor models that grow inside the
body. We address issues of animal handling, setting up the measurement, measurement options, and data
analysis.
Key words Cancer, Magnetic resonance spectroscopic imaging, Animal setup, Shimming, Chemical
shift imaging, Metabolite, Amino acid, Lipid
1 Introduction
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0_20, © Springer Science+Business Media, LLC 2018
331
332 Menglin Cheng and Kristine Glunde
2 Materials
2.1 Mice Growing cells and weakly aggressive human MCF-7 cells can be pur-
Orthotopic Tumor chased. Other cell lines and cancer types are possible and have
Xenografts been widely published.
2. Cell culture media containing fetal bovine serum and antibio-
tics. MDA-MB-231 cells: Maintain cells in RPMI 1640 (Invi-
trogen Corp.) supplemented with 10% fetal bovine serum,
100 units/ml penicillin, and 100 μg/ml streptomycin (Invitro-
gen Corp.). MCF-7 cells: Culture cells in EMEM medium
(Mediatech, Inc.) supplemented with 10% fetal bovine serum
and the same antibiotics as used for MDA-MB-231 cells.
3. All cells require a humidified atmosphere of 5% CO2 in air, at
37 C, which is achieved in a cell culture incubator.
4. A sterile hood is required for mammalian cell culture, which has
to occur under sterile conditions.
5. A mixture of ketamine (20–50 mg/kg) and acepromazine
(0.5–2.5 mg/kg) is injected intraperitoneally to anesthetize
mice for cell inoculation into the mammary fat pad.
6. Hank’s balanced salt solution (HBSS).
7. Matrigel™ (Corning Life Sciences or BD Biosciences).
8. 0.72 mg/pellet 17β-estradiol 60-day release pellets to achieve
growth of estrogen-sensitive xenografts in mice.
9. Female homozygous athymic nude mice or severe combined
immunodeficient (SCID) mice can be purchased from differ-
ent, country-specific vendors and are obtained at 4–6 weeks of
age (see Note 1).
10. A state-of-the-art animal facility is required to house and take
care of mice.
11. An animal protocol that is in compliance with the performance
site’s country and institutional regulations and policies needs
to be in place prior to performing animal experiments. In our
case, all surgical procedures and animal handling are approved
by the Johns Hopkins University Institutional Animal Care and
Use Committee, and conform to the National Institutes of
Health (NIH) Guide for the Care and Use of Laboratory
Animals.
12. Surgical instruments including surgical scissors, scalpels,
razors, trocar, and syringes and needles are required. All surgi-
cal instruments need to be sterilized in an autoclave prior to use
and should only be used once after sterilization. Cell inocula-
tions are performed with a 26 Gauge needle. Gloves are neces-
sary for animal handling. Dedicated space in an animal facility is
needed to perform tumor cell inoculations.
13. Cotton padding, laboratory tape, and thin cardboard are nec-
essary for the mouse setup on the animal holder.
334 Menglin Cheng and Kristine Glunde
2.2 Major Equipment 1. A small animal MR scanner of relatively high field strength is
required. This could be a Bruker Biospec small animal MRI
scanner (Bruker Co., Billerica, MA) operating at 4.7 T, 7.0 T,
9.4 T, or 11.7 T. Higher fields will provide better sensitivity,
but shimming could potentially be more difficult at higher field
strengths.
2. Anesthesia equipment is needed to initially put to sleep the
mouse with isoflurane.
3. An animal holder that carries the surface solenoid or the vol-
ume coil to be used for the MR measurement is required. It is
also advisable to attach a warming pad for the mouse to stay
warm (see Note 2). Such animal holders with adequate coils are
either commercially available from Bruker, or they can be
home-built. Coils can also be obtained from specialized com-
panies such as MRcoils BV, Drunen, The Netherlands. The
holder is typically constructed on a half-shell of a plastic cone
of cylindrical shape, which fits the bore of the MR scanner to be
used. This plastic holder carries the coil, all electronics, tuning
screw sticks, cables, mouse holder, warming pad, and a nose
cone for continuous supply of isoflurane gas anesthesia includ-
ing the gas connection tube to the anesthesia equipment.
4. It is helpful to use animal monitoring equipment to continu-
ously monitor the breathing rate of the mouse with a move-
ment sensor to assess the depth of anesthesia throughout the
experiment. Additionally, it is also good to monitor the body
temperature of the animal with an anal thermometer.
3 Methods
3.1 Generate Breast 1. For MCF-7 tumor xenografts, subcutaneously implant one
Cancer Xenograft 0.72 mg/pellet 17β-estradiol 60-day release pellet per mouse
Models in Mice in the scruff region 24 h prior to tumor cell inoculation.
2. Prepare sterile single cell solutions of breast cancer cells by
suspending 2 107 cells per mL in a 50:50 (v/v) solution of
Hank’s balanced salt solution and Matrigel under a
sterile hood.
MRS of Mouse Models of Cancer 335
3.2 Mouse Setup 1. Prepare mouse holder close to magnet by attaching all cables,
on Animal Holder heating pad, and gas anesthesia line. Log into scanner software
in MR Magnet and open up measurement protocol to be used in software.
2. Measure weight and tumor size of the mouse to be MR
scanned. Tumor sizes of about 500 mm3 and larger will provide
good signal-to-noise ratio (SNR) for MRS measurements (see
Note 4).
3. Anesthetize the tumor-bearing mouse to be scanned by insert-
ing her into the isoflurane apparatus, which infuses into a
closed chamber inhalatable isoflurane anesthetic. Use 3.5–4%
of inhalatable isoflurane anesthetic in this chamber for about
2 min until the mouse is deeply asleep.
4. Quickly transfer the mouse to the animal holder to which all
electronics, circulating water bath lines for heating pad, and gas
line for inhalatable isoflurane anesthetic are already attached.
Position the mouse with the tumor hanging into the surface
coil of about 12 mm in diameter underneath the mouse (see
Note 5). Adjust the nose cone such that the mouse’s nose and
face are well covered with the nose cone mask for breathing
isoflurane. Use laboratory tape to hold the mouse in its posi-
tion. Be careful to not squeeze the mouse too hard. Attach the
movement sensor of the breathing monitor at the mouse’s
abdomen and gently fix it with tape. Apply specialized eye
336 Menglin Cheng and Kristine Glunde
ointment to the eyes of the mouse so that they do not dry out
during the relatively long MRS scanning time. During the MRI
and MRS measurements, which will last for up to 2 h per
mouse, wrap mice in cotton/plastic paper for warmth, and
place a pad circulated with warm water on top of the mouse
to maintain normal animal body temperature.
5. After transfer to the animal holder, continuously apply an
average of 1.5% of inhalatable isoflurane anesthetic by nose
cone mask for maintenance of anesthesia while performing
the MRI and MRS(I) scans on a small animal scanner. Contin-
uously monitor breathing throughout the MRI and MRS
(I) measurement with the attached movement sensor. Check
anesthetic depth via the toe pinch, and adjust the isoflurane
accordingly.
6. Carefully and slowly, with all lines (electronics, water bath,
anesthesia) attached, push the animal holder with the attached
mouse into the magnet, just far enough to be at the correct
depth, which should have been pre-marked from previous
measurements with the same animal holder and coil.
3.3 Noninvasive In 1. Set up the overall MR Scan for this mouse in the Paravision
Vivo 1H MRSI Studies Software (Bruker Co., Billerica, MA) that controls the MR
Scanner using the commands in Paravision Software. Initially,
adjust the spectrometer frequency (auto SF), set the transmit-
ter attenuator for optimal flip angle (auto RF gain), and set the
optimal receiver gain (auto RG), then wobble by adjusting the
wobble signal to be as far at the bottom and in the center as
possible by using the two wobble screws and going back and
forth between them as often as needed for optimal results (see
Note 6).
2. Start the positioning scan and optimally place the animal holder
in the center of the magnet for optimal signal strength by
pushing it further inside the magnet or pulling it out millimeter
by millimeter observing the signal strength on the monitor
while scanning with the GSP command. GSP reads out the
MR signal from one acquisition without adding up signal from
multiple repeats of the pulse program used. Go step-wise from
a large field of view of initially 50 cm for initial positioning,
then to 10 cm, and eventually to 3.2 cm for fine-tuned optimi-
zation of the position of the mouse inside the magnet.
3. Use a three-dimensional (3D) tripilot (also called triplanar)
scan for setting up the geometry of your MRI/MRSI scans.
First optimize the Tx0 and Tx1 pulses with 6 db units in
between them, optimize the gain, and acquire the tripilot
(triplanar) scan with the GOP command, which starts the
actual acquisition.
MRS of Mouse Models of Cancer 337
3.4 Noninvasive In 1. In vivo 31P MRS requires a double tuned coil that is tuned for
Vivo 31P MRS Studies the 1H and 31P frequencies, which was in our case a solenoid
coil with an inner diameter of 12 mm (MRcoils BV, Drunen,
The Netherlands) [15, 16]. Set up the mouse as described in
Subheading 3.2.
2. Set up the overall MR Scan for a new mouse in the Paravision
Software at the MR Scanner and perform steps 1–5 in the same
way as described under Subheading 3.3 for 1H MSRI measure-
ments including shimming.
3. Acquire non-localized pulse-acquire 31P MRS with an adiabatic
excitation (BIR4 45 , 200 μs, 120 ppm band width), TR of 1 s,
and 2000 averages. Use a saturation slab, which is an adiabatic
full passage pulse driven at half the amplitude to achieve
MRS of Mouse Models of Cancer 339
A B mmol/kg C mmol/kg
6.0 0.8
5.25 0.7
4.5 0.5
3.75 0.5
3.0 0.4
2.25 0.3
1.5 1.2
0.75 0.1
0.0 0.0
D E
0 6 mmol/kg 0 1 mmol/kg
Fig. 1 (a) Example of a water-suppressed 1H CSI MRSI spectrum from a representative MDA-MB-231-HRE-
tdTomato tumor (approximately 360 mm3) obtained with a spatial resolution of 0.9 0.9 0.8 mm showing
tCho at 3.2 ppm and lipid CH3 at 0.9 ppm. (b) Example of a two-dimensional tCho concentration map obtained
from the signal at 3.2 ppm in the water-suppressed MRSI data (TE ¼ 82 ms). (c) Example of a
two-dimensional lipid CH3 concentration map obtained from the signal at 0.9 ppm in the water-unsuppressed
MRSI data (TE ¼ 15 ms). Example of (d) a three-dimensional tCho concentration volume (green) and (e) a
three-dimensional lipid CH3 concentration volume (cyan) with the tumor boundary shown as a yellow grid.
Adapted from [21]
Fig. 2 Example of in vivo pulse-acquire (PA, top) and BINEPT (bottom) 31P MR spectra of a representative
MCF-7 (left) and MDA-MB-231 (right) orthotopic human breast tumor xenografts. Lorentzian lines as fitted by
JMRUI are shown below each MR spectrum. All phosphorylated metabolites are visible in the PA spectrum,
whereas the BINEPT spectrum only contains signals from phospholipid metabolites with H-P-coupling such as
PE, PC, GPE, and GPC. Note the broad, uneven baseline in the 0–5 ppm region of the PA spectra, where signals
from mobile membrane phospholipids are resonating. Adapted from [16]
4 Notes
Acknowledgements
References
1. Gatenby RA, Gillies RJ (2004) Why do cancers Nelson MT, Yee D, Garwood M, Bolan PJ
have high aerobic glycolysis? Nat Rev Cancer 4 (2009) Metabolite quantification and high-
(11):891–899. https://doi.org/10.1038/ field MRS in breast cancer. NMR Biomed 22
nrc1478 (1):65–76. https://doi.org/10.1002/nbm.
2. Warburg O (1956) On the origin of cancer 1217
cells. Science 123(3191):309–314 12. Howe FA, Barton SJ, Cudlip SA, Stubbs M,
3. Kaelin WG Jr, Thompson CB (2010) Q&A: Saunders DE, Murphy M, Wilkins P, Opstad
Cancer: clues from cell metabolism. Nature KS, Doyle VL, McLean MA, Bell BA, Griffiths
465(7298):562–564. https://doi.org/10. JR (2003) Metabolic profiles of human brain
1038/465562a tumors using quantitative in vivo 1H magnetic
4. Negendank W (1992) Studies of human resonance spectroscopy. Magn Reson Med 49
tumors by MRS: a review. NMR Biomed 5 (2):223–232. https://doi.org/10.1002/mrm.
(5):303–324 10367
5. Glunde K, Bhujwalla ZM, Ronen SM (2011) 13. Serkova NJ, Brown MS (2012) Quantitative
Choline metabolism in malignant transforma- analysis in magnetic resonance spectroscopy:
tion. Nat Rev Cancer 11(12):835–848. from metabolic profiling to in vivo biomarkers.
https://doi.org/10.1038/nrc3162 Bioanalysis 4(3):321–341. https://doi.org/
10.4155/bio.11.320
6. Hanahan D, Weinberg RA (2011) Hallmarks
of cancer: the next generation. Cell 144 14. Nelson MT, Everson LI, Garwood M,
(5):646–674. https://doi.org/10.1016/j.cell. Emory T, Bolan PJ (2008) MR spectroscopy
2011.02.013 in the diagnosis and treatment of breast cancer.
Semin Breast Dis 11(2):100–105. https://doi.
7. DeBerardinis RJ, Lum JJ, Hatzivassiliou G, org/10.1053/j.sembd.2008.03.004
Thompson CB (2008) The biology of cancer:
metabolic reprogramming fuels cell growth 15. Wijnen JP, Jiang L, Greenwood TR, Cheng M,
and proliferation. Cell Metab 7(1):11–20. Dopkens M, Cao MD, Bhujwalla ZM,
https://doi.org/10.1016/j.cmet.2007.10. Krishnamachary B, Klomp DW, Glunde K
002 (2014) Silencing of the glycerophosphocholine
phosphodiesterase GDPD5 alters the phos-
8. Wise DR, Thompson CB (2010) Glutamine pholipid metabolite profile in a breast cancer
addiction: a new therapeutic target in cancer. model in vivo as monitored by (31) P MRS.
Trends Biochem Sci 35(8):427–433. https:// NMR Biomed 27(6):692–699. https://doi.
doi.org/10.1016/j.tibs.2010.05.003 org/10.1002/nbm.3106
9. Moestue SA, Giskeodegard GF, Cao MD, 16. Wijnen JP, Jiang L, Greenwood TR, van der
Bathen TF, Gribbestad IS (2012) Glyceropho- Kemp WJ, Klomp DW, Glunde K (2014)
sphocholine (GPC) is a poorly understood bio- 1H/31P polarization transfer at 9.4 Tesla
marker in breast cancer. Proc Natl Acad Sci U S for improved specificity of detecting phospho-
A 109(38):E2506; author reply E2507. monoesters and phosphodiesters in breast
https://doi.org/10.1073/pnas.1208226109 tumor models. PLoS One 9(7):e102256.
10. Danishad KK, Sharma U, Sah RG, Seenu V, https://doi.org/10.1371/journal.pone.
Parshad R, Jagannathan NR (2010) Assess- 0102256
ment of therapeutic response of locally 17. Meisamy S, Bolan PJ, Baker EH, Pollema MG,
advanced breast cancer (LABC) patients under- Le CT, Kelcz F, Lechner MC, Luikens BA,
going neoadjuvant chemotherapy (NACT) Carlson RA, Brandt KR, Amrami KK, Nelson
monitored using sequential magnetic reso- MT, Everson LI, Emory TH, Tuttle TM,
nance spectroscopic imaging (MRSI). NMR Yee D, Garwood M (2005) Adding in vivo
Biomed 23(3):233–241. https://doi.org/10. quantitative 1H MR spectroscopy to improve
1002/nbm.1436 diagnostic accuracy of breast MR imaging: pre-
11. Haddadin IS, McIntosh A, Meisamy S, liminary results of observer performance study
Corum C, Styczynski Snyder AL, Powell NJ, at 4.0 T. Radiology 236(2):465–475. https://
MRS of Mouse Models of Cancer 345
Abstract
Magnetic Resonance Imaging (MRI) is an important tool to study various animal models of degenerative
diseases. This chapter describes routine protocols of T1-, T2-, and T2*-weighted and diffusion-weighted
MRI for rodent brain and spinal cord. These protocols can be used to measure atrophy, axonal and myelin
injury and changes in white matter connectivity.
Key words Magnetic Resonance Imaging, Diffusion-weighted imaging, Diffusion tensor imaging,
Animal model of neurodegenerative diseases, Brain, Spinal cord, In vivo MRI, Ex vivo MRI, High-field
MRI
1 Introduction
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0_21, © Springer Science+Business Media, LLC 2018
347
348 Nyoman D. Kurniawan
MRI for such small sample size are often necessary to be performed
using high-field systems (>7 Tesla), as the high magnetic field
provides better signal sensitivity and these scanners are equipped
with strong gradients (>300 mT/m) to enable the acquisition of
high-resolution images [27].
1.1 In Vivo MRI In vivo mouse and rat brain/spinal cord MRI data are often acquired
with two-dimensional (2D) multi-slice sequences with in-plane reso-
lutions in the range of 0.08–0.15 mm and 0.5–1 mm slice thickness.
Recent cryoprobe hardware development for mouse and rat brain
MRI have tremendously improved the image quality and signal-to-
noise ratio (SNR), especially for functional MRI, and allowed the
acquisition of 3D datasets at a higher (0.15–0.20 mm) isotropic re-
solution [28, 29]. The improvement of conventional MRI sequences,
such as a modified 3D-gradient spin-echo (GRASE) sequence, have
enabled zoomed imaging and better reduction of physiological
motion artifacts using gating and navigator sequence [30].
In vivo MRI requires animal anesthesia, maintenance of animal
physiology, and monitoring of respiration and body temperature
[31–34]. The length of in vivo imaging is normally kept under
2–3 h using air–isoflurane gas mixture or ketamine/xylazine injec-
tion [33]. Longer acquisition time can result in deterioration of the
stability of animal physiology, especially increased difficulty to
maintain a stable breathing rate, which may already be compro-
mised in the disease model. For spinal cord imaging, respiratory
gating is especially required to minimize motion artifact. Respira-
tory gating, however, can significantly lengthen the acquisition
time, but its use is often critical for sequences which are very
sensitive to motion such as diffusion-weighted imaging (DWI)
[35]. If required, a MRI-compatible ventilator can be used to
synchronize MRI acquisition and controlled mechanical
respiration [36].
1.2 Ex Vivo MRI Ex vivo imaging using fixed samples allows the acquisition of much
higher resolution images compared to in vivo, as the length of scan
time is much less limited. Depending on sample size and the
scanner field-strength, ex vivo images may be obtained at 30-μm
isotropic resolution in 1–2 h acquisition, or as high as 10 μm 3D
isotropic resolution over a 16 h acquisition using a small 5 mm
solenoid coil [37, 38]. Ex vivo imaging also allows the enhance-
ment of the tissue white matter (WM) and gray matter (GM)
contrast post fixation using a low concentration of gadolinium
MRI contrast agent. MRI contrast agents improve image quality
by reducing tissue T1 relaxation time to enable the use of a short
recycling time (TR) and increased scan averages to increase signal-
to-noise ratio; and also to enhance the T1 and T2 WM/GM con-
trast [39, 40].
MRI in Neurodegenerative Diseases 349
1.3 The Scope of MRI Animal models of neurodegeneration can be imaged using a wide
Protocols array of MR modalities. This chapter limits the description into
and Analyses widely used protocols: high-resolution T1-weighted imaging to
measure volumetric changes; T1, T2, and T2* mapping to measure
changes in tissue relaxation, edema, and magnetic susceptibility
[44–48]; and diffusion-weighted imaging (DWI), which is impor-
tant to measure axonal injury/demyelination and changes in brain
white matter connectivity [49, 50]. Other MRI modalities such as
spectroscopy [51], quantitative susceptibility-weighted imaging
(qSWI) [15, 52], and magnetization transfer imaging (MTI)
[53, 54] can provide quantitative measurements of tissue metabo-
lites, iron, and myelin content, but they are beyond the scope of this
chapter.
This chapter also describes standard approaches to analyze MRI
data using voxel based analysis (VBA) [23, 55], or using manual
segmentation of regions-of-interest (ROIs) [56]. Whole brain VBA
is typically performed on a medium (n ¼ 10–20) and large data sets
(n > 30). VBA involves automatic registration of MRI data into
mouse/rat brain atlases in order to allow for voxel-wise statistical
analyses between groups. In comparison, ROI analyses involve
segmentations of anatomical regions by drawing on the MR images
slice by slice. Subsequent statistical analyses can be done using an
analysis of variance and t-test, with the appropriate corrections for
multiple comparisons. Manual segmentation can be a tedious pro-
cess and thus may be suitable only for small sample studies (n < 10)
with limited ROIs [57, 58]. ROI analyses may also be influenced by
operator bias, and thus the operators are often blinded to the group
assignments.
2 Materials
2.1 In Vivo Mouse Bruker Micro 2.5 gradient coil, 20 mm volume head radiofre-
Brain Imaging quency (RF) coil, an animal bed and a probe base. This setup is
suitable for mouse imaging from approximately P28 (post-natal
2.1.1 MRI Equipment
28 day/weaning age) to adult, with maximum weight of approxi-
mately 35 g.
