(Methods in Molecular Biology 1579) Charles A. Galea (Eds.) - Matrix Metalloproteases - Methods and Protocols-Humana Press (2017)
(Methods in Molecular Biology 1579) Charles A. Galea (Eds.) - Matrix Metalloproteases - Methods and Protocols-Humana Press (2017)
(Methods in Molecular Biology 1579) Charles A. Galea (Eds.) - Matrix Metalloproteases - Methods and Protocols-Humana Press (2017)
Matrix
Metalloproteases
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
Charles A. Galea
Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences,
Monash University, Parkville, VIC, Australia
Editor
Charles A. Galea
Drug Delivery, Disposition and Dynamics
Monash Institute of Pharmaceutical Sciences
Monash University
Parkville, VIC, Australia
The matrix metalloprotease (MMP) field has witnessed enormous advances since Jerome
Gross and Charles Lapière first observed in 1962 an enzymatic activity (collagen degrada-
tion) associated with tadpole tail metamorphosis. Since the identification of this enzyme
(interstitial collagenase or MMP-1), more than 20 closely related and evolutionarily con-
served vertebrate MMPs have been discovered. These MMPs and their endogenous inhibi-
tors (TIMPS) are involved in a diverse range of functions including tissue remodeling,
immunity, inflammation, and angiogenesis. The first part of this book outlines recent
advances in the expression and purification of MMPs in various expression systems, high-
lighting the advantages and disadvantages of each. In Part II we highlight how various
biophysical methods such as X-ray crystallography, NMR spectroscopy, and small angle
X-ray scattering, in combination with computational analyses (Part III), can provide a
detailed understanding of the molecular mechanism of action of these enzymes. Part IV
details how experimental and bioinformatics approaches can be used to define the substrate
specificity of MMPs while Part V discusses methods for detecting MMP activity in vitro and
in vivo. In Part VI we present various methods for the development and characterization of
MMP-based inhibitors as potential therapeutics for the treatment of various diseases. The
final part presents an overview of the involvement of MMPs in various diseases and their
potential as diagnostic biomarkers.
v
Contents
Preface . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
vii
viii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 313
Contributors
ix
x Contributors
Joan Hare • Institute of Molecular Biophysics, Florida State University, Tallahassee, FL, USA
Harvey Hensley • Biological Imaging Facility, Fox Chase Cancer Center, Philadelphia,
PA, USA
Robert A. Holland • Institute and Biomedical and Biomolecular Science (IBBS)
and School of Biological Sciences, University of Portsmouth, Portsmouth, UK
Ruchi Jain • Department of Molecular Reproduction, Development and Genetics, Indian
Institute of Science, Bangalore, India
B. Jayaram • Department of Chemistry, Indian Institute of Technology, HauzKhas,
New Delhi, India; Supercomputing Facility for Bioinformatics & Computational Biology,
Indian Institute of Technology, HauzKhas, New Delhi, India; Kusuma School
of Biological Sciences, Indian Institute of Technology, HauzKhas, New Delhi, India
Ulrich auf dem Keller • ETH Zurich, Department of Biology, Institute of Molecular
Health Sciences, Zurich, Switzerland
Nikul S. Khunti • Institute and Biomedical and Biomolecular Science (IBBS) and School
of Biological Sciences, University of Portsmouth, Portsmouth, UK; Diamond Light Source,
Diamond House, Harwell Science and Innovation Campus, Didcot, Oxfordshire, UK
Anna M. Knapinska • Department of Chemistry and Biochemistry, Florida Atlantic
University, Jupiter, FL, USA
Rama K. Koppisetti • Department of Biochemistry, University of Missouri, Columbia, MO,
USA; Department of Medical Microbiology and Immunology, Life Sciences Center,
University of Missouri, Columbia, MO, USA
Colin A. Kretz • Thrombosis and Atherosclerosis Research Institute and Department
of Medicine, McMaster University, Hamilton, ON, Canada
Zon W. Lai • Institute of Molecular Medicine and Cell Research, University of Freiburg,
Freiburg, Germany; Department of Genetics and Complex Diseases, Harvard T.H. Chan
School of Public Health, Boston, MA, USA
Wolfgang Lubitz • Max Planck Institute for Chemical Energy Conversion, Mülheim an
der Ruhr, Germany
Tara C. Marcink • Department of Biochemistry, University of Missouri, Columbia, MO,
USA
Jennifer S. Myers • Department of Chemistry and Biochemistry, Florida State University,
Tallahassee, FL, USA
Hideaki Ogata • Max Planck Institute for Chemical Energy Conversion, Mülheim an der
Ruhr, Germany
Rutvik S. Patel • Department of Molecular Physiology and Biophysics, Baylor College of
Medicine, Houston, TX, USA
Andrew R. Pickford • Institute of Biomedical and Biomolecular Science (IBBS) and
School of Biological Sciences, University of Portsmouth, Portsmouth, UK
Debra L. Quinn • Institute of Biomedical and Biomolecular Science (IBBS) and School of
Biological Sciences, University of Portsmouth, Portsmouth, UK
Harini Ramanan • Department of Molecular Reproduction, Development and Genetics,
Indian Institute of Science, Bangalore, India
Deepak K. Saini • Department of Molecular Reproduction, Development and Genetics,
Indian Institute of Science, Bangalore, India
Qing-Xiang Amy Sang • Department of Chemistry and Biochemistry, Florida State
University, Tallahassee, FL, USA; Institute of Molecular Biophysics, Florida State
University, Tallahassee, FL, USA
Contributors xi
Abstract
The MMP (matrix metalloproteinases) family of endopeptidases are involved in cleavage induced remodel-
ling of the extracellular matrix including collagen, fibrinogen, elastin, and gelatin. Owing to their proteo-
lytic activity which can cleave and degrade multiple intracellular substrates, the overexpression and
purification of these proteins tends to be toxic. Here we describe a novel “matrix assisted refolding” pro-
tocol to overcome the technical challenges associated with overexpression and purification of full-length
MMPs. The toxicity issue associated with MMP expression, is circumvented by expressing the recombi-
nant protein in Escherichia coli in an inactive insoluble form. The methodology used for obtaining full-
length MMP2 protein from these inclusion bodies, by its subsequent purification and refolding using
affinity chromatography, through a single-step matrix based refolding protocol is presented here. The
protocol described yields high concentrations of pure full-length and active MMP2 protein useful for
downstream applications.
1 Introduction
Charles A. Galea (ed.), Matrix Metalloproteases: Methods and Protocols, Methods in Molecular Biology, vol. 1579,
DOI 10.1007/978-1-4939-6863-3_1, © Springer Science+Business Media LLC 2017
3
4 Krishna K. Singh et al.
2 Materials
All the reagents utilized for protein work are from Sigma-Aldrich
(MO, USA), of molecular biology grade, free of DNase, RNase,
and proteases, unless otherwise mentioned.
Compositions of various media and buffers used in the proto-
cols described herein are listed below:
2.1 Plasmids and 1. MMP2 expression plasmid: cDNA for MMP2 was cloned into
E. coli Strains the pPROEx-HTc (Invitrogen, Carlsbad, CA) expression vec-
tor [14].
2. CFP-2RS expression plasmid: CFP-2RS CDS was cloned into
the pPROEx-HTa expression vector [14].
3. E. coli C43: F − ompT hsdSB (rB − mB − ) gal dcm (DE3) pLysS
(CmR ) [15].
4. E. coli BL21 ArcticExpressTM: E. coli B F− ompT hsdS(rB − mB
−
) dcm+ Tetr gal λ(DE3) endA Hte [cpn10 cpn60 Gentr] [16].
2.3 MMP2 1. 1 M sodium phosphate buffer, pH 7.4: Mix 77.4 ml of 1 M
Purification Na2HPO4 (monobasic) and 22.6 ml of 1 M NaH2PO4 (diba-
and Refolding sic) to obtain 100 ml of 1 M sodium phosphate buffer, pH 7.4.
2. 5 M NaCl: Dissolve 29.25 g of NaCl in 100 ml of water.
3. 2 M imidazole: Dissolve 2.72 g imidazole in 20 ml of water.
Store at 4 °C till use or prepare fresh.
4. 100 mM phenyl methyl sulfonylfluoride (PMSF): Dissolve
174 mg of PMSF in 10 ml of absolute ethanol and store at −20
°C in aliquots until use.
5. Native lysis buffer: 50 mM sodium phosphate buffer pH 7.4,
300 mM NaCl, 10 mM imidazole, 1.0 mM PMSF. (Tris–HCl
pH 8.0 can also be used instead of 50 mM sodium phosphate
buffer pH 7.4).
6 Krishna K. Singh et al.
2.4 MMP2 Activity 1. 5× sample buffer: 250 mM Tris–HCl, pH 6.8, 5% SDS, 50%
Analysis glycerol, 0.5% bromophenol blue, and 6.25% β-mercaptoethanol.
2. 2× sample buffer (for zymography): 100 mM Tris–HCl, pH
6.8, 2% SDS, 20% glycerol, and 0.2% bromophenol blue.
3. 10× activation buffer: 500 mM Tris–HCl, pH 7.4, 50 mM
CaCl2, and 10 μM ZnCl2.
4. ARP100 (Cayman Chemical Co., USA): 5 mM ARP100 dis-
solved in DMSO. Aliquots stored at −20 °C till further use.
5. Staining solution: 0.5% CBB R250, 40% methanol, 10% acetic
acid, and 50% water.
6. Destaining solution: 40% methanol, 10% acetic acid, and 50%
water.
3 Methods
3.1 Cloning MMP2 1. PCR amplify cDNA template coding for MMP2, from total
Gene for Prokaryotic RNA extracted from HEK293 cells, using gene specific prim-
Expression ers (see Note 1).
2. The amplified MMP2 ORF is cloned into the EcoRI and XhoI
sites of the pPROEx-HT expression vector, which contains an
N-terminal 6× His tag and a trc promoter upstream of the
MMP2 ORF allowing for IPTG-dependent overexpression of
the fusion protein.
3. Clones were verified by DNA sequencing to ensure the in-
frame fusion of the purification tag with the MMP2 CDS.
MMP2 Purification from E. coli 7
3.2 Transformation 1. Chemically competent E. coli C43 (DE3) pLysS cells were
and Propagation transformed with the recombinant MMP2 expression plasmid
of the MMP DNA (see Notes 2 and 3).
Expression Strain 2. The transformed cells were plated onto Luria Agar (LA) plates
containing 100 μg/ml carbenicillin and incubate overnight at
37 °C. The colonies obtained were immediately used for pro-
tein overexpression (see Note 4).
3.3 Overexpression Given that inclusion body formation is favoured when the expres-
of Recombinant MMP2 sion of recombinant protein is high, conditions for MMP2 overex-
Protein pression had to be optimized to maximize expression and inclusion
in Inclusion Bodies body formation (see Note 5).
1. Inoculate a single colony of E. coli C43 cells into 10 ml of LB,
containing 100 μg/ml carbenicillin (see Note 4) and incubate
overnight at 37 °C.
2. Inoculate 1% of the overnight culture into 1 liter of terrific
broth (see Note 6) containing 35 μg/ml chloramphenicol and
100 μg/ml ampicillin and grow at 37 °C to an OD600 of
0.8–1.0.
3. Add IPTG to the culture to a final concentration of 1 mM and
incubate at 37 °C for a further 24 h at 180 rpm.
4. Pellet the induced culture by centrifuging at 6,000 × g for
10 min at room temperature.
5. Resuspend the pellet in 20 ml of native lysis buffer and sonicate
on ice (Branson Sonifier model S-450D with 1/8″ tapered
microtip) at 50% amplitude, six cycles of sonication with 10 s
of ON and OFF pulse (see Note 7).
6. Centrifuge the sonicated lysate at 20,000 × g for 30 min at 4
°C to separate the soluble and insoluble fractions.
7. The soluble supernatant and insoluble pellet containing the
inclusion bodies are analyzed by SDS-PAGE or western blotting
using anti-His antibody (Fig. 1). If the yield of MMP2 in inclu-
sion bodies is poor then it may be necessary to repeat steps 1–7
using a different concentration of IPTG (see Note 8).
3.4 Matrix Assisted 1. Resuspend the pellet containing the recombinant MMP2
Purification inclusion bodies in solubilization buffer (5 ml buffer for inclu-
and Refolding sion bodies obtained from 1 l of culture) and incubate at 37 °C
of Denatured MMP2 for 2 h at 180 rpm (see Note 9).
2. Centrifuged at 20,000 × g for 30 min at 15 °C to remove
insoluble debris.
3. Load the supernatant containing the denatured MMP2 pro-
tein onto a column containing Ni2+-NTA resin pre-equilibrated
with solubilization buffer (see Note 10). In general, 1 ml of
packed bead volume was used per litre of bacterial culture in a
column of 12–20 ml of total capacity.
8 Krishna K. Singh et al.
Fig. 1 SDS-PAGE analysis for MMP2 protein expression. Bacterial cell lysates
were analyzed for MMP2 expression in soluble (supernatant) and insoluble (pel-
let) fraction. Lane M: Marker; lane 1, purified MMP2 protein; lane 2, soluble frac-
tion; lane 3, insoluble fraction containing inclusion bodies
Table 1
Washing steps used for on-column refolding of the denatured MMP2 protein
Washing Final urea concentration Volume of solubilization buffer Volume of refolding buffer
step (M) (ml) (ml)
1 6 7.50 2.5
2 5 6.25 3.75
3 4 5.0 5.0
4 3 3.75 6.25
5 2 2.50 7.50
6 1 1.25 8.75
3.5 Activity Analysis The MMP2 protein activity was determined using a conventional
of Purified and Refolded “gelatin zymography” methodology as well as using an advanced
MMP2 Protein “form invariant substrate cleavage assay” as described previously [14].
3.5.1 Gelatin The biological activity of the purified MMP2 protein was assessed
Zymography using gelatin based zymography as reported in various studies [17,
18], which is described below:
1. Add 2 μg of purified and refolded MMP2 protein to an equal
volume of 2× sample buffer (see Note 18).
2. Resolve the sample on a 12% SDS-PAGE gel co-polymerized
with 0.1% gelatin at 100 V for 2 h.
3. Carefully remove the gel and wash with activation buffer con-
taining 2.5% Triton X-100 for 1 h at room temperature (see
Note 19).
4. Briefly rinse the gel with deionized water and incubate in acti-
vation buffer (without Triton X-100) overnight at 37 °C.
10 Krishna K. Singh et al.
Fig. 2 SDS-PAGE analysis of refolded MMP2 protein. MMP2 protein purified and
refolded from IBs using affinity chromatography were analyzed on 12% SDS-
PAGE and stained with Coomassie brilliant blue
3.5.2 Form Invariant The total activity of purified MMP2 protein can be further tested
Substrate Cleavage Assay using an advanced substrate cleavage assay (see Note 21) [14].
1. In brief, a synthetic substrate site for MMP2 proteins com-
posed of CFP fluorescent protein fused with MMP2 substrate
recognition sequence IPVS↓LRSG (CFP-2RS), was overex-
pressed, purified and used for activity assessment as previously
described [14].
2. Incubate 2 μg of purified CFP-2RS protein with 500 ng of
purified MMP2 in the presence of 1× activation buffer with or
without the MMP2 specific inhibitor, ARP-100 [19] for 16 h
at 37 °C.
MMP2 Purification from E. coli 11
4 Notes
Fig. 4 Form invariant substrate cleavage assay. Activity of purified and refolded
MMP2 was monitored using recombinant CFP-2RS protein containing a synthetic
MMP2 substrate recognition sequence. SDS-PAGE analysis of CFP-2RS sub-
strate protein incubated with purified and refolded MMP2 in the absence and
presence of the MMP2 inhibitor, ARP100. Lane 1, CFP-2RS only; lane 2, CFP-2RS
+ MMP2; lane 3: CFP-2RS + MMP2 + 1 μM ARP 100; and lane 4, CFP-2RS +
MMP2 + 10 μM ARP 100
Acknowledgments
References
1. Massova I, Kotra LP, Fridman R, Mobashery S cancer progression and their pharmacological
(1998) Matrix metalloproteinases: structures, targeting. FEBS J 278(1):16–27
evolution, and diversification. FASEB 4. Schmalfeldt B, Prechtel D, Härting K, Späthe
J 12(12):1075–1095 K, Rutke S, Konik E, Fridman R, Berger U,
2. Baker AH, Edwards DR, Murphy G (2002) Schmitt M, Kuhn W, Lengyel E (2001)
Metalloproteinase inhibitors: biological actions Increased expression of matrix metalloprotein-
and therapeutic opportunities. J Cell Sci ases (MMP)-2, MMP-9, and the urokinase-
115(Pt 19):3719–3727 type plasminogen activator is associated with
3. Gialeli C, Theocharis AD, Karamanos NK progression from benign to advanced ovarian
(2011) Roles of matrix metalloproteinases in cancer. Clin Cancer Res 7(8):2396–2404
16 Krishna K. Singh et al.
5. Vihinen P, Kähäri V-M (2002) Matrix metal- and activity assessment using a ‘form invariant’
loproteinases in cancer: prognostic markers and assay for matrix metalloproteinase 2 (MMP2).
therapeutic targets. Int J Cancer Mol Biotechnol 56(12):1121–1132
99(2):157–166 15. Miroux B, Walker JE (1996) Over-production
6. Roy R, Yang J, Moses MA (2009) Matrix of proteins in Escherichia coli: mutant hosts
metalloproteinases as novel biomarkers and that allow synthesis of some membrane pro-
potential therapeutic targets in human cancer. teins and globular proteins at high levels. J Mol
J Clin Oncol 27(31):5287–5297 Biol 260(3):289–298
7. Hadler-Olsen E, Winberg JO, Uhlin-Hansen L 16. Ferrer M, Chernikova TN, Yakimov MM,
(2013) Matrix metalloproteinases in cancer: Golyshin PN, Timmis KN (2003) Chaperonins
their value as diagnostic and prognostic mark- govern growth of Escherichia coli at low tem-
ers and therapeutic targets. Tumour Biol peratures. Nat Biotechnol 21(11):1266–1267
34(4):2041 17. Hawkes SP, Li H, Taniguchi GT (2010)
8. Ra HJ, Parks WC (2007) Control of matrix Zymography and reverse zymography for
metalloproteinase catalytic activity. Matrix Biol detecting MMPs and TIMPs. Methods Mol
26(8):587–596 Biol 622:257–269
9. Nagase H, Visse R, Murphy G (2006) Structure 18. Hu X, Beeton C (2010) Detection of func-
and function of matrix metalloproteinases and tional matrix metalloproteinases by zymogra-
TIMPs. Cardiovasc Res 69(3):562–573 phy. J Vis Exp 45:2445
10. Steffensen B, Chen Z, Pal S, Mikhailova M, Su 19. Rossello A, Nuti E, Orlandini E, Carelli P,
J, Wang Y, Xu X (2011) Fragmentation of Rapposelli S, Macchia M, Minutolo F, Carbonaro
fibronectin by inherent autolytic and matrix L, Albini A, Benelli R, Cercignani G, Murphy G,
metalloproteinase activities. Matrix Biol Balsamo A (2004) New N-arylsulfonyl-N-
30(1):34–42 alkoxyaminoacetohydroxamic acids as selective
11. Wang X, Yi J, Lei J, Pei D (1999) Expression, inhibitors of gelatinase A (MMP-2). Bioorg Med
purification and charaterization of recombi- Chem 12(9):2441–2450
nant mouse MT5-MMP protein products. 20. Saini DK, Pant N, Das TK, Tyagi JS (2002)
FEBS Lett 462(3):261–266 Cloning, overexpression, purification, and
12. Carrió MM, Villaverde A (2002) Construction matrix-assisted refolding of DevS (Rv 3132c)
and deconstruction of bacterial inclusion bod- histidine protein kinase of Mycobacterium
ies. J Biotechnol 96(1):3–12 tuberculosis. Protein Expr Purif
13. Tsumoto K, Ejima D, Kumagai I, Arakawa T 25(1):203–208
(2003) Practical considerations in refolding 21. Turk BE, Huang LL, Piro ET, Cantley LC
proteins from inclusion bodies. Protein Expr (2001) Determination of protease cleavage
Purif 28(1):1–8 site motifs using mixture-based oriented
14. Singh KK, Jain R, Ramanan H, Saini DK peptide libraries. Nat Biotechnol
(2014) Matrix-assisted refolding, purification 19(7):661–667
Chapter 2
Abstract
Membrane tethered matrix metalloproteases are bound to the plasma membrane by a
glycosylphosphatidylinositol-anchor or a transmembrane domain. To date, most studies of membrane-bound
matrix metalloprotease have focused on the globular catalytic and protein–protein interaction domains of
these enzymes. However, the transmembrane domains have been poorly studied even though they are known
to mediate intracellular signaling via interaction with various cellular proteins. The expression and purifica-
tion of the transmembrane domain of these proteins can be challenging due to their hydrophobic nature. In
this chapter we describe the purification of a transmembrane domain for a membrane-bound matrix metal-
loprotease expressed in E. coli and its initial characterization by NMR spectroscopy.
1 Introduction
Charles A. Galea (ed.), Matrix Metalloproteases: Methods and Protocols, Methods in Molecular Biology, vol. 1579,
DOI 10.1007/978-1-4939-6863-3_2, © Springer Science+Business Media LLC 2017
17
18 Charles A. Galea
Fig. 1 Structural domains of the membrane-anchored MMPs. MT1-MMP, MT2-MMP, MT3-MM, MT4-MMP,
MT5-MMP, and MT6-MMP share a common domain structure consisting of an N-terminal signal peptide (SP)
that targets the protease to the cell surface, a pro-domain (Pro), a catalytic domain, a linker domain (L1), a
hemopexin (Hpx) domain involved in protein–protein interactions, and a second linker region (L2). MT1-MMP,
MT2-MMP, MT3-MMP, and MT5-MMP contain a C-terminal single helical transmembrane (TM) domain fol-
lowed by a cytoplasmic (CP) domain while MT4-MMP and MT6-MMP possess a C-terminal GPI anchor. MMP23
differs in its domain structure, possessing an N-terminal signal peptide followed by a single type-II helical
transmembrane domain, a pro-domain, a catalytic domain, a cysteine-rich toxin-like (TxD) domain, and a
protein–protein interaction IgCAM domain. The pro-domain (Pro) in most membrane-anchored MMPs (except
MMP23) contains an unpaired cysteine sulfhydryl group that interacts with the active site zinc atom and main-
tains the enzyme in a latent inactive form (the cysteine switch). The MT-MMPs are transported in their inactive
latent form to the cell surface, where they are cleaved at the recognition motif (RXKR) within the pro-domain
by a furin-like protease, resulting in activation of the enzyme
2 Materials
3 Methods
3.3 Purification 1. Weigh the frozen pellet and add 10 ml of lysis buffer per 1 g of
of Isotopically Labeled pellet.
His6-Tagged TM/CPD 2. Place on a rotary mixer and gently resuspend the pellet over
Peptide 30–60 min.
3. Lyse the cells by using either sonication (see Note 23) or a
French press.
4. Centrifuge the lysate at 16,000 × g for 20 min at 4 °C.
Fig. 2 Expression of 15N-labeled His6-tagged Trx-TM/CPD. Protein expression was induced by (a) the addition
of 1 mM IPTG and the culture was grown for 3 h at 37 °C or (b) autoinduction at 28 °C over 48 h
Purification of MMP Transmembrane Domain 25
3.4 Reverse-Phase 1. Add an equal volume of solvent A and then filter the cleaved
HPLC Purification peptide through a 0.22 μm membrane.
of TM/CPD Peptide 2. Load onto a semi-preparative C8 reverse-phase HPLC column
at a flow-rate of 1 ml/min.
3. The column is developed at 0% solvent B for 5 min followed by
a gradient of 0–40% solvent B over 10 min and 40–80% solvent
B over 30 min. Monitor the absorbance at 214 nm and collect
eluting peaks.
4. Freeze the peak fractions in liquid nitrogen and lyophilize.
3.5 NMR Sample 1. Dissolve the dried TM/CPD peptide in 550 μl of NMR buffer
Preparation (see Notes 29 and 30).
2. Vortex briefly for about 30 s to promote the incorporation of
the TM/CPD peptide into the DPC micelles.
3. Take a 50 μl sample to determine the concentration of the
protein and for analysis by circular dichroism (CD) spectros-
copy (Fig. 4b) (see Note 31).
4. Transfer the remaining 500 μl of material to a 5 mm NMR tube
(see Note 32). Store at 4 °C prior to NMR data acquisition.
Fig. 4 The purified TM/CPD protein. (a) C8 reverse-phase HPLC chromatogram for the purification of the TM/
CPD protein. The blue line indicates the percentage of solvent B in the mobile phase. Inset: SDS-PAGE analysis
of HPLC peaks 1 and 2 showing the presence of the TM/CPD protein in both fractions. Native PAGE gel analysis
shows peak 2 contains the dimeric form of the TM/CPD protein (data not shown). (b) CD spectrum recorded at
25 and 40 °C for the TM/CPD protein prepared in the NMR buffer which contains DPC micelles. The spectrum
indicates that part of the protein adopts a helical structure (minima at 208 and 222 nm)
Purification of MMP Transmembrane Domain 27
4 Notes
Fig. 5 NMR spectra for the TM/CPD protein in DPC micelles. 2D 1H/15N TROSY spectra were acquired at 20, 30,
40, and 50 °C on a Varian Inova 600 MHz NMR spectrometer equipped with a cryogenically cooled triple-
resonance probe. Optimal spectral resolution and peak intensities was obtained at 40 °C
References
intracellular organelles and confers resistance 42. Hoshino D, Tomari T, Nagano M, Koshikawa
to lamin A/C degradation during apoptosis. N (2009) A novel protein associated with
Am J Pathol 166(5):1555–1563 membrane-type 1 matrix metalloproteinase
31. Si-Tayeb K, Monvoisin A, Mazzocco C, binds p27kip1 and regulates RhoA activation,
Lepreux S, Decossas M, Cubel G, Taras D, actin remodeling, and matrigel invasion. J Biol
Blanc J-FF, Robinson DR, Rosenbaum Chem 284(40):27315–27326
J (2006) Matrix metalloproteinase 3 is present 43. Wang Y, McNiven MA (2012) Invasive matrix
in the cell nucleus and is involved in apoptosis. degradation at focal adhesions occurs via prote-
Am J Pathol 169(4):1390–1401 ase recruitment by a FAK-p130Cas complex.