2.2 Ex Vivo Mouse 1. A perfusion pump (e.g., Harvard Peristaltic Pump P-70) or a
Brain Imaging syringe pump (e.g., Harvard Pump 11) for cardiac perfusion
(see Notes 2–5).
2.2.1 Sample
Preparation 2. A small vacuum pump for degassing sample prior to MRI.
3. Chemicals: Lethabarb, pentobarbital or isoflurane (to induce
deep anesthesia prior to cardiac perfusion), phosphate buffer
saline (PBS), and heparin (optional, to avoid blood clotting).
4. MRI contrast agent, e.g., Gd-DTPA (diethylenetriaminepen-
taacetic acid) or Magnevist® (Bayer).
5. Perfluoro ether solution Fomblin Y06/06 (Solvay Solexis,
Italy) medium.
2.2.2 MRI Equipment 1. Bruker Micro 2.5 or Micro 5 gradients and microimaging
probes.
2. Imaging coils.
(a) 15 mm volume RF coil for juvenile and adult mouse brain
imaging.
(b) 10 mm quadrature birdcage volume RF coil for embry-
onic to neonate mouse brain imaging.
MRI in Neurodegenerative Diseases 351
Fig. 1 Sample set up for ex vivo brain imaging. (a) The brain sample is held by two plastic scaffoldings placed
gently above and below the brain. The scaffolding should only touch the brain lightly so they do not distort the
brain shape. The fomblin solution should only be about 2–3 mm above the sample. Do not use excess fomblin
as it produces an upward force that may distort the tissue. A foam is used to keep the sample centered and the
tube is closed with a paraffin membrane. (b) T1/T2* gradient echo image acquired at 30 μm 3D isotropic
resolution at 16.4 T
2.3 In Vivo Mouse 1. A single transmit/receive linear surface coil (1.5 3.0 cm) and
Spinal Cord Imaging micro 2.5 imaging gradient.
Alternatively, a separate whole body transmit only and a receive
2.3.1 MRI Equipment
only surface coil can be used. This setup is favorable as the
volume coil will provide more homogenous excitation of the
sample.
2. Animal respiratory monitoring and gating (e.g., SA Instrument
or Biopac) is essential.
2.4 Ex Vivo Mouse 1. Bruker Micro 2.5 gradient coil and microimaging probe base.
Spinal Cord Imaging (a) 10, 15, or 20 mm volume coils can be used for a single or
2.4.1 MRI Equipment tandem sample imaging.
(b) 5 mm solenoid coil for 3 mm spinal cord slab imaging.
3 Methods
Fig. 2 Animal positioning for mouse brain imaging at 16.4 T. (a) Animal head is placed inside a 20 mm head
coil and restrained with a toothbar. Animal is lightly strapped with surgical tape, and a respiratory breathing
pad (blue tubing) is placed underneath the abdomen just below the diaphragm. Animal is maintained under
anesthesia using an oxygen/isoflurane mixture (clear tubing). (b, c, d) are coronal, sagittal, and axial MRI
slices
3.1.2 Diffusion-Weighted One preferred method to obtain 2D DWI data at high-field is using
Imaging the Stejskal-Tanner pulse-field gradient spin-echo sequence (DWI
SE). This sequence produces only small distortion artifacts but its
acquisition is slow and it is susceptible to motion artifacts
[59]. DWI spin-echo is thus limited for obtaining data with a
small number of diffusion encoding directions (6–12 directions).
To obtain a high angular resolution diffusion imaging
(HARDI) dataset with 30–60 diffusion encoding directions, the
Stejskal-Tanner DWI sequence can be acquired using echo planar
imaging (EPI) read-out sequence (DWI EPI) to achieve fast acqui-
sition [60]. DWI EPI, however, is more susceptible to distortion
artifact than DWI spin echo, which can be improved using high-
order shimming (see Note 11). Additionally, the echo time
(TE) may be reduced using EPI segmentation, but this may result
in increased susceptibility to motion artifact due to imperfect align-
ment of the lines in the segmented k-space data. Typically, 2D DWI
EPI may be acquired without segmentation or with 2–4 EPI seg-
ments at ultra-high field. Respiratory gating can be used to reduce
motion artifacts, which may often be present at the caudal part of
the brain. The read axis may also be placed in the dorso-ventral
plane to reduce motion artefacts.
2D DWI SE Acquisition 1. The field of view (FOV) required to cover the brain axially is
Parameters (See Note 12) typically 1.5 1.5 cm, with a matrix size of 128 128 to
354 Nyoman D. Kurniawan
2D DWI EPI Acquisition Higher-order diffusion models, such as diffusion kurtosis imaging
Parameters (See Note 12) (DKI), constrained spherical deconvolution (CSD), and Neurite
Orientation Dispersion and Density Imaging (NODDI), requires
HARDI or multi-shell HARDI acquisitions (see Note 13). Such
dataset can only be acquired using DWI EPI sequences.
1. 2D DWI EPI may be acquired using 24 contiguous slices at
0.6 mm thickness with FOV ¼ 1.60 0.96 cm and matrix
size ¼ 128 64, to produce 125 150 μm2 in-plane resolu-
tion. A combination of partial Fourier transform (FT) and
zero-fill acceleration factors of 1.35 can be used to reduce TE
and speed up the scan time. If using a cryoprobe, a thinner slice
thickness can be used to obtain a pseudo 3D dataset at
(200 μm)3 isotropic resolution.
2. TR ¼ 6 s, TE ¼ 14 ms, and sampling bandwidth 300–500 kHz.
Diffusion gradients δ/Δ ¼ 2.4/6.4 ms, b-value of
3000 s/mm2, NEX ¼ 2 and DS ¼ 4.
With these parameters, a 64 diffusion, single-shell direc-
tion-encoding measurements can be acquired within 1 h with-
out respiratory triggering, or 2 h with respiratory triggering
[60]. An example of a FA map acquired using this protocol is
shown in Fig. 3.
3. To acquire a multi-shell HARDI dataset, the number of direc-
tions may be reduced to accommodate various shells and
reducing NEX to 1 to keep the acquisition time within 3 h.
The SNR should be tested and the image resolution will need
to be adjusted to ensure good data quality is maintained.
MRI in Neurodegenerative Diseases 355
Fig. 3 FA map of an in vivo mouse brain acquired using DTI-EPI at 16.4 T. The brain white matter structures
are shown. This image is reproduced from [60]
3.1.4 T2-Weighted T2-weighted imaging is often used to determine tissue edema, for
Imaging example, in stroke [65] and traumatic CNS injury [5], and ventric-
ular enlargement. At 16.4 T, the T2 of mouse brain in vivo is
approximately 25–30 ms [60], so that a TE of 40–60 ms can be
used to obtain T2-weighted images.
2D T2-Weighted Acquisition – T2-weighted imaging can be acquired using spin-echo with long
Parameters TR (>5 s) and long TE (>40 ms). This TR setting, however, will
result in a long acquisition time (>20 min). Shorter acquisition
time can be obtained using FSE with a turbo factor of 4–8,
however turbo factor >4 may result in blurring or motion
artifacts. Alternatively, a shorter TR (~2 s) may be used with
some T1-weighting effect in the image.
– Quantitative T2 MR images can be acquired using a multi-echo
spin-echo sequence with TR ¼ 2 s and the TE of 10, 20, 30, 40,
50, 60 ms, using similar FOV/matrix image parameters as
described above (see Note 16). The total acquisition time is
approximately 20 min.
3.2.2 Ex Vivo Mouse 1. Adult mouse brain HARDI: 3D DWI SE sequence with the
Brain MRI parameters TE/TR ¼ 22.8/400 ms, 30 uniformly distributed
DW directions, b ¼ 5000 s/mm2, δ/Δ ¼ 2.5/12.5 ms, NEX ¼ 1.
3D DWI SE Acquisition
FOV ¼ 19.6 11.7 8.4 mm matrix size ¼ 19.6 11.7 8.4,
Parameters
resolution ¼ 100 μm isotropic. The acquisition time ~32 h
(without partial Fourier acceleration), or 15 h with 1.5 partial
Fourier encoding acceleration in the phase dimensions [37].
2. Embryonic and neonate mouse brain HARDI can be acquired
with similar parameters as above, but the FOV is reduced to fit
in the sample size, and the matrix size adjusted to obtain 80 μm
isotropic resolution.
3. Ultra-high resolution brain HARDI can be acquired using
similar parameters as above using a 3 mm brain section, with
the FOV and matrix size adjusted to obtain 48 μm isotropic
resolution, with NEX ¼ 2 and the acquisition time of
~24 h [37].
358 Nyoman D. Kurniawan
Fig. 4 Animal positioning for mouse spinal cord imaging at 16.4 T using a surface coil. (a) Animal head is
placed inside a head restrain with a toothbar. The animal spine is placed in the center of the surface coil, with
the respiratory breathing pad (blue tubing) is placed on the abdomen and lightly secured with a surgical tape. A
foam may be placed on its feet to support its upward position in the vertical scanner. (b, c, d) are sagittal,
coronal, and axial MRI slices. A FOV saturation band (b) may be used to suppress signal from the abdomen
2D T1-, T2-, and T2*- 2D T1-, T2-, and T2*-weighted imaging can be acquired using
Weighted Acquisition similar parameters to that described in Subheadings 3.1.3–3.1.5
Parameters for in vivo brain imaging, but with the FOV and resolution adjusted
as above. The application of T2*-weighted imaging with long TE is
limited in the spinal cord, due to severe inhomogeneity artifacts
from the spine and the lung.
3.3.2 Ex Vivo DTI For tandem imaging, samples must be glued as close as possible
between them to improve shimming and reduce the scan time (see
Note 21).
3D Acquisition Parameters
1. 3D DTI spin-echo sequence with TR/TE ¼ 400/20.8 ms, δ/
Δ ¼ 2.5/10 ms, 12 non-collinear directions, two b0 images,
b ¼ 3000 s/mm2, NEX ¼ 1 and partial Fourier acceleration
factor ¼ 1.5. Images can be acquired at 60 μm isotropic reso-
lution with the acquisition time of 13–24 h depending on the
number of samples imaged [7, 8].
2. Ultra-high resolution HARDI and T1/T2* imaging can be
acquired using similar parameters as described in Subheading
3.2.2.
3.4.2 Data 1. Data conversion. MRI data are typically exported into DICOM
Pre-processing before they are converted into nifti (.nii) format using dcm2nii
program (MRIcron suite), or into .mih/dat format using
mrconvert (MRTrix).
2. Image transform correction. This step is often needed when
working with Bruker data with MRTrix (www.mrtrix.org).
The 3 3 image cosine orientation matrix shown in the .mih
header should be changed to reflect pure axial or sagittal or
coronal image transforms (integers of 0 or 1) (see
Note 23).
3. Masking. Masking is used to remove other parts of the image
which are not part of the study and improve the quality of
subsequent analyses. For DWI, the mask may be derived from
an average of all b-weighted images (excluding the b ¼ 0
images), as they normally have adequate suppression of
non-CNS tissues. The mask may be generated using region
MRI in Neurodegenerative Diseases 361
Fig. 5 ODF map of an ex vivo mouse brain and generated streamlines of the corpus callosum. (a) and (b) are
ODF for the axial and transverse slices. Red, green, blue (RGB) indicate ODF directions in dorso/ventral,
medial/lateral, and rostro/caudal, respectively. Note that although the RGB colors themselves may be
interchangeable, it is the shape and the direction of the ODF (or the tensor) that are critical for fibertracking.
(c) Corpus callosum streamlines generated from the midline ROI using probabilistic iFOD2 fibertracking in
MRTRix 3. HARDI data was acquired ex vivo using 30 diffusion encoding direction at 100 μm isotropic
resolution at 16.4 T
Fig. 6 Ex vivo C56 BL6J mouse brain MRI template and atlas. Cortical and basal ganglia atlas are overlaid on
the Australian Mouse Brain Mapping Consortium (AMBMC) 15 μm resolution template [92, 93]
3.4.3 ROI Analyses ROI analyses may be performed directly using an offline version of
Bruker Paravision. Alternatively, MRI data can be converted into
DICOM for analysis using freely available programs such as MRI-
cron, Osirix, MIPAV, or ITK-SNAP. ROIs are typically segmented
using the freehand tools within these programs. Experts blinded to
group assignments and/or independent raters may be required to
do manual segmentation as this method can be subjective and the
results may vary between operators.
3.4.4 Voxel-Based 1. Choice of MRI atlases. There are several atlases available for
Analyses of Structural ex vivo mouse brain (brainatlas.mbi.ufl.edu; imaging.org.au/
Images AMBMC/AMBMC; mouseimaging.ca/research/mouse_atlas.
html; lbam.med.jhmi.edu) and in vivo mouse brain MRI
(brainatlas.mbi.ufl.edu). An example of the atlas segmentation
showing cortical and basal ganglia segmentations of an adult C57
BL6 brain atlas (AMBMC) is shown in Fig. 6 (see Note 31).
MRI in Neurodegenerative Diseases 363
3.4.5 Analysis of DTI There are several methods that may be used to normalize DTI data
Parametric Maps to the template.
1. Using the transformation matrix derived from the T1-weighted
image structural images to normalize the DTI data to the
template. This method may reduce registration bias due to
the disease, especially if there are significant morphological
changes in the WM structures that can affect registration.
2. Using one of DTI parametric maps (e.g., FA) to create the
study template (using a similar strategy as outlined for the
structural images, e.g., using ANTS buildtemplateparallel.sh),
364 Nyoman D. Kurniawan
3.4.6 Statistical Analyses Statistical analyses may be performed using FSL randomize T-tests.
First, datasets need to be arranged as a 4D data (using FSL
fslmerge) and a design matrix needs to be created accordingly.
Randomize calculation is typically performed using >5000 permu-
tations, and the output is measured using a threshold-free cluster
enhancement with a family-wise error correction and p < 0.05.
4 Notes
rffiffiffisffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffiffi
ffi
1 ðλ1 λ2 Þ2 þ ðλ1 λ3 Þ2 þ ðλ2 λ3 Þ2
FA ¼ ð4Þ
2 λ1 2 þ λ2 2 þ λ3 2
Acknowledgements
References
1. Neumann-Haefelin T, Kastrup A, de cord injury. Brain Sci 7(2). https://doi.org/
Crespigny A, Yenari MA, Ringer T, Sun GH, 10.3390/brainsci7020021
Moseley ME (2000) Serial MRI after transient 10. Kim JH, Loy DN, Wang Q, Budde MD,
focal cerebral ischemia in rats: dynamics of tis- Schmidt RE, Trinkaus K, Song SK (2010) Dif-
sue injury, blood-brain barrier damage, and fusion tensor imaging at 3 hours after trau-
edema formation. Stroke 31(8):1965–1972; matic spinal cord injury predicts long-term
discussion 1972–1963 locomotor recovery. J Neurotrauma 27
2. Yan G, Dai Z, Xuan Y, Wu R (2015) Early (3):587–598. https://doi.org/10.1089/neu.
metabolic changes following ischemia onset in 2009.1063
rats: an in vivo diffusion-weighted imaging and 11. Cowin GJ, Butler TJ, Kurniawan ND,
1H-magnetic resonance spectroscopy study at Watson C, Wallace RH (2011) Magnetic reso-
7.0T. Mol Med Rep 11(6):4109–4114. nance microimaging of the spinal cord in the
https://doi.org/10.3892/mmr.2015.3283 SOD1 mouse model of amyotrophic lateral
3. Hutchinson EB, Schwerin SC, Radomski KL, sclerosis detects motor nerve root degenera-
Irfanoglu MO, Juliano SL, Pierpaoli CM tion. Neuroimage 58(1):69–74. https://doi.
(2016) Quantitative MRI and DTI abnormal- org/10.1016/j.neuroimage.2011.06.003
ities during the acute period following CCI in 12. Underwood C, Kurniawan ND, Butler T,
the ferret. Shock 46(3 Suppl 1):167–176. Cowin GJ, Wallace RA (2011) Non-invasive
https://doi.org/10.1097/SHK. diffusion tensor imaging detects white matter
0000000000000659 degeneration in the spinal cord of a mouse
4. Kharatishvili I, Immonen R, Grohn O, Pitka- model of amyotrophic lateral sclerosis. Neuro-
nen A (2007) Quantitative diffusion MRI of image 55(2):455–461. https://doi.org/10.
hippocampus as a surrogate marker for post- 1016/j.neuroimage.2010.12.044
traumatic epileptogenesis. Brain 130 13. Absinta M, Sati P, Reich DS (2016) Advanced
(Pt 12):3155–3168. https://doi.org/10. MRI and staging of multiple sclerosis lesions.
1093/brain/awm268 Nat Rev Neurol 12(6):358–368. https://doi.
5. Long JA, Watts LT, Chemello J, Huang S, org/10.1038/nrneurol.2016.59
Shen Q, Duong TQ (2015) Multiparametric 14. Hunger M, Budinger E, Zhong K, Angenstein
and longitudinal MRI characterization of mild F (2014) Visualization of acute focal lesions in
traumatic brain injury in rats. J Neurotrauma rats with experimental autoimmune encephalo-
32(8):598–607. https://doi.org/10.1089/ myelitis by magnetic nanoparticles, comparing
neu.2014.3563 different MRI sequences including phase imag-
6. Blomster LV, Cowin GJ, Kurniawan ND, Rui- ing. J Magn Reson Imaging 39(5):1126–1135.
tenberg MJ (2013) Detection of endogenous https://doi.org/10.1002/jmri.24280
iron deposits in the injured mouse spinal cord 15. Nathoo N, Agrawal S, Wu Y, Haylock-Jacobs S,
through high-resolution ex vivo and in vivo Yong VW, Foniok T, Barnes S, Obenaus A,
MRI. NMR Biomed 26(2):141–150. https:// Dunn JF (2013) Susceptibility-weighted imag-
doi.org/10.1002/nbm.2829 ing in the experimental autoimmune encepha-
7. Brennan FH, Cowin GJ, Kurniawan ND, Rui- lomyelitis model of multiple sclerosis indicates
tenberg MJ (2013) Longitudinal assessment of elevated deoxyhemoglobin, iron deposition
white matter pathology in the injured mouse and demyelination. Mult Scler 19
spinal cord through ultra-high field (16.4T) (6):721–731. https://doi.org/10.1177/
in vivo diffusion tensor imaging. Neuroimage 1352458512460602
82:574–585. https://doi.org/10.1016/j. 16. Tambalo S, Peruzzotti-Jametti L, Rigolio R,
neuroimage.2013.06.019 Fiorini S, Bontempi P, Mallucci G,
8. Brennan FH, Kurniawan ND, Vukovic J, Bart- Balzarotti B, Marmiroli P, Sbarbati A,
lett PF, Kasermann F, Arumugam TV, Basta M, Cavaletti G, Pluchino S, Marzola P (2015)
Ruitenberg MJ (2016) IVIg attenuates com- Functional magnetic resonance imaging of
plement and improves spinal cord injury out- rats with experimental autoimmune encephalo-
comes in mice. Ann Clin Transl Neurol 3 myelitis reveals brain cortex remodeling. J
(7):495–511. https://doi.org/10.1002/ Neurosci 35(27):10088–10100. https://doi.
acn3.318 org/10.1523/JNEUROSCI.0540-15.2015
9. Sun P, Murphy RK, Gamble P, George A, Song 17. Colgan N, Siow B, O’Callaghan JM, Harrison
SK, Ray WZ (2017) Diffusion assessment of IF, Wells JA, Holmes HE, Ismail O,
cortical changes, induced by traumatic spinal Richardson S, Alexander DC, Collins EC,
MRI in Neurodegenerative Diseases 371
Fisher EM, Johnson R, Schwarz AJ, Ahmed Z, Longitudinal characterization of brain atrophy
O’Neill MJ, Murray TK, Zhang H, Lythgoe of a Huntington’s disease mouse model by
MF (2016) Application of neurite orientation automated morphological analyses of magnetic
dispersion and density imaging (NODDI) to a resonance images. Neuroimage 49
tau pathology model of Alzheimer’s disease. (3):2340–2351. https://doi.org/10.1016/j.
Neuroimage 125:739–744. https://doi.org/ neuroimage.2009.10.027
10.1016/j.neuroimage.2015.10.043 25. Yang S, Yang Z, Fischer K, Zhong K, Stadler J,
18. Li L, Wang XY, Gao FB, Wang L, Xia R, Li ZX, Godenschweger F, Steiner J, Heinze HJ, Bern-
Xing W, Tang BS, Zeng Y, Zhou GF, Zhou stein HG, Bogerts B, Mawrin C, Reutens DC,
HY, Liao WH (2016) Magnetic resonance T2 Speck O, Walter M (2013) Integration of ultra-
relaxation time at 7 Tesla associated with amy- high field MRI and histology for connectome
loid beta pathology and age in a double- based research of brain disorders. Front Neu-
transgenic mouse model of Alzheimer’s dis- roanat 7:31. https://doi.org/10.3389/fnana.
ease. Neurosci Lett 610:92–97. https://doi. 2013.00031
org/10.1016/j.neulet.2015.10.058 26. Spencer RG, Fishbein KW, Cheng A, Mattson
19. Wells JA, O’Callaghan JM, Holmes HE, MP (2006) Compatibility of Gd-DTPA perfu-
Powell NM, Johnson RA, Siow B, sion and histologic studies of the brain. Magn
Torrealdea F, Ismail O, Walker-Samuel S, Reson Imaging 24(1):27–31. https://doi.org/
Golay X, Rega M, Richardson S, Modat M, 10.1016/j.mri.2005.10.017
Cardoso MJ, Ourselin S, Schwarz AJ, 27. Webb AG, Van de Moortele PF (2016) The
Ahmed Z, Murray TK, O’Neill MJ, Collins technological future of 7 T MRI hardware.
EC, Colgan N, Lythgoe MF (2015) In vivo NMR Biomed 29(9):1305–1315. https://doi.
imaging of tau pathology using multi- org/10.1002/nbm.3315
parametric quantitative MRI. Neuroimage 28. Baltes C, Radzwill N, Bosshard S, Marek D,
111:369–378. https://doi.org/10.1016/j. Rudin M (2009) Micro MRI of the mouse
neuroimage.2015.02.023 brain using a novel 400 MHz cryogenic quad-
20. Antonsen BT, Jiang Y, Veraart J, Qu H, rature RF probe. NMR Biomed 22
Nguyen HP, Sijbers J, von Horsten S, Johnson (8):834–842. https://doi.org/10.1002/nbm.
GA, Leergaard TB (2013) Altered diffusion 1396
tensor imaging measurements in aged trans- 29. Bosshard SC, Baltes C, Wyss MT, Mueggler T,
genic Huntington disease rats. Brain Struct Weber B, Rudin M (2010) Assessment of brain
Funct 218(3):767–778. https://doi.org/10. responses to innocuous and noxious electrical
1007/s00429-012-0427-0 forepaw stimulation in mice using BOLD
21. Blockx I, De Groof G, Verhoye M, Van fMRI. Pain 151(3):655–663. https://doi.
Audekerke J, Raber K, Poot D, Sijbers J, org/10.1016/j.pain.2010.08.025
Osmand AP, Von Horsten S, Van der Linden 30. Wu D, Xu J, McMahon MT, van Zijl PC,
A (2012) Microstructural changes observed Mori S, Northington FJ, Zhang J (2013) In
with DKI in a transgenic Huntington rat vivo high-resolution diffusion tensor imaging
model: evidence for abnormal neurodevelop- of the mouse brain. Neuroimage 83:18–26.
ment. Neuroimage 59(2):957–967. https:// https://doi.org/10.1016/j.neuroimage.
doi.org/10.1016/j.neuroimage.2011.08.062 2013.06.012
22. Sawiak SJ, Wood NI, Williams GB, Morton AJ, 31. Adams S, Pacharinsak C (2015) Mouse anes-
Carpenter TA (2009) Voxel-based morphome- thesia and analgesia. Curr Protoc Mouse Biol 5
try in the R6/2 transgenic mouse reveals dif- (1):51–63. https://doi.org/10.1002/
ferences between genotypes not seen with 9780470942390.mo140179
manual 2D morphometry. Neurobiol Dis 33
(1):20–27. https://doi.org/10.1016/j.nbd. 32. Colby LA, Morenko BJ (2004) Clinical con-
2008.09.016 siderations in rodent bioimaging. Comp Med
54(6):623–630
23. Sawiak SJ, Wood NI, Williams GB, Morton AJ,
Carpenter TA (2013) Voxel-based morphome- 33. Lukasik VM, Gillies RJ (2003) Animal anaes-
try with templates and validation in a mouse thesia for in vivo magnetic resonance. NMR
model of Huntington’s disease. Magn Reson Biomed 16(8):459–467. https://doi.org/10.
Imaging 31(9):1522–1531. https://doi.org/ 1002/nbm.836
10.1016/j.mri.2013.06.001 34. McConville P (2011) Small animal preparation
24. Zhang J, Peng Q, Li Q, Jahanshad N, Hou Z, and handling in MRI. Methods Mol Biol
Jiang M, Masuda N, Langbehn DR, Miller MI, 771:89–113. https://doi.org/10.1007/978-
Mori S, Ross CA, Duan W (2010) 1-61779-219-9_5
372 Nyoman D. Kurniawan
35. Kim JH, Song SK (2013) Diffusion tensor model of Parkinson’s disease. PLoS One 11(3):
imaging of the mouse brainstem and cervical e0151884. https://doi.org/10.1371/journal.
spinal cord. Nat Protoc 8(2):409–417 pone.0151884
36. Hedlund LW, Cofer GP, Owen SJ, Allan John- 46. Kara F, Hofling C, Rossner S, Schliebs R, Van
son G (2000) MR-compatible ventilator for der Linden A, Groot HJ, Alia A (2015) In vivo
small animals: computer-controlled ventilation longitudinal monitoring of changes in the cor-
for proton and noble gas imaging. Magn Reson pus callosum integrity during disease progres-
Imaging 18(6):753–759 sion in a mouse model of Alzheimer’s disease.
37. Kurniawan ND, Richards KL, Yang Z, She D, Curr Alzheimer Res 12(10):941–950
Ullmann JFP, Moldrich RX, Liu S, Yaksic JU, 47. Rudko DA, Klassen LM, de Chickera SN, Gati
Leanage G, Kharatishvili I, Wimmer V, JS, Dekaban GA, Menon RS (2014) Origins of
Calamante F, Galloway GJ, Petrou S, Reutens R2* orientation dependence in gray and white
DC (2014) Visualization of mouse barrel cor- matter. Proc Natl Acad Sci U S A 111(1):
tex using ex-vivo track density imaging. Neuro- E159–E167. https://doi.org/10.1073/pnas.
image 87:465–475. https://doi.org/10. 1306516111
1016/j.neuroimage.2013.09.030 48. Thiessen JD, Zhang Y, Zhang H, Wang L,
38. Ullmann JF, Calamante F, Collin SP, Reutens Buist R, Del Bigio MR, Kong J, Li XM, Martin
DC, Kurniawan ND (2015) Enhanced charac- M (2013) Quantitative MRI and ultrastruc-
terization of the zebrafish brain as revealed by tural examination of the cuprizone mouse
super-resolution track-density imaging. Brain model of demyelination. NMR Biomed 26
Struct Funct 220(1):457–468. https://doi. (11):1562–1581. https://doi.org/10.1002/
org/10.1007/s00429-013-0667-7 nbm.2992
39. Huang S, Liu C, Dai G, Kim YR, Rosen BR 49. Zhang J, Aggarwal M, Mori S (2012) Struc-
(2009) Manipulation of tissue contrast using tural insights into the rodent CNS via diffusion
contrast agents for enhanced MR microscopy tensor imaging. Trends Neurosci 35
in ex vivo mouse brain. Neuroimage 46 (7):412–421. https://doi.org/10.1016/j.
(3):589–599. https://doi.org/10.1016/j. tins.2012.04.010
neuroimage.2009.02.027 50. Mori S, Zhang J (2006) Principles of diffusion
40. Kim S, Pickup S, Hsu O, Poptani H (2009) tensor imaging and its applications to basic
Enhanced delineation of white matter struc- neuroscience research. Neuron 51
tures of the fixed mouse brain using (5):527–539. https://doi.org/10.1016/j.neu
Gd-DTPA in microscopic MRI. NMR Biomed ron.2006.08.012
22(3):303–309. https://doi.org/10.1002/ 51. Choi IY, Lee SP, Guilfoyle DN, Helpern JA
nbm.1324 (2003) In vivo NMR studies of neurodegener-
41. D’Arceuil H, de Crespigny A (2007) The ative diseases in transgenic and rodent models.
effects of brain tissue decomposition on diffu- Neurochem Res 28(7):987–1001
sion tensor imaging and tractography. Neuro- 52. Robinson S, Berglass JB, Denson JL, Berkner J,
image 36(1):64–68. https://doi.org/10. Anstine CV, Winer JL, Maxwell JR, Qiu J,
1016/j.neuroimage.2007.02.039 Yang Y, Sillerud LO, Meehan WP 3rd,
42. Kim TH, Zollinger L, Shi XF, Rose J, Jeong EK Mannix R, Jantzie LL (2016) Microstructural
(2009) Diffusion tensor imaging of ex vivo and microglial changes after repetitive mild
cervical spinal cord specimens: the immediate traumatic brain injury in mice. J Neurosci
and long-term effects of fixation on diffusivity. Res. https://doi.org/10.1002/jnr.23848
Anat Rec (Hoboken) 292(2):234–241. 53. Ou X, Sun SW, Liang HF, Song SK, Gochberg
https://doi.org/10.1002/ar.20823 DF (2009) Quantitative magnetization transfer
43. Schwartz ED, Cooper, Chin CL, Wehrli S, measured pool-size ratio reflects optic nerve
Tessler A, Hackney DB (2005) Ex vivo evalua- myelin content in ex vivo mice. Magn Reson
tion of ADC values within spinal cord white Med 61(2):364–371. https://doi.org/10.
matter tracts. AJNR Am J Neuroradiol 26 1002/mrm.21850
(2):390–397 54. Perez-Torres CJ, Reynolds JO, Pautler RG
44. Alonso-Ortiz E, Levesque IR, Pike GB (2015) (2014) Use of magnetization transfer contrast
MRI-based myelin water imaging: A technical MRI to detect early molecular pathology in
review. Magn Reson Med 73(1):70–81. Alzheimer’s disease. Magn Reson Med 71
https://doi.org/10.1002/mrm.25198 (1):333–338. https://doi.org/10.1002/
45. Cong L, Muir ER, Chen C, Qian Y, Liu J, Biju mrm.24665
KC, Clark RA, Li S, Duong TQ (2016) Multi- 55. Biedermann S, Fuss J, Zheng L, Sartorius A,
modal MRI evaluation of the MitoPark mouse Falfan-Melgoza C, Demirakca T, Gass P,
MRI in Neurodegenerative Diseases 373
Magn Reson Imaging 34(7):889–895. 83. Lepore N, Brun CA, Chiang MC, Chou YY,
https://doi.org/10.1016/j.mri.2016.04.016 Dutton RA, Hayashi KM, Lopez OL, Aizen-
74. Douaud G, Jbabdi S, Behrens TE, Menke RA, stein HJ, Toga AW, Becker JT, Thompson PM
Gass A, Monsch AU, Rao A, Whitcher B, (2006) Multivariate statistics of the Jacobian
Kindlmann G, Matthews PM, Smith S (2011) matrices in tensor based morphometry and
DTI measures in crossing-fibre areas: increased their application to HIV/AIDS. Med Image
diffusion anisotropy reveals early white matter Comput Comput Assist Interv 9
alteration in MCI and mild Alzheimer’s dis- (Pt 1):191–198
ease. Neuroimage 55(3):880–890. https:// 84. Schwarz CG, Reid RI, Gunter JL, Senjem ML,
doi.org/10.1016/j.neuroimage.2010.12.008 Przybelski SA, Zuk SM, Whitwell JL, Vemuri P,
75. Zhang H, Schneider T, Wheeler-Kingshott Josephs KA, Kantarci K, Thompson PM, Peter-
CA, Alexander DC (2012) NODDI: Practical sen RC, Jack CR Jr, Alzheimer’s Disease Neu-
in vivo neurite orientation dispersion and den- roimaging Initiative (2014) Improved DTI
sity imaging of the human brain. Neuroimage registration allows voxel-based analysis that
61(4):1000–1016 outperforms tract-based spatial statistics. Neu-
76. Barazany D, Basser PJ, Assaf Y (2009) In vivo roimage 94:65–78. https://doi.org/10.1016/
measurement of axon diameter distribution in j.neuroimage.2014.03.026
the corpus callosum of rat brain. Brain 132 85. Smith SM, Johansen-Berg H, Jenkinson M,
(Pt 5):1210–1220. https://doi.org/10. Rueckert D, Nichols TE, Miller KL, Robson
1093/brain/awp042 MD, Jones DK, Klein JC, Bartsch AJ, Behrens
77. Dyrby TB, Sogaard LV, Hall MG, Ptito M, TE (2007) Acquisition and voxelwise analysis
Alexander DC (2013) Contrast and stability of multi-subject diffusion data with tract-based
of the axon diameter index from microstruc- spatial statistics. Nat Protoc 2(3):499–503.
ture imaging with diffusion MRI. Magn Reson https://doi.org/10.1038/nprot.2007.45
Med 70(3):711–721. https://doi.org/10. 86. Keihaninejad S, Zhang H, Ryan NS, Malone
1002/mrm.24501 IB, Modat M, Cardoso MJ, Cash DM, Fox
78. Daducci A, Canales-Rodriguez EJ, Zhang H, NC, Ourselin S (2013) An unbiased longitudi-
Dyrby TB, Alexander DC, Thiran JP (2015) nal analysis framework for tracking white mat-
Accelerated Microstructure Imaging via Con- ter changes using diffusion tensor imaging with
vex Optimization (AMICO) from diffusion application to Alzheimer’s disease. Neuro-
MRI data. Neuroimage 105:32–44. https:// image 72:153–163. https://doi.org/10.
doi.org/10.1016/j.neuroimage.2014.10.026 1016/j.neuroimage.2013.01.044
79. Sepehrband F, Alexander DC, Kurniawan ND, 87. DiFrancesco MW, Rasmussen JM, Yuan W,
Reutens DC, Yang Z (2016) Towards higher Pratt R, Dunn S, Dardzinski BJ, Holland SK
sensitivity and stability of axon diameter esti- (2008) Comparison of SNR and CNR for
mation with diffusion-weighted MRI. NMR in vivo mouse brain imaging at 3 and 7T
Biomed 29(3):293–308. https://doi.org/10. using well matched scanner configurations.
1002/nbm.3462 Med Phys 35(9):3972–3978. https://doi.
org/10.1118/1.2968092
80. Wedeen VJ, Wang RP, Schmahmann JD,
Benner T, Tseng WY, Dai G, Pandya DN, 88. van de Ven RC, Hogers B, van den Maagden-
Hagmann P, D’Arceuil H, de Crespigny AJ berg AM, de Groot HJ, Ferrari MD, Frants
(2008) Diffusion spectrum magnetic reso- RR, Poelmann RE, van der Weerd L, Kiihne
nance imaging (DSI) tractography of crossing SR (2007) T(1) relaxation in in vivo mouse
fibers. Neuroimage 41(4):1267–1277. brain at ultra-high field. Magn Reson Med 58
https://doi.org/10.1016/j.neuroimage. (2):390–395. https://doi.org/10.1002/mrm.
2008.03.036 21313
81. Moldrich RX, Pannek K, Hoch R, Rubenstein 89. Boretius S, Kasper L, Tammer R, Michaelis T,
JL, Kurniawan ND, Richards LJ (2010) Com- Frahm J (2009) MRI of cellular layers in mouse
parative mouse brain tractography of diffusion brain in vivo. Neuroimage 47(4):1252–1260.
magnetic resonance imaging. Neuroimage 51 https://doi.org/10.1016/j.neuroimage.
(3):1027–1036. https://doi.org/10.1016/j. 2009.05.095
neuroimage.2010.03.035 90. Harrison M, O’Brien A, Adams L, Cowin G,
82. Liu C, Li Y, Edwards TJ, Kurniawan ND, Ruitenberg MJ, Sengul G, Watson C (2013)
Richards LJ, Jiang T (2016) Altered structural Vertebral landmarks for the identification of
connectome in adolescent socially isolated spinal cord segments in the mouse. Neuro-
mice. Neuroimage 139:259–270. https://doi. image 68:22–29. https://doi.org/10.1016/j.
org/10.1016/j.neuroimage.2016.06.037 neuroimage.2012.11.048
MRI in Neurodegenerative Diseases 375
91. Raffelt D, Tournier JD, Rose S, Ridgway GR, protocol and MRI atlas of the C57BL/6J
Henderson R, Crozier S, Salvado O, Connelly mouse neocortex. Neuroimage 78:196–203.
A (2012) Apparent Fibre Density: a novel mea- https://doi.org/10.1016/j.neuroimage.
sure for the analysis of diffusion-weighted mag- 2013.04.008
netic resonance images. Neuroimage 59 93. Ullmann JFP, Watson C, Janke AL, Kurniawan
(4):3976–3994. https://doi.org/10.1016/j. ND, Paxinos G, Reutens DC (2014) An MRI
neuroimage.2011.10.045 atlas of the mouse basal ganglia. Brain Struct
92. Ullmann JF, Watson C, Janke AL, Kurniawan Funct 219(4):1343–1353. https://doi.org/
ND, Reutens DC (2013) A segmentation 10.1007/s00429-013-0572-0
Chapter 22
Abstract
Stroke consists of the loss of cerebral functions resulting from the interruption of blood supply to a region
of the brain, and represents the second cause of death and the leading cause of major disability in adults in
Europe. Stroke is a very active field of research at preclinical and clinical levels, and Magnetic Resonance
Imaging (MRI) is one of the most powerful tools that scientist and clinicians have for the study of the onset,
evolution and consequences of this devastating disease, as well as for the monitoring of the success of
available treatments, or for the development of novel therapeutic strategies.
MRI can tackle the study of stroke from different points of view, and at scales ranging from subcellular to
systems biology level. Magnetic resonance spectroscopy (MRS) allows the noninvasive measurement of the
levels of principal metabolites in the brain, and how they change during the course of the disease, or in
response to therapy. Glutamate, in particular, is very important in the field of stroke. Several anatomical MR
techniques allow the characterization of the lesion volumes, the formation of cytotoxic and vasogenic
edema, changes in cerebral blood flow and volume, structural changes in gray and white matter, the
obtaining of the vascular architecture and status, etc. At functional level, diverse modalities of functional
MRI (fMRI) allow the assessment of the alteration in the function and organization of neuronal networks of
the subject under study, as a consequence of the disease or in response to treatment. Finally, emerging
imaging modalities that include temperature and pH mapping of the brain, imaging by chemical exchange
saturation transfer effect (CEST), all of them closely related to tissue status, or the use of contrast agents for
the targeting of tissue in theranostic approaches or for cell tracking studies in cell-based therapies, etc., are
only a few examples of the power and versatility of MRI as a definitive tool for the study of stroke.
In this work we will set our focus on preclinical imaging of stroke models, emphasizing the most
commonly used imaging modalities in a stroke-dedicated research laboratory. However, advanced techni-
ques will be briefly discussed, providing references to specialized literature for more advanced readers. Thus,
the aim of this chapter consist in the description of a simple imaging protocol for the study of the most
important and common aspects of stroke in a research laboratory.
Key words MRI, MRA, ADC, DWI, PWI, Stroke, Ischemia, Angiography, Perfusion, Diffusion
1 Introduction
Stroke is the second cause of death in Europe, and the leading cause
of major disability in adults (with 1484 DALY -Disability Adjusted
Life Years- per 100,000 population) [1, 2]. Ischemic stroke consists
of the loss of cerebral functions resulting from the interruption of
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0_22, © Springer Science+Business Media, LLC 2018
377
378 Pedro Ramos-Cabrer and Daniel Padro
Fig. 1 Evolution of T2 weighting contrast in the brain of a rat submitted to a transient (60 min) occlusion of the
middle cerebral artery (MCAo). The lesion is not (or barely) visible during the occlusion and the first hours after
it (hyper-acute and acute phases: t < 6–8 h). At subacute to chronic stages (t > 6–8 h), the lesion is clearly
visible as a hyper-intense area in the brain
MRI in Stroke 381
Table 1
Recommended imaging protocols at different stages of the progression of cerebral ischemia
MCAo stage Main assessments T2w T2* (SWI) DWI/ADC TOF-angio Total time
0 00
Pre- Discard abnormalities 9 36 – – 40 200 130 3800
During Cytotoxic edema, bleeding – 20 5900 80 000 40 200 150 100
<6 h post Reperfusion, hemorrhage – 20 5900 – 40 200 70 100
>6 h post Vasogenic edema, necrosis, 90 3600 20 5900 – – 130 3500
hemorrhage
2 Materials
2.1 Rodent Models 1. Rats (250 50 g), preferred strains: Sprague Dawley, Wistar
of Cerebral Ischemia Kyoto.
2. Anesthetics: Isofluorane or Sevofluorane.
3. Appropriate anesthetic gas vaporizer.
4. Carrier gases for anesthesia: 1–1.5 L/min, N2:02
(70:30–80:20) mixtures, or medical air.
5. Set of general surgical tools.
6. Disposable material (syringes, needles, falcon tubes, etc.)
7. 0.9% NaCl solution for infusion (saline).
8. Betadine® dermic solution 10%.
9. Braided silk suture: 4/0, 6/0, 10/0.
10. Monofilament for MCA occlusion: set with 0.35 mm,
0.37 mm, 0.39 mm, 0.41 mm—4-0 medium MCAO suture
L34 PK10 (Doccol Corporation, Sharon, MA, USA).
11. Disposable low temperature ophthalmic cautery, 28 mm fine
tip (FIAB Spa, icchio—Firenze, Italy, ref. F7255).
12. Feedback controlled heating mat for surgery with rectal PT100
probe (Neoos Biotec S.L., Pamplona, Spain).
382 Pedro Ramos-Cabrer and Daniel Padro
2.3 Image Analysis 1. A general purpose computer (In our case, HP Z230 Worksta-
tion with an Intel® Xenon® CPU E3–1246 v3 @ 3.5 GHz,
8 Gb RAM running a 64-bit Windows 7 professional operative
system).