32. Itoh Y (2015) Membrane-type matrix metal- J Cell Biol 196(3):375–385
loproteinases: Their functions and regulations. 44. Woskowicz AM, Weaver SA, Shitomi Y, Ito N
Matrix Biol 44-46:207–223 (2013) MT-LOOP-dependent localization of
33. Zucker S, Pei D, Cao J, Lopez-Otin C (2003) membrane type I matrix metalloproteinase
Membrane type-matrix metalloproteinases (MT1-MMP) to the cell adhesion complexes
(MT-MMP). Curr Top Dev Biol 54:1–74 promotes cancer cell invasion. J Biol Chem
34. Hernandez-Barrantes S, Bernardo M, Toth M, 288(49):35126–35137
Fridman R (2002) Regulation of membrane 45. Uekita T, Itoh Y, Yana I, Ohno H, Seiki M
type-matrix metalloproteinases. Semin Cancer (2002) Cytoplasmic tail-dependent internal-
Biol 12(2):131–138 ization of membrane-type 1 matrix metallopro-
35. Galea C, Nguyen H, Chandy K, Smith B, teinase is important for its invasion-promoting
Norton R (2014) Domain structure and func- activity. J Cell Biol 155(7):1345–1356
tion of matrix metalloprotease 23 (MMP23): 46. Wang X, Ma D, Keski-Oja J, Pei D (2004)
role in potassium channel trafficking. Cell Mol Co-recycling of MT1-MMP and MT3-MMP
Life Sci 71(7):1191–1210 through the trans-Golgi network identification
36. Itoh Y, Takamura A, Ito N, Maru Y, Sato H of DKV582 as a recycling signal. J Biol Chem
(2001) Homophilic complex formation of 279(10):9331–9336
MT1-MMP facilitates proMMP-2 activation 47. Wang P, Wang X, Pei D (2004) Mint-3 regu-
on the cell surface and promotes tumor cell lates the retrieval of the internalized membrane-
invasion. EMBO J 20(17):4782–4793 type matrix metalloproteinase, MT5-MMP, to
37. Itoh Y, Ito N, Nagase H, Seiki M (2008) The the plasma membrane by binding to its car-
second dimer interface of MT1-MMP, the boxyl end motif EWV. J Biol Chem
transmembrane domain, is essential for 279(19):20461–20470
ProMMP-2 activation on the cell surface. 48. Nguyen H, Galea C, Schmunk G, Smith B,
J Biol Chem 283(19):13053–13062 Edwards R, Norton R, Chandy K (2013)
38. Itoh Y, Ito N, Nagase H, Evans RD (2006) Intracellular trafficking of the K(V)1.3 potas-
Cell surface collagenolysis requires homodi- sium channel is regulated by the prodomain of
merization of the membrane-bound collage- a matrix metalloprotease. J Biol Chem
nase MT1-MMP. Mol Biol Cell 288(9):6451–6464
17(12):5390–5399 49. Cai M, Huang Y, Sakaguchi K, Clore MG,
39. Sakamoto T, Seiki M (2010) A membrane pro- Gronenborn AM, Craigie R (1998) An effi-
tease regulates energy production in macro- cient and cost-effective isotope labeling proto-
phages by activating hypoxia-inducible factor-1 col for proteins expressed in shape Escherichia
via a non-proteolytic mechanism. J Biol Chem coli. J Biomol NMR 11(1):97–102
285(39):29951–29964 50. Studier FW (2005) Protein production by
40. Gonzalo P, Guadamillas MC, Hernández- auto-induction in high-density shaking cul-
Riquer MV (2010) MT1-MMP is required for tures. Protein Expr Purif 41(1):207–234
myeloid cell fusion via regulation of Rac1 sig- 51. Johnson BA (2004) Using NMRView to visu-
naling. Dev Cell 18(1):77–89 alize and analyze the NMR spectra of macro-
41. Gingras D, Michaud M, Tomasso DG, Béliveau molecules. Methods Mol Biol 278:313–352
E (2008) Sphingosine-1-phosphate induces 52. Delaglio F, Grzesiek S, Vuister GW, Zhu G,
the association of membrane-type 1 matrix Pfeifer J, Bax A (1995) NMRPipe: a multidi-
metalloproteinase with p130Cas in endothelial mensional spectral processing system based on
cells. FEBS Lett 582(3):399–404 UNIX pipes. J Biomol NMR 6(3):277–293
Chapter 3
Abstract
Meprins are zinc-dependent proteases of the metzincin superfamily of metalloproteases. The enzymes are
extracellular multi-domain proteins which are stabilized by disulfide bridges, dimerization, and glycosyl-
ation. Due to their complex structure, recombinant expression was first established in mammalian and
insect cells. However, these methods have several disadvantages such as high costs and the low yields. For
this reason, yeast is often considered a preferable expression system. Here, we describe the manipulation
and secretory expression of human meprin β in the methylotrophic yeast P. pastoris. We show that the posi-
tion of the affinity tag strongly influences the yield of expression, favoring fusion of the affinity tag at the
C-terminus.
1 Introduction
Charles A. Galea (ed.), Matrix Metalloproteases: Methods and Protocols, Methods in Molecular Biology, vol. 1579,
DOI 10.1007/978-1-4939-6863-3_3, © Springer Science+Business Media LLC 2017
35
36 Dagmar Schlenzig and Stephan Schilling
2 Materials
If not otherwise stated, all chemicals and solvents used are of gradi-
ent grade. All materials are sterilized prior to use. Procedures are
carried out under semi-sterile conditions.
2.2 Test Expression 1. BMGY medium: 10 g yeast extract, 20 g peptone, 700 ml
mono-distilled water supplemented with 100 ml 1 M K2SO4
pH 6.0, 100 ml YNB (34 mg yeast nitrogen base w/o amino
acids, 100 mg (NH4)2SO4, and 1 l of water), 2 ml 0.02% bio-
tin, and 100 ml 10% glycerol (filter sterilized) after
autoclaving.
2. BMMY medium: the same as BMGY but replace the glycerol
with 100 ml of 5% methanol.
3. Methanol (99% HPLC grade).
2.3 Activity Assay. 1. Assay buffer 1: 50 mM Tris–HCl, pH 7.5, 20 mM CaCl2.
2. Assay buffer 2: 40 mM Tris, pH 8.0.
3. Trypsin: 400 μg/ml trypsin in assay buffer 2 (see Note 1).
4. Substrate: 50 μM solution of Abz-YVADAP(Dnp)G-OH (or
Mca-YVADAPK(Dnp)-OH) in assay buffer 2 (see Note 2).
3 Methods
Scheme 1 Schematic representation of constructs used for expression of human meprins α and β in
P. pastoris. The native N-terminal signal sequence was replaced by the vector-encoded α-factor pre pro-
sequence of Saccharomyces to enable efficient secretion of the protein. The C-terminal transmembrane
and cytosolic regions were omitted. A His6-tag was introduced either N-terminally between the α-factor
and pro-sequence or C-terminally to accelerate the purification process. Domain structure of meprins
according to UniProt Knowledgebase: S signal sequence, Pro pro-sequence, MAM meprin, 5A protein,
tyrosine phosphatase μ domain, MATH meprin and TRAF homology domain, EGF epidermal growth factor-
like domain, T transmembrane region, I insertion in meprin α which contains a cleavage site. Modified
from [9] with permission of Elsevier
3.1 Plasmid 1. Incubate expression vector (2 μg) with 20 U of PmeI and 5 μl
Linearization 10× CutSmart buffer in a total volume of 50 μl at 37 °C for
and Transformation 1 h. Check for complete linearization using agarose gel elec-
of P. pastoris trophoresis (0.8% gel).
Expression and Purification of Merpin β in Pichia pastoris 39
3.3 Activity Assay 1. Pipet 50 μl of media supernatant to a 96-well plate, add 100 μl
of assay buffer 1, 50 μl trypsin and incubate for 30 min at
37 °C.
2. Transfer 50 μl of each well to a black clear bottom 96-well plate,
add 150 μl of assay buffer 2 and incubate for 10 min at 30 °C. Start
kinetic fluorescence measurement immediately after addition of
50 μl of substrate solution and measure time-dependent conver-
sion of substrate at excitation and emission wavelengths of 340
and 420 nm, respectively (Fig. 1, see Note 8).
3. Select samples with the highest RFU/min and save the corre-
sponding colonies as glycerol stocks (Figs. 1 and 2, see Note 9).
40 Dagmar Schlenzig and Stephan Schilling
Fig. 1 (a) Meprin β activity determined in the medium after small-scale expression of C-terminally or
N-terminally His6-tagged meprin β in P. pastoris. Meprin β activity in the media of at least 50 clones after
an activation step with trypsin depicted as logarithmic scatter plot. On average, the activity for NHis-meprin
β is more than 20-fold lower compared with CHis-meprin β. (b) The difference in expression efficiency is
also reflected by Western blot analysis. A total amount of 100 μg protein from the fermentation medium was
applied to each lane. Meprin β was detected using a polyclonal antibody. (c) No enzymatic activity could be
detected after test expression of CHis-meprin α in P. pastoris. On the protein level, a signal of reduced size
was detected with a polyclonal antibody. ServaGelTM TG PrimeTM 4–20%, Modified from [9] with
permission of Elsevier
Fig. 2 Activity measurement of three selected colonies expressing CHis-meprin β or NHis-meprin β (Fig. 1),
respectively. Prior to measurement, samples were activated by incubation with trypsin. Assays were started by
addition of substrate. Samples of CHis-meprin β were diluted 1:10
Expression and Purification of Merpin β in Pichia pastoris 41
3.5 Purification 1. Thaw the supernatant and dilute it with 1:10 of the final total
(Fig. 3, See Note 12) volume with equilibration buffer 3. Centrifuge at 30,000 × g
for 20 min at 4 °C and apply the supernatant to a Streamline
chelating expanded bed column (25 × 280 mm in non-
expanded state; GE Healthcare Life Science, Piscataway, USA,
see Note 13) previously equilibrated with equilibration buffer
4 in reverse flow at a flow rate of 12 ml/min. After a washing
step with wash buffer 5 the enzyme is eluted in forward flow
with elution buffer 6. The eluted protein is stored at 4 °C
overnight.
2. The eluted enzyme is supplemented with CaCl2 (final concen-
tration of 10 mM). One milligram of trypsin is added per
15 mg of protein and incubated with gentle shaking at room
temperature for 15 min. Afterwards, add AEBSF (final concen-
tration of 5 mM) for inhibition of trypsin.
3. The volume of the sample is doubled by the addition of 3 M
(NH4)2SO4 in the cold (see Note 14). Syringe filter through a
0.46 μM filter membrane to remove turbidity. Load the sample
onto a pre-equilibrated Butyl Sepharose column (25 × 100 mm,
GE Healthcare Life Science, Piscataway, USA, equilibrated
with buffer 7) at a flow rate of 3 ml/min. Elute with an ammo-
nium sulfate gradient (1.5–0 M ammonium sulfate acheived by
mixing buffers 7 and 8) for 10 column volumes at the same
flow rate.
4. Eluted protein is concentrated by ultrafiltration (MWCO
30,000 Da) and applied onto a Superdex 200 column
(26 × 850 mm, material GE Healthcare Life Science, Piscataway,
USA) equilibrated with buffer 9 (see Notes 15 and 16). Eluted
42 Dagmar Schlenzig and Stephan Schilling
Fig. 3 Purification of CHis-meprin β expressed in P. pastoris. (a) Chromatogram of purification of the fermenta-
tion broth by immobilized metal chelating chromatography (IMAC). (b) Chromatogram of purification of the
eluate from IMAC using hydrophobic interaction chromatography. (c) Final purification performing size exclu-
sion chromatography. Bars indicate meprin containing fractions
4 Notes
References
1. Dumermuth E, Sterchi EE, Jiang WP et al side specificity for the astacin metalloprotease
(1991) The astacin family of metalloendopepti- family reflected by physiological substrates. Mol
dases. J Biol Chem 266(32):21381–21385 Cell Proteomics 10(9):M111.009233
2. Sterchi EE, Stocker W, Bond JS (2008) 4. Kronenberg D, Bruns BC, Moali C et al (2010)
Meprins, membrane-bound and secreted asta- Processing of procollagen III by meprins: new
cin metalloproteinases. Mol Aspects Med players in extracellular matrix assembly? J Invest
29(5):309–328 Dermatol 130(12):2727–2735
3. Becker-Pauly C, Barre O, Schilling O et al 5. Bao J, Yura RE, Matters GL et al (2013) Meprin
(2011) Proteomic analyses reveal an acidic prime A impairs epithelial barrier function, enhances
Expression and Purification of Merpin β in Pichia pastoris 45
monocyte migration, and cleaves the tight junc- cancer and fibrosis. Biochem J 450(2):
tion protein occludin. Am J Physiol Renal 253–264
Physiol 305(5):F714–F726 8. Biasin V, Marsh LM, Egemnazarov B et al
6. Huguenin M, Muller EJ, Trachsel-Rosmann S (2014) Meprin β, a novel mediator of vascular
et al (2008) The metalloprotease meprinbeta remodelling underlying pulmonary hyperten-
processes E-cadherin and weakens intercellular sion. J Pathol 233(1):7–17
adhesion. PLoS ONE 3(5):e2153 9. Schlenzig D, Wermann M, Ramsbeck D et al
7. Broder C, Becker-Pauly C (2013) The metal- (2015) Expression, purification and initial char-
loproteases meprin α and meprin β: unique acterization of human meprin β from Pichia
enzymes in inflammation, neurodegeneration, pastoris. Protein Expr Purif 116:75–81
Part II
Abstract
Matrix Metalloproteinases (MMPs) are a family of proteolytic enzymes whose endopeptidase activity is
dependent on the presence of specific metal ions. MT1-MMP (or MMP-14), which has been implicated
in tumor progression and cellular invasion, contains a membrane-spanning region located C-terminal to a
hemopexin-like domain and an N-terminal catalytic domain. We recombinantly expressed the catalytic
domain of human MT1-MMP in E. coli and purified it from inclusion bodies using a refolding protocol
that yielded significant quantities of active protein. Crystals of MT1-MMP were obtained using the vapour
diffusion method. Here, we describe the protocols used for crystallization and the data analysis together
with the resulting diffraction pattern.
1 Introduction
Charles A. Galea (ed.), Matrix Metalloproteases: Methods and Protocols, Methods in Molecular Biology, vol. 1579,
DOI 10.1007/978-1-4939-6863-3_4, © Springer Science+Business Media LLC 2017
49
50 Elena Decaneto et al.
2 Materials
2.4 Computer The computer programs for data analysis can be found on the
Programs Internet websites.
1. Linux operation system (Ubuntu12.04).
2. XDS (X-ray Detector Software), XSCALE and XDSCONV
(http://xds.mpimf-heidelberg.mpg.de/).
3. CCP4i (http://www.ccp4.ac.uk/).
4. PHENIX (http://www.phenix-online.org/).
5. COOT (http://www2.mrc-lmb.cam.ac.uk/personal/pems-
ley/coot/).
6. PYMOL (https://www.pymol.org/).
3 Methods
3.1 Purification 1. Transform E. coli BL21 (DE3) chemically competent cells with
the pET-3a plasmid containing the catalytic domain of human
3.1.1 Induction MT1-MMP together with the hinge-linker to the hemopexin-
and Harvest like domain (residues 112–292) [5]. Transfer 1 μL of DNA to
100 μL of E. coli strain BL21 (DE3) competent cells in a sterile
tube and kept on ice for 30 min.
2. Incubate the cells at 40 °C for 90 s and then place on ice for
2 min.
3. Add 60 μL of cells to 1 mL of SOC medium in a 15 mL sterile
tube (see Note 1).
4. Incubate the cells at 37 °C for 1 h at 180 rpm.
5. Streak cells on 2YT agar plates containing ampicillin (100 μg/
mL) and incubate overnight at 37 °C.
6. Inoculate a single colony into LB media containing 100 μg/
mL of ampicillin and grow overnight at 37 °C (see Note 2).
7. Inoculate 1 L of LB media containing ampicillin with an ali-
quot (50–100 mL) of the overnight culture and grow at 37 °C
to an OD600 of 0.8.
8. Induce cells with IPTG (1 mM) for a further 5 h at 37 °C.
9. Following induction, harvest the cells by centrifugation
(6,000 × g, 4 °C, 15 min) and suspend the cell pellet in 0.9%
NaCl (see Note 3).
X-ray Diffraction Studies of Matrix Metalloproteases 53
3.2 Crystallization Screening of crystallization conditions was carried out by the sit-
ting vapour-diffusion method with a liquid dispensing robot,
PHOENIX (Art Robbins Instruments, CA, USA).
54 Elena Decaneto et al.
3.3 Crystal Handling 1. Prepare the cryoprotectant buffer (see Note 10).
for the Cryo 2. Open the seal of the crystallization plate.
Experiments
3. Pick the crystals with a cryo-loop under the microscope.
4. Soak the crystals in the cryoprotectant buffer and wait for sev-
eral minutes (1─5 min).
5. Pick a single crystal and freeze in liquid nitrogen (see Note
11).
6. Put the cryocap on the cryo-loop vial in liquid nitrogen and
position it on the cryo-cane (see Note 12).
7. Store the cryo-cane in the precooled dryshipper for future
analysis (see Note 13).
3.4 X-Ray Diffraction Diffraction data sets were collected on beamline BL14.2 at
Data Collection BESSYII (Hermholtz-Zentrum Berlin, Germany).
1. The CCD detector is a Rayonix MX-225 (Rayonix, USA). Set
the detector-to-crystal distance to 300 mm.
2. Set the cryo nitrogen gas-stream temperature at 100 K.
3. Mount the crystal with the cryo-loop on the goniometer. Then
align the crystal to the center of the X-ray beam position.
4. Set the X-ray wavelength of 0.91841 Å (maximum X-ray beam
intensity).
5. Collect a diffraction image and check the highest resolution
diffraction spot. If the resolution is higher, set the detector-to-
crystal distance shorter.
6. Repeat step 5 until the maximum resolution of the diffraction
spots is covered.
Fig. 2 MT1-MMP crystals. (a) The original MT1-MMP crystals and (b) stained with the blue colored Izit dye
56 Elena Decaneto et al.
3.5 Data Analysis The computer programs described below were used for data analy-
sis on a Linux operating system (Ubuntu12.04).
1. The diffraction images are indexed and integrated using the
program XDS [13]. The input file template to use is supplied
by the beamline facility. The input parameters are as follows;
JOB = XYCORR INIT COLSPOT IDXREF DEFPIX XPLAN
INTEGRATE CORRECT, DETECTOR_DISTANCE =
250.0, OSCILLATION_RANGE = 1.0, X-RAY_
WAVELENGTH = 0.91841, NAME_TEMPLATE_OF_
DATA_FRAMES = mmp_1_???.img, DATA_RANGE = 1100
(XDS will substitute the appropriate image number for the
question marks), SPOT_RANGE = 1 11, SPOT_RANGE =
42 52, SPOT_RANGE = 73 78, SPACE_GROUP_
NUMBERS = 96, UNIT_CELL_PARAMETERS = 63 63
123 90 90 90, INCLUDE_RESOLUTION_RANGE = 50.0
2.24, FRIEDEL’S_LAW = TRUE (see Note 14).
2. Scaling is carried out using XSCALE. In the input file, select
the integrated data (XDS_ASCII.HKL) as the input file. The
output file is named as MT1-MMP.xscale. Set the parameters,
INCLUDE_RESOLUTION_RANGE = 50 2.24, OUTPUT_
FILE = MT1-MMP.xscale.
3. The scaled file (MT1-MMP.xscale) is converted to the MTZ
file format (CCP4) by XDSCONV. Set the parameters:
GENERATE_FRACTION_OF_TEST_REFLECTIONS =
0.05, FRIEDEL’S_LAW = TRUE.
(a) For molecular replacement, an initial model, the MT1-
MMP/TIMP-2 complex (PDB entry 1BQQ) [10], was cho-
sen from the PDB database (http://www.rcsb.org/).
4. Before molecular replacement, remove the coordinates for
TIMP-2 from the PDB file with a text editor and save this
(1BQQ-MT1-MMP.pdb) as the search model.
5. In the program CCP4i [14], create a project (MT1-MMP)
using the “Change Project” tab.
6. Select “Run Molrep –auto MR” [15] in the “Molecular
Replacement” tab.
7. Select the reflection file MT1-MMP.mtz from XDSCONV as
the input file. Set the parameters; Model in “1BQQ-MT1-
MMP.pdb”, Coords out “MT1-MMP-MR.pdb”, Use data to
maximum resolution “2.24” in the Experimental Data tab,
Search for “1” monomers in the asymmetric unit in the Search
Parameters, then run the program.
X-ray Diffraction Studies of Matrix Metalloproteases 57
4 Notes
Acknowledgment
We thank Dr. Moran Grossman and Prof. Irit Sagi for providing
the plasmid containing human MT1-MMP and Yvonne
Brandenburger and Ingeborg Heise for technical assistance. This
work was supported by the Cluster of Excellence RESOLV (grant
No. EXC 1069) funded by the Deutsche Forschungsgemeinschaft
and the Max Planck Society.
References
1. Tallant C, Marrero A, Gomis-Ruth FX (2010) metalloprotease active site. Nat Struct Mol Biol
Matrix metalloproteinases: Fold and function 18:1102–1108
of their catalytic domains. Biochim Biophys 6. Bertini I et al (2012) The catalytic domain of
Acta Molecular Cell Research 1803:20–28 MMP-1 studied through tagged lanthanides.
2. Vandenbroucke RE, Libert C (2014) Is there Febs Lett 586:557–567
new hope for therapeutic matrix metallopro- 7. Pavlovsky AG et al (1999) X-ray structure of
teinase inhibition? Nat Rev Drug Discov human stromelysin catalytic domain com-
13:904–927 plexed with nonpeptide inhibitors: implications
3. Gialeli C, Theocharis AD, Karamanos NK for inhibitor selectivity. Protein Science
(2011) Roles of matrix metalloproteinases in 8:1455–1462
cancer progression and their pharmacological 8. Bertini I et al (2006) Snapshots of the reaction
targeting. FEBS J 278:16–27 mechanism of matrix metalloproteinases.
4. Decaneto E et al (2015) Pressure and tempera- Angew Chem Int Ed 45:7952–7955
ture effects on the activity and structure of the 9. Grossman M et al (2010) The intrinsic protein
catalytic domain of human MT1-MMP. Biophys flexibility of endogenous protease inhibitor
J 109:2371–2381 TIMP-1 controls its binding interface and
5. Grossman M et al (2011) Correlated structural affects its function. Biochemistry
kinetics and retarded solvent dynamics at the 49:6184–6192
60 Elena Decaneto et al.
10. Fernandez-Catalan C (1998) Crystal structure 16. Adams PD, Afonine PV, Bunkoczi G et al
of the complex formed by the membrane type (2010) PHENIX: a comprehensive Python-
1-matrix metalloproteinase with the tissue based system for macromolecular structure
inhibitor of metalloproteinases-2, the soluble solution. Acta Cryst Sec D 66:213–221
progelatinase A receptor. EMBO 17. Emsley P, Lohkamp B, Scott WG et al (2010)
J 18:5238–5248 Features and development of coot. Acta Cryst
11. Bertini I et al (2003) X-ray structures of binary Sec D 66:486–501
and ternary enzyme-product-inhibitor com- 18. Wilkins MR et al (1999) Protein identification
plexes of matrix metalloproteinases. Angew and analysis tools in the ExPASy server.
Chem Int Ed 42:2673–2676 Methods Mol Biol 112:531–552
12. Ogata H et al (2014) Crystalization and pre- 19. Decaneto E et al (2015) A caged substrate pep-
liminary X-ray crystallographic analysis of tide for matrix metalloproteinases. Photochem
the catalytic domain of membrane type 1 Photobiol Sci 14:300–307
matrix metalloproteinase. Acta Cryst F 20. Neumann U et al (2004) Characterization of
70:232–235 M c a - L y s - P r o - L e u - G l y - L e u - D p a - A l a -
13. Kabsch W (2010) XDS. Acta Cryst Arg-NH2, a fluorogenic substrate with
D66:125–132 increased specificity constants for collagenases
14. Winn MD et al (2011) Overview of the CCP4 and tumor necrosis factor converting enzyme.
sute and current developments. Acta Cryst Sec Analytical Biochemistry 328:166–173
D 67:235–242 21. Krug M, Weiss MS, Heinemann U, Mueller U
15. Vagin AA, Teplyakov A (2010) An approach to (2012) XDSAPP: a graphical user interface for
multi-copy search in molecular replacement. the convenient processing of diffraction data
Acta Cryst D 56:1622–1624 using XDS. J Appl Cryst 45:568–572
Chapter 5
Abstract
Peripheral binding of proteins to lipid bilayers is critical not only in intracellular signaling but also in metal-
loproteinase shedding of signaling proteins from cell surfaces. Assessment of how proteins recognize fluid
bilayers peripherally using crystallography or structure-based predictions has been important but incom-
plete. Assay of dynamic protein–bilayer interactions in solution has become feasible and reliable using
paramagnetic NMR and site-directed fluor labeling. Details of preparations and assay protocols for these
spectroscopic measurements of bilayer proximity or contact, respectively, are described.
1 Introduction
*
These authors contributed equally to this work.
Charles A. Galea (ed.), Matrix Metalloproteases: Methods and Protocols, Methods in Molecular Biology, vol. 1579,
DOI 10.1007/978-1-4939-6863-3_5, © Springer Science+Business Media LLC 2017
61
62 Tara C. Marcink et al.
2 Materials
3 Methods
3.1 Navigating Two excellent servers are available to predict the atomic structural
Servers for Predicting coordinates of proteins that peripherally bind to membranes:
Protein Binding Membrane Optimal Docking Area (MODA) [43] and Positioning
Peripherally of Proteins in Membranes (PPM) [44]. MODA is designed for
to Membranes predicting membrane binding sites of peripheral membrane pro-
teins [43]. PPM is designed for predicting the positioning of both
peripheral and integral membrane proteins with respect to bilayers
[44]. PPM was preceded by the well-developed OPM database of
predictions of positions and orientations of proteins in and on
membranes [44–47]. The predictions may be used to determine
whether it is worthwhile undertaking experimental testing of
membrane interactions. For example, a comparison of a PPM pre-
diction for the catalytic domain of MMP-12 with results of experi-
mental docking studies using paramagnetic NMR [13] is shown in
Fig. 1. The PPM prediction failed to predict the α-interface (Fig.
1a). However, it did predict one of the three experimentally deter-
mined loops (the II–III loop) of the β-interface [13]. This predic-
tion bears resemblance to the measured orientation, but approaches
the bilayer at a different angle (Fig. 1b). While MODA predicted
residues for the other two experimentally defined loops (III–IV
and IV–V loops) within the β-interface [13].
3.1.1 MODA and PPM 1. First obtain the PDB file containing the structural coordinates
Predictions for the protein of interest from the Protein Data Bank (www.
rcsb.org).
2. MODA predictions (http://molsoft.com/~eugene/moda/
modamain.cgi).
(a) Enter the PDB accession code or upload the PDB coordi-
nate file. If the PDB file contains multiple chains, indicate
which chain should be used for the prediction, e.g., A, B,
C. Select “Predict” and wait for the results (see Note 5).
(b)
After the server identifies potential sites of membrane
binding, it outputs a tabulated list of MODA scores for
each of the amino acid residues in the sequence. The plain
MODA score is scaled down by curvIndex to the more
conservative curvMODA score.
(c) If the curvMODA score for a residue is above 40 [48] and
it has one or more neighbors (in sequence or space) with a
high curvMODA score, that patch of residues is likely to
66 Tara C. Marcink et al.
Fig. 1 Comparison of the PPM predicted and experimentally determined mode of bilayer binding by MMP-12.
The protein backbone shown is used both to illustrate the experimental dockings and the predicted docking.
The lipid chains plotted are from the experimental docking. The blue grid indicates the predicted location of
bilayer head groups where the bilayer contacts the II–III loop of MMP-12. (a) The experimental structural model
bound to the membrane via the α-interface (PDB: 2MLR, with lipid chains) is superposed with predicted inter-
faced marked by the blue grid. Molecules comprising the bilayer are shown and phosphorous atoms of the
DMPC head groups are illustrated as orange spheres. The protein chain is rainbow colored from blue at the
N-terminus to red at the C-terminus. (b) The experimental structural model bound to the membrane via the
β-interface (PDB: 2MLS, with lipid chains) which includes not only the II–III loop but also the III–IV and IV–V
loops, resulting in a different tilt angle than predicted by PPM (blue grid)
3.3 Assignment 1. Initially check to determine whether the assignments for the
of NMR Spectral Peaks backbone amide NMR peaks are not available in the literature
or in the BioMagResBank database (http://www.bmrb.wisc.
edu/).
2. If de novo NMR peak assignments are required, prepare the
15
N/13C labeled protein at a concentration of at least 150 μM
using the procedure outlined in Subheading 3.2 (see Notes 7
and 8).
3. Acquire standard NMR spectra for spectral peak assignments
(including HNCA, HN(CO)CA, CBCA(CO)NH and
HNCACB triple resonance spectra) (see Note 9).
4. Semi-automated assignments for a majority of the backbone
resonance peaks can be readily determined by non-specialists
using the program PINE, a downloadable, integrated software
environment which comes with a set of explanatory tutorials
[58]. The software relies heavily upon NMR spectral analysis
software NMRFAM-Sparky for visualizing the spectra [59]
and PONDEROSA for automation of peak assignments [60,
61].
5. Assignments of methyl peaks for hydrophobic side chains are
optional, but are desirable in order to provide additional dis-
tance constraints between protein residues and atoms within
the lipid bilayer. This typically involves selective “ILV” labeling
of isoleucine, leucine, and valine residues in a perdeuterated
medium supplemented with α-ketoacids containing 1H/13C–
labeled methyl group(s) and optional deuteration [62] (see
Note 10).
6. Methyl peaks can be assigned for smaller proteins using
HMCM[CG]CBCA (HMCACB) [63] and CC(CO)NH or
HNCACB triple resonance spectra, supplemented with
NOESY-derived NOEs to the backbone [64] and methyl-
amides [56]. This strategy works well for single domain pro-
teins. To extend assignments of methyl peaks to multiple
domain protein constructs such as MMPs, the assignments of
methyl peaks from the single domains can simply be combined
with NOEs from methyl groups in the larger construct [64,
65]. Software designed to aid in the determination of methyl
peak assignments that utilizes additional experimental data
such as structural coordinates, methyl-methyl NOEs, and
PREs is also available [66].