2. Image-J V1.50b, the open-source and publically available
image processing software package for JAVA of the National
Institutes of Health (Bethesda, MD, USA), developed by W. S:
Rasband (available at http://imagej.nih.gov/ij/, 1997–2016).
3. Bruker’s ParaVision 6® software suit (Bruker Biospin GmbH,
Ettlingen, Germany).
4. To build ADC maps (Fig. 2), DWI images acquired with b ¼ 0
were divided by DWI images acquired with b ¼ 800 mm2 s.
Then, the natural logarithm of the resulting image was calcu-
lated and divided by 0.8 (b ¼ 800 divided by 103). The result-
ing maps provide a pixel-by-pixel representation of 103 ADC
(mm2 s1).
3 Methods
3.1 Stroke Model: A detailed description of surgical procedures and different models
Occlusion of middle cerebral artery occlusion can be found elsewhere
of the Medial Cerebral [21, 22].
Artery (MCAo)
MRI in Stroke 383
Fig. 2 Diffusion-weighted images (DWI, top row) and Apparent Diffusion Coefficient maps (ADC, bottom row) at
the same planes and acquisition times of the rat brain shown in Fig. 1. During the occlusion (hyper-acute
phase) areas suffering cytotoxic edema are visible as hyper-intensities in DWI, or as areas of decreased ADC
value (darker blue color). In models of transient occlusion, the contrast in DWI and ADC vanishes briefly after
reperfusion of the MCA (acute phase), showing the false impression that the lesion is disappearing. At
subacute to chronic stages (t > 6–8 h) the lesion becomes visible again, but with opposite contrast (hypo-
intense signal in DWI and elevated ADC values in ADC maps). DWI/ADC imaging is particularly useful during
the occlusion, but may lead to false negatives during periods of reperfusion
step). The knot at the stump of the ECA is strongly tighten, the
clamp of the ACA is retrieved, two or three stitches are used to
close the neck of the animal, and the animal is allowed to
recover in its cage.
11. A laser Doppler flowmeter may be used in the head of the
animal to assess the drop of blood flow during the MCA0
procedure (see [22, 23] for details).
12. Typical complications during surgery include excessive interac-
tion with the vagus nerve, internal bleeding by rupture of a
vessel during the introduction, but most likely during retriev-
ing of the thread, excessive bleeding during the transient loos-
ening of the ECA knot (to introduce/retrieve the threat from
the stump) and intensive increase of intracranial pressure due
to excessive brain edema (consequent to the occlusion of the
MCA). Most of them will result on the death of the animal
either during surgery or in the first hours after it.
13. The use of a pain killer such as buprenorphine (twice a day
during 48–72 h, 0.05–0.1 mg/kg) is recommended to treat
pain from surgical procedures.
14. Preparation of a food paste with pellet powder mixed with
water, and hydratation of the animal via i.p. or s.c. injections
of saline may be necessary to facilitate the feeding of the animal
during the first 78 h, since bad general condition is expected
for the animal (usually due to vasogenic edema in the brain
with displacement of the midline), which starts to resolve
48–72 h after occlusion.
4 Notes
Fig. 3 Susceptibility-weighted images (SWI) showing enhanced T2* contrast of the brain. Areas of elevated
oxygen extraction (reduced blood supply) lead to marked susceptibility effects due to high levels of
deoxyhemoglobin. This sequence is very sensible to the presence of small bleedings or to hemorrhages
(not present in the images shown here), as strong hypo-intense signal. Otherwise, T2* contrast follows a
similar pattern as T2 contrast in cerebral ischemia. Nevertheless, during the occlusion of the MCA, the hypo-
perfused areas of the brain show a marked hypo-intense signal (see panel “occlusion” in Fig. 4). Although DWI
is the most powerful image modality for detecting acute cerebral ischemia, SWI may be used as an adjunct to
characterize the affected vascular territory during this phase
388 Pedro Ramos-Cabrer and Daniel Padro
Fig. 4 Axial (top row, caudal to head view) and coronal (bottom row, top to bottom view) maximal intensity
projections (MIP) of MR angiographies of the head of a rat submitted to transient MCAo surgery. Arrows signal
the right MCA of the animal (the one occluded) while stars show the branching place of the MCA to the circle or
Willis. During the occlusion the right MCA is not seen, confirming a successful occlusion of this artery. When
the thread is withdrawn from the vessel, the angiography confirms a successful reperfusion of the MCA
Acknowledgements
References
1. Nichols M, Townsend N, Scarborough P, Ray- de Vaccaro KC, Couser W, Cowie BC, Criqui
ner M (2013) Cardiovascular disease in Eur- MH, Cross M, Dabhadkar KC, Dahiya M,
ope: epidemiological update. Eur Heart J 34 Dahodwala N, Damsere-Derry J, Danaei G,
(39):3028–3034. https://doi.org/10.1093/ Davis A, De Leo D, Degenhardt L,
eurheartj/eht356 Dellavalle R, Delossantos A, Denenberg J,
2. Murray CJ, Vos T, Lozano R, Naghavi M, Flax- Derrett S, Des Jarlais DC, Dharmaratne SD,
man AD, Michaud C, Ezzati M, Shibuya K, Dherani M, Diaz-Torne C, Dolk H, Dorsey
Salomon JA, Abdalla S, Aboyans V, ER, Driscoll T, Duber H, Ebel B, Edmond K,
Abraham J, Ackerman I, Aggarwal R, Ahn SY, Elbaz A, Ali SE, Erskine H, Erwin PJ,
Ali MK, Alvarado M, Anderson HR, Anderson Espindola P, Ewoigbokhan SE, Farzadfar F,
LM, Andrews KG, Atkinson C, Baddour LM, Feigin V, Felson DT, Ferrari A, Ferri CP,
Bahalim AN, Barker-Collo S, Barrero LH, Bar- Fevre EM, Finucane MM, Flaxman S,
tels DH, Basanez MG, Baxter A, Bell ML, Flood L, Foreman K, Forouzanfar MH,
Benjamin EJ, Bennett D, Bernabe E, Fowkes FG, Fransen M, Freeman MK, Gabbe
Bhalla K, Bhandari B, Bikbov B, Bin BJ, Gabriel SE, Gakidou E, Ganatra HA,
Abdulhak A, Birbeck G, Black JA, Garcia B, Gaspari F, Gillum RF, Gmel G,
Blencowe H, Blore JD, Blyth F, Bolliger I, Gonzalez-Medina D, Gosselin R, Grainger R,
Bonaventure A, Boufous S, Bourne R, Grant B, Groeger J, Guillemin F, Gunnell D,
Boussinesq M, Braithwaite T, Brayne C, Gupta R, Haagsma J, Hagan H, Halasa YA,
Bridgett L, Brooker S, Brooks P, Brugha TS, Hall W, Haring D, Haro JM, Harrison JE,
Bryan-Hancock C, Bucello C, Buchbinder R, Havmoeller R, Hay RJ, Higashi H, Hill C,
Buckle G, Budke CM, Burch M, Burney P, Hoen B, Hoffman H, Hotez PJ, Hoy D,
Burstein R, Calabria B, Campbell B, Canter Huang JJ, Ibeanusi SE, Jacobsen KH, James
CE, Carabin H, Carapetis J, Carmona L, SL, Jarvis D, Jasrasaria R, Jayaraman S,
Cella C, Charlson F, Chen H, Cheng AT, Johns N, Jonas JB, Karthikeyan G,
Chou D, Chugh SS, Coffeng LE, Colan SD, Kassebaum N, Kawakami N, Keren A, Khoo
Colquhoun S, Colson KE, Condon J, Connor JP, King CH, Knowlton LM, Kobusingye O,
MD, Cooper LT, Corriere M, Cortinovis M, Koranteng A, Krishnamurthi R, Laden F,
MRI in Stroke 391
Lalloo R, Laslett LL, Lathlean T, Leasher JL, 3. Dyken ML (2010) What lessons have we
Lee YY, Leigh J, Levinson D, Lim SS, Limb E, learned in the past 40 years? Stroke 41
Lin JK, Lipnick M, Lipshultz SE, Liu W, (6):1073–1075. https://doi.org/10.1161/
Loane M, Ohno SL, Lyons R, Mabweijano J, STROKEAHA.110.584011
MacIntyre MF, Malekzadeh R, Mallinger L, 4. Hachinski V (2007) The 2005 Thomas Willis
Manivannan S, Marcenes W, March L, Margo- Lecture: stroke and vascular cognitive
lis DJ, Marks GB, Marks R, Matsumori A, impairment: a transdisciplinary, translational
Matzopoulos R, Mayosi BM, McAnulty JH, and transactional approach. Stroke 38
McDermott MM, McGill N, McGrath J, (4):1396. https://doi.org/10.1161/01.STR.
Medina-Mora ME, Meltzer M, Mensah GA, 0000260101.08944.e9
Merriman TR, Meyer AC, Miglioli V, 5. Hossmann KA (2008) Cerebral ischemia:
Miller M, Miller TR, Mitchell PB, Mock C, models, methods and outcomes. Neurophar-
Mocumbi AO, Moffitt TE, Mokdad AA, macology 55(3):257–270. https://doi.org/
Monasta L, Montico M, Moradi-Lakeh M, 10.1016/j.neuropharm.2007.12.004
Moran A, Morawska L, Mori R, Murdoch
ME, Mwaniki MK, Naidoo K, Nair MN, 6. Runck F, Steiner RP, Bautz WA, Lell MM
Naldi L, Narayan KM, Nelson PK, Nelson (2008) MR imaging: influence of imaging
RG, Nevitt MC, Newton CR, Nolte S, technique and postprocessing on measurement
Norman P, Norman R, O’Donnell M, of internal carotid artery stenosis. AJNR Am J
O’Hanlon S, Olives C, Omer SB, Ortblad K, Neuroradiol 29(9):1736–1742. https://doi.
Osborne R, Ozgediz D, Page A, Pahari B, Pan- org/10.3174/ajnr.A1179
dian JD, Rivero AP, Patten SB, Pearce N, 7. Ewing JR, Knight RA, Nagaraja TN, Yee JS,
Padilla RP, Perez-Ruiz F, Perico N, Nagesh V, Whitton PA, Li L, Fenstermacher
Pesudovs K, Phillips D, Phillips MR, Pierce K, JD (2003) Patlak plots of Gd-DTPA MRI data
Pion S, Polanczyk GV, Polinder S, Pope CA yield blood-brain transfer constants concor-
3rd, Popova S, Porrini E, Pourmalek F, dant with those of 14C-sucrose in areas of
Prince M, Pullan RL, Ramaiah KD, blood-brain opening. Magn Reson Med 50
Ranganathan D, Razavi H, Regan M, Rehm (2):283–292. https://doi.org/10.1002/
JT, Rein DB, Remuzzi G, Richardson K, Rivara mrm.10524
FP, Roberts T, Robinson C, De Leon FR, 8. Abo-Ramadan U, Durukan A, Pitkonen M,
Ronfani L, Room R, Rosenfeld LC, Marinkovic I, Tatlisumak E, Pedrono E,
Rushton L, Sacco RL, Saha S, Sampson U, Soinne L, Strbian D, Tatlisumak T (2009)
Sanchez-Riera L, Sanman E, Schwebel DC, Post-ischemic leakiness of the blood-brain bar-
Scott JG, Segui-Gomez M, Shahraz S, Shepard rier: a quantitative and systematic assessment
DS, Shin H, Shivakoti R, Singh D, Singh GM, by Patlak plots. Exp Neurol 219(1):328–333.
Singh JA, Singleton J, Sleet DA, Sliwa K, https://doi.org/10.1016/j.expneurol.2009.
Smith E, Smith JL, Stapelberg NJ, Steer A, 06.002
Steiner T, Stolk WA, Stovner LJ, Sudfeld C, 9. Barbier EL, Lamalle L, Decorps M (2001)
Syed S, Tamburlini G, Tavakkoli M, Taylor Methodology of brain perfusion imaging. J
HR, Taylor JA, Taylor WJ, Thomas B, Thom- Magn Reson Imaging 13(4):496–520
son WM, Thurston GD, Tleyjeh IM, 10. Campos F, Sobrino T, Ramos-Cabrer P,
Tonelli M, Towbin JA, Truelsen T, Tsilimbaris Argibay B, Agulla J, Perez-Mato M,
MK, Ubeda C, Undurraga EA, van der Werf Rodriguez-Gonzalez R, Brea D, Castillo J
MJ, van Os J, Vavilala MS, (2011) Neuroprotection by glutamate oxaloac-
Venketasubramanian N, Wang M, Wang W, etate transaminase in ischemic stroke: an exper-
Watt K, Weatherall DJ, Weinstock MA, imental study. J Cereb Blood Flow Metab 31
Weintraub R, Weisskopf MG, Weissman MM, (6):1378–1386. https://doi.org/10.1038/
White RA, Whiteford H, Wiebe N, Wiersma jcbfm.2011.3
ST, Wilkinson JD, Williams HC, Williams SR,
Witt E, Wolfe F, Woolf AD, Wulf S, Yeh PH, 11. Ramos-Cabrer P, Campos F, Sobrino T, Cas-
Zaidi AK, Zheng ZJ, Zonies D, Lopez AD, tillo J (2011) Targeting the ischemic penum-
AlMazroa MA, Memish ZA (2012) Disability- bra. Stroke 42(1 Suppl):S7–11. https://doi.
adjusted life years (DALYs) for 291 diseases org/10.1161/STROKEAHA.110.596684
and injuries in 21 regions, 1990–2010: a sys- 12. Weber R, Ramos-Cabrer P, Hoehn M (2006)
tematic analysis for the Global Burden of Dis- Present status of magnetic resonance imaging
ease Study 2010. Lancet 380 and spectroscopy in animal stroke models. J
(9859):2197–2223. https://doi.org/10. Cereb Blood Flow Metab 26(5):591–604.
1016/S0140-6736(12)61689-4 https://doi.org/10.1038/sj.jcbfm.9600241
392 Pedro Ramos-Cabrer and Daniel Padro
13. Cai K, Haris M, Singh A, Kogan F, Greenberg Use of fluid attenuated inversion recovery
JH, Hariharan H, Detre JA, Reddy R (2012) (FLAIR) pulse sequences in MRI of the brain.
Magnetic resonance imaging of glutamate. Nat J Comput Assist Tomogr 16(6):841–844
Med 18(2):302–306. https://doi.org/10. 19. Wegener S, Weber R, Ramos-Cabrer P,
1038/nm.2615 Uhlenkueken U, Sprenger C, Wiedermann D,
14. Stejskal EO, Tanner JE (1965) Spin diffusion Villringer A, Hoehn M (2006) Temporal pro-
measurements: spin echoes in the presence of a file of T2-weighted MRI distinguishes between
time dependent field gradient. J Chem Phys pannecrosis and selective neuronal death after
42:288–292 transient focal cerebral ischemia in the rat. J
15. Heidenreich JO, Hsu D, Wang G, Jesberger Cereb Blood Flow Metab 26(1):38–47.
JA, Tarr RW, Zaidat OO, Sunshine JL (2008) https://doi.org/10.1038/sj.jcbfm.9600166
Magnetic resonance imaging results can affect 20. Van der Linden A, Van Camp N, Ramos-
therapy decisions in hyperacute stroke care. Cabrer P, Hoehn M (2007) Current status of
Acta Radiol 49(5):550–557. https://doi.org/ functional MRI on small animals: application
10.1080/02841850801958320 to physiology, pathophysiology, and cognition.
16. Chen F, Ni YC (2012) Magnetic resonance NMR Biomed 20(5):522–545. https://doi.
diffusion-perfusion mismatch in acute ischemic org/10.1002/nbm.1131
stroke: an update. World J Radiol 4(3):63–74. 21. Dirnagl U (2016) Rodent models of stroke. In:
https://doi.org/10.4329/wjr.v4.i3.63 Neuromethods, vol 47. Humana Press,
17. Tournier JD, Mori S, Leemans A (2011) Dif- New York
fusion tensor imaging and beyond. Magn 22. Wang-Fischer Y (2008) Manual of stroke mod-
Reson Med 65(6):1532–1556. https://doi. els in rats. CRC Press, Boca Raton, FL
org/10.1002/mrm.22924 23. Le Bihan D, Mangin JF, Poupon C, Clark CA,
18. Hajnal JV, Bryant DJ, Kasuboski L, Pattany Pappata S, Molko N, Chabriat H (2001) Dif-
PM, De Coene B, Lewis PD, Pennock JM, fusion tensor imaging: concepts and applica-
Oatridge A, Young IR, Bydder GM (1992) tions. J Magn Reson Imaging 13(4):534–546
Part VI
Other Applications
Chapter 23
Abstract
The integrity of the blood–brain barrier (BBB) can be noninvasively monitored by magnetic resonance
imaging (MRI). Conventional MR contrast agents (CAs) containing gadolinium are used in association
with MRI in routine clinical practice to detect and quantify BBB leakage. Under normal circumstances CAs
do not cross the intact BBB. However due to their small size they extravasate from the blood into the brain
tissue even when the BBB is partially compromised. Here we describe an MR method based on
T1-weighted images taken prior to and after CA injection. This MR method is useful for investigating
BBB permeability in in vivo mouse models and can be easily applied in a number of experimental disease
conditions including neuroinflammation disorders, or to assess (un)wanted drug effects.
Key words Magnetic resonance imaging (MRI), Mouse, Blood brain barrier (BBB), contrast agent
(CA)
1 Introduction
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0_23, © Springer Science+Business Media, LLC 2018
395
396 Min-Chi Ku et al.
2 Materials
2.1 Animals Mice. All mice are housed under standard conditions according to
the animal regulations in our institute. We used female mice
(C57BL/6 J) with a body weight of 16–20 g. Other strains, ages,
and genders might require some changes in the experimental
design (see Note 1).
MRI assessment of BBB 397
2.3 Magnetic This method requires access to an high field MRI system in addi-
Resonance Imaging tion suitable accessories for the MR acquisition such as radio fre-
(MRI) quency (RF) antennas; equipment for animal positioning,
anesthetizing, warming, and monitoring of physiological para-
meters of the animals to be imaged; and trained personnel for
operating the MRI system.
1. MRI system: A dedicated small animal MR system with a
magnetic field strength of 4.7 Tesla or higher is recommended.
Here we describe the use of a 9.4 Tesla 20 cm bore system
(Biospec 94/20, Bruker Biospin, Ettlingen, Germany)
equipped with a gradient system integrated with shim set
(B-GA12S2, Bruker Biospin, Ettlingen, Germany; gradient
amplitude 440 mT/m, max. Slew rate 3440 T/m/s).
2. Radio frequency (RF) coils: Use RF volume coils (reception
house made birdcage) suitable for mouse brain imaging, such
as a 16 mm volume resonator (custom-made), or surface RF
coils, such as a 2 2 element mouse brain RF coil array linear
reception-only surface coil (e.g., model: T11071 V3, Bruker
Biospin, Ettlingen, Germany) combined with a volume reso-
nator for transmit (model: T10325_V3, Bruker Biospin,
Ettlingen, Germany); for high resolved imaging cryogenically
cooled RF coil (CryoProbe, Bruker) is recommended
[18, 19]. Here we used the first mentioned RF coil setup, as
it also provides high spatial resolution.
398 Min-Chi Ku et al.
3 Methods
3.1 Preparation 1. Start the ParaVision software (here we used ParaVision 5.0)
of MRI and—only once prior to the first experiment—create and store
the following MR protocols.
(a) Protocol_TriPilot (pilot scan for checking the mouse head
position): conventional FLASH pilot with three slices in
each direction (axial, coronal, sagittal).
(b) Protocol_Pilot_Axial (axial pilot scan): FLASH sequence,
repetition time (TR) ¼ 560 ms, effective echo time
(TE) ¼ 3 ms, the geometry define as an axial field of
view (FOV) ¼ 25 25 mm2, matrix size ¼ 256 256,
ten slices with a thickness of 0.7 mm.
(c) Protocol_T1 map (for T1-mapping): RARE sequence,
TR ¼ 9000, 2400, 1480, 940, 650, 418 ms (set up six
separate scans or use the RAREVTR protocol, which
automatically sets six different TRs for the 6 consecutive
scans). Echo times ¼ 8.1, rare factor ¼ 4, define as
MRI assessment of BBB 399
3.3 MRI Pre-scans 1. Start the ParaVision software and register a new subject and
study. For the first scan select the Protocol_TriPilot.
MRI assessment of BBB 401
2. Tune and match the mouse brain RF coils using the Wobble
function.
3. Start the first pilot scan and verify on the acquired images
whether the brain is in the center of the magnet (field of
view) and well positioned within the signal intensity profile. If
necessary correct the animal or/and laser marker position.
Repeat the tuning/matching and redo this pilot scan (see
Note 9).
4. Load the Protocol_Pilot_Axial (axial pilot scan), edit the geom-
etry based on the TriPilot scan to position the brain slices
correctly in the center (Fig. 1) and run the scan.
5. Clone the Protocol_Pilot_Axial, change the slice orientation to
coronal, and run the scan.
6. Clone the Protocol_Pilot_Axial, change the slice orientation to
sagittal, and run the scan.