Mapping Peripheral Membrane Binding by Matrix Metalloproteases 69
3.4 Preparation Bilayered micelles or bicelles that are small, discoidal, and isotropic
of Small Bicelles in tumbling are preferred for solution NMR [30, 32, 33, 67].
for NMR Studies These are composed of 2–3 equivalents of detergent-like short
chain lipids per equivalent of long chain phospholipid, which are
designated q = 0.5 to q = 0.33 (ratio of long to short chains),
respectively. Short chain lipids are amendable to forming curved
surfaces, in this case, the rims of the bicelle disks [30] (Fig. 2b).
Long chain phospholipids, such as DMPC, form the planar bilay-
ers in the interior of the disks (Fig. 2b). Dihexanoylphosphatidylcholine
(D6PC) with its 6-carbon acyl chains has dominated biophysical
studies [30] but suffers the limitation of not aggregating into
micelles or bicelles until reaching a critical micelle concentration
(CMC) as high 14 mM in monomers [33]. D7PC with its 7-carbon
acyl chains has the much lower and more favorable CMC of 1.2
mM allowing D7PC-DMPC bicelles (q ≤ 0.5) to form at much
lower lipid concentrations with only 1.2 mM monomers in solu-
tion [33]. Preparation of bicelles incorporating the latter and using
our recommended formulation is outlined below.
1. Weigh 20 mg of the powder form of D7PC into a clean 5 ml
glass tube and dissolve in 1.5 ml of chloroform.
2. Blow a gentle stream of argon or nitrogen gas into the glass
tube for 30–45 min to evaporate the chloroform. Disturbance
Fig. 2 Phosphatidylcholine molecules with 7- and 14-carbon acyl chains (a) form the annulus and center,
respectively, of disk-like bicelles (b)
70 Tara C. Marcink et al.
3.5 Preparation This is a synopsis of the previously reported protocol for preparing
of Nanodiscs for NMR nanodiscs [42, 68, 69]. This protocol uses the MSP1D1 variant of
or Fluorescence the molecular scaffolding protein to encircle a DMPC phospho-
Assays lipid disc. Other lipids and MSP variants may also be assembled
into nanodiscs.
Mapping Peripheral Membrane Binding by Matrix Metalloproteases 71
3.6 NMR to Measure The main idea here is to estimate distances from protein amide and
Proximity to Mimics methyl groups to doxyl spin labeled DPPC, with around one or
of Membrane Bilayers two such DPPC added per leaflet of the bilayer mimetic. PRE
(paramagnetic relaxation enhancement) measurements from the
unpaired electron of the doxyl group to the amide or methyl pro-
ton of the protein depends strongly (α r−6) on the distance r
between them and less strongly on the time constant τc of their
rotational diffusion. The PREs are measured from the increased
paramagnetic relaxation rate enhancement Γ2 resulting from the
addition of an unpaired electron.
1. Prepare a 15N-2H labeled protein sample according to
Subheading 3.1.1 and concentrate to between 200 and 600
μM in NMR buffer in a volume of at least 400 μl, typically
using a centrifugal membrane concentrator (Millipore or
Vivaspin).
72 Tara C. Marcink et al.
d N = g N B0 Dd N / (3 2) (3)
Using Eq. (5), ηxy is plotted versus τc and is almost linear. At the
measured ηxy rate, the τc value corresponding to this point on
the line is read from this plot. This estimate of τc represents the
hydrodynamics. It can be monitored throughout the titration.
An increase in τc during the titration indicates the binding of
protein to the membrane and slower tumbling of the protein
due to its association with the membrane.
74 Tara C. Marcink et al.
r = 4K t c / G 2 (6)
Fig. 5 The site-directed fluor labeling (SDFL) approach for defining peripheral membrane-binding sites for a
protein. When the IANBD (red) conjugated to the protein (blue) inserts in a hydrophobic compartment such as
the bilayer, its fluorescence emission increases
78 Tara C. Marcink et al.
3.8.2 Fluorescent 1. In a 96-well plate prepare a series of 200 μl solutions (in qua-
Binding Assay druplicate) containing 10–100 nM fluorophore-conjugated
protein. Measure fluorescence at 541 nm using an excitation
wavelength of 478 nm to establish sensitivity and fluorescence
linearity (see Note 30).
2. Each 200 μl solution should contain 250–300 μM liposome
monomers, typically prepared by diluting 5–6 μl from a 10
mM stock solution. Measure fluorescence intensity every
10–30 min for 2–3 h in order to allow the protein time to
equilibrate (i.e., insert into the liposomes), before photo-
bleaching takes a toll on emission from the IANBD. Normalize
the fluorescence intensity in the presence of the liposomes
(FSUV) by dividing it by the fluorescence in the absence of
membranes (F0), (i.e., F0/FSUV). Bilayer insertion of the
NBD moiety is accompanied by an increase in fluorescence
intensity with FSUV/F0 ≥ 1.5 [13, 73] (see Note 31).
3.8.3 Confocal Imaging 1. The IANBD-tagged protein may also be used to investigate
of Binding to Live Cells interactions with cell membranes and their compartments.
We have used human HeLa cells or Raw264.7 murine macro-
phages grown in suspension [13, 14], (Macrophages secrete
4 Notes
15. The 1:1 assemblies should tumble faster and have sharper
NMR peaks than 2:1 assemblies.
16. Note residues with NMR peaks broadened or shifted by addi-
tion of the membrane mimics. Such spectral perturbations
suggest that the protein is binding to the membrane mimic
and the general location of binding or conformational pertur-
bations linked to binding.
17. This is performed before doping the bilayers with doxyl-
substituted DPPC, which is paramagnetic.
18. We have had success with D7PC/DMPC (q = 0.5) bicelles at
300 μM discoidal aggregates, corresponding to 100 mM in
total lipid monomers. These conditions require addition of
the doxyl-DPPC to 600 μM for an average of one per leaflet.
19. The R2 1H NMR relaxation rate constants fitted to the series
should be insensitive to experimental choices of length of sig-
nal averaging, spectral window, and exact concentrations.
20. The R2 values do depend on the hydrodynamics, which are
sensitive to temperature and degree of molecular association,
which should be kept uniform for making comparisons. Using
similar protein and lipid concentrations for the diamagnetic
(control) and paramagnetic experiments ensures similar
degrees of molecular association are obtained in both
experiments.
21. Minimize exposure to air while weighing the dry lipids. Weigh
out a 10% excess of DMPC in order to obtain the desired con-
centration of SUVs since ~10% of the DMPC will form MUVs
which will subsequently be removed by microcentrifugation.
22. Lipids dissolved in chloroform can be used to prepare the
SUVs, provided an additional day is allowed to evaporate the
chloroform and dry the lipids overnight under vacuum.
23. The aggregates could be sonicated to recover SUVs, but it is
not recommended.
24. If one or more such cysteine already exists in the protein at an
experimentally undesirable location, it should first be removed
by site-directed mutagenesis.
25. If mapping by NMR (preferably paramagnetic NMR) is not
available, the interfaces predicted by MODA and PPM should
be regarded as the hypotheses to test, realizing that these algo-
rithms have each missed one of the dual membrane-binding
sites that we located experimentally [13, 14] (Fig. 1a).
26. Since basic and hydrophobic residues are frequently found
within peripheral membrane interfaces, care should be taken
not to substitute Arg, Lys, Phe, Tyr, Leu, and Ile residues
within this region as this could disrupt binding at the protein–
membrane interface.
82 Tara C. Marcink et al.
Acknowledgment
References
1. Moravcevic K, Oxley CL, Lemmon MA (2012) metalloproteinase expressed on the surface of
Conditional peripheral membrane proteins: invasive tumour cells. Nature 370:61–65
facing up to limited specificity. Structure 7. Strongin AY, Collier I, Bannikov G, Marmer
20:15–27 BL, Grant GA, Goldberg GI (1995) Mechanism
2. Sternlicht MD, Werb Z (2001) How matrix of cell surface activation of 72-kDa type IV col-
metalloproteinases regulate cell behavior. Annu lagenase. Isolation of the activated form of the
Rev Cell Dev Biol 17:463–516 membrane metalloprotease. J Biol Chem
3. Clark P (2014) Protease-mediated ectodomain 270:5331–5338
shedding. Thorax 69:682–684 8. Yu WH, Woessner JF Jr, McNeish JD,
4. Rose-John S (2013) ADAM17, shedding, Stamenkovic I (2002) CD44 anchors the
TACE as therapeutic targets. Pharmacol Res assembly of matrilysin/MMP-7 with heparin-
71:19–22 binding epidermal growth factor precursor and
5. Edwards DR, Handsley MM, Pennington CJ ErbB4 and regulates female reproductive organ
(2008) The ADAM metalloproteinases. Mol remodeling. Genes Dev 16:307–323
Aspects Med 29:258–289 9. Berton A, Selvais C, Lemoine P, Henriet P,
6. Sato H, Takino T, Okada Y, Cao J, Shinagawa Courtoy PJ, Marbaix E, Emonard H (2007)
A, Yamamoto E, Seiki M (1994) A matrix Binding of matrilysin-1 to human epithelial
Mapping Peripheral Membrane Binding by Matrix Metalloproteases 83
cells promotes its activity. Cell Mol Life Sci (2006) Hsc70 contacts helix III of the
64:610–620 J domain from polyomavirus T antigens:
10. Owen CA, Hu Z, Lopez-Otin C, Shapiro SD addressing a dilemma in the chaperone hypoth-
(2004) Membrane-bound matrix metallopro- esis of how they release E2F from pRb.
teinase-8 on activated polymorphonuclear cells Biochemistry 45:6917–6929
is a potent, tissue inhibitor of metalloproteinase- 21. Palmier MO, Fulcher YG, Bhaskaran R, Duong
resistant collagenase and serpinase. J Immunol VQ, Fields GB, Van Doren SR (2010) NMR
172:7791–7803 and bioinformatics discovery of exosites that
11. Owen CA, Hu Z, Barrick B, Shapiro SD tune metalloelastase specificity for solubilized
(2003) Inducible expression of tissue inhibitor elastin and collagen triple helices. J Biol Chem
of metalloproteinases–resistant matrix metallo- 285:30918–30930
proteinase-9 on the cell surface of neutrophils. 22. Gobl C, Madl T, Simon B, Sattler M (2014)
Am J Respir Cell Mol Biol 29:283–294 NMR approaches for structural analysis of
12. Cobos-Correa A, Trojanek JB, Diemer S, Mall multidomain proteins and complexes in solu-
MA, Schultz C (2009) Membrane-bound tion. Prog Nucl Magn Reson Spectrosc
FRET probe visualizes MMP12 activity in pul- 80:26–63
monary inflammation. Nat Chem Biol 23. Hennig J, Warner LR, Simon B, Geerlof A,
5:628–630 Mackereth CD, Sattler M (2015) Structural
13. Koppisetti RK, Fulcher YG, Jurkevich A, Prior analysis of protein–RNA complexes in solution
SH, Xu J, Lenoir M, Overduin M, Van Doren using NMR paramagnetic relaxation enhance-
SR (2014) Ambidextrous binding of cell and ments. In: Sarah AW, Frédéric HTA (eds)
membrane bilayers by soluble matrix metallo- Methods in Enzymology. Academic, New York,
proteinase-12. Nat Commun 5:5552 NY, pp 333–362
14. Prior SH, Fulcher YG, Koppisetti RK, Jurkevich 24. Prior, S. H., Byrne, T. S., Tokmina-Roszyk, D.,
A, Van Doren SR (2015) Charge-triggered Fields, G. B., and Van Doren, S. R. (2016)
membrane insertion of matrix metalloprotein- Path to collagenolysis: collagen V triple-helix
ase-7, supporter of innate immunity and model bound productively and in encounters
tumors. Structure 23:2099–2110 by matrix metalloproteinase-12. J Biol Chem
15. Kutateladze TG, Capelluto DG, Ferguson CG, 291:7888
Cheever ML, Kutateladze AG, Prestwich GD, 25. Iwahara J, Clore GM (2006) Detecting transient
Overduin M (2004) Multivalent mechanism of intermediates in macromolecular binding by
membrane insertion by the FYVE domain. paramagnetic NMR. Nature 440:1227–1230
J Biol Chem 279:3050–3057 26. Fawzi NL, Doucleff M, Suh JY, Clore GM
16. Hilty C, Wider G, Fernandez C, Wuthrich K (2010) Mechanistic details of a protein-protein
(2004) Membrane protein-lipid interactions in association pathway revealed by paramagnetic
mixed micelles studied by NMR spectroscopy relaxation enhancement titration
with the use of paramagnetic reagents. measurements. Proc Natl Acad Sci U S A
Chembiochem 5:467–473 107:1379–1384
17. Arumugam S, Van Doren SR (2003) Global 27. Schilder J, Ubbink M (2013) Formation of
orientation of bound MMP-3 and N-TIMP-1 in transient protein complexes. Curr Opin Struct
solution via residual dipolar couplings. Biol 23:911–918
Biochemistry 42:7950–7958 28. Bashir Q, Volkov AN, Ullmann GM, Ubbink
18. Arumugam S, Hemme CL, Yoshida N, Suzuki M (2010) Visualization of the encounter
K, Nagase H, Berjanskii M, Wu B, Van Doren ensemble of the transient electron transfer
SR (1998) TIMP-1 contact sites and perturba- complex of cytochrome c and cytochrome c
tions of stromelysin 1 mapped by NMR and a peroxidase. J Am Chem Soc 132:241–247
paramagnetic surface probe. Biochemistry 29. Dancea F, Kami K, Overduin M (2008) Lipid
37:9650–9657 interaction networks of peripheral membrane
19. Takeda M, Terasawa H, Sakakura M, proteins revealed by data-driven micelle dock-
Yamaguchi Y, Kajiwara M, Kawashima H, ing. Biophys J 94:515–524
Miyasaka M, Shimada I (2003) Hyaluronan 30. Glover KJ, Whiles JA, Wu G, Yu N, Deems R,
recognition mode of CD44 revealed by cross- Struppe JO, Stark RE, Komives EA, Vold RR
saturation and chemical shift perturbation (2001) Structural evaluation of phospholipid
experiments. J Biol Chem 278:43550–43555 bicelles for solution-state studies of membrane-
20. Garimella R, Liu X, Qiao W, Liang X, associated biomolecules. Biophys
Zuiderweg ER, Riley MI, Van Doren SR J 81:2163–2171
84 Tara C. Marcink et al.
31. Morrison EA, DeKoster GT, Dutta S, PPM web server: resources for positioning of
Vafabakhsh R, Clarkson MW, Bahl A, Kern D, proteins in membranes. Nucleic Acids Res
Ha T, Henzler-Wildman KA (2012) 40:D370–D376
Antiparallel EmrE exports drugs by exchang- 45. Lomize MA, Lomize AL, Pogozheva ID,
ing between asymmetric structures. Nature Mosberg HI (2006) OPM: orientations of pro-
481:45–50 teins in membranes database. Bioinformatics
32. Liu Y, Kahn RA, Prestegard JH (2010) 22:623–625
Dynamic structure of membrane-anchored 46. Lomize AL, Pogozheva ID, Lomize MA,
Arf*GTP. Nat Struct Mol Biol 17:876–881 Mosberg HI (2007) The role of hydrophobic
33. Lu Z, Van Horn WD, Chen J, Mathew S, Zent interactions in positioning of peripheral pro-
R, Sanders CR (2012) Bicelles at low concen- teins in membranes. BMC Struct Biol 7:44
trations. Mol Pharm 9:752–761 47. Lomize AL, Pogozheva ID, Lomize MA,
34. Song Y, Mittendorf KF, Lu Z, Sanders CR Mosberg HI (2006) Positioning of proteins in
(2014) Impact of bilayer lipid composition on membranes: a computational approach. Protein
the structure and topology of the transmem- Sci 15:1318–1333
brane amyloid precursor C99 protein. J Am 48. Kufareva, I., and Overduin, M. (2015) http://
Chem Soc 136:4093–4096 w w w. s l i d e s h a r e . n e t / o v e r d u i n /
35. Schuler MA, Denisov IG, Sligar SG (2013) moda-slideshare.
Nanodiscs as a new tool to examine lipid- 49. Spurlino JC, Smallwood AM, Carlton DD,
protein interactions. Methods Mol Biol Banks TM, Vavra KJ, Johnson JS, Cook ER,
974:415–433 Falvo J, Wahl RC, Pulvino TA et al (1994)
36. Denisov IG, Sligar SG (2016) Nanodiscs for 1.56 A structure of mature truncated human
structural and functional studies of membrane fibroblast collagenase. Proteins 19:98–109
proteins. Nat Struct Mol Biol 23:481–486 50. Bertini I, Fragai M, Luchinat C, Melikian M,
37. Raschle T, Hiller S, Yu TY, Rice AJ, Walz T, Mylonas E, Sarti N, Svergun DI (2009)
Wagner G (2009) Structural and functional Interdomain flexibility in full-length matrix
characterization of the integral membrane pro- metalloproteinase-1 (MMP-1). J Biol Chem
tein VDAC-1 in lipid bilayer nanodiscs. J Am 284:12821–12828
Chem Soc 131:17777–17779 51. Ye Q-Z, Johnson LJ, Baragi V (1992) Gene
38. Gluck JM, Wittlich M, Feuerstein S, Hoffmann synthesis and expression in E. coli for PUMP, a
S, Willbold D, Koenig BW (2009) Integral human matrix metalloproteinase. Biochem
membrane proteins in nanodiscs can be studied Biophys Res Commun 186:143–149
by solution NMR spectroscopy. J Am Chem 52. Fulcher YG, Sanganna Gari RR, Frey NC,
Soc 131:12060–12061 Zhang F, Linhardt RJ, King GM, Van Doren
39. Yu TY, Raschle T, Hiller S, Wagner G (2012) SR (2014) Heparinoids activate a protease,
Solution NMR spectroscopic characterization secreted by mucosa and tumors, via tethering
of human VDAC-2 in detergent micelles and supplemented by allostery. ACS Chem Biol
lipid bilayer nanodiscs. Biochim Biophys Acta 9:957–966
1818:1562–1569 53. Zheng X, Ou L, Tong X, Zhu J, Wu H (2007)
40. Hagn F, Etzkorn M, Raschle T, Wagner G Over-expression and refolding of isotopically
(2013) Optimized phospholipid bilayer nano- labeled recombinant catalytic domain of human
discs facilitate high-resolution structure deter- macrophage elastase (MMP-12) for NMR
mination of membrane proteins. J Am Chem studies. Protein Expr Purif 56:160–166
Soc 135:1919–1925 54. Bertini I, Calderone V, Fragai M, Jaiswal R,
41. Studier FW (2005) Protein production by Luchinat C, Melikian M, Mylonas E, Svergun
auto-induction in high-density shaking cul- DI (2008) Evidence of reciprocal reorientation
tures. Protein Expr Purif 41:207–234 of the catalytic and hemopexin-like domains of
42. Denisov IG, Grinkova YV, Lazarides AA, Sligar full-length MMP-12. J Am Chem Soc
SG (2004) Directed self-assembly of monodis- 130:7011–7021
perse phospholipid bilayer Nanodiscs with con- 55. Lovejoy B, Welch AR, Carr S, Luong C, Broka
trolled size. J Am Chem Soc 126:3477–3487 C, Hendricks RT, Campbell JA, Walker KA,
43. Kufareva I, Lenoir M, Dancea F, Sridhar P, Martin R, Van Wart H, Browner MF (1999)
Raush E, Bissig C, Gruenberg J, Abagyan R, Crystal structures of MMP-1 and -13 reveal
Overduin M (2014) Discovery of novel mem- the structural basis for selectivity of collagenase
brane binding structures and functions. inhibitors. Nat Struct Biol 6:217–221
Biochem Cell Biol 92:555–563 56. Zhao Y, Marcink TC, Sanganna Gari RR,
44. Lomize MA, Pogozheva ID, Joo H, Mosberg Marsh BP, King GM, Stawikowska R, Fields
HI, Lomize AL (2012) OPM database and GB, Van Doren SR (2015) Transient collagen
Mapping Peripheral Membrane Binding by Matrix Metalloproteases 85
triple helix binding to a key metalloproteinase 69. Bayburt, T. H., Dennisov, I. G., Grinkova,
in invasion and development. Structure Y. V., and Sligar, S. G. Nanodisc technology:
23:257–269 protocols for preparation of nanodiscs.
57. Udi Y, Fragai M, Grossman M, Mitternacht S, University of Illinois, Urbana-Champaign, IL.
Arad-Yellin R, Calderone V, Melikian M, http://sligarlab.life.uiuc.edu/nanodisc/pro-
Toccafondi M, Berezovsky IN, Luchinat C, tocols.html
Sagi I (2013) Unraveling hidden regulatory 70. Liu Y, Prestegard JH (2008) Direct measure-
sites in structurally homologous metalloprote- ment of dipole-dipole/CSA cross-correlated
ases. J Mol Biol 425:2330–2346 relaxation by a constant-time experiment.
58. Lee W, Cornilescu G, Dashti H, Eghbalnia J Magn Reson 193:23–31
HR, Tonelli M, Westler WM, Butcher SE, 71. Lee D, Hilty C, Wider G, Wuthrich K (2006)
Henzler-Wildman KA, Markley JL (2016) Effective rotational correlation times of pro-
Integrative NMR for biomolecular research. teins from NMR relaxation interference.
J Biomol NMR 64:307–332 J Magn Reson 178:72–76
59. Lee W, Tonelli M, Markley JL (2015)
72. Aguilar JA, Nilsson M, Bodenhausen G,
NMRFAM-SPARKY: enhanced software for Morris GA (2012) Spin echo NMR spectra
biomolecular NMR spectroscopy. without J modulation. Chem Commun
Bioinformatics 31:1325–1327 48:811–813
60. Lee W, Kim JH, Westler WM, Markley JL 73. Schulz TA, Choi MG, Raychaudhuri S, Mears
(2011) PONDEROSA, an automated JA, Ghirlando R, Hinshaw JE, Prinz WA
3D-NOESY peak picking program, enables (2009) Lipid-regulated sterol transfer between
automated protein structure determination. closely apposed membranes by oxysterol-
Bioinformatics 27:1727–1728 binding protein homologues. J Cell Biol
61.
Lee W, Stark JL, Markley JL (2014) 187:889–903
PONDEROSA-C/S: client-server based 74. Kim YE, Chen J, Chan JR, Langen R (2010)
software package for automated protein 3D Engineering a polarity-sensitive biosensor for
structure determination. J Biomol NMR time-lapse imaging of apoptotic processes and
60:73–75 degeneration. Nat Methods 7:67–73
62. Ruschak AM, Kay LE (2010) Methyl groups as 75. Song Y, Hustedt EJ, Brandon S, Sanders CR
probes of supra-molecular structure, dynamics (2013) Competition between homodimeriza-
and function. J Biomol NMR 46:75–87 tion and cholesterol binding to the C99
63. Tugarinov V, Kay LE (2003) Ile, Leu, and Val domain of the amyloid precursor protein.
methyl assignments of the 723-residue malate Biochemistry 52:5051–5064
synthase G using a new labeling strategy and 76. Löhr F, Tumulka F, Bock C, Abele R, Dötsch
novel NMR methods. J Am Chem Soc V (2015) An extended combinatorial 15N,
125:13868–13878 13
Cα, and 13C′ labeling approach to protein
64. Sprangers R, Kay LE (2007) Quantitative backbone resonance assignment. J Biomol
dynamics and binding studies of the 20S pro- NMR 62:263–279
teasome by NMR. Nature 445:618–622 77. Ikeya T, Takeda M, Yoshida H, Terauchi T, Jee
65. Sinha K, Jen-Jacobson L, Rule GS (2013) JG, Kainosho M, Guntert P (2009) Automated
Divide and conquer is always best: sensitivity of NMR structure determination of stereo-array
methyl correlation experiments. J Biomol isotope labeled ubiquitin from minimal sets of
NMR 56:331–335 spectra using the SAIL-FLYA system. J Biomol
66. Chao FA, Kim J, Xia Y, Milligan M, Rowe N, NMR 44:261–272
Veglia G (2014) FLAMEnGO 2.0: an enhanced 78. Hefke F, Bagaria A, Reckel S, Ullrich SJ,
fuzzy logic algorithm for structure-based Dotsch V, Glaubitz C, Guntert P (2011)
assignment of methyl group resonances. Optimization of amino acid type-specific 13C
J Magn Reson 245:17–23 and 15N labeling for the backbone assignment
67. Poget SF, Cahill SM, Girvin ME (2007) Isotropic of membrane proteins by solution- and solid-
bicelles stabilize the functional form of a small state NMR with the UPLABEL algorithm.
multidrug-resistance pump for NMR structural J Biomol NMR 49:75–84
studies. J Am Chem Soc 129:2432–2433 79. Opitz C, Isogai S, Grzesiek S (2015) An eco-
68. Bayburt TH, Grinkova YV, Sligar SG (2002) nomic approach to efficient isotope labeling in
Self-assembly of discoidal phospholipid bilayer insect cells using homemade 15N-, 13C- and
nanoparticles with membrane scaffold pro-
2
H-labeled yeast extracts. J Biomol NMR
teins. Nano Lett 2:853–856 62:373–385
86 Tara C. Marcink et al.
80. Sattler M, Fesik SW (1996) Use of deuterium NMR experiments to a few minutes. J Am
labeling in NMR: overcoming a sizeable prob- Chem Soc 128:9042–9043
lem. Structure 4:1245–1249 85. Velyvis A, Ruschak AM, Kay LE (2012) An
81. Tugarinov V, Muhandiram R, Ayed A, Kay LE economical method for production of (2)H,
(2002) Four-dimensional NMR spectroscopy (13)CH3-threonine for solution NMR stud-
of a 723-residue protein: chemical shift assign- ies of large protein complexes: application to
ments and secondary structure of malate syn- the 670 kDa proteasome. PLoS One
thase g. J Am Chem Soc 124:10025–10035 7:e43725
82. Revington M, Zuiderweg ER (2004) TROSY- 86. Gennis RB (1989) Biomembranes: molecular
driven NMR backbone assignments of the structure and function. Springer, New York,
381-residue nucleotide-binding domain of the NY
Thermus Thermophilus DnaK molecular chap- 87. Palmier MO, Van Doren SR (2007) Rapid
erone. J Biomol NMR 30:113–114 determination of enzyme kinetics from fluores-
83. Lescop E, Schanda P, Brutscher B (2007) A set cence: overcoming the inner filter effect. Anal
of BEST triple-resonance experiments for Biochem 371:43–51
time-optimized protein resonance assignment. 88. Li L, Shi X, Guo X, Li H, Xu C (2014) Ionic
J Magn Reson 187:163–169 protein-lipid interaction at the plasma mem-
84. Schanda P, Van Melckebeke H, Brutscher B brane: what can the charge do? Trends Biochem
(2006) Speeding up three-dimensional protein Sci 39:130–140
Chapter 6
Abstract
Small angle X-ray scattering (SAXS) provides information about the conformation and flexibility of pro-
teins in solution, and hence provides complementary structural information to that obtained from X-ray
crystallography and nuclear magnetic resonance spectroscopy. In this chapter, we describe the methods for
the preparation of matrix metalloproteinase (MMP) samples for SAXS analyses, and for the acquisition,
processing and interpretation of the SAXS data.
1 Introduction
Charles A. Galea (ed.), Matrix Metalloproteases: Methods and Protocols, Methods in Molecular Biology, vol. 1579,
DOI 10.1007/978-1-4939-6863-3_6, © Springer Science+Business Media LLC 2017
87
88 Louise E. Butt et al.
AT-
C linker-
HPX arrangement is supplemented by additional
domains for substrate binding (e.g., the fibronectin type 2 domains
in MMP-2 and -9), or cell surface localization, as seen in the
membrane-type (MT) MMPs. The linker connecting the CAT and
HPX domains varies in length from 14 residues in the prototypic
collagenase MMP-1, to 68 residues in the gelatinase MMP-9. In
their zymogenic proMMP forms, a small pro-peptide (PRO)
domain docks into the active site of the CAT domain sterically
blocking substrate access, and chemically blocking hydrolytic activ-
ity by chelation of the reactive Zn2+ ion with a Cys residue (the
so-called “cysteine switch”).
1.3 Small Angle In MX, diffraction of X-rays from well-ordered atoms in oriented
X-Ray Scattering molecules within the crystal lattice gives rise to discrete reflections
(SAXS) of varying intensity on the two-dimensional (2D) detector; trans-
formation of this diffraction pattern gives rise to the electron den-
sity map, which can precisely locate atomic positions within the
protein. However, in small-angle X-ray scattering (SAXS), the ran-
domized orientation of the isotropically tumbling molecules in the
sample gives rise to a circularly symmetric pattern of elastically scat-
tered X-rays [21, 22]. As with MX, a 2D detector is utilized, but
the rotational symmetry means the SAXS data is, in effect, a one-
dimensional profile, varying in intensity with angular deviation
(2θ) out from the direction of the incident collimated X-ray beam
(of wavelength λ). The resultant scattering profile, which is
described as intensity I(q) versus q, where q = (4π sin θ)/λ (see
Note 1), can be interpreted in terms of the protein conformation
but the rotational averaging in solution means that both the infor-
mation content and resolution (typically about 10 Å) are much
lower than that obtained from MX [21, 22]. Nevertheless, SAXS
provides an effective way not only of validating those protein struc-
tures solved by MX, but also of characterizing the mixed popula-
tions of the varying conformations that exist in solution.
1.5 Interpretation Scattering at the lower end of the q range gives information about
of the Scattering the size of the protein in solution, that is the apparent molecular
Profile weight (MW) and the approximate radius of gyration (Rg) [21–
24]. The MW reveals the oligomeric state of the sample, and the
Rg provides a quantitative measure of the protein’s compactness.
These parameters can be used to reveal any monomer-dimer
transitions, for example as exhibited by the HPX domain of MMP-
14 in solution [27]. Scattering at higher q values gives information
about the molecular shape, albeit at low resolution compared to
MX. A Kratky plot (q2I(q) vs. q), a mathematical manipulation of
the scattering profile, can distinguish between folded, unfolded
and flexible proteins.
An indirect Fourier transform of the reciprocal-space scatter-
ing profile (I(q) vs. q) over a finite q range yields the real-space
pairwise distance function (P(r) vs. r), which is in effect a histo-
gram of the possible interatomic distances (r) in the protein
92 Louise E. Butt et al.
1.6 Disclaimer This chapter describes generalized methods for assessing the confor-
mational state of (pro)MMPs in solution using SAXS. The strategies
employed are adapted from those published previously [9, 16, 18,
20, 27]. However, they remain “first-shot” approaches, and may
require some optimization (e.g., sample buffer conditions) in order
to obtain the highest quality data. As with other structural methods,
the quality of results will depend on the MMP (or proMMP) under
study, and the purity of the sample. In recent years, the amount of
available SAXS experiment time has grown with the expansion of
dedicated synchrotron beamlines, and data acquisition is becoming
more automated and routine. However, we strongly advise anyone
interested in performing SAXS analyses to consult their local beam-
line scientist and identify any potential pitfalls prior to planning
experiments and production of their MMP samples.
2 Materials
3 Methods
3.1 Procedures To date, all SAXS analyses on MMPs have been performed on
for Preparation recombinant proteins, expressed either from E. coli (proMMP-1
of Protein Samples [9], MMP-1 [9, 18, 20], MMP-12 [16] and the HPX domain of
and Buffers MMP-14 [27]) or Sf9 insect cells (for proMMP-9 [30]).
Recombinant expression more readily provides the substantial
94 Louise E. Butt et al.
3.2 Procedure 1. Dialyse the sample overnight at 4 °C against the SAXS buffer.
for Data Acquisition The precise buffer used will depend on the (pro)MMP under
Using the BioSAXS study. Do not discard the dialysate (see Note 10).
Robot 2. Clarify the dialysate using a 0.2 μm filter and keep as the SAXS
reference buffer.
SAXS Studies of Matrix Metalloproteases 95
3.3 Procedure In SEC-SAXS, as the sample (or BSA standard) passes through the
for Data Acquisition SEC column it becomes more dilute resulting in a lower scattering
Using SEC-SAXS signal. Hence, the initial concentration required is slightly higher
than that for the BioSAXS™ robot.
1. Measure the protein concentration of the dialysed sample by
spectrophotometry using the Nanodrop 2000. If the concen-
tration is below 7 mg/mL (or the solubility limit, if that is
lower), concentrate the sample by ultrafiltration as in
Subheading 3.1, step 1 above.
2. Prior to the first analysis, pass at least five column volumes of
SAXS buffer through the SEC column to ensure that it is thor-
oughly equilibrated (see Note 9).
3. Program the HPLC to inject 40-50 μL of (pro)MMP sample
onto the SEC column and into the X-ray beam once the shut-
ter is open. Start monitoring the A280 with the sample running
onto the column at a flowrate of 0.16 mL/min, and begin
acquiring tandem 3-s exposures (frames) of raw scattering
data.
96 Louise E. Butt et al.
3.4 Procedures Parameters for data normalization and reduction will be beamline
for Processing dependent, and some (e.g., beam center position) may need calibrat-
of SAXS Data ing. Data reduction and normalization is usually performed at the
synchrotron beamline using bespoke software (i.e., neither ScÅtter
3.4.1 Data Normalization nor the ATSAS package); in many cases it will be an automated step
and Reduction with no user input necessary. If unsure, consult your local beamline
scientist for assistance and advice with the following steps.
3.4.2 Buffer Subtraction Data reduction provides us with a one-dimensional profile of the
and Data Quality scattering from both protein and buffer [21–24]. For both the
(pro)MMP sample and the BSA standard, we obtain the protein’s
scattering profile by subtracting the contribution from the buffer
(Fig. 1). Buffer subtraction and inspection of the data quality can
be performed in either PRIMUS [34] or ScÅtter.
Fig. 1 Buffer subtraction and Guinier analysis. SAXS data were acquired on proMMP-1 at DLS beamline B21.
(a) Overlay of buffer-only (grey) and proMMP-1 (black) scattering profiles; (b) Buffer-subtracted scattering
profile for proMMP-1; (c) Guinier analysis of proMMP-1 (above) with residuals (below), showing the reciprocal-
space approximation to Rg and extrapolation of I(0). Graphs were produced using ScÅtter
98 Louise E. Butt et al.
3.4.3 Determination In the absence of aggregation, the forward scattering, I(0), is one of
of Forward Scattering, I(0), the most accurate parameters that can be derived from SAXS data
and Reciprocal-Space Rg [21–24]. However, I(0) is not measured directly (the beamstop pre-
by Guinier Analysis vents us from doing so), but instead must be obtained by backward
extrapolation of the scattering profile to zero q. Guinier analysis (i.e.,
inspecting ln(I(q)) vs. q2 at low q2) is a convenient way of obtaining
I(0). The same process yields the reciprocal-space Rg, an excellent
approximation of the real-space Rg (see Subheading 3.4.5). For globu-
lar proteins, Guinier approximation of Rg is only robust when utiliz-
ing the range of q-values where q.Rg ≤ 1.3 [21–24]. Interactive
Guinier analysis can be performed in either PRIMUS [34] or ScÅtter.
3.4.4 Determination The forward scattering, I(0), has been shown to scale linearly with
of Apparent MW MW if one assumes a constant protein density [21–24]. It may be
obtained from the Guinier region (see Subheading 3.4.3) or the
P(r) distribution (see Subheading 3.4.5). If using the ATSAS suite,
use PRIMUS [34] for Guinier analysis, and GNOM [35] for P(r)
generation. ScÅtter can perform both of these functions.
3.4.5 Generating the P(r) The real-space pairwise distance function, P(r), is generated by
Distribution indirect Fourier transformation of the reciprocal-space scattering
profile over a finite q range [21–24]. From the P(r), we can deduce
the Dmax (the r where P(r) returns to zero) and a more accurate
(real-space) measurement of the Rg than that obtained from
Guinier approximation (see Subheading 3.4.3). In addition, the
P(r) profile gives an indication of the global shape of the protein in
solution, and can be used for volumetric modeling (see Subheading
3.4.8). Dmax is actually a “soft” parameter that must be iteratively
optimized by the user in the original GNOM [35] software, or by
a recursive algorithm in the more recent AUTOGNOM program
[28], and in ScÅtter. The following instructions apply to GNOM
and ScÅtter:
1. Enter the required Protein Data Bank (PDB) file as the initial
input file. CRYSOL will read the atomic coordinates and evalu-
ate scattering profiles for all models and chains present, unless
otherwise specified.
2. If comparison with experimental data is required, specify the
buffer-subtracted experimental scattering data (see Subheading
3.4.2), and select the appropriate angular units (Å−1 or nm−1)
for this data.
3. After execution, multiple output files are generated (see Note
19). The file with extension “.log” contains the experimental
and theoretical Rg values. The fit to the experimental scatter-
ing data and the χ2 value are located in the file with extension
“.fit”.
4. Inspect the χ2 values; low values indicate good agreement
between the known atomic structure and the experimental
SAXS data. However, for highly flexible proteins, one expects
variations throughout the sampled q range. Examine the data
for signs of any deviation; upward deviation of the experimen-
tal data at low q range (< 0.2 Å−1) is suggestive of molecular
elongation, whilst deviation within the medium q range (> 0.2
Å−1) may indicate a difference in relative domain orientation/
location between the two structures.
Fig. 2 Optimizing Dmax in the P(r) distribution. SAXS data were acquired on proMMP-1ΔHPX at DLS beamline B21.
Pairwise distance distributions are shown for (a) Dmax = 60 Å; (b) Dmax = 80 Å, the correct value; and (c) Dmax =
100 Å. In (a), the Dmax is too low, giving rise to a truncated P(r) (vertical arrow). In (c), the Dmax is too high, making
the P(r) descend below the x-axis (vertical arrow). Data displayed using GNOM
SAXS Studies of Matrix Metalloproteases 101
4 Notes
Fig. 3 Ensemble optimization modeling of proMMP-1. (a) Preparing the crystal structure for an EOM calcula-
tion. The PDB file was divided into four separate ordered “domains”: the first helix of the PRO domain, the
remainder of the structured PRO domain, the CAT domain and the HPX domain. All except the HPX domain are
fixed in space during the EOM calculation. The program generates Cα atomic positions for the unstructured
regions that connect these, i.e., the N-terminal tail, “bait” region, and PRO-CAT linker (white spheres in the
diagram). Although precisely positioned in the crystal structure, the CAT-HPX linker (black spheres) was
allowed to flex during the EOM calculation to accommodate varying positions/orientations of the HPX domain;
(b) Results of the EOM calculation showing the Rg distribution for the starting pool of proMMP-1 conformers
(dashed line) and the ensemble of selected conformers (solid line). Unlike the mature enzyme, proMMP-1
remains compact in solution [9]; (c) Superposition of the theoretical scattering from the ensemble of selected
conformers (black line) over the experimental data (grey crosses)
Acknowledgments
References
HD, Konarev PV, Svergun DI (2012) New 36. Svergun D, Barberato C, Koch MHJ (1995)
developments in the program package for CRYSOL – a program to evaluate x-ray solu-
small-angle scattering data analysis. J Appl tion scattering of biological macromolecules
Cryst 45(Pt 2):342–350 from atomic coordinates. J Appl Cryst
30. Rosenblum G, Van den Steen PE, Cohen SR, 28:768–773
Grossmann JG, Frenkel J, Sertchook R, Slack 37. Franke D, Svergun DI (2009) DAMMIF, a
N, Strange RW, Opdenakker G, Sagi I (2007) program for rapid ab-initio shape determina-
Insights into the structure and domain flexibil- tion in small-angle scattering. J Appl Cryst
ity of full-length pro-matrix metalloproteinase- 42(2):342–346
9/gelatinase B. Structure 15(10):1227–1236 38. Volkov VV, Svergun DI (2003) Uniqueness of
31. Nie J, Pei D (2010) Expression and purifica- ab initio shape determination in small-angle scat-
tion of membrane-type MMPs. Methods Mol tering. J Appl Crystallogr 36(3 Part 1):860–864
Biol 622:99–110 39. Kozin MB, Svergun DI (2001) Automated
32. Rodgers UR, Clark IM (2010) Expression of matching of high- and low-resolution struc-
recombinant MMP-28 in mammalian cells. tural models. J Appl Cryst 34(1):33–41
Methods Mol Biol 622:55–65 40. Jeffries CM, Graewert MA, Svergun DI,
33. Windsor LJ, Steele DL (2010) Expression of Blanchet CE (2015) Limiting radiation dam-
recombinant matrix metalloproteinases in age for high-brilliance biological solution scat-
Escherichia coli. Methods Mol Biol 622:67–81 tering: practical experience at the EMBL P12
34. Konarev PV, Volkov VV, Sokolova AV, Koch beamline PETRAIII. J Synchrotron Radiat
MHJ, Svergun DI (2003) PRIMUS: a Windows 22(Pt 2):273–279
PC-based system for small-angle scattering data 41. Biasini M, Bienert S, Waterhouse A, Arnold K,
analysis. J Appl Cryst 36:1277–1282 Studer G, Schmidt T, Kiefer F, Cassarino TG,
35. Svergun DI (1992) Determination of the regu- Bertoni M, Bordoli L, Schwede T (2014) SWISS-
larization parameter in indirect-transform MODEL: modelling protein tertiary and quater-
methods using perceptual criteria. J Appl Cryst nary structure using evolutionary information.
25:495–503 Nucleic Acids Res 42(W1):W252–W258
Part III
Abstract
Matrix metalloproteases are multidomain enzymes with a remarkable proteolytic activity located in the
extracellular environment. Their catalytic activity and structural properties have been intensively studied
during the last few decades using both experimental and theoretical approaches, but many open questions
still remain. Extensive molecular dynamics simulations enable the sampling of the configurational space of
a molecular system, thus contributing to the characterization of the structure, dynamics, and ligand bind-
ing properties of a particular MMP. Based on previous computational experience, we provide in this chap-
ter technical and methodological guidelines that may be useful to and stimulate other researchers to
perform molecular dynamics simulations to help address unresolved questions concerning the molecular
mode of action of MMPs.
Key words Free energy calculations, Ligand binding, Molecular dynamics, Molecular modeling,
Multidomain enzymes, Peptide hydrolysis
1 Introduction
Charles A. Galea (ed.), Matrix Metalloproteases: Methods and Protocols, Methods in Molecular Biology, vol. 1579,
DOI 10.1007/978-1-4939-6863-3_7, © Springer Science+Business Media LLC 2017
111
112 Natalia Díaz and Dimas Suárez
2 Materials
2.1 Initial Structure To start an MD simulation, initial coordinates for all the atoms in
the desired MMP should be provided to the minimization/simula-
tion packages.
1. Check the Protein Data Bank for available structures and select
a suitable X-ray crystal structure with a high resolution.
Currently, structural information is only available for 12 of the
23 human MMPs: MMP-1 (catalytic domain and full-length
enzyme), MMP-2 (catalytic domain, fibronectin modules,
hemopexin domain, and full-length enzyme), MMP-3 (cata-
lytic domain), MMP-7 (catalytic domain), MMP-8 (catalytic
domain), MMP-9 (catalytic-fibronectin domains, hemopexin
domain, and a model for the full-length enzyme), MMP-10
(catalytic domain), MMP-12 (catalytic domain, hemopexin
domain, and full-length enzyme), MMP-13 (catalytic domain,
hemopexin domain, and full-length enzyme), MMP-14 (cata-
lytic and hemopexin domains), MMP-16 (catalytic domain),
and MMP-20 (catalytic domain). For the remaining MMPs,
homology modeling may provide a convenient initial structure
provided the homology models are carefully validated (see
Note 1).
2. Edit the initial structure by removing additional molecules,
reverse mutations (e.g., the catalytic glutamic acid), and select-
ing appropriate protonation states for the titratable amino-
acids (see Note 2). Take into account that the catalytic Zn1 is
coordinated by three histidines through their side chain Nε
atom, but Zn2 is bound to two Nε and one Nδ atoms of three
different histidines.
3. Refine the structure by adding missing atoms/residues and
optimize the conformational state of some side chains (i.e.,
His, Asn, Gln) (see Note 3).
4. The use of explicit solvent in extended molecular simulations
of MMPs is highly recommended to realistically account for
both entropic and enthalpic solvent effects, which can influ-
ence inter and intra-domain conformational changes as well as
ligand binding. Therefore, the MMP molecule should be
placed in an appropriate solvent box that extends at least 15 Å
from the protein atoms. Neutralize the system by randomly
adding Na+/Cl− counterions.
2.2 Software Fortunately, a variety of programs are readily available for prepar-
ing and running MD simulations of biomolecules assuming an all
atom representation. Our preferred choice for studying MMPs is
the AMBER (Assisted Model Building with Energy Refinement)
package, which integrates a collection of programs to set up, per-
form, and analyze MD simulations [32]. AMBER can be used with
several types of molecular mechanics (MM) methods or force fields
Molecular Dynamics Studies of Matrix Metalloporteases 115
3 Methods
3.1 MM The MMP catalytic domain binds two zinc ions (Zn1 catalytic and
Parametrization Zn2 structural) and a number of calcium ions that are structurally
of the Coordination and functionally relevant. To describe these metal ions in classical
Environment of Metal force fields for extended MD simulations, we assign these metal
Ions ions to either a nonbonded or a bonded representation.
Molecular Dynamics Studies of Matrix Metalloporteases 117
1. In the nonbonded form, only the total charge (+2) and van der
Waals parameters available in the literature are required to
describe these ions. We suggest using the nonbonding param-
eters developed by Li et al. that reproduce experimental ion-
oxygen distance values and coordination number for the first
solvation shell [33]. This works well for calcium ions but it is
not recommended for describing the coordination environ-
ment surrounding the zinc ions.
2. Different coordination spheres have been observed for zinc
ions in MMPs (e.g., tetrahedral for the structural and the pro-
peptide bound catalytic zincs, distorted trigonal bipyramid for
the hydroxamate inhibited catalytic zinc, etc.) which, in prin-
ciple, can be well represented in the bonded metal-ligand con-
figuration (see Note 4).
3. If there is incomplete experimental data to define the proton-
ation state of the zinc-binding groups (ZBGs) and the essential
Glu residue, and/or the number and position of water mole-
cules, preliminary QM/MM studies should be carried out to
determine the most likely configuration of the zinc sites [34].
For example, by initially assigning a nonbonded conformation
for the zinc ion in AMBER, it is relatively simple to define a
QM region comprising the catalytic Zn1 ion, closest residues,
and ligand/water molecules for each configuration. The QM
region is described by either one of the semiempirical QM
methods implemented in sander or a more sophisticated
method using an external QM code (Gaussian, ORCA,
GAMESS; see Note 5). Subsequent energy minimization of
the QM region followed by structural comparisons with the
experimental structures and/or energy comparison allow for
discrimination between various configurations.
4. Equilibrium values and force constants for all zinc-ligand
bonds and angles should be provided in the bound configura-
tion. In order to do this we usually carry out QM calculations
on realistic cluster models of the zinc coordination spheres
(Fig. 1). For example, the selected cluster model for the struc-
tural Zn2 should comprise the side chains of the coordinating
ligands (three histidines and an aspartic acid). On the other
hand, for the active site Zn1, it is advisable to include the cata-
lytic glutamic acid located in the second coordination shell and
other relevant ligands (e.g., water molecules, inhibitor ZBGs,
etc.). Bond and angle reference values involving zinc can be
directly taken from the optimized cluster models. To compute
force constants, perturbed structures should be generated by
selectively modifying all the bond distances and angles involv-
ing the zinc ion and fitting the relative energies to a second-
order polynomial [35]. The torsion parameters associated with
the zinc-ligand interactions can be set to zero.
118 Natalia Díaz and Dimas Suárez
Fig. 1 Ball-and-stick representation of a cluster model used to parametrize the Zn1 ion following the bonded
approach. Selected atomic charges and MM parameters (r: reference bond distance, θ: reference bond angle,
Kr: bond force constant, Kθ: angle force constant for Zn1–L bonds and the corresponding angles) as obtained
from quantum mechanical (B3LYP/6-31G*) calculations are included. An example of curve fitting to obtain a
force constant is also shown for the His2-Nε⋯Zn1 bond (y: relative energy of the perturbed structure with
respect to the unperturbed one; x: distance perturbation)
3.2 Simulation of To date, most MD simulations of MMPs have focused on the cata-
the Catalytic Domain lytic domain, mainly examining the binding of active site inhibi-
tors. We believe that future studies of the catalytic domain should
carefully address the following points:
1. The N-terminal coil in most MMP catalytic domain crystal
structures is either longer or shorter than the biologically rel-
evant form because its length depends on the protocol used for
the activation of the pro-MMP. This disparity prevents the
binding of the N-terminal portion to the surface of the cata-
lytic domain via the formation of a salt bridge with a conserved
aspartic acid residue (Fig. 2) and leads to a reduction in the
catalytic activity of the enzyme. Thus, for MD simulations of
MMPs it is important to select a structure with the correct
N-terminus, usually a tyrosine or a phenylalanine, and a prop-
erly positioned N-terminal coil. If this physiologically relevant
(superactivated) structure is not available for a particular MMP,
it can be easily generated by superposition with structures for
other MMPs.
2. The number of bound calcium ions fluctuates between one
and three, depending on the crystal structure chosen (Fig. 3).
These calcium ions interact with several loops and turns and
contribute toward generating a catalytically competent enzyme.
For MMP-2, previous MD simulations followed by approxi-
mate free energy calculations have found that the binding of
Fig. 2 Ribbon representation of the MMP-3 catalytic domain generated for different crystal structures with PDB
codes 1SLM (a), 1CAQ (b), and 1HFS (c) illustrating the different positions of the N-terminal coil (in orange).
The side chains of the conserved aspartic acid (Asp237) and N-terminal residues are shown as sticks. Zinc and
calcium ions are depicted as spheres (magenta and green, respectively) and the active site histidines as sticks
120 Natalia Díaz and Dimas Suárez
3.3 Binding of Small Numerous MD simulations have previously analyzed the binding
Ligands to the Active of small peptide substrates and inhibitors within the active site of
Site various MMPs. Moreover, MD simulations of the Michaelis com-
plex formed between the MMP catalytic domain and a substrate of
interest must be considered a prerequisite prior to attempting to
study the mechanism of catalysis (see Note 7).
1. The initial MMP/peptide complex can be built taking into
account previously published data showing that small peptide
substrates extend within the active site and make contact via a
number of polar and hydrophobic interactions with the S3–S3′
binding sites. Several crystal structures are available that can
act as a template for initially binding a short peptide around
the catalytic zinc ion (Fig. 4). For this purpose, use a program
Fig. 3 Ribbon representation of the MMP-13 catalytic domain generated using different crystal structures with
PDB codes 1YOU (a), 1XUC (b), and 1ZTQ (c). Zinc and calcium ions are depicted as spheres (magenta and
green, respectively) and the side chains of peptide-binding residues as sticks
Molecular Dynamics Studies of Matrix Metalloporteases 121
Fig. 4 Ribbon representation of the catalytic domains of MMP-12 (a, b) and MMP-2 (c) generated from crystal
structures with PDB codes 2OXW, 2OXZ, and 1CK7, respectively. Ligands bound within the active site are illus-
trated as sticks with carbon atoms colored green. The ligand sequence and position within the active site is
also indicated below each structure. Zinc and calcium ions are depicted as spheres (magenta and green,
respectively) and the side chains of the Zn1 binding protein residues as sticks
122 Natalia Díaz and Dimas Suárez
3.4 Binding Large and structurally complex biomolecules like those based on
of Collagen-like the collagen triple helix are among the natural substrates of the
Substrates MMPs. For collagen-like substrates the complexity of both the
substrate and the receptor (enzyme) increases the level of difficulty
in the preparation and analysis of molecular simulations involving
MMP complexes. Although such studies are particularly challeng-
ing, they are potentially more interesting. Therefore, in this sub-
section, we give some broad guidelines that will hopefully
complement procedures available in the literature.
1. Initial coordinates for triple helical peptides (THPs) contain-
ing standard amino-acid residues can be obtained from the
amino acid sequence using an interactive triple helical collagen
building script termed THe BuScr. (http://structbio.biochem.
dal.ca/jrainey/THeBuScr.html). Nevertheless, an initial MD
simulation of the solvated helix is required to properly relax
the system, especially if nonstandard amino-acid residues need
to be included in the THP model.
2. The triple helix must locally unfold to accommodate one of its
chains within the narrow active site of a MMP. Targeted MD
simulations can be used to locally force one of the three chains
into a more extended configuration and relax the whole triple
helix. This transition can be smoothly driven along the tar-
geted MD simulation by defining a harmonic penalty based on
the root mean squared deviation of the backbone residues
located around the scissile peptide bond. The reference struc-
ture used for the constraint can be taken from a previous MD
simulation of the complex formed between the MMP and a
small linear peptide substrate with the appropriate sequence.
3. We recommend implementing a semiflexible docking protocol
combining various software tools (e.g., cpptraj, sander,
Mmpbsa.py) using linux shell scripts to dock the distorted triple
Molecular Dynamics Studies of Matrix Metalloporteases 123
Fig. 5 Schematic representation of the semiflexible protocol proposed for docking a THP molecule within the
active site of an MMP. Initial coordinates for the MMP/THP complexes (11, 12, …,1N ,…, MN) come from the
superposition of M snapshots obtained from the MD simulation of the distorted THP onto N snapshots extracted
from the trajectory computed for the MMP bound to a short linear peptide. After relaxing the steric clashes and
scoring the complexes with MMPBSA energies, the most stable MMP/THP complexes are structurally evalu-
ated using the whatcheck software tool (http://swift.cmbi.ru.nl/gv/whatcheck/) prior to run extended MD
simulations
124 Natalia Díaz and Dimas Suárez
3.6 Running the MD To properly run an MD simulation for MMP systems in explicit
Simulations solvent, the following steps should be sequentially performed to
relax and thermalize the system:
1. Solvent boxes around macromolecules are usually generated
by replicating the coordinates of a small and pre-equilibrated
box in three directions. As a result, the large solvent box sur-
rounding an MMP molecule contains vacuum bubbles at the
corners of the interfaces formed by the smaller boxes. Thus,
before starting the simulation for the entire system we first
relax the large solvent box. This can be done by performing an
initial energy minimization, a short MD simulation at constant
pressure, and a final minimization of the solvent molecules
(counterions can be also included in these steps). In this way,
the solvent molecules are allowed to adapt to the solute (i.e.,
the protein) and the volume of the box changes to the correct
density.
2. The whole system should be minimized to remove potentially
bad contacts in the initial MMP structure. If needed, this can
be done in several steps relaxing first those parts of the system
with more severe steric clashes.
3. The system is then gradually heated from 0 K to the desired
simulation temperature, usually 300 K. To avoid any problem,
heating is best done by running a number of short MD simula-
tions to progressively increase the target temperature of the
system. We usually employ six constant volume simulations of
10 ps each to increase the temperature according to the fol-
lowing sequence 0 K → 50 K → 100 K → 150 K → 200 K →
250 K → 300 K.
4. After thermalization, the actual MD simulation can be started
by choosing either constant volume (NVT) or constant pres-
sure (NPT) conditions. If using NVT conditions, we recom-
mend running a preliminary pressurization job under NPT
conditions (1–2 ns).
5. During the MD simulation of the MMP, it is necessary to dis-
criminate between equilibration and production phases. In
general simulations begin in a state (edited X-ray structure,
homology model, docking model, etc.) that can be structurally
different from the one that you wish to sample and, therefore,
the first part of the MD trajectory is rather sensitive to the ini-
tial conditions and to singular transitions occurring during the
structural relaxation of the system. Thus, during the equilibra-
tion phase, which can extend for tens or even hundreds of ns in
the case of full-length MMPs, the structure and energy of the
system normally evolve to reach a relatively stable plateau.
Sampling during the production phase of the simulation occurs
along this plateau where the system is in equilibrium (Fig. 6).
126 Natalia Díaz and Dimas Suárez
Fig. 6 Time evolution of (a) RMSD and (b) MM-PB energy for the MD simulation of full-length MMP-2 (PDB ID:
1CK7). The vertical blue line separates the equilibration and production phases. The red and green curves
represent the evolution of the accumulated average and the adjacent average, respectively
side chain groups located within the active site cleft. The radius
of accessibility [21] is defined as the maximum radius of a
spherical ligand that can touch the desired target and its deter-
mination is based on the rapid calculation of the molecular
surface using programs such as MSMS (http://mgl.scripps.
edu/people/sanner/html/msms_home.html).