3.4 Baseline 1. Load the Protocol_T1 map and edit the geometry based on the
(Pre-contrast) Images coronal and sagittal views such that the slice package encloses
the brain (Fig. 2).
2. Clone Protocol_T1 map and run scans with GOP. Result from
pre-CA scans are shown in Fig. 5 (upper row) (see Note 10).
3. Load the Protocol_IR-GRE and copy the geometry from pre-
vious scan. Run the scan and check image geometry and
quality.
4. Load the Protocol_T1w-GRE and position one axial slice such
that it crosses the brain in the center. Run the scan.
Fig. 1 Coronal and sagittal image of a mouse brain from the Protocol_TriPilot
(pilot scan) and show how to place the slice position for Protocol_Pilot_Axial
402 Min-Chi Ku et al.
Fig. 2 Position planning for running Protocol_T1 map based on the coronal and
sagittal pilot scans. Adjust the angle according to the brain orientation (see Note 11)
Table 1
Step-down injection method for injecting the CA over 20 mins
For 1 mmol/kg
20 g Rate (μL/min) Duration (mins) Volume (μL)
50 0.5 25
40 0.5 20
30 1 30
20 1 20
16 1 16
14 1 14
10 2 20
8 3 24
6 5 30
4 5 20
Total 20 219
3.5 Post-contrast 1. There are two ways of CA injection. The most common one is a
Images bolus injection (single fast inject) and the other is an infusion in
a step-down manner. Table 1 shows an example (for a 20 g
mouse) for injecting CA (1 mmol/kg) in a step-down manner
over 20 min via the tail vein using a syringe pump, which can
regulate the injection volume and injection rates. For mouse
with different weights, adjust the CA amount and flow rates
accordingly (see Note 12).
MRI assessment of BBB 403
Fig. 3 Axial view images acquired prior to (Pre-CA) and after a successful
injection (Post-CA) with CA. Protocol_T1w-GRE images (right panel; Post-CA)
depict a succeeded injection as shown by the significant signal intensity
changes within the head vessels and muscle tissue but not in the healthy
brain (see Note 14)
Fig. 4 Axial view images acquired prior to (Pre-CA) and after (Post-CA) a failed
injection of CA. Protocol_T1w-GRE images (right panel; Post-CA) depict an
incomplete CA injection, the hyperintensity is only seen in bigger vessels of
the head but not in muscle tissue (see Note 15)
Fig. 5 Axial view images acquired prior to (Pre-CA; upper row) and after (Post-CA; lower row) CA injection and
scanned with six different TRs. Protocol_T1map images show the signal intensity changes between the scans
acquired with different TR values. Lower row shows increased signal intensity for each TR value in the Post-CA
images, compared to Pre-CA images
Fig. 6 Axial view images acquired prior to (Pre-CA) and after (Post-CA) injection. Protocol_IR-GRE provides
contrast between white/gray matters without CA (left panel; Pre-CA). This scan is informative about the
integrity of BBB following a successful CA injection, and can be performed prior to a T1 map, which is usually
more time consuming. The presence of CA is shown as increased signal intensity within the head region and
here also in the brain, indicating BBB leakage labeled with yellow arrows (right panel; Post-CA)
7. Clone Protocol_T1 map from previous scans and run scan with
GOP. Results from post-CA scans shown in Fig. 5 (lower row).
8. Clone Protocol_IR-GRE and run with GOP. Scan result shown
in Fig. 6.
Fig. 7 Representative T1 maps before (Pre-CA) and after (Post-CA) CA injection. T1 maps were created from
images by running Protocol_T1 map and post processed. T1 maps were calculated by fitting a mono-
exponential curve to the signal intensities of the six T1-weighted images using custom-made software
program (: MATLAB). Color coded ΔT1 map of relative change in T1 were calculated as ΔT1 ¼ |T1,postCA
/ T1,preCA| - 1 100. The result revealed that the BBB leakage detected by post-pre CA approach can be
seen in paraventricular space
3.7 Data Analysis 1. Open the reconstructed MR images in MATLAB (:; see Mate-
rials) by loading the binary 2dseq files of the RAREVTR scan
and import TR parameters from the method text file.
2. Perform pixel-wise exponential curve fitting of the equation:
4 Notes
1. When doing a study, make sure the body weight of each mouse
within a study group is similar.
2. A syringe pump is not necessary when injecting contrast agents
as bolus.
406 Min-Chi Ku et al.
Acknowledgement
References
12. Absinta M, Sati P, Reich DS (2016) Advanced blood volume, and blood-brain barrier perme-
MRI and staging of multiple sclerosis lesions. ability, using dynamic contrast-enhanced T(1)-
Nat Rev Neurol 12(6):358–368. https://doi. weighted MRI at 3 tesla. Magn Reson Med 62
org/10.1038/nrneurol.2016.59 (5):1270–1281. https://doi.org/10.1002/
13. Lin Y, Pan Y, Wang M, Huang X, Yin Y, mrm.22136
Wang Y, Jia F, Xiong W, Zhang N, Jiang JY 17. Feng Y, Jeong EK, Mohs AM, Emerson L, ZR
(2012) Blood-brain barrier permeability is pos- L (2008) Characterization of tumor angiogen-
itively correlated with cerebral microvascular esis with dynamic contrast-enhanced MRI and
perfusion in the early fluid percussion-injured biodegradable macromolecular contrast agents
brain of the rat. Lab Invest 92 in mice. Magn Reson Med 60(6):1347–1352.
(11):1623–1634. https://doi.org/10.1038/ https://doi.org/10.1002/mrm.21791
labinvest.2012.118 18. Niendorf T, Pohlmann A, Reimann HM,
14. Heye AK, Culling RD, Valdes Hernandez Waiczies H, Peper E, Huelnhagen T,
Mdel C, Thrippleton MJ, Wardlaw JM (2014) Seeliger E, Schreiber A, Kettritz R, Strobel K,
Assessment of blood-brain barrier disruption Ku MC, Waiczies S (2015) Advancing cardio-
using dynamic contrast-enhanced MRI. A sys- vascular, neurovascular, and renal magnetic res-
tematic review. NeuroImage Clin 6:262–274. onance imaging in small rodents using
https://doi.org/10.1016/j.nicl.2014.09.002 cryogenic radiofrequency coil technology.
15. Barnes SR, Ng TS, Montagne A, Law M, Zlo- Front Pharmacol 6:255. https://doi.org/10.
kovic BV, Jacobs RE (2016) Optimal acquisi- 3389/fphar.2015.00255
tion and modeling parameters for accurate 19. Wagenhaus B, Pohlmann A, Dieringer MA,
assessment of low Ktrans blood-brain barrier Els A, Waiczies H, Waiczies S, Schulz-Menger J,
permeability using dynamic contrast-enhanced Niendorf T (2012) Functional and morpho-
MRI. Magn Reson Med 75(5):1967–1977. logical cardiac magnetic resonance imaging of
https://doi.org/10.1002/mrm.25793 mice using a cryogenic quadrature radiofre-
16. Larsson HB, Courivaud F, Rostrup E, Hansen quency coil. PLoS One 7(8):e42383. https://
AE (2009) Measurement of brain perfusion, doi.org/10.1371/journal.pone.0042383
Chapter 24
Abstract
Over the past few years, many papers have been published on the nanomedical applications of magnetic
nanoparticles. However, most studies lack important information about the in vivo behavior of these
nanoparticles, which is a critical aspect for their rational design. In this chapter we describe a simple
protocol for the in vivo characterization of the pharmacokinetics of magnetic nanoparticles intravenously
injected in mice, using basic MRI sequences.
Key words Magnetic nanoparticles, Dynamic T2-weighted MRI, T2 mapping, CPMG, Pharmacoki-
netics, Relative enhancement
1 Introduction
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0_24, © Springer Science+Business Media, LLC 2018
409
410 Carlos Caro et al.
2 Materials
2.1 Animals 1. Balb/c mice with ca. 22 g in weight, provided by Janvier Labs
and Small Material (Le Genest-Saint-Isle, France).
2. 26G catheters for tail vein cannulation.
3. Heparinized saline (50 U/mL).
2.2 Magnetic The studies reported in this chapter were performed with 6 nm
Nanoparticles manganese ferrite nanoparticles (MnFe2O4) coated with a 3 kDa
polyethylene glycol (PEG) chain.
2.4 Other Equipment 1. Vaporizer and induction chamber to anesthetize the animals
outside the MR spectrometer.
2. Small Animal Monitoring and Gating system (Small Animal
Instruments Inc. Stony Brook, NY, USA), provided with respi-
ration and temperature sensors.
3. Animal physiology monitoring software (PC-SAM 32 v8.02,
Small Animal Instrument Inc.).
3 Methods
3.2 Animal Handling 1. Turn on the gas flow (oxygen or air at a flow rate of 1–1.5 mL/
min) and anesthesia (4–5% isoflurane), directed to the induc-
tion chamber, and place the mouse inside.
2. Once the animal is asleep, redirect the anesthesia to the outside
animal bed, reduce the isoflurane to 2–2.5%, and place the
mouse on the outside animal bed for tail vein cannulation.
3. Proceed with tail vein cannulation using a 26G catheter and
heparinized saline.
4. Redirect the anesthesia to the animal holder already placed on
the scanner, and quickly move the animal to the animal cradle,
which should have a temperature control system (water or air)
to maintain the temperature between 36.5 and 37.5 C during
the experimental procedure.
5. Once sensor for breathing and rectal temperature probe are
positioned correctly, check the animal monitoring unit. Anes-
thesia should be adjusted throughout the experiment to main-
tain a breathing rate between 50 and 80 breaths per minute.
6. At the end of the experiment, remove the animal from the
animal holder and place it on a warm location until it is awake
(2–3 min). Then return the animal to its cage.
3.4 Pharma- The acquisition scheme used for the pharmacokinetics studies is as
cokinetics Study follows: high resolution T2-weighted images, T2 map, intravenous
injection of magnetic nanoparticles, dynamic T2-weighted
sequence (35 min), T2-weighted images and T2 map (see Note 3).
Pharmacokinetics of Magnetic Nanoparticles 413
Fig. 1 Representative T2-weighted images with respiratory gating acquired at different experimental times
3.5 Data Analysis 1. Load the T2-weighted sequence in the Paravision image display
and processing program and look for the image on the series
3.5.1 Dynamic
that gives good contrast between the different tissue regions
T2-Weighted Sequence
(see Note 6).
414 Carlos Caro et al.
Fig. 3 Pharmacokinetics of magnetic nanoparticles injected intravenously in mice. The upper panel shows the
signal intensity in arbitrary units, and the lower panel shows the normalized signal expressed as relative
enhancement (RE)
Mðt Þ ¼ M0 eTE=T2 þ n
4 Notes
Fig. 4 Long-term pharmacokinetics by quantitative T2 measurements. Selected regions of interest (ROIs) are
outlined with dashed lines on one of the series images that provides good contrast. The right panel shows the
exponential fittings for T2 calculation in the different ROIs. Table at the bottom shows the average T2 values
(standard deviation) and the average increments in T2 (standard deviation) of three different animals at
different experimental times
Acknowledgements
The MRI system used in this work has been funded by the Spanish
Ministry of Science and Innovation (National Plan for Scientific
Research, Development and Technological Innovation 2008-
2011) and the European Regional Development Fund
(PCT-420000-2010-3).
References
1. Petros RA, Desimone JM (2010) Strategies in 5. Pernia Leal M, Torti A, Riedinger A, La
the design of nanoparticles for therapeutic Fleur R, Petti D, Cingolani R, Bertacco R, Pel-
applications. Nat Rev Drug Discov 9 legrino T (2012) Controlled release of doxo-
(8):615–627 rubicin loaded within magnetic thermo-
2. Lee JH, Jang JT, Choi JS, Moon SH, Noh SH, responsive nanocarriers under magnetic and
Kim JW, Kim JG, Kim IS, Park KI, Cheon J thermal actuation in a microfluidic channel.
(2011) Exchange-coupled magnetic nanopar- ACS Nano 6(12):10535–10545
ticles for efficient heat induction. Nat Nano- 6. Gazeau F, Lévy M, Wilhelm C (2008) Opti-
technol 6(7):418–422 mizing magnetic nanoparticle design for
3. Mornet S, Vasseur S, Grasset F, Duguet E nanothermotherapy. Nanomedicine 3
(2004) Magnetic nanoparticle design for med- (6):831–844
ical diagnosis and therapy. J Mater Chem 14 7. Figuerola A, Di Corato R, Manna L, Pellegrino
(14):2161–2175 T (2010) From iron oxide nanoparticles
4. Lee JH, Huh YM, Jun YW, Seo JW, Jang JT, towards advanced iron-based inorganic materi-
Song HT, Kim S, Cho EJ, Yoon HG, Suh JS, als designed for biomedical applications. Phar-
Cheon J (2007) Artificially engineered mag- macol Res 62(2):126–143
netic nanoparticles for ultra-sensitive molecular 8. Challenor M, Gong P, Lorenser D, House MJ,
imaging. Nat Med 13(1):95–99 Woodward RC, St Pierre T, Fitzgerald M,
418 Carlos Caro et al.
Dunlop SA, Sampson DD, Iyer KS (2014) The 20. Garcı́a KP, Zarschler K, Barbaro L, Barreto JA,
influence of NaYF4:Yb,Er size/phase on the O’Malley W, Spiccia L, Stephan H, Graham B
multimodality of co-encapsulated magnetic (2014) Zwitterionic-coated "stealth" nanopar-
photon-upconverting polymeric nanoparticles. ticles for biomedical applications: Recent
Dalton Transactions (Cambridge, England: advances in countering biomolecular corona
2003) 43(44):16780–16787. https://doi. formation and uptake by the mononuclear
org/10.1039/c4dt01597e phagocyte system. Small 10(13):2516–2529.
9. Wang B, He X, Zhang Z, Zhao Y, Feng W https://doi.org/10.1002/smll.201303540
(2013) Metabolism of nanomaterials in vivo: 21. Moyano DF, Saha K, Prakash G, Yan B,
blood circulation and organ clearance. Acc Kong H, Yazdani M, Rotello VM (2014) Fab-
Chem Res 46(3):761–769 rication of corona-free nanoparticles with tun-
10. Karakoti AS, Das S, Thevuthasan S, Seal S able hydrophobicity. ACS Nano 8
(2011) PEGylated inorganic nanoparticles. (7):6748–6755. https://doi.org/10.1021/
Angewandte Chemie - International Edition nn5006478
50(9):1980–1994 22. Jiang Y, Huo S, Mizuhara T, Das R, Lee YW,
11. Tong S, Hou S, Zheng Z, Zhou J, Bao G Hou S, Moyano DF, Duncan B, Liang XJ,
(2010) Coating optimization of superpara- Rotello VM (2015) The interplay of size and
magnetic iron oxide nanoparticles for high T2 surface functionality on the cellular uptake of
relaxivity. Nano Lett 10(11):4607–4613 sub-10 nm gold nanoparticles. ACS Nano 9
12. Howard MD, Jay M, Dziubla TD, Lu X (2008) (10):9986–9993. https://doi.org/10.1021/
PEGylation of nanocarrier drug delivery sys- acsnano.5b03521
tems: state of the art. J Biomed Nanotechnol 23. Elci SG, Jiang Y, Yan B, Kim ST, Saha K,
4(2):133–148 Moyano DF, Yesilbag Tonga G, Jackson LC,
13. Yoo JW, Chambers E, Mitragotri S (2010) Rotello VM, Vachet RW (2016) Surface charge
Factors that control the circulation time of controls the suborgan biodistributions of gold
nanoparticles in blood: Challenges, solutions nanoparticles. ACS Nano 10(5):5536–5542.
and future prospects. Curr Pharm Des 16 https://doi.org/10.1021/acsnano.6b02086
(21):2298–2307 24. Gustafson HH, Holt-Casper D, Grainger DW,
14. Pernia Leal M, Rivera-Fernandez S, Franco Ghandehari H (2015) Nanoparticle uptake:
JM, Pozo D, de la Fuente JM, Garcia-Martin the phagocyte problem. Nano Today 10
ML (2015) Long-circulating PEGylated man- (4):487–510. https://doi.org/10.1016/j.
ganese ferrite nanoparticles for MRI-based nantod.2015.06.006
molecular imaging. Nanoscale 7 25. Alexis F, Pridgen E, Molnar LK, Farokhzad
(5):2050–2059. https://doi.org/10.1039/ OC (2008) Factors affecting the clearance and
c4nr05781c biodistribution of polymeric nanoparticles.
15. Peer D, Karp JM, Hong S, Farokhzad OC, Mol Pharm 5(4):505–515
Margalit R, Langer R (2007) Nanocarriers as 26. Pelaz B, Del Pino P, Maffre P, Hartmann R,
an emerging platform for cancer therapy. Nat Gallego M, Rivera-Fernández S, De La Fuente
Nanotechnol 2(12):751–760 JM, Nienhaus GU, Parak WJ (2015) Surface
16. Dobrovolskaia MA, McNeil SE (2007) Immu- functionalization of nanoparticles with poly-
nological properties of engineered nanomater- ethylene glycol: effects on protein adsorption
ials. Nat Nanotechnol 2(8):469–478 and cellular uptake. ACS Nano 9
(7):6996–7008. https://doi.org/10.1021/
17. Dobrovolskaia MA, Aggarwal P, Hall JB, acsnano.5b01326
McNeil SE (2008) Preclinical studies to under-
stand nanoparticle interaction with the 27. Suk JS, Xu Q, Kim N, Hanes J, Ensign LM
immune system and its potential effects on (2016) PEGylation as a strategy for improving
nanoparticle biodistribution. Mol Pharm 5 nanoparticle-based drug and gene delivery.
(4):487–495 Adv Drug Deliv Rev 99(Part A):28–51.
https://doi.org/10.1016/j.addr.2015.09.
18. Verma A, Stellacci F (2010) Effect of surface 012
properties on nanoparticle-cell interactions.
Small 6(1):12–21. https://doi.org/10.1002/ 28. Ye L, Zhang Y, Yang B, Zhou X, Li J, Qin Z,
smll.200901158 Dong D, Cui Y, Yao F (2016) Zwitterionic-
modified starch-based stealth micelles for
19. Zhu M, Nie G, Meng H, Xia T, Nel A, Zhao Y prolonging circulation time and reducing mac-
(2013) Physicochemical properties determine rophage response. ACS Appl Mater Interfaces
nanomaterial cellular uptake, transport, and 8(7):4385–4398. https://doi.org/10.1021/
fate. Acc Chem Res 46(3):622–631. https:// acsami.5b10811
doi.org/10.1021/ar300031y
Pharmacokinetics of Magnetic Nanoparticles 419
29. Huang P, Liu J, Wang W, Zhang Y, Zhao F, 31. Sun S, Zeng H, Robinson DB, Raoux S, Rice
Kong D, Liu J, Dong A (2016) Zwitterionic PM, Wang SX, Li G (2004) Monodisperse
nanoparticles constructed from bioreducible MFe2O4 (M ¼ Fe, Co, Mn) nanoparticles. J
RAFT–ROP double head agent for shell shed- Am Chem Soc 126(1):273–279. https://doi.
ding triggered intracellular drug delivery. Acta org/10.1021/ja0380852
Biomater 40:263–272. https://doi.org/10. 32. Pernia Leal M, Munoz-Hernandez C, Berry
1016/j.actbio.2015.11.038 CC, Garcia-Martin M (2015) In vivo pharma-
30. Pernia Leal M, Caro C, Garcia-Martin ML cokinetics of T2 contrast agents based on iron
(2017) Shedding light on zwitterionic mag- oxide nanoparticles: optimization of blood cir-
netic nanoparticles: limitations for in vivo culation times. RSC Adv 5(94):76883–76891.
applications. Nanoscale 9(24):8176–8184. https://doi.org/10.1039/C5RA15680G
https://doi.org/10.1039/c7nr01607g
Part VII
Abstract
The use of imaging represents a major impact on the refinement and the reduction of in vivo studies in
animal models, in particular for allowing longitudinal monitoring of the onset and the progression of
disease within the same animal, and studying the biological effects of drug candidate and their therapeutic
effectiveness. But the use of imaging procedures can affect animal physiology, and the need to anesthetize
the animals for imaging entails potential health risks. During anesthesia, there is an inevitable autonomic
nervous system depression which induces cardiovascular depression, respiratory depression, and hypother-
mia. Also other procedures associated with imaging such as animal preparation (e.g., fasting, premedica-
tion), blood sampling, and dosage/contrast agent injections can also affect physiology and animal welfare.
All these factors are likely to have confounding effect on the outcome of the imaging studies and pose
important concerns regarding the animal’s well-being, particularly when imaging immune deprived animals
or diseased animals. We will discuss these challenges and considerations during imaging to maximize
efficacious data while promoting animal welfare.