4. The evolution of the size and shape of cavities in the MMPs
like the S1′ pocket during the MD simulation can be moni-
tored using a program like mdpocket (http://fpocket.source-
forge.net/).
5. Interdomain and enzyme-ligand contacts include H-bonds
and hydrophobic interactions. H-bonds can be characterized
on the basis of geometrical criteria (e.g., X⋯Y distance <3.5 Å
and X–H⋯Y angle >120°; cpptraj has different commands to
search for and characterize H-bond contacts). In addition,
nonpolar interactions can be detected by measuring the dis-
tance between the centers of mass of two interacting groups.
We have also used in-house developed software (available upon
request) to evaluate the empirical dispersion attraction between
pairs of atoms belonging to two hydrophobic groups.
3.8 Energetic Relative free energies can be estimated from conventional MD sim-
Analysis of MD ulations using end-point free energy methods [38, 39] to evaluate
Trajectories the inhibitory ability of structurally dissimilar compounds toward
MMPs and/or to discriminate among different configurations or
inter-domain arrangements of full-length MMPs. In particular, the
MM-PB(GB)SA (molecular mechanics-Poisson Boltzmann or
generalized Born surface area) method offers a good balance
between speed and accuracy and has been widely used to compute
relative binding affinities [11, 40, 41]. However, there are differ-
ent variants of this method and various protocols describing its
application. We comment below on options that particularly apply
to MMPs.
1. Extract a minimum of 500 snapshots separated by 25–50 ps
from the production phase. Compute solvation energies using
the Poisson-Boltzman (PB) method (this method is especially
applicable for large systems). Both linear and nonlinear solvers
of the PB equation are implemented in the program pbsa that
is included in the AMBER package.
2. The calculation of binding energies for ligands bearing ZBGs
with a strong affinity for the catalytic Zn ion can be problem-
atic. First, a nonbonded approach for the Zn environment
should be used for the MM-PBSA calculations. However, the
purely MM representation of the Zn···ZBG interaction can
result in unbalanced interaction energies and, consequently, a
careful validation of the relative MM-PBSA energies would be
128 Natalia Díaz and Dimas Suárez
4 Supporting Information
5 Notes
References
1. Butler GS, Overall CM (2009) Updated bio- 4. Fabre B, Ramos A, de Pascual-Teresa B (2014)
logical roles for matrix metalloproteinases and Targeting matrix metalloproteinases: exploring
new “intracellular” substrates revealed by the dynamics of the S1′ pocket in the design of
degradomics. Biochemistry 48:10830–10845 selective, small molecule inhibitors. J Med
2. Fingleton B (2008) MMPs as therapeutic tar- Chem 57:10205–10219
gets—still a viable option? Semin Cell Dev Biol 5. Gioia M, Fasciglione GF, Marini S, D’Alessio
19:61–68 S, de Sanctis G, Diekmann O, Pieper M, Politi
3. Bode W, Maskos K (2003) Structural basis of V, Tschesche H, Coletta M (2002) Modulation
the matrix metalloproteinases and their physi- of the catalytic activity of neutrophil collage-
ological inhibitors, the tissue inhibitors of nase MMP-8 on bovine collagen I. J Biol
metalloproteinases. Biol Chem 384:863–872 Chem 277:23123–23130
Molecular Dynamics Studies of Matrix Metalloporteases 133
Abstract
A continuous assay method, such as the one that utilizes an increase in fluorescence upon hydrolysis, allows
for rapid and convenient kinetic evaluation of proteases. To better understand MMP behaviors toward
native substrates, a variety of fluorescence resonance energy transfer (FRET)/intramolecular fluorescence
energy transfer (IFET) triple-helical substrates have been constructed to examine the collagenolytic activ-
ity of MMP family members. Results of these studies have been valuable for providing insights into (a) the
relative triple-helical peptidase activities of the various collagenolytic MMPs, (b) the collagen preferences
of these MMPs, and (c) the relative roles of MMP domains and specific residues in efficient collagenolysis.
The present chapter provides an overview of MMP FRET triple-helical substrates and describes how to
construct and utilize these substrates.
1 Introduction
1.1 Development A continuous assay method, such as the one that utilizes an increase
of Peptide Substrates in fluorescence upon hydrolysis, allows for rapid and convenient
for Matrix kinetic evaluation of proteases, both in solution and cell surface
Metalloproteinases bound. For this reason, significant research efforts have focused on
designing substrates for matrix metalloproteinase (MMP) family
members [1]. Sequence specificity studies for MMPs have been
performed with peptides based on protein sequences surrounding
MMP cleavage sites, and the results of these studies have been
comprehensively reviewed [2, 3]. In addition, phage display and
peptide libraries have been utilized to identify MMP-selective sub-
strate sequences [4–10]. Results of the aforementioned studies
were subsequently used to design fluorogenic MMP substrates.
Charles A. Galea (ed.), Matrix Metalloproteases: Methods and Protocols, Methods in Molecular Biology, vol. 1579,
DOI 10.1007/978-1-4939-6863-3_8, © Springer Science+Business Media LLC 2017
137
138 Maciej J. Stawikowski et al.
1.2.1 Resonance Energy Förster resonance energy transfer (FRET) may occur between a
Transfer fluorescent donor group and a quenching acceptor group when
the fluorophore has a high quantum yield (ΦF) and a fluorescence
emission spectrum that is exactly overlapped by a strongly absorb-
ing acceptor (quencher) [11]. The efficiency of transfer will also
depend upon the distance between the donor and acceptor [11,
12]. Resonance energy transfer quenched fluorogenic substrates
are thus created by incorporating the donor and acceptor on oppo-
site sides of the scissile bond, at a distance that allows for highly
efficient energy transfer. When the substrate is cleaved by an
enzyme, diffusion of the donor-containing substrate fragment
away from the acceptor-containing substrate fragment results in
loss of energy transfer and subsequent fluorescence. The creation
of an optimal MMP fluorogenic substrate thus depends upon (1)
incorporation of a fluorescent donor that has a high quantum yield,
(2) incorporation of an acceptor that absorbs at the donor fluores-
cence emission wavelength (λem), and (3) the number of amino
acid residues between the donor and the acceptor.
1.2.2 Chemical Moieties There are a wide variety of donor and acceptor groups that have
Used for Donor/Acceptor been used in fluorogenic substrates [11, 12]. MMP substrates have
Pairs used one of eight different fluorophores. The first was Trp [13],
which has an ε280 value of 5600 M−1 cm−1 and ΦF of 0.2. The
quencher group was N-2,4-dinitrophenyl (Dnp) [13, 14], which
has an absorption maximum at λ = 363 nm and a prominent shoul-
der at λ = 410 nm [15]. Alternatively, a fluorogenic substrate for
the Leishmania surface metalloproteinase used a dansyl group to
quench Trp fluorescence [16]. It should be noted that dansyl
quenching of Trp decreases rapidly with intervening residue dis-
tance [11]. Knight and colleagues proposed the use of
(7-methoxycoumarin-4-yl)acetyl (Mca; ε325 = 14,500 M−1 cm−1
and ΦF = 0.49) as a fluorophore for MMP substrates [15]. Mca is
efficiently quenched by Dnp moieties [15, 17], as the shoulder in
the Dnp absorption spectrum overlaps the Mca fluorescence emis-
sion spectrum [15]. Dnp is also used as a quencher for MMP sub-
strates containing the fluorophore N-methylanthranilic acid (Nma)
[18]. 5-[(2-Aminoethyl)amino]naphthalene-1-sulfonic acid
(Edans; ε336 = 5400 M cm and ΦF = 0.13) has been used as a
−1 −1
1.2.3 Practical Aspects Trp and Tyr(NO2) can be incorporated easily by solid-phase pep-
of Donor/Acceptor Groups tide synthesis and Dnp or dansyl acylated to the N-terminus of the
peptide. Other moieties require additional synthetic steps. For
example, to use the Mca/Dnp pair, a Dnp derivative such as N-3-
(2,4-dinitrophenyl)-l-2,3-diaminopropionic acid (Dpa) [15] or
Lys(Dnp) [17] must be synthesized (see below) and incorporated
into a peptide by solid-phase methods, while Mca is acylated to the
peptide N-terminus. Alternatively, the Mca derivative l-2-amino-
3-(7-methoxy-4-coumaryl)propionic acid (Amp) can be synthe-
sized (see below) and incorporated by solid-phase methods [30].
For Dabcyl/Edans substrates, derivatives of Glu(Edans) must be
prepared (see below). Substrates containing Nma, Fam, or Cy3
can be synthesized using “on-resin” reactions to create Lys(Nma),
Lys(Fam), or Lys(Cy3) moieties (see below).
In contrast to solid-phase assembly of fluorogenic substrates,
“post-synthesis” approaches have been used for the preparation of
peptides containing LY and Ctmr [20, 21] or Cy5Q [29]. Although
they are more laborious, post-synthesis approaches do alleviate one
potential purification problem of fluorogenic peptides. Typically,
fluorophores and quenchers are hydrophobic, and thus the reverse-
phase (RP) HPLC separation of the desired peptide from deletion
peptides containing these groups can be difficult.
1.3 Collagen-Model Collagen is the most abundant protein in animals, and is the major
Triple-Helical structural protein found in the connective tissues such as basement
Substrates membranes, tendons, ligaments, cartilage, bone, and skin. The
most defining feature of collagen is the supersecondary structure,
composed of three parallel extended left-handed polyproline type
II alpha chains of primarily repeating Gly-Xxx-Yyy triplets. Three
140 Maciej J. Stawikowski et al.
Table 1
Hydrolysis of fTHPs by MMPs
Substrate
Enzyme abbreviation Substrate sequence kcat (s−1) KM (μM) kcat/kMa (s−1 M−1) Reference
(continued)
142 Maciej J. Stawikowski et al.
Table 1
(continued)
Substrate
Enzyme abbreviation Substrate sequence kcat (s−1) KM (μM) kcat/kMa (s−1 M−1) Reference
(continued)
Fluorogenic Peptide Substrates for Matrix Metalloproteases 143
Table 1
(continued)
Substrate
Enzyme abbreviation Substrate sequence kcat (s−1) KM (μM) kcat/kMa (s−1 M−1) Reference
(continued)
144 Maciej J. Stawikowski et al.
Table 1
(continued)
Substrate
Enzyme abbreviation Substrate sequence kcat (s−1) KM (μM) kcat/kMa (s−1 M−1) Reference
(continued)
Fluorogenic Peptide Substrates for Matrix Metalloproteases 145
Table 1
(continued)
Substrate
Enzyme abbreviation Substrate sequence kcat (s−1) KM (μM) kcat/kMa (s−1 M−1) Reference
(continued)
146 Maciej J. Stawikowski et al.
Table 1
(continued)
Substrate
Enzyme abbreviation Substrate sequence kcat (s−1) KM (μM) kcat/kMa (s−1 M−1) Reference
(continued)
Fluorogenic Peptide Substrates for Matrix Metalloproteases 147
Table 1
(continued)
Substrate
Enzyme abbreviation Substrate sequence kcat (s−1) KM (μM) kcat/kMa (s−1 M−1) Reference
(continued)
148 Maciej J. Stawikowski et al.
Table 1
(continued)
Substrate
Enzyme abbreviation Substrate sequence kcat (s−1) KM (μM) kcat/kMa (s−1 M−1) Reference
(continued)
Fluorogenic Peptide Substrates for Matrix Metalloproteases 149
Table 1
(continued)
Substrate
Enzyme abbreviation Substrate sequence kcat (s−1) KM (μM) kcat/kMa (s−1 M−1) Reference
(continued)
150 Maciej J. Stawikowski et al.
Table 1
(continued)
Substrate
Enzyme abbreviation Substrate sequence kcat (s−1) KM (μM) kcat/kMa (s−1 M−1) Reference
MMP-9 CAT fTHP-15 (Gly-Pro-Hyp)5-Gly-Pro- 1.05 ± 0.14 15.1 ± 69,566 ± 9138 [52, 64]
Lys(Mca)-Gly-Pro-Gln- 1.4
Gly~Leu-Arg-Gly-Gln-
Lys(Dnp)-Gly-Val-Arg-
(Gly-Pro-Hyp)5-NH2
MMP-9 CAT-FN fTHP-15 (Gly-Pro-Hyp)5-Gly-Pro- 1.75 ± 0.15 143.0 ± 12,227 ± 1077 [52, 64]
Lys(Mca)-Gly-Pro-Gln- 13.9
Gly~Leu-Arg-Gly-Gln-
Lys(Dnp)-Gly-Val-Arg-
(Gly-Pro-Hyp)5-NH2
(continued)
Fluorogenic Peptide Substrates for Matrix Metalloproteases 151
Table 1
(continued)
Substrate
Enzyme abbreviation Substrate sequence kcat (s−1) KM (μM) kcat/kMa (s−1 M−1) Reference
(continued)
152 Maciej J. Stawikowski et al.
Table 1
(continued)
Substrate
Enzyme abbreviation Substrate sequence kcat (s−1) KM (μM) kcat/kMa (s−1 M−1) Reference
(continued)
Fluorogenic Peptide Substrates for Matrix Metalloproteases 153
Table 1
(continued)
Substrate
Enzyme abbreviation Substrate sequence kcat (s−1) KM (μM) kcat/kMa (s−1 M−1) Reference
(continued)
154 Maciej J. Stawikowski et al.
Table 1
(continued)
Substrate
Enzyme abbreviation Substrate sequence kcat (s−1) KM (μM) kcat/kMa (s−1 M−1) Reference
(continued)
Fluorogenic Peptide Substrates for Matrix Metalloproteases 155
Table 1
(continued)
Substrate
Enzyme abbreviation Substrate sequence kcat (s−1) KM (μM) kcat/kMa (s−1 M−1) Reference
(continued)
156 Maciej J. Stawikowski et al.
Table 1
(continued)
Substrate
Enzyme abbreviation Substrate sequence kcat (s−1) KM (μM) kcat/kMa (s−1 M−1) Reference
(continued)
Fluorogenic Peptide Substrates for Matrix Metalloproteases 157
Table 1
(continued)
Substrate
Enzyme abbreviation Substrate sequence kcat (s−1) KM (μM) kcat/kMa (s−1 M−1) Reference
(continued)
158 Maciej J. Stawikowski et al.
Table 1
(continued)
Substrate
Enzyme abbreviation Substrate sequence kcat (s−1) KM (μM) kcat/kMa (s−1 M−1) Reference
(continued)
Fluorogenic Peptide Substrates for Matrix Metalloproteases 159
Table 1
(continued)
Substrate
Enzyme abbreviation Substrate sequence kcat (s−1) KM (μM) kcat/kMa (s−1 M−1) Reference
(continued)
160 Maciej J. Stawikowski et al.
Table 1
(continued)
Substrate
Enzyme abbreviation Substrate sequence kcat (s−1) KM (μM) kcat/kMa (s−1 M−1) Reference
(continued)
Fluorogenic Peptide Substrates for Matrix Metalloproteases 161
Table 1
(continued)
Substrate
Enzyme abbreviation Substrate sequence kcat (s−1) KM (μM) kcat/kMa (s−1 M−1) Reference
(continued)
162 Maciej J. Stawikowski et al.
Table 1
(continued)
Substrate
Enzyme abbreviation Substrate sequence kcat (s−1) KM (μM) kcat/kMa (s−1 M−1) Reference
(continued)
Fluorogenic Peptide Substrates for Matrix Metalloproteases 163
Table 1
(continued)
Substrate
Enzyme abbreviation Substrate sequence kcat (s−1) KM (μM) kcat/kMa (s−1 M−1) Reference
MMP-9 α1(V)436– (Gly-Pro-Hyp)5-Gly-Pro- 3.99 ± 0.13 77.8 ± 51,360 ± 1676 [52, 64]
447 Lys(Mca)-Gly-Pro-Pro- 5.8
fTHP Gly~Val-Val-Gly-Glu-
Lys(Dnp)-Gly-Glu-Gln-
(Gly-Pro-Hyp)5-NH2
MMP-9 CAT α1(V)436– (Gly-Pro-Hyp)5-Gly-Pro- 1.17 ± 0.21 22.5 ± 52,249 ± 9192 [52, 64]
447 Lys(Mca)-Gly-Pro-Pro- 1.7
fTHP Gly~Val-Val-Gly-Glu-
Lys(Dnp)-Gly-Glu-Gln-
(Gly-Pro-Hyp)5-NH2
(continued)
164 Maciej J. Stawikowski et al.
Table 1
(continued)
Substrate
Enzyme abbreviation Substrate sequence kcat (s−1) KM (μM) kcat/kMa (s−1 M−1) Reference
1.4 Dissecting MMP A series of fTHPs has been assembled that incorporate sequences
Collagenolytic based on a consensus types I-III collagen 769–783 region (Table
Activities 1). The fTHP substrates have been utilized to determine individual
kinetic parameters (Table 1) and activation energies for hydrolysis
of triple-helices [46, 47, 49–53, 63–69]. MMP-1, MMP-2, MMP-
8, MMP-9, MMP-13, and MT1-MMP hydrolysis of the consensus
types I-III collagen sequence (designated fTHP-15) occurred at
the Gly~Leu bond, the analogous bond cleaved by these MMPs in
the native collagen chains [70]. Thus, the incorporation of the
fluorophore/quenching pair of Mca and Dnp did not affect the
ability of the enzyme to recognize or cleave the substrate. A modi-
fied version of the consensus types I-III collagen fTHP, in which
the P1′ subsite incorporated a Cys(4-methoxybenzyl) group
(fTHP-9), was hydrolyzed efficiently by MT1-MMP, MMP-8, and
MMP-13, but poorly by other MMPs [49, 50].
Analysis of MMP-1/THP interactions by hydrogen/deute-
rium exchange mass spectrometry (HDX MS) followed by evalua-
tion of wild-type and mutant MMP-1 kinetics using fTHPs led to
166 Maciej J. Stawikowski et al.
2 Materials
2.1 Manual Peptide Standard peptide reaction vessels (Chemglass Life Sciences or
Synthesis Peptides International) are used for manual peptide synthesis.
2.3 Solid Phase Resins for solid-phase synthesis, including Fmoc-Rink amide
for Peptide Synthesis 4-methylbenzhydrylamine (MBHA) (Rapp-Polymere, EMD
Millipore or Advanced ChemTech).
Fig. 1 The design of MMP triple-helical substrates. Illustration of a MMP/peptide complex showing that mul-
tiple contacts are formed between a triple-helical collagen-model peptide and the MMP CAT and HPX domains.
The optimal positions for donor/acceptor moieties when designing synthetic fluorescent peptides were found
to be at the P5 subsite (behind the active site) and the P5′ subsite (within the hinge domain). The active site is
marked with a dashed line
5. N,N-diisopropylethylamine (DIEA).
6. 1,8-diazabicyclo[5.4.0]undec-7-ene (DBU).
7. Hydrazine hydrate.
8. [dimethylamino(triazolo[4,5-b]pyridin-3-yloxy)methylidene]-
dimethylazanium hexafluorophosphate (HATU).
9. 4-dimethylaminopyridine (DMAP).
10. Trifluoroacetic acid (TFA, peptide synthesis grade).
11. Piperidine.
12. Ammonia solution.
13. Bromoacetic acid.
14. Acetic acid.
15. Ninhydrin test kit (Anaspec).
16. Amines of choice (commercial sources for these chemical can
be found using an online chemical directory search, such as
ChemExper Chemical Directory, www.chemexper.com).
2.6.1 Fmoc Deprotection Mix 20 mL DBU, 100 mL of piperidine, and 880 mL of NMP.
Solution
2.6.3 Peptide Cleavage To prepare 5 mL of such a solution mix 250 μL of H2O and 4.75
Cocktail mL of TFA. For every 100 mg of resin use 1.5 mL cleavage cock-
tail (see Note 1).
C = A / (l × 3 × ε ) .
3 Methods
3.1.1 General Amino Acid For Fmoc-amino acid couplings the following molar excess is rec-
Coupling Cycles ommended: Fmoc-AA = 3 equiv., HCTU = 2.5 equiv., and DIEA
= 5.5 equiv. Recommended coupling time using a conventional
automated peptide synthesizer is 1 h.
3.1.2 General Fmoc The Fmoc protecting group is removed by gentle treatment of
Deprotection Cycle peptidyl-resin using standard Fmoc deprotection solution. This
step can be performed on an automated peptide synthesizer using
an appropriate deprotection program or manually as follows:
1. Place the resin in a peptide synthesis reaction vessel.
2. Add the appropriate amount of Fmoc deprotection solution
(three to five times the resin volume).
3. Agitate the resin for 5 min. Decant the solution.
4. Repeat step 2.
5. Agitate the resin for another 15 min. Decant the solution.
6. Wash resin (three times) using 5 mL of DMF.
3.1.3 Removal The Dde protecting group is removed from Lys residues according
of the Dde Protecting to the following steps:
Group 1. Place the resin in a peptide synthesis reaction vessel.
2. Add the appropriate amount of Dde deprotection solution
(four times the resin volume).
3. Agitate the resin for 5 min and then decant the solution.
4. Repeat the treatment with Dde deprotection solution two
more times before decanting off the solution.
5. Wash the resin three times using 5 mL of DMF.
Fig. 2 Flowchart depicting MMP triple-helical peptidase activities. The nine MMPs examined in this study were
initially subdivided based upon their ability to cleave a triple-helical substrate. Triple-helical peptidase MMPs
were further subdivided upon their ability to process more thermally stable substrates and finally, based on
their specificity for residues within the P1′ and P2 subsites. Ultimately, the flowchart illustrates that “collageno-
lytic” MMPs display distinct proteolytic behaviors. Figure modified from [51]
(c) Add this mixture to the resin and agitate for 16 h. Filter
the resin and sequentially wash with 5 mL of DMF, DCM,
and MeOH.
(d) Perform a ninhydrin test. A negative result indicates com-
plete coupling of 5-FAM.
(e) Cleave the peptide from the resin using a solution contain-
ing 5% thioanisole and 90% TFA in water (use 1.5 mL of
cleavage cocktail per 100 mg of dry resin). Incubate at
room temperature for 3 h to ensure complete cleavage and
side-chain deprotection of the peptide. Perform this step
under the fume hood (see Note 2).
(f) Collect the filtrate and add 10–15 mL of water. Perform
this step under the fume hood.
(g) Freeze and then lyophilize.
(h) Purify the peptide by preparative RP-HPLC. Then analyze
by analytical HPLC and MALDI-TOF mass spectrometry
to check the purity and mass of the peptide.
(i) The concentration of the 5-FAM-containing THP peptide
can be determined by absorption spectroscopy (λ = 495
nm, ε = 75,000 cm−1 M−1) on a NanoDrop
spectrophotometer.
3.1.5 “On-Resin” This protocol is adapted from [76]. DABCYL is fairly soluble in
Approach for Synthesis DMF, but has much greater solubility in NMP.
of THPs Containing 1. Remove side chain Dde protecting groups using the Dde
DABCYL deprotection solution and conditions (see above).
Table 2
Fmoc fluorophore and quencher building block synthesis and commercial availability
3.1.6 Synthesis of THP This protocol (Fig. 3) is adapted from [68]. Peptoid residues can
Substrates Containing be conveniently introduced during a standard peptide synthesis
Peptoid Residues using the submonomeric approach. The incorporation of each
peptoid unit requires two steps: acylation using chloro- or bromo-
acetic acid followed by substitution with the appropriate amine
[77] as outlined below:
1. Prepare all solvents and reagents in a fume hood.
2. Place the resin in a dry reaction vessel.
3. If necessary, remove Fmoc protecting groups by agitating the
resin with 20% piperidine in DMF (3 × 6 min) using four to
five times the resin volume.
4. Wash the resin (3 min each) three times with 10 mL of DMF
followed by three times with 10 mL of DCM.
5. Dissolve bromoacetic acid (10 equiv. relative to resin loading)
and DIC (10 equiv) in an amount of DMF which equals 2–3
volumes of resin and add this solution to the resin.
6. Allow the resin to agitate at room temperature for 30 min.
7. Wash the resin three times with 10 mL of DMF for 3 min
each.
8. Dissolve the amine (25 equiv) in DMSO, NMP, or DMF.
9. Upon addition of the amine solution to the resin, allow the
resin to agitate at room temperature for 2 h. In some cases,
this displacement step may take up to 12 h (overnight).
10. Wash the resin (3 min each) three times with 10 mL of DMF
followed by three times with 10 mL of DCM.
11. Perform the ninhydrin test when necessary.
Fluorogenic Peptide Substrates for Matrix Metalloproteases 175
3.3 Enzyme 1. Dilute proMMP-1 to 100 μg/mL in TS buffer (see Note 4).
Activation 2. Activate proMMP-1 by adding APMA to a final concentration
3.3.1 Activation of 1 mM.
of proMMP-1
3. Incubate at 37 °C for 2–3 h.
4. Immediately after incubation dilute MMP-1 to 20 μg/mL and
store at −80 °C until ready to use.
5. Activity is initially measured with the Knight substrate (MMP-1
exhibits a weak activity toward fTHP-15).
3.3.2 Activation 1. Dilute proMMP-2 to 100 μg/mL in TS buffer (see Note 4).
of proMMP-2 2. Activate proMMP-2 by adding APMA to a final concentration
of 1 mM.
3. Incubate at 37 °C for 24 h.
4. Immediately after incubation dilute MMP-2 to 20 μg/mL and
store at −80 °C until ready to use.
5. MMP-2 can also be obtained in active form from EMD
Millipore (cat # PF023).
3.3.3 Activation 1. Activate proMMP-3 at 20 μg/mL in EAB containing 5 μg/
of proMMP-3 mL chymotrypsin.
2. Incubate at 37 °C for 30 min.
176 Maciej J. Stawikowski et al.
3.3.4 Activation 1. Dilute proMMP-8 to 100 μg/mL in TS buffer (see Note 4).
of proMMP-8 2. Activate proMMP-8 by adding APMA to a final concentration
of 1 mM.
3. Incubate at 37 °C for 1 h.
4. Immediately after incubation dilute MMP-8 to 20 μg/mL and
store at −80 °C until ready to use.
3.3.5 Activation 1. Dilute proMMP-9 to 100 μg/mL in TS buffer (see Note 4).
of proMMP-9 2. Activate proMMP-9 by adding APMA to a final concentration
of 1 mM.
3. Incubate at 37 °C for 24 h.
4. Immediately after incubation dilute MMP-9 to 20 μg/mL and
store at −80 °C until ready to use.
5. MMP-9 can also be obtained in active form from EMD
Millipore (cat # PF140).
3.3.6 Activation 1. Dilute proMMP-12 to 100 μg/mL in TS buffer (see Note 4).
of proMMP-12 2. Activate proMMP-12 by adding APMA to a final concentra-
tion of 1 mM.
3. Incubate at 37 °C for 24 h.
4. Immediately after incubation dilute MMP-12 to 20 μg/mL
and store at −80 °C until ready to use.
3.3.7 Activation 1. Dilute proMMP-13 to 100 μg/mL in TS buffer (see Note 4).
of proMMP-13 2. Activate proMMP-13 by adding APMA to a final concentra-
tion of 1 mM.
3. Incubate at 37 °C for 2 h.
4. Immediately after incubation dilute MMP-13 to 20 μg/mL
and store at −80 °C until ready to use.
3.4 Enzymatic Manual assays are performed in black low volume 384-well plates
Assays Using fTHPs (Greiner Bio-One, Monroe, NC) using a microplate reader such as
BioTek Synergy 4 running Gen5 1.11, Gen5 2.00, and Gen5 2.01
software as described previously [64, 81]. MMP-3 should not
cleave fTHP substrates and can be used as a negative control. Its
activity can be tested with the Knight SSP. Enzymatic assays are
performed as follows:
1. Prepare 100 μM fTHP-15 and α1(V)436–447 fTHP stock
solutions in TS buffer and incubate overnight at 4 °C to allow
for the proper folding of the peptide triple-helix. The working
stock is stable for 1 week.
2. Prepare a 12-point serial dilution of fTHP substrate with the
highest concentration being 50 μM.
3. Add 5 μL of each peptide solution to 12 wells of the 384-well
plate.
4. Measure the initial fluorescence in relative fluorescence units
(RFUs).
5. Add 5 μL of a twofold enzyme stock solution to each well.
6. Monitor fluorescence for 15 min at 25 °C to determine the
initial reaction rate.