1 Introduction
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0_25, © Springer Science+Business Media, LLC 2018
423
424 Jordi L. Tremoleda et al.
2 Methods
2.2 Transport Animals often have to be transported from general holding rooms
of Animals to the imaging facility. One consequence is that this will require full
to the Imaging Unit compliance with the institutional health screening controls to min-
and Acclimatization imize disease transfer to any other animals in the facility. A second
consequence is that for the transported animals themselves experi-
ence stress leading to an increase of the glucocorticoid level, a loss
in body weight and immune response suppression which have been
observed for up to 48 h after transport in rodents [13]. Therefore
an acclimatization period, typically between 3 and 7 days depend-
ing on the procedure and transport duration, is required after
animal transport. Imaging facilities should make housing capacity
available to allow for the acclimatization of transferred animals and
reduce stress before imaging takes place. Housing in the imaging
facility is also useful for monitoring animal recovery after anesthe-
sia. A sufficiently long recovery period should also be included after
imaging, depending on the type of anesthetic, the length of anes-
thesia, and any experimental procedures carried out. If an imaging
unit is integrated within a centralized animal unit, transport time
and acclimatization may be considerably reduced.
MRI and Anaesthesia 427
2.5 Anesthesia Injectable and inhaled anesthetics are commonly used in preclinical
Regimes imaging. Gas anesthetics are most suitable and recommended for
imaging due to their rapid onset and recovery times, and faster
elimination. Inhaled anesthetics with medical air or medical oxygen
as gas carrier allow for a better control of depth of anesthesia and
degree of oxygenation than injectables [15]. This is particularly
critical when undertaking long-term imaging procedures due to
the risk of developing hypoxia, respiratory depression, hypercapnia,
and acidosis. Dosing with injectables remains challenging due to
the lack of interventional management and when additional dosing
is required which can lead to overdosing and intermittent changes
on the depth of anesthesia. Injectables also often cause a prolonged
recovery time, in particular opioids with their strong residual effect
[16]. Some injectables can be administered via infusion which may
allow to achieve a more steady plasma concentration over the
course of imaging.
Anesthetic dose rates for injectable agents will depend on spe-
cies used, administration route, age, sex, strain, body condition,
environment, experimental setup, previous drug treatments, and
the level of anesthesia required. During the initial period of use, it is
important to monitor animals closely and make any adjustments
necessary in the protocol for subsequent experiments.
2.5.1 Injectables The most commonly used injectable anesthesia agents are listed in
Anesthetics Table 1 with their individual advantages and disadvantages and
typical dosing regimes.
(continued)
Table 1
430
(continued)
l Suitable for long-term anesthesia. l Slow induction and recovery 50 mg/kg iv bolus followed by 25–40 mg/kg/
l Minimal CVS and respiratory associated with involuntary hr (rats)
depression excitement
l Terminal/non-recovery work only
Urethane l Suitable for long-term anesthesia. l Carcinogenic: only allowed to be 0.8–1.3 g/kg, i.p. (mouse and rats) duration of
l Minimal CVS and respiratory used with special justification! action 8–10 h (nonrecovery only)
depression l Terminal/non-recovery work only
® l l
Avertin Wide safety margin Local irritation/peritonitis 0.015 mL/g body wt of 2.5% i.p.
(tribromoethanol) l Good muscle relaxation l Handling and storage safety issues 30 min—supplemental doses of anesthesia:
l Rapid induction and recovery l Toxic effects minimum of one-half of the initial does up to
l Pharmaceutical-grade TBE (e.g., 1 mL max vol per animal
Avertin) is no longer available:
IACUC/AWERB
MRI and Anaesthesia 431
2.6 Monitoring Most anesthetic drugs will impact on respiration, the cardiovascular
and Impact system, and/or thermoregulation. It is important to monitor the
of Anesthesia During animals during imaging and provide any physiologic support to
Imaging Procedures ensure animal welfare and also to minimize any confounding effects
on data acquisition. The recommended approach is to monitor
respiratory function and body temperature, as the minimum stan-
dard applicable during imaging to control depth of anesthesia.
Cardiac function is also highly recommended. Direct visualization
is not possible and it is greatly suggested to use the existing moni-
toring equipment for laboratory rodents which is tailored to their
small body size, fast cardiac and respiratory rates, and importantly
in the case of MRI, is non-ferromagnetic to avoid any interference
with the magnetic field [17, 18]. Most MRI-compatible systems are
based on fiber-optic or carbon fiber equipment and power source or
batteries are adequately filtered/isolated to avoid magnetic
interference [19].
While protocols do not specifically define how frequently one
should monitor the animals during anesthesia, it is obvious that the
more invasive the procedure and/or the longer time under anes-
thesia, the more likely it is to interfere with normal homeostasis and
thus greater the need for more frequent/constant monitoring. Also
it is instrumental to support the acquisition of gated imaging,
minimizing the effects due to biological motion and also targeting
specific imaging sequences, in a given respiratory or cardiac phase.
The position of the animals and the imaging bed systems used
are also crucial, to ensure that the neck and head are well positioned
to avoid restricting the breathing, and also the body and extremities
to avoid restricting the circulation and to avoid any bruising, strains
or avulsion in the body structures.
Isoflurane l Similar physical properties to halothane l Decreases arterial blood pressure (vasodilatation) Induction: 3–4%
l Rapid induction and recovery l More expensive than halothane Maintenance 1–2% (mice);
l Low toxicity and metabolic activity: highly safe l Strong smell; aversive response for repetitive use 1.5–2.5% (rats)
l Suitable for high frequency and log-term anesthesia l More potent respiratory depressant than halothane
l Minimal cardiovascular depression
l Moderate respiratory depression
l Good muscle relaxation
Sevoflurane l Faster induction and recovery times (3 times faster l Expensive but cost coming down Induction: 1–8%
than isoflurane) l Costly specific vaporizers Maintenance: 3–5% (mice
l Less respiratory depression that isoflurane l Provides an equally reliable anesthesia in and rats)
l Less struggling and excitement during induction laboratory mice.
l Metabolism similar to isoflurane: good for repetitive
and/or long-term anesthesia
l Blood glucose homeostasis is better maintained
l Method of choice for PET FDG myocardium uptake
studies
MRI and Anaesthesia 433
2.6.2 Respiratory System Most anesthetics are known to cause respiratory depression. There-
fore it is important to reduce variability due to poor ventilation
including hypoxia, hypercapnia, and acidosis [21, 22]. Respiratory
monitoring is generally carried out via detecting the breathing
motion registered as compressions of a respiratory sensor placed
in contact with the animal’s chest. These systems are extensively
used during imaging and highly compatible with MRI. Motion
artifacts due to breathing can be eliminated from the images by
employing gated imaging. Typically the most prominent motion of
the diaphragm and abdomen occurs during inspiration, and acqui-
sitions are generally carried out during expiration.
Other more advanced approaches include the use of arterial
blood gas analysis, which is very valuable during functional MRI.
The partial measurements of oxygen (pO2) and carbon dioxide
(pCO2) and the pH of the blood are detected from a single blood
sample allowing levels of oxygenation, imbalance of CO2 produc-
tion and acid-base balance to be monitored. Impaired gas
exchange, or hyper- or hypoventilation can be corrected by chang-
ing the anesthetic regime, and when critical through using artificial
ventilation. Indeed, some studies will require the animal to be
mechanically ventilated and it is important to ensure that the animal
does not develop hypercapnia or hypoxia. Other advanced respira-
tory monitoring systems include digitized systems such as capno-
graphs that measure the CO2 level through a highly sensitive
infrared spectroscopy CO2 sensor in the inhaled and exhaled gas,
based on the CO2 values in the venous return to the heart and the
efficacy of breathing. This equipment provides a very good indica-
tor of the respiratory function by continuous measurements of the
CO2 level. Capnographs that measure the CO2 level in the inhaled
and exhaled gas between inspiration and expiration at the endotra-
cheal tube connector or face masks level are very good indicator of
the respiratory function [23].
2.6.3 Cardiovascular Basic cardiovascular monitoring, including heart rate and blood
System pressure, is highly recommended. Electrocardiograms (ECG) are
generally used to monitor heart rate and rhythm and help to detect
434 Jordi L. Tremoleda et al.
2.6.4 Repeated Some imaging protocols may require long acquisition times and/or
and Long-Term Anesthesia high frequency imaging with considerable impact on cumulative
time of anesthesia. In these circumstances it is recommended to use
minimally metabolized volatile anesthetics like isoflurane or sevo-
flurane, which allow for a quick induction, good surgical anesthesia
and fast recovery, so that animals can regain full physiological
functions quickly. Repeat imaging is likely to induce some
hypothalamic-pituitary-adrenal (HPA) axis and autonomic nervous
system responses to the chronic stress with prolonged elevation of
corticosterone and impairments to homeostasis [27]. It is of
utmost importance to monitor body temperature. Hypo- or hyper-
thermia will interfere with many electrophysiological outcome
measurements. The extremities and tail should be covered when-
ever possible. Long imaging procedures with lengthy anesthesia
may seriously compromise the hydration statues of the animals.
Therefore it is important to compensate for the fluid lost and
prevent animal dehydration. Parental administration of warmed
fluids (0.9% saline or lactated Ringer’s) may be applied and ideally
also the inspired gases may be humidified to avoid desiccation via
the airways. Eye protection may also be used to help maintain good
lubrication of the cornea and also to protect the eyes from exposure
keratitis.
MRI and Anaesthesia 435
2.6.5 Ventilation Some studies will require the animal to be mechanically ventilated,
which may have important effects on thoracic hemodynamics and
may also override the autonomic reflex control of breathing, which
normally maintains blood gas homeostasis. When monitoring a
ventilated animal, ideally, one should have oxygen/carbon dioxide
monitoring equipment (e.g., pulse oximeter, end tidal capno-
graph). The ventilation settings can then be adjusted to try to
maintain physiological levels of O2/CO2. Blood gas and electrolyte
analysis are useful but confer intermittent monitoring, and a local
analyzer is needed.
The most commonly observed physiological effects and health
risks during anesthesia and troubleshooting management are listed
in Table 3.
2.8 Recovery Monitoring should be carried out until the animal has fully recov-
ered from anesthesia, i.e., until the animal regained full conscious-
ness with essential physiological functions back to normal. Checks
should include in particular respiratory and cardiovascular function
and the ability to control body temperature. Only then the animal
may be transferred back to its housing cage.
During recovery, it is important to maintain the core tempera-
ture of the animal. Good practice is to place the animals on their
right side or in sternal recumbent position in a warm recovery cage
with no sawdust or suchlike bedding that may be inhaled. If the
animals have been under anesthesia for a relatively long time, it may
be worth giving oxygen, fluids and nutrition supplement. This can
include oral or parenteral support, e.g., high-energy moist foods
such as nutrient agar, jelly or crushed rodent pellets mixed with
water, presented in a Petri dish or other manner that does not
require the animal to reach up high. Additional analgesia should
be provided if the animal has undergone a painful procedure or if
there appears to be any sign of pain [28].
Table 3
Commonly physiological effects and management during anesthesia
reversing anesthesia
l Hypothermia: Check temperature
l Hyperventilation in oxygen
thiopentone)
Respiration fails:
l Manual ventilation if animal is intubated
reverse anesthesia
l Animal too cold: Control body temperature
by anesthetic drugs
l Atropine may be indicated
Cardiac arrest
l Gently massage the chest side to side-not very
effective
l Stop anesthesia and transfer to O supply only
2
l Some drugs are effective in larger animals
Table 4
Exemplary systems for MRI-compatible physiological monitoring
References
1. Driehuys B, Nouls J, Badea A et al (2008) 10. Directive 2010/63/EU (2016) Legislation for
Small animal imaging with magnetic resonance the protection of animals used for scientific
microscope. ILAR J 49:35–53 purposes. Available via http://ec.europa.eu/
2. Bakermans AJ, Abdurrachim D, Moonen RP environment/chemicals/lab_animals/legisla
et al (2015) Small animal cardiovascular MR tion_en.htm. Accessed 7 Nov 2016
imaging and spectroscopy. Prog Nucl Magn 11. Public Health Service Policy on Humane Care
Reson Spectrosc 88-89:1–47. https://doi. and Use of Laboratory Animals National Insti-
org/10.1016/j.pnmrs.2015.03.001 tutes of Health Office of Laboratory Animal
3. Febo M, Foster TC (2016) Preclinical mag- Welfare (2016) Available via http://grants.
netic resonance imaging and spectroscopy nih.gov/grants/olaw/references/
studies of memory, aging, and cognitive phspolicylabanimals.pdf. Accessed 7 Nov 2016
decline. Front Aging Neurosci 8:158. https:// 12. The ARRIVE Guidelines: Animal Research:
doi.org/10.3389/fnagi.2016.00158 Reporting of In Vivo Experiments (2016)
4. Sasser TA, Bahadur A, Frederick E et al (2016) Available via https://www.nc3rs.org.uk/sites/
Considerations When Setting Up a Preclinical default/files/documents/Guidelines/NC3Rs
In Vivo Imaging Laboratory. Available via %20ARRIVE%20Guidelines%202013.pdf
https://www.bruker.com/fileadmin/user_ Accessed 7 Nov 2016
upload/8-PDF-Docs/PreclinicalImaging/ 13. Obernier JA, Baldwin RL (2016) Establishing
Brochures/Considerations_Preclinical_Lab_ an appropriate period of acclimatization fol-
T158053.pdf. Accessed 7 Nov 2016 lowing transportation of laboratory animals.
5. Sack M, Wetterling F, Sartorius A et al (2014) ILAR J 47(4):364–369
Signal-to-noise ratio of a mouse brain (13) C 14. LASA (2010) Guiding Principles for Preparing
CryoProbe™ system in comparison with room for and Undertaking Aseptic Surgery. In:
temperature coils: spectroscopic phantom and Jennings M, Berdoy M (eds) A report by the
in vivo results. NMR Biomed 27(6):709–715. LASA Education, Training and Ethics section.
https://doi.org/10.1002/nbm.3110 www.lasa.co.uk/publications.html. Accessed
6. Ferrari L, Turrini G, Crestan V et al (2012) A 7 Nov 2016
robust experimental protocol for pharmaco- 15. Wolfensohn S, Lloyd M (2003) Anaesthesia of
logical fMRI in rats and mice. J Neurosci laboratory animals. In: Wolfensohn S, Lloyd M
Methods 204:9–18. https://doi.org/10. (eds) Handbook of laboratory animal manage-
1016/j.jneumeth.2011.10.020 ment and welfare, 3rd edn. Blackwell Publish-
7. Tremoleda JL, Kerton A, Gsell W (2012) ing Ltd, Oxford. https://doi.org/10.1002/
Anaesthesia and physiological monitoring dur- 9780470751077.ch7
ing in vivo imaging of laboratory rodents: con- 16. Lovell DP (1986) Variation in pentobarbitone
siderations on experimental outcomes and sleeping time in mice. 2. Variables affecting test
animal welfare. EJNMMI Res 2(1):44. results. Lab Anim 20:91–96. https://doi.org/
https://doi.org/10.1186/2191-219X-2-44 10.1258/002367786780865089
8. Berdoy M, Hawkins P et al (2015) Guiding 17. Data Acquisition System – BIOPAC Systems,
principles on good practice for animal welfare Inc. (2016) Available via http://www.biopac.
and ethical review bodies. In: Jennings M com/magnetic-resonance-imaging-mri-com
(ed) A report by the RSPCA Research Animals patible. Accessed 7 Nov 2016
Department and LASA Education, Training 18. SAII Small Animal Instruments, Inc. (2016)
and Ethics Section. Available via http://www. Available via http://www.i4sa.com/web_app/
lasa.co.uk/PDF/AWERB_Guiding_ main/defaultProduct.aspx?ID¼76&PT¼3.
Principles_2015_final.pdf. Accessed 7 Nov Accessed 7 Nov 2016
2016 19. Peden CJ, Menon DK, Hall AS et al (1992)
9. IACUC Resources (2016) Available via Magnetic resonance for the anaesthetist. Part
https://www.aalas.org/iacuc/iacuc_res II: Aanaesthesia and monitoring in MR units.
ources#.WADzWfkrJD8 Accessed 7 Nov 2016 Anaesthesia 47:508–517
MRI and Anaesthesia 439
20. Colby LA, Morenko BJ (2004) Clinical con- 25. Wagner AE, Brodbelt DC (1997) Arterial
siderations in rodent bioimaging. Comp Med blood pressure monitoring in anesthetized ani-
54(6):623–630 mals. J Am Vet Med Assoc 210:1279–1285
21. Hauber HP, Karp D, Goldmann T et al (2010) 26. Thal SC, Plesnila N (2007) Non-invasive
Effect of low tidal volume ventilation on lung intraoperative monitoring of blood pressure
function and inflammation in mice. BMC Pulm and arterial pCO2 during surgical anesthesia
Med 10:21. https://doi.org/10.1186/1471- in mice. J Neurosci Methods 159(2):261–267
2466-12-7 27. Wu XY, Hua YT, Lua L et al (2015) Effect of
22. Pecchiari M, Monaco A, Koutsoukou A et al pentobarbital and isoflurane on acute stress
(2014) Effects of various modes of mechanical response in rat. Physiol Behav 145:118–121
ventilation in normal rats. Anesthesiology 120 28. Kohn DF, Martin TE (eds) (2006) Guidelines
(4):943–950 for the assessment and management of pain in
23. Larach DR, Schuler G, Skeehan TM et al rodents and rabbits. Available via https://
(1988) Mass spectrometry for monitoring wwwaclamorg/Content/files/files/Public/
respiratory and anaesthetic gas waveforms in Active/position_pain-rodent-rabbitpdf
rats. J Appl Physiol 65:955–963 Accessed 7 Nov 2016
24. Available via http://www.starrlifesciences.
com/conscious-monitoring. Accessed 7 Nov
2016
Chapter 26
Abstract
Clinical imaging modalities have reached a prominent role in medical diagnosis and patient management in
the last decades. Different image methodologies as Positron Emission Tomography, Single Photon Emis-
sion Tomography, X-Rays, or Magnetic Resonance Imaging are in continuous evolution to satisfy the
increasing demands of current medical diagnosis. Progress in these methodologies has been favored by the
parallel development of increasingly more powerful contrast agents. These are molecules that enhance the
intrinsic contrast of the images in the tissues where they accumulate, revealing noninvasively the presence of
characteristic molecular targets or differential physiopathological microenvironments. The contrast agent
field is currently moving to improve the performance of these molecules by incorporating the advantages
that modern nanotechnology offers. These include, mainly, the possibilities to combine imaging and
therapeutic capabilities over the same theranostic platform or improve the targeting efficiency in vivo by
molecular engineering of the nanostructures. In this review, we provide an introduction to multimodal
imaging methods in biomedicine, the sub-nanometric imaging agents previously used and the development
of advanced multimodal and theranostic imaging agents based in nanotechnology. We conclude providing
some illustrative examples from our own laboratories, including recent progress in theranostic formulations
of magnetoliposomes containing ω-3 poly-unsaturated fatty acids to treat inflammatory diseases, or the use
of stealth liposomes engineered with a pH-sensitive nanovalve to release their cargo specifically in the acidic
extracellular pH microenvironment of tumors.
Key words Imaging agents, Image Guided Drug Delivery, Magnetic Resonance Imaging, Nanotech-
nology, Positron Emission Tomography, Single Photon Emission Tomography, Theranostic agents, X-
Ray computed tomography
1 Introduction
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0_26, © Springer Science+Business Media, LLC 2018
441
442 Daniel Calle et al.
Fig. 1 The electromagnetic spectrum expressed as energy (top) or associated wavelength (bottom). The
middle panel shows some of the initial images obtained from each modality including radiofrequency (MRI)
[17], infrared [18], X-Rays [19], and γ-rays (PET) [20]
gamma rays with 140 meV (SPECT) or 511 meV (PET), enabling
the detection. The targeting moieties are vectorial reagents, includ-
ing either small chelates or even peptides, proteins, or antibodies
with molecular specificity for the desired target[34]. The radio-
nuclides involve mainly99mTc,68Ga, 111In or131I for SPECT
or18F,11C,15O for PET, among others.
A large variety of SPECT probes are commercially available to
monitor noninvasively tissue perfusion [35], hypoxia [36], inflam-
mation [37], thyroid function [38], or even cerebral activation
[39]. Their main advantage is the possibility to use relatively low
cost gamma cameras for detection, available in many hospitals and
the relatively long half-lives of the SPECT emitters (in the hour
range), while their main limitation is the reduced resolution of the
SPECT method [21, 22]. This limitation has been overcome, in
large part, through the use of PET probes, that offer increase spatial
and temporal resolution, at the expense of the more expensive PET
equipment and the reduced half-life (in the min. range) of the
positron emitters.18F-2-deoxiglucose (FDG) is probably the most
widely used PET tracer, despite its poor selectivity. FDG is taken up
by virtually all tissues in an analogous manner to glucose, but it is
not degraded by glycolysis, resulting in FDG accumulation in those
regions with enhanced glucose uptake. FDG reveals spectacularly
the presence of tumors or metastases, regions of cerebral activation
or even inflammatory lesions, providing probably the most success-
ful tracer in nuclear medicine. However, since glucose uptake is a
universal process, the use of FDG is hampered by its lack of selec-
tivity, making very difficult to discriminate tumoral from inflamed
regions, resulting in a non-negligible number of false positives
(up to 30% [40]) and requiring frequently complementary exam-
inations with additional tracers or modalities.