178 Maciej J. Stawikowski et al.
4 Notes
Fig. 3 Synthesis of fTHP substrates. (a) Peptidic fTHPs and (b) fTHP containing a peptoid residue
Fluorogenic Peptide Substrates for Matrix Metalloproteases 179
Acknowledgments
References
10. Pan W, Arnone M, Kendall M, Grafstrom RH, 22. Rakhmanova V, Meyer R, Tong X (2007) New
Seitz SP, Wasserman ZR, Albright CF (2003) substrates for FRET-based assays: Use of a
Identification of peptide substrates for human long-wavelength fluorophore to detect the
MMP-11 (stromelysin-3) using phage display. activity of MMPs. Gen Eng Biotechnol News
J Biol Chem 278:27820–27827 15:40
11. Knight CG (1995) Fluorometric assays of pro- 23. Moss ML, Rasmussen FH (2007) Fluorescent
teolytic enzymes. Methods Enzymol substrates for the proteinases ADAM17,
248:18–34 ADAM10, ADAM8, and ADAM12 useful for
12. Gershkovich AA, Kholodovych VV (1996) high-throughut inhibitor screening. Anal
Fluorogenic substrates for proteases based on Biochem 366:144–148
intramolecular fluorescence energy transfer 24. Buchardt J, Ferreras M, Krog-Jensen C,
(IFETS). J Biochem Biophys Methods Delaisse J-M, Foged NT, Meldal M (1999)
33:135–162 Phosphinic peptide matrix metalloproteinase-9
13. Stack MS, Gray RD (1989) Comparison of inhibitors by solid-phase synthesis using a
vertebrate collagenase and gelatinase using a building block approach. Chem A Eur
new fluorogenic substrate peptide. J Biol Chem J 5:2877–2884
264(8):4277–4281 25. Buchardt J, Schiodt CB, Krog-Jensen C,
14. Niedzwiecki L, Teahan J, Harrison RK, Stein Delaissé J-M, Foged NT, Meldal M (2000)
RL (1992) Substrate specificity of the human Solid phase combinatorial library of phos-
matrix metalloproteinase stromelysin and the phinic peptides for discovery of matrix metal-
development of continuous fluorometric loproteinase inhibitors. J Comb Chem
assays. Biochemistry 31:12618–12623 2:624–638
15. Knight CG, Willenbrock F, Murphy G (1992) 26. Ito AS, Turchiello RDF, Hirata IY, Cezari
A novel coumarin-labelled peptide for sensitive MHS, Meldal M, Juliano L (1999) Fluorescent
continuous assays of the matrix metalloprotein- properties of amino acids labeled with ortho-
ases. FEBS Lett 296(3):263–266 aminobenzoic acid. Biospectroscopy
16. Bouvier J, Schneider P, Malcolm B (1993) A 4:395–402
fluorescent peptide substrate for the surface 27. de Souzaa ES, Hiratab IY, Julianob L, Itoa AS
metalloprotease of Leishmania. Exp Parasitol (2000) End-to-end distance distribution in
76:146–155 bradykinin observed by Förster resonance
17. Nagase H, Fields CG, Fields GB (1994) Design energy transfer. Biochim Biophys Acta
and characterization of a fluorogenic substrate 1474:251–261
selectively hydrolyzed by stromelysin 1 (matrix 28. Takara M, Ito AS (2005) General and specific
metalloproteinase-3). J Biol Chem solvent effects in optical spectra of ortho-
269:20952–20957 aminobenzoic acid. J Fluoresc 15:171–177
18. Bickett DM, Green MD, Berman J, Dezube 29. George J, Teear ML, Norey CG, Burns DD
M, Howe AS, Brown PJ, Roth JT, McGeehan (2003) Evaluation of an imaging platform dur-
GM (1993) A high throughput fluorogenic ing the development of a FRET protease assay.
substrate for interstitial collagenase (MMP-1) J Biomol Screen 8:72–80
and gelatinase (MMP-9). Anal Biochem 30. Knight CG (1991) A quenched fluorescent
212:58–64 substrate for thimet peptidase containing a new
19. Beekman B, Wouter J, Bloemhoff W, Ronday fluorescent amino acid, DL-2-amino-3-(7-
K, Tak PP, te Koppele JM (1996) Convenient methoxy-4-coumaryl)propionic acid. Biochem
fluorometric assay for matrix metalloproteinase J 274:45–48
activity and its application in biological media. 31. Woodhead-Galloway J (1980) In: Collagen:
FEBS Lett 390:221–225 The anatomy of a protein. Edward Arnold
20. Geoghegan KF, Emery MJ, Martin WH, Limited, London, pp. 10–19
McColl AS, Daumy GO (1993) Site-directed 32. Shoulders MD, Raines RT (2009) Collagen
double fluorescent tagging of human renin and structure and stability. Annu Rev Biochem
collagenase (MMP-1) substrate peptides using 78:929–958
the periodate oxidation of N-terminal serine. 33. Fields GB, Prockop DJ (1996) Perspectives on
An apparently general strategy for provision of the synthesis and application of triple-helical,
energy-transfer substrates for proteases. collagen-model peptides. Biopolymers (Pept
Bioconjug Chem 4:537–544 Sci) 40:345–357
21. Geoghegan KF (1996) Improved method for 34. Fields GB (2010) Synthesis and biological
converting an unmodified peptide to an energy applications of collagen-model triple-helical
transfer substrate for proteinase. Bioconjug peptides. Org Biomol Chem 8:1237–1258
Chem 7:385–391
Fluorogenic Peptide Substrates for Matrix Metalloproteases 181
35. Jenkins CL, Raines RT (2002) Insights on the teinase-2 and -9. J Biol Chem
conformational stability of collagen. Nat Prod 278:18140–18145
Rep 19:49–59 48. Lauer-Fields JL, Kele P, Sui G, Nagase H,
36. Brodsky B, Shah NK (1995) The triple-helix Leblanc RM, Fields GB (2003) Analysis of
motif in proteins. FASEB J 9:1537–1546 matrix metalloproteinase activity using triple-
37. Koide T (2005) Triple helical collagen-like helical substrates incorporating fluorogenic L-
peptides: engineering and applications in or D-amino acids. Anal Biochem
matrix biology. Connect Tissue Res 321:105–115
46:131–141 49. Minond D, Lauer-Fields JL, Nagase H, Fields
38. Koide T (2007) Designed triple-helical pep- GB (2004) Matrix metalloproteinase triple-
tides as tools for collagen biochemistry and helical peptidase activities are differentially
matrix engineering. Phil Trans R Soc B regulated by substrate stability. Biochemistry
362:1281–1291 43:11474–11481
39. Brodsky B, Thiagarajan G, Madhan B, Kar K 50. Minond D, Lauer-Fields JL, Cudic M, Overall
(2008) Triple-helical peptides: an approach to CM, Pei D, Brew K, Visse R, Nagase H, Fields
collagen conformation, stability, and self- GB (2006) The roles of substrate thermal sta-
association. Biopolymers 89:345–353 bility and P2 and P1' subsite identity on matrix
40. Lauer-Fields JL, Tuzinski KA, Shimokawa K, metalloproteinase triple-helical peptidase activ-
Nagase H, Fields GB (2000) Hydrolysis of ity and collagen specificity. J Biol Chem
triple-helical collagen peptide models by matrix 281:38302–38313
metalloproteinases. J Biol Chem 51. Minond D, Lauer-Fields JL, Cudic M, Overall
275:13282–13290 CM, Pei D, Brew K, Moss ML, Fields GB
41. Lauer-Fields JL, Nagase H, Fields GB (2000) (2007) Differentiation of secreted and
Use of Edman degradation sequence analysis membrane- type matrix metalloproteinase
and matrix-assisted laser desorption/ionization activities based on substitutions and interrup-
mass spectrometry in designing substrates for tions of triple-helical sequences. Biochemistry
matrix metalloproteinases. J Chromatogr A 46:3724–3733
890:117–125 52. Lauer-Fields JL, Chalmers MJ, Busby SA,
42. Ottl J, Battistuta R, Pieper M, Tschesche H, Minond D, Griffin PR, Fields GB (2009)
Bode W, Kühn K, Moroder L (1996) Design Identification of specific hemopexin-like
and synthesis of heterotrimeric collagen pep- domain residues that facilitate matrix metallo-
tides with a built-in cystine-knot. FEBS Lett proteinase collagenolytic activity. J Biol Chem
398:31–36 284:24017–24024
43. Ottl J, Gabriel D, Murphy G, Knäuper V, 53. Lauer-Fields JL, Fields GB (2002) Triple-
Tominaga Y, Nagase H, Kröger M, Tschesche helical peptide analysis of collagenolytic prote-
H, Bode W, Moroder L (2000) Recognition ase activity. Biol Chem 383:1095–1105
and catabolism of synthetic heterotrimeric col- 54. Lauer-Fields JL, Juska D, Fields GB (2002)
lagen peptides by matrix metalloproteinases. Matrix metalloproteinases and collagen catabo-
Chem Biol 7:119–132 lism. Biopolymers (Pept Sci) 66:19–32
44. Ottl J, Moroder L (1999) Disulfide-bridged 55. Tokmina-Roszyk M, Tokmina-Roszyk D,
heterotrimeric collagen peptides containing Fields GB (2013) The synthesis and applica-
the collagenase cleavage site of collagen type I: tion of Fmoc-Lys(FAM) building blocks.
synthesis and conformational properties. J Am Biopolymers (Pept Sci) 100:347–355
Chem Soc 121:653–661 56. Berndt P, Fields GB, Tirrell M (1995) Synthetic
45. Bertini I, Fragai F, Luchinat C, Melikian M, lipidation of peptides and amino acids: mono-
Toccafondi M, Lauer JL, Fields GB (2012) layer structure and properties. J Am Chem Soc
Structural basis for matrix metalloproteinase 1 117:9515–9522
catalyzed collagenolysis. J Am Chem Soc 57. Yu Y-C, Berndt P, Tirrell M, Fields GB (1996)
134:2100–2110 Self-assembling amphiphiles for construction
46. Lauer-Fields JL, Broder T, Sritharan T, Nagase of protein molecular architecture. J Am Chem
H, Fields GB (2001) Kinetic analysis of matrix Soc 118:12515–12520
metalloproteinase triple-helicase activity using 58. Yu Y-C, Tirrell M, Fields GB (1998) Minimal
fluorogenic substrates. Biochemistry lipidation stabilizes protein-like molecular
40:5795–5803 architecture. J Am Chem Soc 120:9979–9987
47. Lauer-Fields JL, Sritharan T, Stack MS, Nagase 59. Yu Y-C, Roontga V, Daragan VA, Mayo KH,
H, Fields GB (2003) Selective hydrolysis of Tirrell M, Fields GB (1999) Structure and
triple-helical substrates by matrix metallopro- dynamics of peptide-amphiphiles incorporating
182 Maciej J. Stawikowski et al.
tides following synthesis by fmoc chemistry. labeling of peptides on solid phase. Org Biomol
Methods Enzymol 289:67–83 Chem 6:4582–4586
83. Liu Y, Kati W, Chen C-M, Tripathi R, Molla A, 86. Malkar NB, Fields GB (2001) Synthesis of Nα-
Kohlbrenner W (1999) Use of a fluorescence (fluoren-9-ylmethoxycarbonyl)-Nε-[(7-
plate reader for measuring kinetic parameters methoxycoumarin-4-yl)acetyl]-L-lysine for use
with inner filter effect correction. Anal Biochem in solid-phase synthesis of fluorogenic sub-
267:331–335 strates. Lett Pept Sci 7:263–267
84. Amar S, Fields GB (2012) Production and 87. Anastasi A, Knight CG, Barrett AJ (1993)
characterization of matrix metalloproteinases Characterization of the bacterial metalloendo-
implicated in multiple sclerosis. In: Kokotos G, peptidase pitrilysin by use of a continuous fluo-
Constantinou-Kokotou V, Matsoukas J (eds) rescence assay. Biochem J 290:601–607
Peptides 2012 (proceedings of the t hirty-second 88. Wang GT, Krafft GA (1992) Automated syn-
european peptide symposium). European thesis of fluorogenic protease substrates: design
Peptide Society, Athens, pp 102–103 of probes for Alzheimer's disease-associated
85. Katritzky AR, Yoshioka M, Narindoshvili T, proteases. Bioorg Med Chem Lett
Chung A, Johnson JV (2008) Fluorescent 2:1665–1668
Chapter 9
Abstract
Identification of physiological substrates is the key to understanding the pleiotropic functions of matrix
metalloproteinases (MMPs) in health and disease. Quantitative mass spectrometry-based proteomics has
revolutionized current approaches in protease substrate discovery and helped to unravel many new MMP
activities in complex biological systems. Multiplexing further extended the capabilities of these techniques
and facilitated more complicated experimental designs that include multiple proteases or monitoring the
activity of a single protease at more than one concentration or at multiple time points with a complex test
proteome. In this chapter, we provide a protocol for time-resolved iTRAQ-based Terminal Amine Isotopic
Labeling of Substrates (TAILS), with the focus on MMP substrate identification and characterization in
cell culture supernatants and introduce an automated procedure for the interpretation of time-resolved
iTRAQ-TAILS datasets.
1 Introduction
Charles A. Galea (ed.), Matrix Metalloproteases: Methods and Protocols, Methods in Molecular Biology, vol. 1579,
DOI 10.1007/978-1-4939-6863-3_9, © Springer Science+Business Media LLC 2017
185
186 Pascal Schlage et al.
Fig. 1 Overview of the time-resolved iTRAQ-TAILS workflow. Cell culture supernatant (CCS) is collected from
MMP-deficient cells (MMP KO) and incubated with the test MMP. Aliquots are removed at given time points and
subjected to the 8plex-iTRAQ-TAILS protocol. Including control samples at time zero and at the end of the
incubation time controls for basal proteolysis in the sample proteome. Peptides are analyzed by LC-MS/MS
prior to (preTAILS sample) and after (TAILS sample) N-terminal enrichment to enable stringent categorization
of true protease cleavages and non-cleavage events with the help of the automated analysis pipeline
188 Pascal Schlage et al.
2 Materials
2.1 Secretome 1. Cell culture medium (free of protein, serum, phenol red, and
Collection antibiotics).
and Concentration 2. Phosphate-buffered saline (PBS).
3. Filter with 0.22 μm pore size.
4. Phenylmethylsulfonylfluoride (PMSF).
5. Amicon Ultra-15 Centrifugal Filter Units (3 kDa MWCO,
Millipore).
6. Amine-free buffer with optimized pH for MMP activity, e.g.,
50 mM HEPES, pH 7.8.
7. Bradford protein assay (Bio-Rad).
2.7 Data Analysis 1. Search engine for spectrum to peptide assignment (e.g.,
Mascot, X! Tandem).
2. Trans-Proteomic Pipeline (TPP) (v7.1 POLAR VORTEX rev 1)
[10] (http://sourceforge.net/projects/sashimi/files/Trans-
Proteomic%20Pipeline%20%28TPP%29/TPP%20v4.7%20
%28polar%20vortex%29%20rev%201/).
190 Pascal Schlage et al.
3 Methods
3.1 Secretome 1. Wash 70% confluent MMP knockout (KO) cells (see Note 2)
Collection three times with PBS.
and Concentration 2. Incubate MMP KO cells with cell culture media (free of pro-
tein, serum, phenol red, and antibiotics).
3. After 24–48 h (see Note 3) collect the culture media and cen-
trifuge for 5 min at 400 × g at room temperature to remove
dead cells.
4. Collect supernatant and add serine protease inhibitor (0.5 mM
PMSF final) on ice.
5. Centrifuge supernatant for 30 min at 3,000 × g at 4 °C to
remove cell debris.
6. Collect supernatant and filter it through a 0.22 μm filter.
7. Freeze supernatants at −20 °C for storage or continue with
step 8.
8. Add water to Amicon Ultra-15 Centrifugal Filter Units and
wash filter to remove glycerol by centrifugation for 35 min at
4 °C (for optimal speed see manufacturer’s instructions).
9. Concentrate supernatants by multiple centrifugation steps at
4 °C.
10. Exchange buffer by diluting the concentrated supernatants
with 10 volumes of 50 mM HEPES, pH 7.8 and
centrifugation.
11. Repeat step 10 two times.
12. Reduce the volume of the concentrated supernatant to 1 ml by
further centrifugation.
13. If precipitates occur, try to dissolve them or remove them by
centrifugation.
14. Take an aliquot and determine protein concentration using the
Bradford protein assay.
15. Freeze concentrated secretome at −80 °C or continue with
test protease treatment.
Measuring Matrix Metalloprotease Activity in Complex Samples 191
3.5 Desalting, 1. Reduce volumes of peptide fractions under vacuum to ≤100 μl.
Pooling, and Cleanup 2. (Optional) If samples are dried or their volumes are too low
of Fractionated (≪100 μl), resuspend samples and adjust volumes to 100 μl by
Peptides adding equilibration solution.
3. Wet the C18 OMIX tip by aspirating twice 100 μl 50%
acetonitrile.
Measuring Matrix Metalloprotease Activity in Complex Samples 193
3.6 LC-MS/MS Analyze pooled peptide fractions for the preTAILS and TAILS
Analysis samples by tandem mass spectrometry on a hybrid mass spectrom-
eter coupled to a C18 in-line chromatography system. Settings will
depend on the mass spectrometer used, but in general we apply the
same parameters as for standard iTRAQ-based analyses. For exam-
ple, results from our test dataset (see Subheading 3.7) were obtained
on a Thermo Scientific Orbitrap Velos instrument coupled to an
Eksigent-Nano-HPLC system utilizing a self-made tip column (75
μm × 150 mm; C18 material: AQ, 3 μm 200 Å) and the following
settings: 1 μg peptide loaded; flowrate of 250 nl/min, gradient 0
to 35% acetonitrile in 62 min; full scan: 300–1700 m/z with reso-
lution of 30,000 at 400 m/z, target value 1E6; collision-induced
dissociation (CID) scan: eight most intense precursors, normalized
collision energy of 35%, activation time of 10 ms; higher energy
collision dissociation (HCD) of same precursors for iTRAQ: nor-
malized collision energy of 42%, resolution of 7500 at 400 m/z,
exclusion time of 45 s, exclusion window of 20 ppm (see Note 8).
The detailed settings for a Thermo Scientific Q Exactive instru-
ment have been published previously [12].
3.7 Data Analysis In general, time-resolved TAILS datasets are analyzed using the
CLIPPER analysis pipeline [9] according to a previously described
procedure [12]. Here, we will describe modifications needed for
time-resolved studies and focus the detailed discussion on steps
required to exploit time-resolved information for effective
discrimination of cleavage from non-cleavage events and the clas-
sification of cleavages by efficiencies. Central to our analysis strat-
egy is the use of soft clustering to stringently filter out true cleavage
events from true non-cleavage events and non-interpretable pep-
tide abundance patterns. In contrast to hard clustering (e.g.,
k-means) soft clustering algorithms (e.g., fuzzy c-means in the
Mfuzz R package [11]) do not definitively assign each object to a
single cluster, but calculate a membership value alpha (0 < alpha <
1) that defines to what extent an object belongs to a cluster.
194 Pascal Schlage et al.
4 Notes
Acknowledgments
References
1. Schlage P, auf dem Keller U (2015) Proteomic ase 10 substrate degradome. Mol Cell
approaches to uncover MMP function. Matrix Proteomics 13:580–593
Biol 44–46:232–238 6. Plasman K, Van Damme P, Kaiserman D et al
2. Rogers LD, Overall CM (2013) Proteolytic (2011) Probing the efficiency of proteolytic
post-translational modification of proteins: events by positional proteomics. Mol Cell
proteomic tools and methodology. Mol Cell Proteomics 10(M110):003301
Proteomics 12:3532–3542 7. Prudova A, auf dem Keller U, Butler GS et al
3. Plasman K, Van Damme P, Gevaert K (2013) (2010) Multiplex N-terminome analysis of
Contemporary positional proteomics strategies MMP-2 and MMP-9 substrate degradomes by
to study protein processing. Curr Opin Chem iTRAQ-TAILS quantitative proteomics. Mol
Biol 17:66–72 Cell Proteomics 9:894–911
4. auf dem Keller U, Prudova A, Gioia M et al 8. Schlage P, Kockmann T, Sabino F et al (2015)
(2010) A statistics-based platform for quantita- Matrix metalloproteinase 10 degradomics in
tive N-terminome analysis and identification of keratinocytes and epidermal tissue identifies
protease cleavage products. Mol Cell bioactive substrates with pleiotropic functions.
Proteomics 9:912–927 Mol Cell Proteomics 14:3234–3246
5. Schlage P, Egli FE, Nanni P et al (2014) Time- 9. auf dem Keller U, Overall CM (2012)
resolved analysis of the matrix metalloprotein- CLIPPER-An add-on to the Trans-Proteomic
198 Pascal Schlage et al.
Abstract
Differential proteolytic processing, for example by matrix metalloproteases (MMPs), has been recognized
as an important hallmark in numerous pathological conditions. One crucial challenge in the present studies
of proteases is system-wide identification of endogenous biological substrates. In this chapter, we highlight
a robust method for the identification of bioactive substrates and their sites of MMP cleavage, as well as by
other proteases and peptidases, in a system-wide manner. This approach enriches for putative protein
N-termini by removal of internal peptides using a charge reversal strategy. In addition, this straightforward
method can be used in combination with gel-based pre-separation of proteins to allow better estimation
of the molecular weight of the identified cleavage product of a given bioactive substrate.
1 Introduction
Charles A. Galea (ed.), Matrix Metalloproteases: Methods and Protocols, Methods in Molecular Biology, vol. 1579,
DOI 10.1007/978-1-4939-6863-3_10, © Springer Science+Business Media LLC 2017
199
200 Zon W. Lai and Oliver Schilling
Fig. 1 Workflow for the enrichment of protein N-termini for protease cleavage site identification. Unmodified
primary amines of protein N-termini and lysine side chains are first protected by chemical dimethylation fol-
lowed by digestion with trypsin. Internal peptides bearing unmodified neo-N-termini are chemically modified
using sodium 4-formylbenzene-1,3-disulfonate. Endogenously modified (e.g., acetylated Protein N-termini)
and dimethylated peptides are subsequently enriched by strong cation exchange chromatography and identi-
fied by mass spectrometry analysis
Identification of Matrix Metalloprotease Cleavage Sites 201
2 Materials
3 Methods
3.3 N-Terminal 1. Check tryptic digest by running 10% SDS-PAGE and follow
Modification silver staining procedure to compare samples from pre- and
of Internal Peptides post-digest. No protein bands (except trypsin) should appear
(i.e., Trypsin in the digested sample.
Cleavage Sites) 2. Add 20 mM (final concentration) sodium cyanoborohydride
and 20 mM (final concentration) 4-formyl-1,3-
benzenedisulfonic acid.
204 Zon W. Lai and Oliver Schilling
3.4 Removal 1. Add 0.1 M Tris (final concentration) for 1.5 h at 37 °C.
of Internal (Tryptic) 2. Acidify digested peptides using 0.3% (v/v) trifluoroacetic acid
Peptides Using to achieve pH of less than 3.0. If needed, hydrochloric acid can
Strong-Cation also be used.
Exchange HPLC 3. Desalt digested peptides using C18 solid phase extraction col-
umn (55–105 μm particle size and 125 Å pore size, up to 3 mg
binding capacity, e.g., Waters Sep-Pak C18 Cartridges):
(a) Equilibrate C18 column using one volume of 80% (v/v)
acetonitrile.
(b) Wash C18 column using three volumes of 0.1% (v/v) tri-
fluoroacetic acid.
(c) Apply digested peptides into C18 column. Reapply the
flow-through once to ensure all peptides are captured.
(d) Wash C18 column using three volumes of 0.1% (v/v) tri-
fluoroacetic acid.
(e) Elute captured peptides using one volume of C18 elution
Buffer.
4. Evaporate acetonitrile using vacuum evaporator.
5. Determine peptide concentration using bicinchoninic acid
assay. Adjust peptide concentration to approximately 1 mg/
mL using Strong Cation Exchange Chromatography Buffer A.
6. Resolve peptide mixture by strong cation exchange high per-
formance liquid chromatography (HPLC). Recommended
column specification: 5 μm particle size and 300 Å pore size,
column length 100 mm, inner diameter 4.6 mm, e.g., PolyLC
Polysulfoethyl A. Elute peptides using a linear gradient with
increasing concentration of Strong Cation Exchange
Chromatography Buffer B up to 45% (v/v) over a period of
90 min. (Due to a relatively large volume of samples, a larger
injection loop (>2.0 mL) is recommended. See Note 3.)
7. Collect between 10–14 peptide fractions and evaporate aceto-
nitrile using vacuum evaporator.
8. Desalt fractionated peptides using micro C18 solid phase
extraction column or self-packed C18 STAGE-tips [8].
9. Analyze fractionated peptides using high resolution LC-MS/
MS mass spectrometry analyzer (see Note 4).
3.5 LC-MS/MS Data 1. Convert raw LC-MS/MS data to a format compatible with the
Analysis X! Tandem search engines. Vendor-specific converters as
Identification of Matrix Metalloprotease Cleavage Sites 205
4 Notes
Acknowledgment
References
platform utilizing open XML file formats. Mol derived peptide libraries improved using
Syst Biol 1:2005.0017 WebPICS, a resource for proteomic identifica-
11. Han DK, Eng J, Zhou HL, Aebersold R tion of cleavage sites. Biol Chem
(2001) Quantitative profiling of differentiation- 392:1031–1037
induced microsomal proteins using isotope- 13. Rappsilber J, Mann M, Ishihama Y (2007)
coded affinity tags and mass spectrometry. Nat Protocol for micro-purification, enrichment,
Biotechnol 19:946–951 pre-fractionation and storage of peptides for
12. Schilling O, Keller UAD, Overall CM (2011) proteomics using StageTips. Nat Protoc
Factor Xa subsite mapping by proteome- 2:1896–1906
Chapter 11
Abstract
Protease specificity is controlled by exosites, which capture and orient the substrate, and the active site,
which binds substrate residues near the P1–P1′ scissile bond and catalyzes peptide hydrolysis. Techniques
used to identify critical contact points between a protease and its substrate can be time consuming and
labor-intensive. Screening tools such as phage display have been revitalized with the emergence of high-
throughput sequencing technology, and can be used to interrogate protease substrate specificity. This
article will outline a method for creating and screening a comprehensive mutagenesis substrate phage
display library. High-throughput sequencing of uncleaved phage at various reaction time points enables
kcat/KM determination for every possible single amino acid substitution at each position of the substrate,
providing unprecedented resolution for the interaction between a protease and its substrate.
1 Introduction
Charles A. Galea (ed.), Matrix Metalloproteases: Methods and Protocols, Methods in Molecular Biology, vol. 1579,
DOI 10.1007/978-1-4939-6863-3_11, © Springer Science+Business Media LLC 2017
209
210 Colin A. Kretz
Table 1
Traditional vs high-throughput phage display
Fig. 1 High-throughput substrate phage display. (a) In substrate phage display, a random peptide library is
displayed as an NH2-terminal fusion to the PIII protein along with an epitope tag (such as FLAG). Phage display-
ing a peptide that is cleaved by the protease is separated from the epitope tag, allowing purification of remain-
ing uncleaved phage particles. (b) This procedure is performed for multiple rounds of selection. The selected
phage is re-amplified in bacteria to iteratively evolve the library. Following multiple rounds of selection and
amplification, individual colonies are picked off agar plates, and a number of clones are identified by Sanger
sequencing. (c) High-throughput sequencing mitigates the need for multiple rounds of selection and amplifica-
tion. Millions of reads are obtained from the starting library and from phage following a single round of
selection
2 Materials
2.1 Designing The design of a mutant substrate phage display library should
Mutagenesis Library account for HTS read length. The Illumina HiSeq platform cur-
rently delivers up to 300 million sequences of 50, 100, or 150 base
pair lengths, whereas the MiSeq platform currently delivers 30 mil-
lion reads up to 300 base pairs in length. Both formats include a
paired-end read option, thus allowing for longer sequence designs.
Because the quality of sequence data decreases toward the end of
longer reads, the library should be designed such that paired-end
212 Colin A. Kretz
3 Method
Table 2
Colin A. Kretz
Primer Sequence
NGS S1a ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNNNgaatcTCAATGACAGGAGGACAACAAATG
NGS S1b ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNNNtctgaTCAATGACAGGAGGACAACAAATG
NGS S1c ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNNNcagtcTCAATGACAGGAGGACAACAAATG
NGS S1d ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNNNagtctTCAATGACAGGAGGACAACAAATG
NGS S1e ACACTCTTTCCCTACACGACGCTCTTCCGATCTNNNNNNtgcaaTCAATGACAGGAGGACAACAAATG
NGS AS1a CTCGGCATTCCTGCTGAACCGCTCTTCCGATCTNNNNNNgtctaACTTTCAACAGTTTCGGCCC
NGS AS1b CTCGGCATTCCTGCTGAACCGCTCTTCCGATCTNNNNNNcagagACTTTCAACAGTTTCGGCCC
NGS AS1c CTCGGCATTCCTGCTGAACCGCTCTTCCGATCTNNNNNNttcacACTTTCAACAGTTTCGGCCC
NGS AS1d CTCGGCATTCCTGCTGAACCGCTCTTCCGATCTNNNNNNagtctACTTTCAACAGTTTCGGCCC
NGS AS1e CTCGGCATTCCTGCTGAACCGCTCTTCCGATCTNNNNNNtcaggACTTTCAACAGTTTCGGCCC
PESeq S1 CAAGCAGAAGACGGCATACGAGATCGGTCTCGGCATTCCTGCTGAACCGCTCTTCCGATCT
PESeq AS1 AATGATACGGCGACCACCGAGATCTACACTCTTTCCCTACACGACGCTCTTCCGATCT
Defining the Protease Substrate Recognition Landscape 217
3.2 Phage 1. From Subheading 3.1, step 12. Pellet bacteria using a two-
Preparation step centrifugation. (Step 1) 10 min at 2400 × g, 4 °C. Transfer
supernatant containing phage to fresh bottle. (Step 2) 10 min
at 6200 × g, 4 °C.
2. Store aliquots of final library. Resuspend bacterial pellet in 50 ×
1 mL aliquots of 2× YT with 20% glycerol, and store in −80 °C.
3. To the phage-containing culture supernatant, add 0.15× vol-
ume (150 mL for 1 L culture) of PEG/NaCl. Gently mix by
inverting bottles several times (up to 100 times). Incubate at
4 °C overnight to precipitate.
Defining the Protease Substrate Recognition Landscape 219
3.3 Initial Reaction Experiments conducted using phage displaying the wild-type sub-
Time Course strate are important for informing the appropriate conditions for
with Phage Displaying the reaction between the protease and the mutant substrate phage
Wild-Type Substrate display library.
and AlphaLISA 1. Assemble 500 μL reaction containing 400 μL wild-type sub-
strate displaying phage (from Subheading 3.2, step 6), 100 μL
5× reaction buffer. Remove 50 μL of solution into 50 μL stop
buffer (0 min).
2. Initiate the reaction by adding remaining 450 μL of phage
solution into 5 μL of 455 nM protease stock solution, achiev-
ing a final concentration of 5 nM protease (see Note 8).
3. Remove 50 μL reaction time points into 50 μL of 4 °C stop
buffer. Repeat at each of the remaining reaction time points for
a total of eight time points. Recommended initial time points
(min): 0, 1, 5, 10, 50, 100, 500, overnight (see Note 9).
4. During reaction time course, perform a dilution series for a
standard curve. Repeat Subheading 3.3, step 1, omitting pro-
tease. Perform a 1:2 dilution series, by adding 250 μL of phage
to 250 μL 1× Reaction Buffer (1 mL 5× reaction buffer + 4 mL
TBS). Add 250 μL Stop Buffer to each dilution (see Note 10).
5. Test the selected time course for rates of phage cleavage using
AlphaLISA (see Note 11). Prepare a 1100 μL Master Mix
solution of 20 μg/mL anti-FLAG coated Acceptor beads and
4 nM biotinylated anti-fdTET in AlphaLISA Buffer. Add 25
μL of Master Mix solution to 32 wells of a 96 half-well plate.
In duplicate, add 5 μL of each standard curve dilution, or 5 μL
of each reaction time point to each well. Incubate at room
temperature for 1 h. Prepare a 700 μL solution of 40 μg/mL
220 Colin A. Kretz
streptavidin-
coated donor beads in AlphaLISA Buffer, and
add 20 μL to each well. Protect from light, and incubate at
room temperature for 30 min (see Note 12). Read the plates
on an EnSpire 2300 Multilabel Plate Reader at an excitation
wavelength of 680 nm and emission wavelength of 615 nm.
6. Construct a standard curve of AlphaLISA signal as a function
of the natural log of the molar phage concentration. Determine
the concentration of uncleaved phage at each time point from
the protease reacted phage, and calculate kcat/KM value.
3.4 Reaction 1. Repeat Subheading 3.3, steps 1–6 using mutant substrate
of Phage Displaying phage display library.
Mutant Substrate 2. Selection of reaction time points (t0–t5) for analysis by HTS
Phage Display Library should be based on time points exhibiting less than 75% sub-
with Protease strate consumption (see Note 13). A single time point repre-
senting maximal library cleavage (Δ[substrate] → 0) should also be
taken (tfinal).
3. Add 500 μL Stop Buffer and 50 μL anti-FLAG agarose beads
to select mutant substrate phage library reaction time points.
Incubate at 4 °C overnight with end-over-end mixing.
4. Centrifuge at 1,000 × g for 2 min to pellet beads. Wash beads
5× with Stop Buffer.
5. Elute phage from beads at each reaction time point by adding
500 μL of 0.15 mg/mL 3× FLAG peptide in Stop Buffer.
Incubate 30 min at room temperature with end-over-end
mixing.
6. Centrifuge at 1,000 × g for 2 min to pellet beads. Remove
supernatant (containing selected phage) into fresh tube.
7. Add 5 μL of 10 mg/mL proteinase K. Incubate at 50 °C for 4
h (or overnight) to digest phage coat protein, exposing ssDNA.
8. Add 500 μL phenol/chloroform/isoamyl alcohol, and vortex.
Centrifuge at 14,000 × g for 10 min at room temperature.
Remove top aqueous layer into clean microcentrifuge tube.
Add 200 μL chloroform to remove excess phenol, vortex, and
then centrifuge for 10 min at 14,000 × g. Remove top aqueous
layer into a fresh tube.
9. Precipitate and concentrate ssDNA as in Subheading 3.1, step 7.
10. Resuspend pelleted ssDNA in 20 μL TE Buffer.
3.5 Preparation 1. Assemble a 50 μL PCR reaction for each ssDNA sample col-
of High-Throughput lected from reaction time points (Subheading 3.4, step 10)
Sequencing Samples containing 5 μL template, 0.5 μM NGS-S1(a → e), 0.5 μM
NGS-AS1(a → e), and Herculase reaction components accord-
ing to manufacturer’s instructions. The thermal profile is: (1)
98 °C (2 min), (2) 98 °C (20 s), (3) 60 °C (20 s), (4) 72 °C (30
s), (5) 72 °C (5 min), repeat 2–4 for 8–16 cycles (see Note 14).
Defining the Protease Substrate Recognition Landscape 221
3.6 Data Analysis Data analysis pipelines for high-throughput sequencing of phage
display experiments are still largely dependent on in-house coding
algorithms. Although some formal pipelines are available to calcu-
late mutation frequency and assess enrichment [16, 20], they do
not provide the flexibility for customized analysis. A general strat-
egy is provided for analyzing the performance of mutant substrate
sequences (Fig. 3), as quantified from high-throughput sequenc-
ing data.
3.6.1 Raw Data 1. Parse sequencing data into bins according to barcodes, repre-
sented by nucleotide reads 7–11. Each bin now contains the
sequencing data collected from the uncleaved phage at each
reaction time point. Check for comparable sequence read
depth across reaction time points.
2. Trim sequences of the 6 N wobble, barcode, and primer, leav-
ing only the mutant library.
3. FASTQ files from paired-end reads are aligned to wild-type
sequence using Bowtie2 [21], yielding a paired SAM output
file (http://bowtie-bio.sourceforge.net).
4. Assemble paired end reads into a single sequence. If paired
reads overlap and alignments show discordant base calls, the
base with the higher quality score is kept. If the discordant
222 Colin A. Kretz
Fig. 3 High-throughput sequencing analysis scheme. An outline for the analysis of high-throughput sequencing
data is shown. Following high-throughput sequencing of the uncleaved phage at various reaction time points,
raw sequencing reads are separated into bins. Paired-end (PE) reads are assembled into single sequences, and
the frequency (f) of wild-type sequences is initially analyzed. Next, the frequency of each amino acid (AA) at
each position of the substrate is calculated by dividing the count by the total number of reads assigned to that
bin. Alternatively, the frequency of each amino acid is only calculated from the subset of clones that only con-
tain a single amino acid substitution. This can be used to mitigate the effects of multiple substitutions within a
clone affecting the results. Ultimately, the frequency of each amino acid at each time point is multiplied by the
concentration of uncleaved phage at that time point, before fitting the data to the first order rate law
Defining the Protease Substrate Recognition Landscape 223
base calls have identical quality scores, the forward read (read
1) should be preferred.
3.6.2 Analysis of Wild- 1. Assess the frequency of wild-type substrate in each reaction
Type Sequences time point. In the starting library (t0), wild-type should be ~(1
Within the Library − μ)n, where μ is the mutation frequency at each nucleotide
position and n is the number of nucleotides in the sequence
(see Note 17).
2. Multiply the frequency of wild-type sequence, by the molar
concentration of uncleaved phage at the corresponding reac-
tion time point (from Subheading 3.4, step 1).
3. Fit the data to the first order rate law: ln([A]t) = −kt + ln([A]0).
Perform a regression of the natural log of molar concentration
of WT phage as a function of time. The slope, k, divided by the
molar concentration of enzyme yields kcat/KM. Compare this
value to previously determined values (from Subheading 3.3)
to assess the reliability of the data (see Note 18).
4. Determine the effect of multiple substitutions within a clone
on the rate of proteolysis measured for a given mutant at a
position (i.e., epistasis), using the WT amino acid frequency at
each position. These counts are derived from substrate clones
containing multiple amino acid substitutions, but share the
WT amino acid at this position in common. As in Subheading
3.6.2, steps 2–3, calculate the kcat/KM for each wild-type
amino acid at each position. If the value for each WT amino
acid at each position is comparable to each other, it suggests
that the aggregate effect of multiple substitutions on a given
peptide is mostly averaged across sequences sharing a given
amino acid. Thus, strong epistasis is not common within the
dataset. If needed, more formal and rigorous analysis of epista-
sis should be explored [15, 22].
3.6.3 Analysis of Mutant 1. If Subheading 3.6.2, step 4 indicates limited epistasis within
Sequences the library, then the counts for each amino acid at each posi-
Within the Library tion of the substrate (Subheading 3.6.2, step 3, above) can be
used to calculate the kcat/KM for each mutant sequence.
2. If Subheading 3.6.2, step 4 suggests multiple interactions
between mutations at certain positions within the library, then
the mutation frequency calculated from Subheading 3.6.2,
step 3, cannot reliably estimate the effect of a single amino
acid substitution on the rate of cleavage in the library. Instead,
the frequency of each amino acid substitution at each position
of the peptide should be calculated from the subset of sequences
that contain only a single amino acid substitution. This will
only cover a fraction of the potential sequence space of the
peptide. However, this subset of mutations will likely contain
224 Colin A. Kretz
4 Conclusion
5 Notes
7. Much of the success of the library build comes from this step.
Poor transformation efficiency can lead to poor library depth,
and ultimately incomplete diversity. Typically, ten electropora-
tions per library are sufficient to yield 1 × 10 [7] independent
clones. However, more may be required. 30–40 electropora-
tions can be needed to yield sufficient diversity. Keep an eye on
the time constant, optimally at 4.2 ms, if drops below 4 ms
may indicate salts in the ligation product that increase conduc-
tivity. Try additional 70% ethanol washes at room temperature
(including a 20 min incubation) to extract excess salts from the
precipitation reaction. Also, too much glycogen can impede
transformation efficiency.
8. It is important to perform these reactions under pseudo-first
order reaction conditions, where the concentration of sub-
strate is ≪KM. Leave lots of room here, in case mutations are
discovered that enhance cleavage relative to WT. But an initial
reaction setup to the conditions for a WT substrate will suffice
for an initial analysis.
9. The reaction time course will need to be determined empiri-
cally if the interaction is not already well defined. Adjust prote-
ase concentration and time points accordingly.
10. Stop buffer contains EDTA, which we have found to interfere
with the AlphaLISA signal. This must be accounted for in the
standard curve.
11. AlphaLISA is a rapid alternative to standard ELISA protocols.
However, if appropriate plate readers are not available, then
standard ELISA can also be used. In this case, precoat plates
with 10 μg/mL anti-fd-TET antibody (Sigma) and detection
with 1/10,000 HRP-anti-FLAG antibody (Sigma). Detection
is achieved using 1 Step Ultra TMB-ELISA substrate (Thermo
Scientific #34028). Filamentous phage exhibits a high degree
of nonspecific binding to plastic surfaces, which can be lever-
aged by capturing with an anti-phage antibody.
12. Donor beads are highly susceptible to photo-bleaching. Turn
off overhead fluorescent lights when working with donor
beads.
13. This may need to be determined empirically, as it depends on
the KM of the interaction. Dogmatically, initial rates of a reac-
tion are determined with less than 10% substrate consumption.
However, fitting the data to a first order rate law accounts for
decreased rates of cleavage at later time points caused by sub-
strate consumption.
14. The number of cycles should be determined empirically to
avoid PCR recombination (product laddering) that can cause
bias in library distribution unrelated to the protease selection.
This phenomenon tends to occur when PCR reactions are
Defining the Protease Substrate Recognition Landscape 227
Acknowledgments
References
1. Chan CE, Lim AP, MacAry PA, Hanson BJ 5. Hills R, Mazzarella R, Fok K, Liu M,
(2014) The role of phage display in therapeutic Nemirovskiy O, Leone J, Zack MD, Arner
antibody discovery. Int Immunol 26:649–657 EC, Viswanathan M, Abujoub A,
2. Chen S, Heinis C (2015) Phage selection of Muruganandam A, Sexton DJ, Bassill GJ, Sato
bicyclic peptides based on two disulfide bridges. AK, Malfait AM, Tortorella MD (2007)
Methods Mol Biol 1248:119–137 Identification of an ADAMTS-4 cleavage
3. Whitney M, Crisp JL, Olson ES, Aguilera TA, motif using phage display leads to the devel-
Gross LA, Ellies LG, Tsien RY (2010) Parallel opment of fluorogenic peptide substrates and
in vivo and in vitro selection using phage display reveals matrilin-3 as a novel substrate. J Biol
identifies protease-dependent tumor- targeting Chem 282:11101–11109
peptides. J Biol Chem 285:22532–22541 6. Eckhard U, Huesgen PF, Schilling O, Bellac
4. Kridel SJ, Chen E, Kotra LP, Howard EW, CL, Butler GS, Cox JH, Dufour A, Goebeler V,
Mobashery S, Smith JW (2001) Substrate Kappelhoff R, Keller UA, Klein T, Lange PF,
hydrolysis by matrix metalloproteinase-9. Marino G, Morrison CJ, Prudova A, Rodriguez
J Biol Chem 276:20572–20578 D, Starr AE, Wang Y, Overall CM (2016)
228 Colin A. Kretz
Active site specificity of the matrix metallopro- FA, Meng F, Ginsburg D (2015) Massively
teinase family: proteomic identification of 4300 parallel enzyme kinetics reveals the substrate
cleavage sites by nine MMPs explored with recognition landscape of the metalloprotease
structural and synthetic peptide cleavage analy- ADAMTS13. Proc Natl Acad Sci U S A
ses. Matrix Biol 49:37–60 112:9328–9333
7. Chen EI, Kridel SJ, Howard EW, Li W, Godzik 16. Fowler DM, Araya CL, Gerard W, Fields S
A, Smith JW (2002) A unique substrate recog- (2011) Enrich: software for analysis of protein
nition profile for matrix metalloproteinase-2. function by enrichment and depletion of vari-
J Biol Chem 277:4485–4491 ants. Bioinformatics 27:3430–3431
8. Ratnikov BI, Cieplak P, Gramatikoff K, Pierce 17. Glenn TC (2011) Field guide to next-
J, Eroshkin A, Igarashi Y, Kazanov M, Sun Q, generation DNA sequencers. Mol Ecol Resour
Godzik A, Osterman A, Stec B, Strongin A, 11:759–769
Smith JW (2014) Basis for substrate recogni- 18. Desch KC, Kretz C, Yee A, Gildersleeve R,
tion and distinction by matrix metalloprotein- Metzger K, Agrawal N, Cheng J, Ginsburg D
ases. Proc Natl Acad Sci U S A (2015) Probing ADAMTS13 substrate speci-
111(40):E4148–E4155 ficity using phage display. PLoS One
9. Huxley-Jones J, Clarke TK, Beck C, Toubaris 10:e0122931
G, Robertson DL, Boot-Handford RP (2007) 19. Scott JK, Smith GP (1990) Searching for pep-
The evolution of the vertebrate metzincins; tide ligands with an epitope library. Science
insights from Ciona intestinalis and Danio 249:386–390
rerio. BMC Evol Biol 7:63 20. Love MI, Huber W, and Anders S (2014).
10. Shendure J, Lieberman Aiden E (2012) The Moderated estimation of fold change and dis-
expanding scope of DNA sequencing. Nat persion for RNA-seq data with DESeq2.
Biotechnol 30:1084–1094 Genome Biology 15:550
11. Tinberg CE, Khare SD, Dou J, Doyle L, 21. Langmead B, Salzberg SL (2012) Fast gapped-
Nelson JW, Schena A, Jankowski W, Kalodimos read alignment with Bowtie 2. Nat Methods
CG, Johnsson K, Stoddard BL, Baker D (2013) 9:357–359
Computational design of ligand-binding pro- 22. Araya CL, Fowler DM, Chen W, Muniez I,
teins with high affinity and selectivity. Nature Kelly JW, Fields S (2012) A fundamental pro-
501:212–216 tein property, thermodynamic stability, revealed
12. Kitzman JO, Starita LM, Lo RS, Fields S, solely from large-scale measurements of pro-
Shendure J (2015) Massively parallel single- tein function. Proc Natl Acad Sci U S A
amino- acid mutagenesis. Nat Methods 109:16858–16863
12:203–206 23. Hochberg YBY (1995) Controlling the false
13. Fowler DM, Araya CL, Fleishman SJ, Kellogg discovery rate: a practical and powerful
EH, Stephany JJ, Baker D, Fields S (2010) approach to multiple testing. J R Stat Soc
High-resolution mapping of protein sequence- Series B 57:289–300
function relationships. Nat Methods 7:741–746 24. Shendure J, Akey JM (2015) The origins,
14. Starita LM, Young DL, Islam M, Kitzman JO, determinants, and consequences of human
Gullingsrud J, Hause RJ, Fowler DM, Parvin mutations. Science 349:1478–1483
JD, Shendure J, Fields S (2015) Massively par- 25. Gnad F, Baucom A, Mukhyala K, Manning G,
allel functional analysis of BRCA1 RING Zhang Z (2013) Assessment of computational
domain variants. Genetics 200:413–422 methods for predicting the effects of missense
15. Kretz CA, Dai M, Soylemez O, Yee A, Desch mutations in human cancers. BMC Genomics
KC, Siemieniak D, Tomberg K, Kondrashov 14(Suppl 3):S7
Part V
Abstract
Matrix metalloproteinases (MMPs) represent more than 20 zinc-containing endopeptidases that cleave
internal peptide bonds, leading to protein degradation. They play a critical role in many physiological cell
functions, including tissue remodeling, embryogenesis, and angiogenesis. They are also involved in the
pathogenesis of a vast array of diseases, including but not limited to systemic inflammation, various can-
cers, and cardiovascular, neurological, and autoimmune diseases. Here, we describe gel zymography to
detect MMPs in cell and tissue samples and in cell culture supernatants.
1 Introduction
Charles A. Galea (ed.), Matrix Metalloproteases: Methods and Protocols, Methods in Molecular Biology, vol. 1579,
DOI 10.1007/978-1-4939-6863-3_12, © Springer Science+Business Media LLC 2017
231
232 Rajeev B. Tajhya et al.
2 Materials
2.2 Zymogram Gel 1. Separating gel buffer: Add 80 mL water to a 100 mL glass
cylinder. Weigh 4.63 g ammediol∙HCl and 0.02 g sodium
azide and transfer to the cylinder. Mix and adjust pH to 8.96
with HCl. Add water to 100 mL. Store at 4 °C for up to a
month.
2. Casein substrate solution (10×) (see Note 5): Add 100 mL
0.1 N NaOH to a glass beaker. Weigh 800 mg casein and
transfer to the beaker. Heat the solution to 37 °C with occa-
sional vortexing until full dissolution (see Note 6). Store at
−20 °C for up to 6 months.
Zymography of Matrix Metalloproteases 233
Table 1
Recipes for preparing zymogram separating gels with different percentages of acrylamide
2.3 Protease 1. Renaturing buffer (see Note 13): Add 50 mL of water to a 100
Detection mL graduated cylinder. Add 2.5 mL of Triton X-100 to the
cylinder (see Note 4). Add water to 100 mL and mix thor-
oughly. Do not store for more than a day.
2. Developing buffer (10×): To prepare 1 L of developing buffer,
add 100 mL water to a 1 L graduated cylinder. Weigh 12.1 g
Tris Base, 63 g Tris–HCl, 117 g NaCl, 7.4 g CaCl2 and add all
four to the cylinder. Add 2 mL Brij 35 to the cylinder (see Note
14). Add water to 1 L and mix thoroughly.
3. Coomassie Blue staining solution (see Note 15): Add 800 mL
water to a graduated cylinder. Add 50 mL methanol and 100
mL acetic acid (see Note 16). Weigh 5 g Coomassie Blue
R-250 and add to the graduated cylinder. Add water to 1 L.
4. Coomassie Blue destaining solution (see Note 15): Add 800
mL water to a 1 L beaker. Add 100 mL methanol and 50 mL
acetic acid (see Note 16). Add water to 1 L.
3 Methods
3.1 Sample 1. If the cells to be tested are adherent, plate in complete growth
Preparation media and allow time to adhere (see Note 17).
3.1.1 Preparation of Cell 2. Wash the cells three times with sterile PBS or serum-free cul-
Culture Supernatants ture media.
3. Incubate the cells at 37 °C with serum-free culture media (see
Note 18) for the optimal duration dependent on the cells (see
Note 19).
4. Collect the culture supernatants.
5. Proceed to measuring protein concentration (Subheading 3.2)
or freeze at −80 °C until use (see Note 20).
3.1.2 Preparation of Cell 1. If the cells to be tested are adherent, plate in complete growth
Lysates media and allow time to adhere (see Note 21).
2. Wash the cells three times with sterile PBS or culture media.
Zymography of Matrix Metalloproteases 235
3.1.3 Preparation 1. Place a bottle of NP-40 lysis buffer on ice for a minimum of
of Tissue Extracts— 20 min to prepare ice-cold buffers.
Technique 1: NP-40 Lysis 2. Collect the tissue and process immediately.
Buffer
3. Cut approximately 50 mg of tissue into small pieces and place
in a 1.5 mL microcentrifuge tube.
4. Add 0.5 mL of cold NP-40 lysis buffer (see Note 22).
5. Homogenize the tissue with a pestle over ice.
6. Centrifuge the homogenate at 16,000 × g for 20 min at 4 °C
in a microcentrifuge.
7. Collect the supernatant.
8. Proceed to measuring protein concentration (Subheading 3.2)
or freeze at −80 °C until use (see Note 20).
3.1.4 Preparation 1. Collect the tissue and immediately freeze with dry ice.
of Tissue Extracts— 2. Pulverize the frozen tissue with a micro-dismembrator.
Technique 2: Pulverization
3. Weigh 50 mg of pulverized tissue and resuspend in 0.15 mL
Triton X-100 lysis buffer under gentle rotation at 4 °C.
4. Centrifuge the homogenate at 16,000 × g for 20 min at 4 °C
in a microcentrifuge.
5. Collect the supernatant.
6. Proceed to measuring protein concentration (Subheading 3.2)
or freeze at −80 °C until use (see Note 20).
3.2 Measuring 1. Remove the bovine serum albumin (BSA) standard reagent
Protein Concentration (see Note 23) from 4 °C storage and let it warm to room tem-
perature (see Note 24). Invert a few times before use.
236 Rajeev B. Tajhya et al.
3.3 Gel Preparation 1. Assemble the electrophoresis plates (see Note 27).
2. Prepare the separating gel and immediately pour (see Note 28)
between the electrophoresis plates up to 2 cm from the top.
3. Overlay with water-saturated n-butanol and allow the gel to
polymerize for approximately 30 min at room temperature (see
Note 29).
4. Prepare the stacking gel (see Note 28).
5. Decant the n-butanol from the gel and gently rinse with water
(see Note 30).
6. Pour the stacking gel on the top of the separating gel.
7. Immediately insert the well comb (see Note 31).
8. Allow the stacking gel to polymerize for approximately 20 min
at room temperature.
3.4 Loading 1. Remove the well comb from the gel and rinse gently with 1×
and Electrophoresis running buffer.
2. Install the zymography gel in the electrophoresis setup.
3. Fill the inside chamber with 1× running buffer and the outside
chamber with 500 mL 1× running buffer (see Note 32).
4. Place 10 μL of 2× sample buffer on a Parafilm membrane and
add 10 μL of sample. Pipet a few times to mix and load the mix-
ture into each well using gel loading pipet tips (see Note 33).
5. Load a protein molecular weight standard in at least one well
per gel.
6. Load appropriate recombinant MMP or a control known to
contain the MMP of interest to identify the sample MMPs
(optional) (see Note 34).
Zymography of Matrix Metalloproteases 237
7. Run the gel at 125 V for 90 min or until the loading dye
reaches the bottom of the gel.
3.6 Gel Development 1. Remove the renaturing buffer by gently pouring it into a waste
container (see Note 36).
2. Add 100 mL of 1× developing buffer to the container with the
gel.
3. Incubate for 30 min at room temperature with gentle agitation
on a bench rocker.
4. Remove the developing buffer by gently pouring it into a waste
container.
5. Add 100 mL of 1× developing buffer to the container with the
gel. Close the lid tightly.
6. Incubate overnight (16–18 h) at 37 °C (see Note 37).
3.7 Gel Staining 1. Remove the developing buffer by gently pouring it into a waste
and Destaining container.
2. Add 100 mL of water to the container containing the gel.
3. Incubate for 5 min at room temperature with gentle agitation
on a bench rocker.
4. Remove the water by gently pouring it into a waste container.
5. Repeat steps 2–4 twice.
6. Scan the gel to record the exact position of the protein stan-
dard bands as they will become difficult to see after gel staining
and destaining.
3.7.1 Staining 1. Add 20 mL of Coomassie blue staining solution (see Note 15)
with Coomassie Blue to the container containing the gel.
R-250 2. Incubate for 1 h at room temperature with gentle agitation on
a bench rocker (see Note 38).
3. Remove the staining buffer by gently pouring it into a waste
container.
238 Rajeev B. Tajhya et al.
3.8 Data Acquisition 1. Remove the gel from the water, place in a plastic sheet protec-
and Analysis tor, and gently remove air bubbles.
2. Scan the gel with a digital scanner (see Note 41).
3. The integrative intensity of the MMP bands can be quantified
with ImageJ (freely available from the National Institutes of
Health) or similar software.
4 Notes
Fig. 1 Gelatin zymography gel loaded with different dilutions of horse serum (HS),
goat serum (GS), and human serum (HuS) (1%, 5%, 10%). Stronger bands of
MMP-2 (~65 kDa) and MMP-9 (~85 kDa) are visible with increasing concentra-
tions of the different sera
Zymography of Matrix Metalloproteases 241
Fig. 2 Gelatin zymography gel comparing MMP-2 and MMP-9 production in the
supernatant of untreated fibroblast-like synoviocytes isolated from a patient with
rheumatoid arthritis (RA-FLS; control) and paxilline-treated RA-FLS incubated in
the presence of 0%, 1%, 5%, or 10% of fetal bovine serum. Paxilline-treated
RA-FLS exhibit a decrease in MMP-2 production when incubated without serum.