The first generation of contrast agents for MRI, involved
mainly Gd(III) chelates, able to enhance water relaxation rates in
those tissues where they accumulated [27]. Gd(III) is used because
it has seven unpaired, slow relaxing electrons, and depicts the
largest magnetic moment among the rare earth series. The ligands
most frequently used are either linear, derived from diethylendia-
mino pentaacetic acid (DTPA), or cyclic, derived from tetraazacy-
clododecane-1,4,7,10-tetraacetic acid (DOTA). In all these cases,
the ligand provides eight binding sites anchoring the Gd(III)
within the chelate, leaving free one the nine chelating sites of the
metal, for water contact. The contact between water in the solution
or tissue with the Gd(III), and the fast exchange of this water
molecule with the bulk solution, reduces very significantly the
relaxation times of tissue water, resulting in clearly enhanced
image intensity in those regions containing the chelate [27]. The
use of other lanthanides as Dy(III) may transform the same chelates
in T2 enhancing probes, due to the inherent T2 relaxing properties
of Dy(III) [41]. MRI contrast agents include additionally a large
446 Daniel Calle et al.
Fig. 2 The development of recent nanotechnology approaches has driven the creation of new families of
imaging contrast agents. Classic contrast agents as Gd(III) DOTA are now competing with newer, more
sensitive and more selective, contrast agents based on PLGA nanoparticles (top left), SPIO nanoparticles (top
center), Lipid Nanoparticles (top right), Quantum Dots (bottom left), Dendrimers (bottom center), or Graphene
structures (bottom right)
Multimodal Imaging Agents Using Nanotechnology 449
Fig. 3 Fluorescence imaging of liposomes. In vitro (a, b) and in vivo (c) fluorescence images of liposomes
loaded with nanoparticles and rhodamine-100 as acquired with the IVIS-Lumina camera. (a) Visible picture of
the 96 well plate used as phantom, showing the columns of wells loaded with decreasing (top to bottom)
concentrations of empty liposomes (left column), liposomes loaded with rhodamine-100 (central column) and
liposomes loaded with rhodamine-100 and Nanotex (right column). (b) Fluorescence image of the same
phantom. Note the larger fluorescence of the liposomes containing rhodamine-100 only as compared to those
containing rhodamine-100 and Nanotex. (c) In vivo images reveal the presence of the liposomal preparations
injected i.p., arrow left: liposomes containing rhodamine-100 and Nanotex, arrow right: empty liposomes.
Taken from [75]. Reproduced with permission of the publisher
450 Daniel Calle et al.
Fig. 4 PET-CT/FDG images of mice subjected to 5 days of DSS administration untreated (a), treated with
placebo (b) or liposomal suspensions containing Nanotex (c) with or without ω-3 PUFA-EE (d) [76]. Note that
FDG uptake ( full arrows) is higher in the untreated animals and those treated with placebo, than in those
treated with magnetoliposomes (dotted arrows), suggesting a higher inflammatory response in untreated
animals
Multimodal Imaging Agents Using Nanotechnology 451
Fig. 5 Effect of magnetoliposomal preparations containing (bottom) or not (top) ω-3 PUFA-EE on the time
course of glioma development after implantation of C6 cells in the mouse brain. Left panels (day 0), central
panels (day 3), right panels (day 6). Orange arrow shows accumulation of Nanotex nanoparticle. Taken from
[75]. Reproduced with permission of the publisher
Fig. 6 Image guided drug release in vivo from pH-sensitive liposomes into a C6 glioma implanted in the mouse
brain. (a) The C6 tumor-bearing animal was anesthetized, positioned in the magnet isocenter, and 0.4 mL of
1 M ISUCA (blue solution) was injected i.p [49]. (b) Successive multivoxel spectroscopy grids acquired (c), the
Henderson-Hasselbalch calibration curve was generated (d), and the measured ISUCA chemical shift in every
voxel was transformed into an extracellular pH value generating a pHe map (e). After ISUCA was cleared from
the tumor and its resonances became no longer detectable, T1-weighted baseline images were acquired (f).
The pH-sensitive or pH-insensitive liposomal preparations containing Gd-DTPA (red solution) were injected
into the tail vein (g). T1-weighted images acquired for the next 60 min and relative changes of T1-weighted
images in signal intensity (SI) were calculated in a pixel-by-pixel manner using the baseline images as
reference (h). The maps representing the mean change in SI (ΔSImean) were calculated by averaging the
changes occurring in SI during 60 min after liposome administration and presented overlaid with the
anatomical T1 image. Taken from [80]. Reproduced with permission of the publisher
5 Conclusion
Acknowledgements
References
1. Suetens P (2009) Fundamentals of medical 2. Medical imaging: technology ad applications
imaging, 2nd edn. Cambridge University (2013). CRC Press
Press, Cambridge
454 Daniel Calle et al.
3. Textbook of contrast media (1999). CRC 17. Lauterbur PC (1973) Image formation by
Press, Boca Raton, FL induced local interactions: examples employing
4. Contrast agents I. Magnetic resonance imaging nuclear magnetic resonance. Nature 242
(2002) Topics in current chemistry, vol 221. (5394):190–191
Springer, Berlin 18. Farrell C, Wallace JD, Mansfield CM (1971)
5. Contrast agents II: optical, ultrasound, x-ray The use of thermography in detection of meta-
and radiopharmaceutical imaging (2002) static breast cancer. Am J Roentgenol 111
Topics in current chemistry, vol 222. Springer, (1):148–152
Berlin 19. Rontgen WC (1896) On a new kind of rays.
6. Contrast agents III: radiopharmaceuticals: Science 3(59):227–231. https://doi.org/10.
from diagnosis to therapeutics (2002) Topics 1126/science.3.59.227
in current chemistry, vol 252. Springer, Berlin 20. Brownell GL, Sweet WH (1953) Localization
7. Key J, Leary JF (2014) Nanoparticles for mul- of brain tumors with positron emitters. Nucle-
timodal in vivo imaging in nanomedicine. Int J onics 11(11):40–45
Nanomedicine 9:711–726. https://doi.org/ 21. Rudin M, Weissleder R (2003) Molecular
10.2147/IJN.S53717 imaging in drug discovery and development.
8. Heidt T, Nahrendorf M (2013) Multimodal Nat Rev Drug Discov 2(2):123–131. https://
iron oxide nanoparticles for hybrid biomedical doi.org/10.1038/nrd1007
imaging. NMR Biomed 26(7):756–765. 22. Bushberg JT, Boone JM (2011) The essential
https://doi.org/10.1002/nbm.2872 physics of medical imaging. Lippincott Wil-
9. Kim J, Piao Y, Hyeon T (2009) Multifunc- liams & Wilkins, Philadelphia, PA
tional nanostructured materials for multimodal 23. Weissleder R, Mahmood U (2001) Molecular
imaging, and simultaneous imaging and ther- imaging. Radiology 219(2):316–333. https://
apy. Chem Soc Rev 38(2):372–390. https:// doi.org/10.1148/radiology.219.2.
doi.org/10.1039/b709883a r01ma19316
10. Lee DE, Koo H, Sun IC, Ryu JH, Kim K, 24. Hounsfield GN (1973) Computerized trans-
Kwon IC (2012) Multifunctional nanoparticles verse axial scanning (tomography): Part
for multimodal imaging and theragnosis. 1. Description of system. Br J Radiol 46
Chem Soc Rev 41(7):2656–2672. https:// (552):1016–1022
doi.org/10.1039/c2cs15261d 25. Hutton BF (2011) Recent advances in iterative
11. Lee N, Yoo D, Ling D, Cho MH, Hyeon T, reconstruction for clinical SPECT/PET and
Cheon J (2015) Iron oxide based nanoparticles CT. Acta Oncol 50(6):851–858
for multimodal imaging and magnetorespon- 26. Antoch G, Freudenberg LS, Beyer T,
sive therapy. Chem Rev 115 Bockisch A, Debatin JF (2004) To enhance or
(19):10637–10689. https://doi.org/10. not to enhance? 18F-FDG and CT contrast
1021/acs.chemrev.5b00112 agents in dual-modality 18F-FDG PET/CT. J
12. Ding H, Wu F, Nair MP (2013) Image-guided Nucl Med 45(Suppl 1):56S–65S
drug delivery to the brain using nanotechnol- 27. Caravan P, Ellison JJ, McMurry TJ, Lauffer RB
ogy. Drug Discov Today 18 (1999) Gadolinium (III) chelates as MRI con-
(21–22):1074–1080. https://doi.org/10. trast agents: structure, dynamics, and applica-
1016/j.drudis.2013.06.010 tions. Chem Rev 99(9):2293–2352
13. Ding H, Wu F (2012) Image guided biodistri- 28. Pacheco-Torres J, Calle D, Lizarbe B, Negri V,
bution of drugs and drug delivery. Theranos- Ubide C, Fayos R, Lopez Larrubia P,
tics 2(11):1037–1039. https://doi.org/10. Ballesteros P, Cerdan S (2011) Environmen-
7150/thno.5321 tally sensitive paramagnetic and diamagnetic
14. Lammers T, Kiessling F, Hennink WE, Storm contrast agents for nuclear magnetic resonance
G (2010) Nanotheranostics and image-guided imaging and spectroscopy. Curr Top Med
drug delivery: current concepts and future Chem 11(1):115–130
directions. Mol Pharm 7(6):1899–1912 29. Elsinga PH, Dierckx RA (2014) Small mole-
15. Yu MK, Park J, Jon S (2012) Magnetic nano- cule PET-radiopharmaceuticals. Curr Pharm
particles and their applications in image-guided Des 20:2268–2274
drug delivery. Drug Deliv Transl Res 2 30. Cai J, Li F (2013) Single-photon emission
(1):3–21. https://doi.org/10.1007/s13346- computed tomography tracers for predicting
011-0049-8 and monitoring cancer therapy. Curr Pharm
16. Webb S (1988) The physics of medical imag- Biotechnol 14(7):693–707
ing. CRC Press, Boca Ratón, FL
Multimodal Imaging Agents Using Nanotechnology 455
31. Thomsen HS, Morcos SK (2000) Radio- 42. Hancu I, Dixon WT, Woods M, Vinogradov E,
graphic contrast media. BJU Int 86(Suppl Sherry AD, Lenkinski RE (2010) CEST and
1):1–10 PARACEST MR contrast agents. Acta Radiol
32. Pavel DG, Zimmer M, Patterson VN (1977) In 51(8):910–923. https://doi.org/10.3109/
vivo labeling of red blood cells with 99mTc: a 02841851.2010.502126
new approach to blood pool visualization. J 43. Sun PZ, Sorensen AG (2008) Imaging pH
Nucl Med 18(3):305–308 using the chemical exchange saturation transfer
33. Hahn MA, Singh AK, Sharma P, Brown SC, (CEST) MRI: correction of concomitant RF
Moudgil BM (2011) Nanoparticles as contrast irradiation effects to quantify CEST MRI for
agents for in-vivo bioimaging: current status chemical exchange rate and pH. Magn Reson
and future perspectives. Anal Bioanal Chem Med 60(2):390–397
399(1):3–27 44. McRae R, Bagchi P, Sumalekshmy S, Fahrni CJ
34. Pimlott SL, Sutherland A (2011) Molecular (2009) In situ imaging of metals in cells and
tracers for the PET and SPECT imaging of tissues. Chem Rev 109(10):4780–4827.
disease. Chem Soc Rev 40(1):149–162. https://doi.org/10.1021/cr900223a
https://doi.org/10.1039/b922628c 45. Zhang S, Malloy CR, Sherry AD (2005) MRI
35. Heinle SK, Noblin J, Goree-Best P, Mello A, thermometry based on PARACEST agents. J
Ravad G, Mull S, Mammen P, Grayburn PA Am Chem Soc 127(50):17572–17573.
(2000) Assessment of myocardial perfusion by https://doi.org/10.1021/ja053799t
harmonic power doppler imaging at rest and 46. Portman MA, Lassen NA, Cooper TG, Sills
during adenosine stress comparison with AM, Potchen EJ (1991) Intra- and extracellu-
99mTc-Sestamibi SPECT imaging. Circulation lar pH of the brain in vivo studied by
102(1):55–60 31P-NMR during hyper- and hypocapnia. J
36. Urtasun RC, Parliament MB, McEwan AJ, Appl Physiol 71(6):2168–2172
Mercer JR, Mannan RH, Wiebe LI, Morin C, 47. Fisher MJ, Dillon PF (1987) Phenylphospho-
Chapman JD (1996) Measurement of hypoxia nate: a 31P-NMR indicator of extracellular pH
in human tumours by non-invasive spect imag- and volume in the isolated perfused rabbit
ing of iodoazomycin arabinoside. Br J Cancer bladder. Circ Res 60(4):472–477
Suppl 27:S209–S212 48. Garcia-Martin ML, Herigault G, Remy C,
37. Leitha T, Glaser C, Pruckmayer M, Rasse M, Farion R, Ballesteros P, Coles JA, Cerdan S,
Millesi W, Lang S, Nasel C, Backfrieder W, Ziegler A (2001) Mapping extracellular pH in
Kainberger F (1998) Technetium-99m-MIBI rat brain gliomas in vivo by 1H magnetic reso-
in primary and recurrent head and neck nance spectroscopic imaging: comparison with
tumors: contribution of bone SPECT image maps of metabolites. Cancer Res 61
fusion. J Nucl Med 39(7):1166–1171 (17):6524–6531
38. Tharp K, Israel O, Hausmann J, Bettman L, 49. Provent P, Benito M, Hiba B, Farion R, Lopez-
Martin W, Daitzchman M, Sandler M, Delbeke Larrubia P, Ballesteros P, Remy C,
D (2004) Impact of 131I-SPECT/CT images Segebarth C, Cerdan S, Coles JA, Garcia-
obtained with an integrated system in the Martin ML (2007) Serial in vivo spectroscopic
follow-up of patients with thyroid carcinoma. nuclear magnetic resonance imaging of lactate
Eur J Nucl Med Mol Imaging 31 and extracellular pH in rat gliomas shows
(10):1435–1442 redistribution of protons away from sites of
39. Fukuyama H, Ouchi Y, Matsuzaki S, glycolysis. Cancer Res 67(16):7638–7645.
Nagahama Y, Yamauchi H, Ogawa M, https://doi.org/10.1158/0008-5472.CAN-
Kimura J, Shibasaki H (1997) Brain functional 06-3459
activity during gait in normal subjects: a 50. Gallagher FA, Kettunen MI, Day SE, DE H,
SPECT study. Neurosci Lett 228(3):183–186 Ardenkjaer-Larsen JH, Zandt R, Jensen PR,
40. Strauss LG (1996) Fluorine-18 deoxyglucose Karlsson M, Golman K, Lerche MH, Brindle
and false-positive results: a major problem in KM (2008) Magnetic resonance imaging of pH
the diagnostics of oncological patients. Eur J in vivo using hyperpolarized 13C-labelled
Nucl Med 23(10):1409–1415 bicarbonate. Nature 453(7197):940–943.
41. Aime S, Barge A, Delli Castelli D, Fedeli F, https://doi.org/10.1038/nature07017
Mortillaro A, Nielsen FU, Terreno E (2002) 51. Pacheco-Torres J, Calle D, Lizarbe B, Negri V,
Paramagnetic lanthanide (III) complexes as Ubide C, Fayos R, Lopez Larrubia P,
pH-sensitive chemical exchange saturation Ballesteros P, Cerdan S (2011) Environmen-
transfer (CEST) contrast agents for MRI appli- tally sensitive paramagnetic and diamagnetic
cations. Magn Reson Med 47(4):639–648 contrast agents for nuclear magnetic resonance
456 Daniel Calle et al.
72. Torchilin VP (2005) Recent advances with (2009) Bicarbonate increases tumor pH and
liposomes as pharmaceutical carriers. Nat Rev inhibits spontaneous metastases. Cancer Res
Drug Discov 4(2):145–160 69(6):2260–2268. https://doi.org/10.1158/
73. Hilgenbrink AR, Low PS (2005) Folate 0008-5472.CAN-07-5575
receptor-mediated drug targeting: from thera- 78. Raghunand N, Mahoney BP, Gillies RJ (2003)
peutics to diagnostics. J Pharm Sci 94 Tumor acidity, ion trapping and chemothera-
(10):2135–2146 peutics. II. pH-dependent partition coeffi-
74. Danhier F, Feron O, Preat V (2010) To exploit cients predict importance of ion trapping on
the tumor microenvironment: passive and pharmacokinetics of weakly basic chemothera-
active tumor targeting of nanocarriers for peutic agents. Biochem Pharmacol 66
anti-cancer drug delivery. J Control Release (7):1219–1229
148(2):135–146 79. Mahoney BP, Raghunand N, Baggett B, Gillies
75. Calle D, Negri V, Ballesteros P, Cerdan S RJ (2003) Tumor acidity, ion trapping and
(2015) Magnetoliposomes loaded with poly- chemotherapeutics. I. Acid pH affects the dis-
unsaturated fatty acids as novel theranostic tribution of chemotherapeutic agents in vitro.
anti-inflammatory formulations. Theranostics Biochem Pharmacol 66(7):1207–1218
5(5):489–503. https://doi.org/10.7150/ 80. Pacheco-Torres J, Mukherjee N, Walko M,
thno.10069 Lopez-Larrubia P, Ballesteros P, Cerdan S,
76. Calle D (2014) Novel contrast agents for mul- Kocer A (2015) Image guided drug release
timodal biomedical imaging based in nano- from pH-sensitive Ion channel-functionalized
technology. Dissertation, Universidad stealth liposomes into an in vivo glioblastoma
Autónoma de Madrid model. Nanomedicine 11(6):1345–1354.
77. Robey IF, Baggett BK, Kirkpatrick ND, Roe https://doi.org/10.1016/j.nano.2015.03.
DJ, Dosescu J, Sloane BF, Hashim AI, Morse 014
DL, Raghunand N, Gatenby RA, Gillies RJ
INDEX
Marı́a Luisa Garcı́a-Martı́n and Pilar López-Larrubia (eds.), Preclinical MRI: Methods and Protocols, Methods in Molecular Biology,
vol. 1718, https://doi.org/10.1007/978-1-4939-7531-0, © Springer Science+Business Media, LLC 2018
459
PRECLINICAL MRI: METHODS AND PROTOCOLS
460 Index
Cardiac MRI (cont.) coefficient ............................ 3, 90, 92, 104–106, 136,
CMR42........................................................... 272, 280 137, 141, 143, 144, 253, 354, 379, 383
ejection fraction (EF).............................................. 279 gradient..................91, 354, 360, 361, 368, 379, 386
end-diastolic volume (EDV) .................................. 279 time ................................................................... 91, 103
endo-cardiac border ....................................... 279, 280 Diffusion tensor imaging (DTI) ......................... 379, 388
end-systolic volume (ESV) ..................................... 279 DtiStudio ........................................................ 108, 111
epi-cardiac border .......................................... 279, 280 Diffusion weighted imaging (DWI) .................... 90, 136,
intragate (Ig) ........................................................... 271 348, 349, 379
long axes (LAX) .......... 271, 273, 275, 276, 278, 279 Diffusivity .................................................... 105, 106, 112
short axes (SAX)............................ 273, 275–278, 280 Directional-encoded-color (DEC) maps ............ 107, 294
Cardiovascular diseases (CVD) .................................... 269 Dynamic contrast enhanced (DCE)........... 301, 387, 396
Cardiovascular system ................................. 431, 433, 434 Tofts model .................................................. 73, 81–83
Carotid artery ................................................................ 384 two-compartments exchange model (2CXM) ....... 73,
Carr Purcell Meiboom Gill (CMPG)..............26, 28, 337 81, 83
Central nervous system (CNS).................... 35, 347, 356, Dynamic susceptibility contrast (DSC)....................42, 59
359, 360, 364, 365, 395, 396 Dynamic T2.......................................................... 412–415
Cerebral perfusion
cerebral blood flow (CBF)........................... 43, 45, 55 E
cerebral blood volume (CBV) ............................43, 55 Echo planar imaging (EPI)...................... 43, 63, 96, 110,
mean transit time (MTT) ............................ 43, 45, 55 122, 301, 353
Cerebrospinal fluid (CSF) ......................... 23, 24, 30, 31, Echo time (TE) ............................... 22, 63, 91, 130, 153,
54, 92, 200, 299, 356, 360, 380, 389, 390
158, 181, 261, 271, 290, 301, 337, 354, 398,
Chelate............................... 24, 25, 47, 55, 444, 445, 447 412, 413
Chemical exchange saturation transfer (CEST)3, 25, 378, Eddy-currents correction (ECC) ................................. 160
446
Edema ................................... 4, 23, 24, 31, 35, 269, 349,
Chemical shift. 7, 8, 114, 151, 158, 159, 163, 171, 189, 356, 359, 379–381, 383, 385, 388
237, 242, 243, 246, 247, 255, 260, 264, 337, Eigenvalues ........................................................... 105, 369
378, 446, 452
Eigenvectors ................................................ 107, 110, 292
Chemical shift displacement error (CSDE)158, 159, 163, Electric-stimulation....................................................... 117
171 Electrocardiogram (ECG) ......................... 108, 109, 114,
Choline (Cho) ............................................. 160, 331, 332 239, 281, 364, 427, 433, 437
Complexone .................................................................... 47
Electrodes ......................... 108, 109, 120–128, 131–133,
Connectivity ........................................117, 131, 349, 361 239, 298, 434
Contrast agent (CA) ..................................................... 321 Embryo .............................. 285, 288, 293, 294, 350, 357
diethylenetriaminepentaacetic acid (DTPA).....24, 47,
Encoding ............................ 9–13, 19, 22, 27, 28, 31, 34,
350, 357, 367, 445 44, 112, 114, 136, 141, 145, 255, 292, 309,
DOTA .......................................................24, 445, 448 353, 354, 357, 358, 361, 368
dysprosium (Dy) .................................................42, 47
EPI ghost....................................................................... 141
gadolinium (Gd) .............................. 24, 47, 350, 357, Ernst angle.............................................................. 34, 227
367, 445, 447, 448 Estimated glucose disposal rate (eGDR) ............ 183, 184
Coupling......................................66, 118, 120, 123, 151,
Ex vivo imaging ............................................................. 348
155, 170, 175, 182
Creatine (Cr) ..................... 155, 160, 162, 190, 191, 332 F
Cryoprobe ......................... 209, 214, 271, 348, 366, 397
19
F ..............................235–257, 298–300, 304, 308, 309
D FA (fractional anisotropy)................. 106, 107, 110, 112,
114, 285, 292, 294, 354, 355, 358–360, 363,
Decoupling ...................................................171–174, 182
364, 369
Demyelination ............................................. 349, 356, 359 Fast diffusion coefficient (Dfast) .......................... 137, 143
Deoxyhemoglobin ................ 34, 35, 299, 305, 322, 387 Fast diffusion phase (FDP).................137, 138, 141, 144
Dexmedetomidine......................................................... 123
Fast low angle shot (FLASH)...........................33, 34, 94,
Diffusion............................v, 3, 4, 41, 45, 135–146, 154, 139, 162, 207, 216, 219, 241, 271, 273, 281,
264, 286, 289–292, 294, 298, 299, 348, 349, 282, 323, 383, 386, 389, 398, 399, 412, 414
353, 354, 358–361, 364, 366, 368, 379, 383, Fastmap........................................... 51, 96, 113, 141, 163
386, 388
PRECLINICAL MRI : METHODS AND PROTOCOLS
Index 461
Fast spin echo (FSE) ........................ 30, 31, 35, 356, 389 Gradient recalled echo (GRE)............................ 206, 228,
Fe(iron)................................... 8, 25, 34, 35, 42, 47, 205, 389, 399, 401, 403, 404, 406
221, 237, 315–317, 325, 349, 356, 411, 447 GRASE ................................ 26, 290, 292, 294, 295, 348
Ferritin ........................................................................... 206 Gray matter (GM)............................. 29, 35, 68, 92, 294,
Fibre orientation distribution(FOD) .................. 358, 369 348, 359, 360
Fibre tracking ................................................................ 365 Gyromagnetic ratio ...................................... 6, 10, 32, 91,
Field of excitation (FOE) .................................... 290, 291 151, 152, 169–172, 264, 308
Field of view (FOV) ...................... 11, 13, 22, 51, 62–65,
110, 113, 130, 133, 157, 190, 192, 194, 197, H
215, 222, 225, 227, 242, 243, 250, 255, 1
H ........................................... 6, 10, 16, 47, 62, 95, 121,
261–263, 271, 273, 290, 301, 322–325, 336, 140, 151, 153, 155, 158, 160, 169–175, 179,
337, 353, 355–359, 365, 383, 386, 387, 398,
180, 182, 189, 207, 209, 210, 220, 222, 235,
399, 401, 412, 413 237, 240–243, 245–250, 252, 255, 299, 300,
Flip angle (FA) .......................... 7, 10, 22, 23, 32–34, 63, 304, 308, 331, 332, 336–339, 446
130, 172, 243, 261, 271, 288, 301, 323, 324,
Hemoglobin ...................................................34, 299, 305
336, 337, 339, 355, 357, 399 Hexafluorobenzene (HFB) .......299, 300, 305, 307, 309
Fluid-attenuated inversion recovery (FLAIR).30, 31, 35, High angular resolution diffusion imaging
380, 389, 390
(HARDI) .................................. 107, 353, 354,
Fluorine (19F) MRI..................................... 242, 244, 252 357–360, 362
Fomblin ......................................222, 350, 351, 357, 367 Huntington’s disease .................................................... 347
Fourier-transform (FT).......................... 7–9, 51, 99, 145, Hybrid echo .................................................................... 26
151, 158, 290, 291, 338, 354, 358
Hypercapnia.........................................427, 428, 433, 436
Fractional anisotropy (FA) ............... 106, 107, 112, 174, Hyperthermia ......................................215, 409, 433, 434
262, 263, 273, 292, 354, 355, 359, 360, 363, Hypothermia ..................................... 210, 239, 424, 427,
364, 369
431, 433, 434, 436
Fractional enrichment (FR) ................................. 172, 176 Hypoxia ............................................. 297, 298, 300, 427,
Free induction decay (FID)............................7, 8, 11, 13, 428, 433, 436, 445
158, 164, 196, 198, 366
Frequency encoding...............10–13, 19, 22, 27, 44, 255 I
FSL software.................................................................. 122
Full-width at half maximum (FWHM) ............... 163, 194 Initial area under the curve (IAUC) .............................. 71
Functional diffusion MRI (fDMRI) ................... 142, 143 Intracerebral microstimulation............................ 119, 120
Functional diffusion weighted imaging (fDWI) ........140, In utero MRI ........................................................ 285–295
142, 145, 146 Inversion recovery (IR) ............................. 16, 26, 29–31,
Functional MRI (fMRI) ............................ 117–133, 205, 264, 301, 356, 366, 380, 389, 399, 404
222, 299, 348, 380, 433, 434 Inversion time (TI) ...........................22, 29–31, 356, 399
Iron ....................................... 8, 34, 35, 42, 47, 206, 237,
G 315–317, 325, 349, 356, 411, 447
Iron oxide nanoparticle ......................................... 25, 447
Gamma-variate function ................................................. 43 Ischemia................................................45, 155, 189, 378,
Ghost artifacts ...................................................... 110, 114
381, 387, 388, 434
Glucose .............................................. 118, 146, 162, 170, Isotopic enrichment (IE).............................................. 176
175–177, 182–184, 259, 331, 395, 431, 445
Glutamate (Glu)................ 155, 160, 162, 181, 378, 380 K
Glutamine (Gln)........................155, 160, 162, 175, 181,
190, 254, 331 K-space .............................11, 13, 14, 27, 31, 56, 95, 96,
Glycerophosphocholine (GPC)................. 155, 160, 162, 99, 114, 140, 141, 145, 190, 194, 218, 225,
331, 332, 340 229, 290, 291, 337, 353, 358
Glycerophosphoethanolamine (GPE)................. 332, 340 Ktrans ................................................................... 73, 82, 83
Glycolysis .............................................................. 175, 445
L
Gradient echo (GE) ............................. 13–15, 18, 23, 25,
32–36, 43, 44, 50, 51, 63, 122, 132, 162, 207, Lactate (Lac)............................................... 155, 162, 175,
242, 252, 261, 288, 292, 294, 300, 306, 308, 177, 184, 190, 331
322, 355, 357, 358, 367, 389, 399, 412, 414 Larmor frequency ....................6, 7, 9, 10, 163, 182, 206
Gradient echo with flow compensation (GEFC) ......... 33, Liposomes............................................442, 447, 449–453
322, 323
PRECLINICAL MRI: METHODS AND PROTOCOLS
462 Index
M Motion artifacts................................. 108, 110, 111, 114,
215, 288, 433
Magnetic field gradient................................. v, 6, 8, 9, 11, Multimodal imaging ...........................442–444, 449, 452
34, 90, 103, 137, 163, 220, 299, 424 Myelin .............................................................90, 107, 349
Magnetic moment.................................... 16, 24, 25, 152, Myocardial ................................................... 269, 279, 434
445, 447 Myo-inositol (myo-Ins) ........................155, 160, 162, 181
Magnetic nanoparticle (MNP) ............................ 409, 414
Magnetic resonance angiography (MRA) N
time-of-flight (TOF)............................................... 386
Magnetic resonance imaging (MRI)............................v, 3, N-acetyl aspartic acid (NAA)..................... 154, 160, 162,
21–36, 41–56, 118, 270–272, 285, 300, 301, 190, 332
315, 332, 347, 378, 382–386, 397, 398, 423, Nanoemulsion ................... 235, 236, 238–241, 251, 254
441, 444 Nanoparticle .................................. 25, 47, 239, 254, 409,
Magnetic resonance spectroscopy (MRS).......... 157, 158, 412, 414, 417, 446–449, 451
162, 164, 165, 171, 174, 191, 198 Neurite orientation dispersion and density imaging
chemical shift imaging (CSI).............................6, 189, (NODDI) ........................................... 354, 360
193, 237, 242, 243, 260, 264, 337, 378 Neurodegeneration ........................................45, 349, 359
13
C MRS Neurodegenerative disease ................................... 59, 297,
distorsionless enhancement by polarization 347–369, 444
transfer (DEPT) ................................. 171, 174 Neurodegenerative disorder ........................................... 35
echo planar spectroscopic imaging (EPSI) ................ 6 Neuronal activation.............................118, 119, 122, 144
heteronuclear MRS .............................. 169, 171, 174, Neurovascular coupling .............................. 118, 120, 123
176, 179, 182, 185 Nuclear Overhauser effect (NOE) ............................... 171
1
H MRS Null point ..................................................................29, 31
LCModel ........................ 157, 158, 162, 164, 165 Nyquist ................................................................. 110, 114
image selected in vivo spectroscopy (ISIS)............ 181
O
J-coupling ................................................................ 181
jMRUI Obesity......................................................... 259, 260, 267
QUEST..................................................... 191, 198 Orthotopic......................... 305, 307, 333, 335, 340–342
localized spectroscopy............................................. 158 Osirix ...................................................................... 75, 362
magnetic resonance spectroscopic imaging Outer volume suppression (OVS) ..................... 159, 164,
(MRSI).......................... 6, 189, 191, 331, 446 181, 194, 196
multivoxel ....................................................... 152, 452 Oximetry......................62, 298, 299, 301, 305–308, 437
31
P MRS .................................................................. 152 Oxygenation .................................. 34, 62, 176, 206, 212,
point resolved spectroscopy (PRESS) ................... 153, 214, 215, 297–309, 424, 428, 433, 434
189, 190 Oxyhemoglobin .......................................... 299, 308, 322
PRESS-MRSI .......................................................... 194
single voxel .............................................152–154, 178 P
spin echo full intensity acquired localized spectroscopy 31
P .............................................. 152, 169–171, 176, 332,
(SPECIAL) ................................ 154, 178, 181
337–340, 343, 446
stimulated echo acquisition mode (STEAM) ....... 154,
Parallel imaging .................................................... 145, 281
163, 181
Paramagnetic compounds............................................... 42
Magnetic susceptibility ............................ 32, 35, 42, 113,
Parametric imaging .................................... 43, 46, 50, 53,
205, 206, 219, 222, 322, 349
94–96, 98, 139, 142, 143
Magnetoliposomes .......................................442, 447–452
Parametric map........................................... 112, 140, 143,
Manganese ...........................................286, 287, 410, 411
144, 363, 364, 380
Manganese-enhanced MRI (MEMRI) ........................ 286
Parkinson’s disease ................................................. 36, 189
Mapshim ......................................... 51, 96, 113, 128, 141
Partial parallel imaging (PPI) ................................ 99, 145
Mean arterial pressure (MAP) ...................................... 434
pCO2 .............................................................................. 433
Mean diffusivity............................................................. 106
Perflurocarbon (PFC) ................................ 235–246, 248,
Medial cerebral artery occlusion (MCAO).................. 381
249, 251–255, 298–300, 303, 305, 309
Metabolic rate ...................................................... 169, 427
Perfusion..............................................v, 4, 34–36, 41–45,
Micro-sized particles of iron oxide (MPIO) ..... 316–320,
47, 50–53, 55, 145, 211, 222, 227, 269, 298,
322–325
302, 319, 350, 364, 365, 378, 379, 434, 445
Molecular MRI (mMRI) ..................................... 315–325
PRECLINICAL MRI : METHODS AND PROTOCOLS
Index 463
Perfusion-diffusion mismatch (PDM) ........................... 45 Receiver gain (RG)........................................................ 164
Perfusion weighted imaging (PWI) ....................... 41, 43, Region of interest (ROI) ............... 47, 98, 143, 303, 405
45, 378, 379 Relaxation ........................................... v, 4, 13–19, 23, 24,
Permeability........................... 4, 316, 395, 396, 406, 449 29–32, 35, 42, 43, 46, 89, 122, 157, 163, 165,
Permeability surface area product .................................. 83 171, 181, 196, 205, 227, 241, 242, 264, 290,
pH ....................................... 25, 189, 247, 318, 319, 323, 303, 305, 337, 340, 348, 349, 356, 366, 380,
325, 442, 446, 452 386, 389, 409, 413, 429, 430, 445
Pharmacodynamics........................................................ 298 longitudinal relaxation .................... 15–17, 23, 29, 65
Pharmacokinetics (PK) .......................381, 384, 409–417 longitudinal relaxation time, (see T1
Phase encoding.............................. 11, 13, 22, 27, 28, 31, relaxation rate, (see R1; R2; R2*
34, 96, 99, 114, 140, 145, 215, 227, 309, 358 transverse relaxation .................................... 17, 18, 23,
Phase image .............. 206, 207, 218, 220, 226, 229, 264 32, 205, 380
Phase mask............................................................ 206, 207 transverse relaxation time, (see T2; T2*
Phosphocholine (PC) ................................ 109, 155, 156, Relaxivity ................................................... 4, 25, 309, 447
160, 162, 190, 191, 237, 331, 332, 339, 340 Repetition time (TR) ................................. 11, 14, 22, 63,
Phosphocreatine (PCr) ..................... 155, 160, 162, 332, 91, 130, 157, 190, 218, 261, 271, 290, 300,
340, 343 337, 348, 354, 398, 412, 413
Phosphodiester (PDE)......................................... 332, 340 Respiratory system ........................................................ 433
Phosphoethanolamine (PE) ................................ 332, 340 Resting state ................................................ 119, 121, 144
Phospholipids .............................................. 236, 251, 340 RF channel..................................................................... 174
Phosphomonoester (PME) ................................. 332, 340 RF coil ........................................... 45, 50, 61, 62, 65, 91,
Physiological monitoring........................... 47, 62, 63, 95, 128, 138, 157, 174, 190, 207–209, 270, 271,
121, 126, 140, 260, 261, 301–303, 424, 426, 273, 278, 280, 282, 286, 303, 350, 352, 397,
427, 437 401, 412
Pixel-by-pixel analysis ................................................... 382 birdcage resonator.......................................... 350, 397
PO2 ............................298–303, 305, 307–309, 364, 433 phased array coil .....................................286–288, 293
pO2 mapping surface coil ......................................45, 47, 65, 67, 91,
DOCENT.................... 298, 300–303, 305, 306, 309 95, 132, 138, 140, 157, 162, 174, 185, 271,
FREDOM.............................................. 301, 307, 308 286–288, 351, 397
Poly-methyl methacrylate (PMM) ...........................46, 47 volume coil ........................................... 45, 47, 65, 91,
Post labeling delay ............................................. 63, 67, 68 95, 138, 140, 157, 286, 287, 351, 397
Proton density (PD) .................................. 18, 23, 24, 28,
33, 34, 63, 65, 67 S
Pulse sequence..................................4–15, 18, 21, 22, 29, Segmentation .................................... 260, 264, 265, 271,
43, 97, 142, 170, 207, 219, 242, 270, 281,
349, 353, 361, 366, 369, 399
369, 386 Segmented EPI ........................................ 96, 97, 99, 113,
Pyruvate ......................................................................... 175 141, 142, 145
Self-gating...................................................................... 281
Q
Short TI inversion recovery (STIR).................. 30, 31, 35
Quantitative imaging biomarkers (QIB) ....................... 14 Signal-to-noise ratio (SNR).................................... 55, 96,
Quantitative susceptibility mapping (QSM)...............219, 108, 121, 124, 141, 270, 300, 335, 348
226, 357 Single-shot EPI ............................................................... 96
Slice selection .........................................9, 10, 14, 22, 27,
R 44, 113, 243, 303
R1 ..................................................... 4, 17, 298, 299, 303, Slow diffusion coefficient (Dslow)............... 137, 141, 143
305, 308, 309 Slow diffusion phase (SDP)........................ 141, 143, 144
Spatial response function (SRF) ................................... 190
R2 ..................................................................................... 46
R2* ............................. 43, 206, 219, 227, 299, 303, 308 Specific absorption rate (SAR) .................................23, 30
Radial diffusivity.............................................13, 106, 354 Spin density (NH).................................................. 89, 241
Spin dephasing ....................................................... 89, 205
Radio-frequency (RF) .................................. 5, 21, 60, 61,
207, 270, 271, 290, 339, 350, 397 Spin-echo (SE) .....................................14, 23, 27, 90, 96,
Rapid acquisition using radiofrequency echoes 130, 136, 140
(RARE) ............ 122, 190, 194, 290, 337, 389 Spoiled gradient-echo ............................. 26, 66, 206, 300
PRECLINICAL MRI: METHODS AND PROTOCOLS
464 Index
Statistical parametric mapping (SPM) ..........63, 122, 281 Tissue oxygen level dependent (TOLD) ....................299,
Steady state ...........................................34, 123, 126, 176, 300, 303, 306, 308
182, 184, 396 Transit time ................................................. 43, 44, 67, 68
Stejskal-Tanner (ST) ...................... 91, 95, 136, 353, 379 Tricarboxylic acid cycle (TCA) ..................................... 175
Stimulated echo (STE) ................................................. 154 Tridimensional (3D) ............................................. 6, 9, 51,
Stroke.................................. 36, 45, 54, 59, 67, 104, 154, 96, 141, 215, 219–221, 226, 227, 229, 230,
243, 297, 316, 347, 356, 359, 377–390, 395 243, 246, 247, 263, 285, 286, 288–292, 322,
Stroke volume (SV)....................................................... 279 323, 336–338, 343, 348, 357–359, 386, 424
Superparamagnetic nanoparticles.......................... 25, 447 Tumor ....................................................... 4, 5, 25, 36, 45,
Susceptibility artifacts ................113, 207, 222, 389, 416 59, 154, 155, 297–300, 303, 305–308, 316,
Susceptibility-weighted imaging (SWI) ........................ 34, 332, 333, 335, 337, 339–343, 355, 396, 445,
205–207, 209, 213, 218–221, 224–227, 230, 449, 451–453
357, 381, 386, 387 Tumor oxygenation ...................................................... 299
Turbo spin echo (TSE) ....................................30, 31, 262
T
U
T1 .....................................................................3, 4, 15–18,
23–25, 28, 30, 32–35, 42, 52, 60, 67, 68, 89, Ultrafast gradient echo ................................................... 26
90, 146, 165, 171, 200, 222, 242, 288, 289, Ultra-fast spin echo ......................................................... 26
298, 300, 301, 303, 307–309, 337, 340, 348, Ultra-short echo time (UTE).............270, 273, 281, 282
351, 356, 358, 359, 361, 363, 378, 380, 396,
404, 405, 407, 416, 447, 452 V
T1 relaxation .......................... 15, 30, 340, 348, 356, 366 Ventilation ...........................................251, 433, 435, 436
T1-weighted.................................4, 18, 23, 66, 163, 252, Ventricular ............................................................ 279, 356
288, 289, 294, 306, 337, 343, 349, 355, 356,
Volume of interest (VOI) ............ 96, 141, 190, 193, 243
363, 366, 396, 405, 412, 453 Voxel based analysis (VBA) ................................. 349, 363
T2 ................................................... 3, 23, 24, 42, 89, 122,
140, 157, 171, 193, 242, 262, 289, 305, 348, W
380, 412, 445
T2 relaxation .............................................. 16, 17, 32, 380 Water suppression
T2-weighted............................................4, 14, 17, 19, 64, chemical shift selective (CHESS) .................. 163, 181
122, 131, 157, 158, 178, 252, 288–292, 294, variable power and optimized relaxation delays
356, 366, 389, 412, 413, 415, 416, 451 (VAPOR) ......... 163, 164, 181, 196, 198, 343
T2* ...........................................23, 24, 32–35, 42, 43, 46, White matter (WM) ................................. 29, 35, 65, 106,
50, 52, 56, 89, 125, 225, 264, 288, 289, 107, 200, 226, 294, 348, 349, 355, 359, 360,
298–300, 303, 305, 308, 316, 356, 380, 381, 363, 364, 379, 380, 388
387, 447
X
T2* relaxation ..................................................43, 90, 367
T2*-weighted ............................................. 206, 219, 288, Xenograft .................. 305, 307, 308, 333, 335, 340–342
289, 299, 301, 303, 306, 316, 380, 389, 416 X-nuclei................................................................. 169–171
Target.........................................112, 119, 126, 175, 176, Xylazine................................ 65, 221, 301, 302, 348, 429
182, 183, 211, 236, 237, 239–242, 245, 248,
249, 254, 287, 290, 315, 316, 318–322, 325, Z
409, 410, 425, 431, 442, 444–447, 449
Zero crossing point ......................................................... 29
Theranostic ...........................................442, 447, 449, 452