This effect is masked by incubation with 1%, 5%, 10% serum
21. The number of cells to use for preparing cell culture superna-
tants for zymography will vary depending on the type of cells
and must be tested prior to assaying a new cell system. For
example, MMP-2 was detectable in the culture supernatants of
human fibroblast-like synoviocytes isolated from patients with
rheumatoid arthritis when cells were plated at a density of
50,000 cells per well with 0.3 mL culture medium per well in
a 24-well plate [14].
22. The volume of lysis buffer to add will depend on the type of
tissue to be homogenized as protein extractability varies with
specimens.
23. While BSA is commonly used, any known protein standard
can be used as a control.
24. Cold solutions can condense atmospheric moisture on the
outside of the cuvette and scatter light giving erroneous
readings.
25. Absorbance of protein-dye conjugate increases over time and
affects the accuracy of protein concentration measurements.
26. If cell supernatants or tissue extracts contain low levels of gela-
tinases, a phase extraction with Triton X-114 is recommended
[19].
27. The electrophoresis plates must be completely clean and dry at
this time or the gel could incompletely polymerize or polym-
erize in a heterogeneous manner.
28. Once the polymerization agent (TEMED) is added to the gel,
immediately mix and pour as the gel will start polymerizing
rapidly. If the gel polymerizes too fast, reduce the ambient
temperature as this will delay polymerization.
29. Ensure that the electrophoresis system is perfectly horizontal
during gel pouring and polymerization.
242 Rajeev B. Tajhya et al.
Acknowledgment
References
PS, Beeton C (2015) KCa1.1 inhibition atten- 18. Puente J, Jaque M, Carrasco C, Cruz C,
uates fibroblast-like synoviocyte invasiveness Valenzuela M, Wolf M, Mosnaim A (2008)
and ameliorates rat models of rheumatoid Triptan drugs, natural killer cell cytotoxicity,
arthritis. Arthritis Rheumatol 67:96–106 and neutrophils pro-matrix metalloproteinase-
17. Pan H, Chen J, Xu J, Chen M, Ma R (2009) 9 secretion. Headache 48:1482–1489
Antifibrotic effect by activation of peroxisome 19. Toth M, Sohail A, Fridman R (2012) Assessment
proliferator-activated receptor-gamma in cor- of gelatinases (MMP-2 and MMP-9) by gelatin
neal fibroblasts. Mol Vis 15:2279–2286 zymography. Methods Mol Biol 878:121–135
Chapter 13
Abstract
The use of fluorescent probes in conjunction with white-light colonoscopy is a promising strategy for
improving the detection of precancerous colorectal lesions, in particular flat (sessile) lesions that do not
protrude into the lumen of the colon. We describe a method for determining the sensitivity and specificity
of an enzymatically activated near-infrared probe (MMPSense680) for the detection of colon lesions in a
mouse model (APC+/Min-FCCC) of spontaneous colorectal cancer. Fluorescence intensity correlates directly
with the activity of matrix metalloproteinases (MMPs). Overexpression of MMPs is an early event in the
development of colorectal lesions. Although the probe employed serves as a reporter of the activity of
MMPs, our method can be applied to any fluorescent probe that targets an early molecular event in the
development of colorectal tumors.
Key words Colorectal cancer, Mice, Min mice, Imaging, Fluorescence imaging, Matrix metallopro-
teinases, Early cancer detection, Colonoscopy, Activated imaging probes, Near-infrared imaging
1 Introduction
Charles A. Galea (ed.), Matrix Metalloproteases: Methods and Protocols, Methods in Molecular Biology, vol. 1579,
DOI 10.1007/978-1-4939-6863-3_13, © Springer Science+Business Media LLC 2017
245
246 Harvey Hensley et al.
2 Materials
2.3 Probes Although this review focuses on our work using MMPsense680 as
probe, the procedures described can be used to study any fluores-
cent probe in the mouse colon, or any similar organ in small
animals.
We have imaged a variety of probes with fluorophores in the
red or near-infrared spectrum including MMPSense680,
ProSense680, ProSense750, IntegriSense680, and IntegriSense750
(PerkinElmer, Hopkinton, MA), as well as PSVUE794 (Molecular
Targeted Technologies, Malvern, PA) using the following
methodology.
2.4 IVIS Images are captured on an IVIS Spectrum (PerkinElmer). This sys-
Imaging System tem consists of a highly sensitive, cooled (−80 °C) CCD camera,
and a versatile filter set for imaging fluorophores. The system also
contains a set of LEDs for white light illumination, permitting the
acquisition of both fluorescent and conventional (black and white)
images, and displays the fluorescence intensity as an overlay on the
white light image. Exposure time, binning, fstop, light illumina-
tion level, imaging field of view, focal point, and excitation and
emission filters can be adjusted to optimize image acquisition.
2.5 Image IVIS images are processed with routines written in the Matlab
Processing (Mathworks) programming environment with the Image
Processing Toolkit. A large number of free and commercial image
processing software programs are available. However, customized
routines are necessary to implement a convenient method for pre-
cisely pairing each fluorescent image with a corresponding paraffin
embedded section.
3 Methods
3.1 Injection To maximize the use of the probe and ensure the ethical treatment
of MMP Probe of animals, endoscopy, MRI, CT, or other noninvasive imaging
methods should be employed prior to injection to confirm the
tumor-bearing status of the experimental animals. If at least one
colon lesion has been identified, inject the mouse intravenously
(via either the tail vein or retro-orbital sinus) with an appropriate
amount of the fluorescent imaging agent (~2 nM for MMPSense680)
in 100–150 μl of isotonic PBS (see Note 1). The inoculation time
in vivo will differ depending upon the pharmacokinetic properties
(plasma half-life) of the probe to be imaged. Time course analyses
revealed 66 h post-injection as the optimal time to image
MMPSense680 ex vivo in C57BL/6J Apc+/Min-FCCC mice. At the
predetermined time point, euthanize the mice according to the
American Veterinary Medical Association Guidelines for Euthanasia
(slow inhalation of CO2, followed by confirmation of death).
Imaging MMPs in Colon Tumors 249
3.3 Ex Vivo Imaging 1. Place the transparent plastic sheet containing the excised colon
in the imaging system on a black plastic mat (see Note 4).
Image settings for our work [4] employing the IVIS Spectrum
250 Harvey Hensley et al.
3.4.2 Identification 1. Using the locations marked on the hard copy printout as a
of Tissue Sections guide (see step 4 of Subheading 3.4.1), identify the position of
with Fluorescence Image each 2 mm segment of the colon on both white light and
Features and Image fluorescence/white light image overlays as displayed on a com-
Display puter screen.
2. Mark a rectangular region of interest on the images as dis-
played on the computer with an image processing software
program.
3. Since up to 60 separate regions of interest (ROIs) must be
placed on the images to cover the entire colon, we have found
it convenient to write routines to help do this using the
MATLAB Imaging Processing Toolbox (MathWorks, Inc.,
Natick, MA). 30–60 ROIs (2 mm in width; the number
depending on the length of the particular colon) are placed on
Imaging MMPs in Colon Tumors 251
the image (Fig. 1). The ROI positions are then adjusted indi-
vidually according to the precise position of each segment on
the ruler, which were recorded when the colons were cut into
segments for embedding into paraffin.
4. Display the fluorescence images as overlays on the white light
images, with the white light intensities set to a grayscale colort-
able, while the fluorescence image is set to a different color
lookup table. We employed a “hot colortable” for fluorescence
intensities (Fig. 1), with the most intense signals correspond-
ing to a display color of white. The least intense is set to a mini-
mum indicative of the signal background level in the colon and
presented as dark red, while the intermediate values display as
a range of shades of red to yellow (Fig. 1). Images were
acquired with a 16 bit CCD camera, producing a total of
65,536 intensity levels. In order to take advantage of this large
252 Harvey Hensley et al.
3.4.3 Use Use of a subset of the data as a training set is essential to: (1) develop
of a Training Set experience identifying lesions; (2) establish rules and guidelines for
scoring images; and (3) identify features on fluorescence images
that correspond to histologically confirmed adenomas.
1. Set aside 10 of the annotated fluorescence/white light images
to be used as a training set.
2. Identify regions of high fluorescence intensity relative to the
local background, and compare these regions with results from
H&E stained sections as reviewed and annotated by a trained
pathologist (Fig. 2).
3. Conversely, determine which features on the fluorescence
images correspond to the presence of a pathologically con-
firmed lesion.
4. Identify which areas have pathologically confirmed lesions and
determine whether or not there is an identifiable fluorescence
signal on the image in that location.
Fig. 2 Micrographs of colorectal lesions in an Apc+/Min-FCCC mouse. (a) 40× view of a peduncular adenoma that
extends above the surface of the normal mucosa compared with a nonpolypoid lesion (b). (b) 40× view of a
sessile adenoma (blue arrow) with a height less than twice that of the adjacent normal mucosa (yellow arrow)
Imaging MMPs in Colon Tumors 253
Fig. 3 Immunohistochemical staining of MMP-7 and -9 in colorectal lesions of Apc+/Min-FCCC mice. (a) 400×
view of an adenoma stained with anti-MMP-7 antibodies. MMP-7 is strongly and diffusely expressed in the
adenomatous epithelium. (b) 400× view of an adenoma stained with anti-MMP-9 antibodies. MMP-9 is
strongly expressed in the polymorphonuclear leukocytes (PMNLs) in the stroma/lamina propria of the adenoma
but not in the epithelial cells
4 Notes
2. When laying the colon flat on the plastic ruler, be sure to keep
track of the proximal and distal ends. This can be done by not-
ing the distinct three-dimensional architecture (V-shaped) of
the proximal colonic mucosa, and placing the colon so it is
always oriented in the same direction. Always lay the colon
with the luminal surface facing up, with no folds or twists.
3. Use a plain black colored marker when recording the animal
number on the plastic sheets, as the dyes in some colored
markers may have some fluorescent signal.
4. The black plastic mat is usually supplied by the manufacturer of
the imaging system.
5. The focal distance on the IVIS Spectrum is set to a default of
1.5 cm for in vivo imaging. When making high spatial resolu-
tion images with a low f-number setting in excised organs, this
setting must be decreased to approximately 1.5 mm and
adjusted carefully.
6. Imaging times (from dissection to image acquisition) should
not exceed 5 min to prevent dehydration.
7. The ruler markings photocopied onto the transparent sheet
can be used as a guide for cutting the colon.
8. To increase the dynamic range of the images, an alternative to
displaying multiple images with different contrast settings
would be to employ a nonlinear mapping (e.g., logarithmic) of
intensity to color. In practice, we find it preferable to examine
multiple representations of the same data with a linear map-
ping of intensity to color.
Acknowledgments
References
1. Cornett D, Barancin C, Roeder B et al (2008) 3. Burggraaf J, Kamerling IM, Gordon PB et al
Findings on optical colonoscopy after positive (2015) Detection of colorectal polyps in
CT colonography exam. Am J Gastroenterol humans using an intravenously administered
103:2068–2074 fluorescent peptide targeted against c-Met. Nat
2. Heresbach D, Barrioz T, Lapalus MG et al Med 21:955–961
(2008) Miss rate for colorectal neoplastic pol- 4. Clapper ML, Hensley HH, Chang WC et al
yps: a prospective multicenter study of back-to- (2011) Detection of colorectal adenomas using
back video colonoscopies. Endoscopy a bioactivatable probe specific for matrix metal-
40:284–290 loproteinase activity. Neoplasia 13:685–691
Imaging MMPs in Colon Tumors 255
5. Cooper HS, Chang WC, Coudry R et al cancer: is it worth talking about? Cancer
(2005) Generation of a unique strain of mul- Metastasis Rev 23:119–135
tiple intestinal neoplasia (Apc(+/Min-FCCC)) 8. Zucker S, Vacirca J (2004) Role of matrix
mice with significantly increased numbers of metalloproteinases (MMPs) in colorectal can-
colorectal adenomas. Mol Carcinog 44: cer. Cancer Metastasis Rev 23:101–117
31–41 9. Newell KJ, Witty JP, Rodgers WH et al (1994)
6. Weissleder R, Ntziachristos V (2003) Shedding Expression and localization of matrix-
light onto live molecular targets. Nat Med degrading metalloproteinases during colorectal
9:123–128 tumorigenesis. Mol Carcinog 10:199–206
7. Wagenaar-Miller RA, Gorden L, Matrisian LM 10. Kinzler KW, Vogelstein B (1996) Lessons from
(2004) Matrix metalloproteinases in colorectal hereditary colorectal cancer. Cell 87:159–170
Part VI
Abstract
Structure-based virtual screening (SBVS) is a common method for the fast identification of hit structures
at the beginning of a medicinal chemistry program in drug discovery. The SBVS, described in this manu-
script, is focused on finding small molecule hits that can be further utilized as a starting point for the
development of inhibitors of matrix metalloproteinase 13 (MMP-13) via structure-based molecular design.
We intended to identify a set of structurally diverse hits, which occupy all subsites (S1′–S3′, S2, and S3)
centering the zinc containing binding site of MMP-13, by the virtual screening of a chemical library com-
prising more than ten million commercially available compounds. In total, 23 compounds were found as
potential MMP-13 inhibitors using Glide docking followed by the analysis of the structural interaction
fingerprints (SIFt) of the docked structures.
Key words Matrix metalloproteinase, Structure-based virtual screening, Docking, Structural interac-
tion fingerprints, Zn-chelating inhibitor
1 Introduction
Charles A. Galea (ed.), Matrix Metalloproteases: Methods and Protocols, Methods in Molecular Biology, vol. 1579,
DOI 10.1007/978-1-4939-6863-3_14, © Springer Science+Business Media LLC 2017
259
260 Jun Yong Choi and Rita Fuerst
1.1 Matrix MMP activity was described for the first time in 1962 as an enzy-
Metalloproteinases matic player in the metamorphosis process of tadpoles. Since then,
as Drug Targets 24 structurally and functionally related MMPs have been found in
mammals. MMPs are characterized by a Zn2+-cation in their
enzyme active site, which is coordinated by three histidines fol-
lowed by a conserved methionine residue. Under healthy condi-
tions, MMPs are very important regulators of cellular activities and
physiological processes including reproduction, tissue remodeling,
embryogenesis, and angiogenesis via the degradation of extracel-
lular matrix (ECM) components. MMPs can also influence the
immune system by directly cleaving signaling molecules including
the tumor necrosis factor (TNF) and other cytokines. MMP activ-
ity is very low under healthy conditions but can be detected during
repair or remodeling processes including angiogenesis, bone devel-
opment, or wound healing, as well as in inflamed tissue [7].
Originally, MMPs attracted the attention of numerous research
groups due to their involvement in tumor angiogenesis and metas-
tasis. First-generation MMP inhibitors, developed in the early
1990s, were generally not selective for any particular MMP. Drug
candidates resultant from these early programs were designed to
mimic peptide sequences of collagen. These peptidomimetics also
consisted of a functional group capable of chelating the active site
zinc within the MMP active site in a competitive manner. Zinc che-
lating functionalities include the carboxylate-, carbamate-, thiol-,
and hydroxamate group as well as phosphoric acid derivatives. The
Virtual High-Throughput Screening for Matrix Metalloproteinase Inhibitors 261
1.2 MMP-13 in Drug During the past 15 years, several highly potent and selective non-
Discovery zinc chelating inhibitors targeting the S1′/S1′* subsite of MMP-
13 were developed and several have entered clinical trials for the
treatment of osteoarthritis. In the early 2000s, the first highly
selective non-zinc chelating MMP-13 inhibitors, 3 and 4 (Fig. 2),
were identified as part of a HTS campaign followed by an extensive
structure activity relationship (SAR) study [12]. X-ray co-crystal
structures revealed that the dynamic movement of the S1′ pocket
O O
H
HO N CH3 O O
N N H
H H HO N CH3
O N N
S H H
OH O
S
1, IC50 2, IC50
MMP-1 = 3 nM MMP-1 = 5 nM
MMP-2 = 4 nM MMP-2 = 6 nM
MMP-3 = 20 nM MMP-3 = 230 nM
MMP-7 = 6 nM MMP-7 = 16 nM
MMP-9 = 4 nM MMP-9 = 3 nM
O
Ph
O
N
O N N OH
N O
-
O S O CH3 O
3, IC50 4, IC50
MMP-13 = 30 nM MMP-13 = 0.67 nM
MMP-1, -2, -3, -7, -9, -12, -14 MMP-1, -2, -3, -7, -9, -12, -14
>100,000 nM >30,000 nM
COOH
H O O
CH3
O N O O
N N O
H H HN
N N H
O N OH N S
MeO N
O
5, IC50 6, IC50
MMP-13 = 0.03 nM MMP-13 = 3.9 pm
MMP-1, -2, -3, -7, -8, -9, -12, -14 > 20,000 nM MMP-8 = 720 nM
MMP-10 = 160 nM
MMP-1, -2, -3, -7, -9, -14 > 4000 nM
2 Materials
2.1 Software Two different programs were used in the SBVS focused on finding
new inhibitors for MMP-13. The program Glide from the
Schrödinger Small Molecule Drug Discovery Suite [17] was uti-
lized for ligand docking and the program MOE developed by
Chemical Computing Group [18] was used for chemical library
composition and ligand-based search. We chose these programs
due to their availability in our research laboratory and their popu-
larity within the research community reflected by a myriad of cited
references. The use of Glide and MOE requires the purchase of
their commercial licenses from Schrödinger Inc. and Chemical
Computing Group, respectively. Alternatively, other docking pro-
grams are freely available such as AutoDock [19], UCSF-DOCK
[20], GOLD [21], and SwissDock [22], which have been devel-
oped and maintained by academic institutions.
2.2 Database The virtual chemical library collection was obtained from the
ZINC12 database [23, 24], which comprises around ten million
commercially available compounds filtered for drug-like physical
properties [25]: 150 ≤ molecular weight ≤ 500, xlogP ≤ 5, num-
ber of rotatable bonds ≤ 7, polar surface area < 150 Å2, number of
hydrogen bond donors ≤ 5, and number of hydrogen bond accep-
tors ≤ 10. The structures were downloaded in an SDF file format
264 Jun Yong Choi and Rita Fuerst
3 Methods
3.1 Workflow The virtual screening comprised five steps as outlined below (Fig. 3):
1. Ligand-based search.
2. Analysis of X-ray structures and preparation of a target
structure.
3. Grid generation.
4. Glide docking.
5. Data analysis.
3.2 Ligand- Initially, all chemical structures possessing one or more carboxylic
Based Search acid functionalities, which can chelate the catalytically active Zn-ion
of MMPs, were selected from the MOE database collection gener-
ated in Subheading 2.2. In the Select panel of the MOE database,
Ligand-based Searching
(ca. 1.3 million hits)
Glide SP docking
(604 docked structures)
Hit Selection
3.3 Analysis Structure coordinates were downloaded for all MMP isozymes
of X-Ray Structures found in the Protein Data Bank (PDB). The sequence alignment
and Preparation for these MMP X-ray crystal structures showed about 50% sequence
of the Target Structure identity between each isozyme. Superimposition of the MMP
structures, based on the sequence alignment, also showed that the
catalytic domain of different isozymes is highly conserved, and the
binding sites of co-crystallized Zn-chelating inhibitors are located
around the metal ion and extended toward all subsites (S1′–S3′,
S2, and S3). Furthermore, the chelating moieties of inhibitors that
interact with the Zn-ion are located around the metal ion at a dis-
tance of approximately 2.1 Å (Fig. 4).
The X-ray co-crystal structure of MMP-13 (PDB ID: 3ELM)
[26] was used as the target for the SBVS within this study. First,
the protein structure was prepared for docking by refining the tar-
get protein structure. This step included: assigning bond orders,
adding hydrogen atoms, creating zero-order bonds to metals, cre-
ating disulfide bonds, filling in missing side chains and loops, and
deleting water molecules beyond 5 Å from hetero groups. Chain A
of the target protein was used for further refinement that com-
prised assigning hydrogen bond interactions using the PROPKA
module (pH = 7.4). Finally, the structure was subjected for
restrained minimization using the OPLS3 force field.
Fig. 4 Superimposed X-ray co-crystal structures of MMP isozymes (a) and Zn-chelating inhibitors (b). Substrate
mimetic (decapeptide) is represented as a surface (PDB ID: 3AYU) (a). Each subsite in the substrate-binding
cleft is annotated (a) [29]
266 Jun Yong Choi and Rita Fuerst
3.4 Receptor Grid A receptor grid was generated for the ligand-binding site of the
Generation target protein. The grid was set to a 25 Å3 box centered on the
Zn-chelating reference ligand of the MMP-13 co-crystal structure
(3ELM) and the metal coordination to the zinc ion was assigned
as a constraint.
3.5 Glide Docking The Virtual Screening Workflow module was used for Glide dock-
ing. The compound collection, containing at least one carboxylic
acid moiety from the ligand-based search described in Subheading
3.2, was directly used without further ligand filtering or prepara-
tion. Ligand docking was restricted by the comparison of core pat-
terns, which required the carboxylic acid unit of docked ligands to
align with that of the reference compound with a tolerance of
0.7 Å. Initially, the ligand structures were sequentially docked into
the target grid in the high-throughput virtual screening (HTVS)
mode, which reduced about 1.3 million carboxylic acid-containing
compounds to 1325 docked structures. This process was followed
by Glide standard precision (SP) mode docking of the remaining
1325 structures, which generated 604 ligands that occupied the
zinc-binding site of MMP-13. All 604 docked structures were
scored and saved for data analysis and hit triage (see Note 2).
3.6 Data Analysis The 604 docked structures occupied the entire area surrounding
and Hit Triage the Zn-ion. Since the ligand interactions with the subsites S1′–S3′,
S2, and S3 are known to improve selectivity within the MMP fam-
ily, the goal of this virtual screen was to identify diverse sets of
Zn-chelating molecules that occupied these regions centered
around the Zn-containing active site of MMP-13. Docking post-
processing procedures were applied in Maestro to calculate struc-
tural interaction fingerprints (SIFt) to analyze intermolecular
binding interactions between the protein and the docked struc-
tures. After the analysis of the SIFt matrix, a number of structurally
diverse compounds, which show interaction with the five subsites,
were selected as potential MMP-13 inhibitors [27, 28].
The computed matrix map shows the clustering of SIFts cor-
related to the docking score of the screened compounds (Fig. 5).
Various positions on the map represent ligand occupancy of differ-
ent binding sites. Ligand binding poses in each cluster of SIFts
were visually inspected and potential hits were selected using the
following selection criteria: the Glide docking score, the conforma-
tional stability of the bound inhibitor, ligand rigidity, and unique-
ness. Furthermore, compounds were triaged by comparing their
binding poses to currently known inhibitors exhibiting interactions
considered to be important for potency and selectivity for MMP-
13, such as a π-stacking interaction with His222 in the S1′ site,
hydrophobic contacts with Tyr176, His187, and Phe189 in the S3
site, and a hydrogen bond interaction with the protein backbone in
the S2′ site [26, 29–31].
Virtual High-Throughput Screening for Matrix Metalloproteinase Inhibitors 267
Fig. 6 Representative hits and their binding poses in MMP-13. (a) ZINC62430943, (b) ZINC82305218, (c)
ZINC41498653, and (d) ZINC57989822 are shown in green sticks. Amino acids in the active site of MMP-13
are shown in gray sticks, and protein structures are shown in a gray ribbon diagram. Figures were generated
using the program PyMol [31]
Virtual High-Throughput Screening for Matrix Metalloproteinase Inhibitors 269
4 Notes
References
1. Cheng T, Li Q, Zhou Z, Wang Y, Bryant SH 3. Ghosh S, Nie A, An J, Huang Z (2006)
(2012) Structure-based virtual screening for Structure-based virtual screening of chemical
drug discovery: a problem- centric review. libraries for drug discovery. Curr Opin Chem
AAPS J 14(1):133–141 Biol 10(3):194–202
2. Ripphausen P, Nisius B, Peltason L, Bajorath J 4. Takaishi H, Kimura T, Dalal S, Okada Y,
(2010) Quo vadis, virtual screening? A compre- D’Armiento J (2008) Joint diseases and matrix
hensive survey of prospective applications. J Med metalloproteinases: a role for MMP-13. Curr
Chem 53(24):8461–8467 Pharm Biotechnol 9(1):47–54
270 Jun Yong Choi and Rita Fuerst
5. Zigrino P, Kuhn I, Bauerle T, Zamek J, Fox Taveras AG, Baragi VM (2011) Discovery and
JW, Neumann S, Licht A, Schorpp-Kistner evaluation of a non-Zn chelating, selective
M, Angel P, Mauch C (2009) Stromal expres- Matrix Metalloproteinase 13 (MMP-13) inhib-
sion of MMP-13 is required for melanoma itor for potential intra-articular treatment of
invasion and metastasis. J Invest Dermatol osteoarthritis. J Med Chem 55(2):709–716
129(11):2686–2693 16. Nara H, Sato K, Naito T, Mototani H, Oki H,
6. Morrison C, Mancini S, Cipollone J, Yamamoto Y, Kuno H, Santou T, Kanzaki N,
Kappelhoff R, Roskelley C, Overall C (2011) Terauchi J, Uchikawa O, Kori M (2014)
Microarray and proteomic analysis of breast Discovery of novel, highly potent, and selective
cancer cell and osteoblast co-cultures: role of quinazoline-2-carboxamide-based Matrix
osteoblast Matrix Metalloproteinase (MMP)- Metalloproteinase (MMP)-13 inhibitors with-
13 in bone metastasis. J Biol Chem 286(39): out a zinc binding group using a structure-
34271–34285 based design approach. J Med Chem
7. Vandenbroucke RE, Libert C (2014) Is there 57(21):8886–8902
new hope for therapeutic matrix metallopro- 17. Friesner RA, Banks JL, Murphy RB, Halgren
teinase inhibition? Nat Rev Drug Discov TA, Klicic JJ, Mainz DT, Repasky MP, Knoll
13(12):904–927 EH, Shelley M, Perry JK, Shaw DE, Francis P,
8. Rothenberg ML, Nelson AR, Hande KR (1999) Shenkin PS (2004) Glide: a new approach for
New drugs on the horizon: matrix metallopro- rapid, accurate docking and scoring: 1. Method
teinase inhibitors. Stem Cells 17(4):237–240 and assessment of docking accuracy. J Med
9. Engel CK, Pirard B, Schimanski S, Kirsch R, Chem 47(7):1739–1749
Habermann J, Klingler O, Schlotte V, 18. Molecular Operating Environment (MOE),
Weithmann KU, Wendt KU (2005) Structural 2013.08 (2016). Chemical Computing Group
basis for the highly selective inhibition of Inc., 1010 Sherbooke St. West, Suite #910,
MMP-13. Chem Biol 12(2):181–189 Montreal, QC, Canada H3A 2R7
10. Li JJ, Johnson AR (2011) Selective MMP13 19. Morris GM, Huey R, Lindstrom W, Sanner
inhibitors. Med Res Rev 31(6):863–894 MF, Belew RK, Goodsell DS, Olson AJ (2009)
11. Dormán G, Cseh S, Hajdu I, Barna L, Konya AutoDock4 and AutoDockTools4: automated
D, Kupai K, Kovacs L, Ferdinandy P (2010) docking with selective receptor flexibility.
Matrix metalloproteinase inhibitors: A critical J Comput Chem 30(16):2785–2791
appraisal of design principles and proposed 20. Allen WJ, Balius TE, Mukherjee S, Brozell SR,
therapeutic utility. Drugs 70(8):949–964 Moustakas DT, Lang PT, Case DA, Kuntz ID,
12. Johnson AR, Pavlovsky AG, Ortwine DF, Prior Rizzo RC (2015) DOCK 6: impact of new fea-
F, Man C-F, Bornemeier DA, Banotai CA, tures and current docking performance.
Mueller WT, McConnell P, Yan C, Baragi V, J Comput Chem 36(15):1132–1156
Lesch C, Roark WH, Wilson M, Datta K, 21. Jones G, Willett P, Glen RC, Leach AR, Taylor
Guzman R, Han H-K, Dyer RD (2007) R (1997) Development and validation of a
Discovery and characterization of a novel genetic algorithm for flexible docking. J Mol
inhibitor of matrix metalloprotease- 13 that Biol 267(3):727–748
reduces cartilage damage in vivo without joint 22. Grosdidier A, Zoete V, Michielin O (2011)
fibroplasia side effects. J Biol Chem SwissDock, a protein-small molecule docking
282(38):27781–27791 web service based on EADock DSS. Nucleic
13. Li JJ, Nahra J, Johnson AR, Bunker A, O’Brien Acids Res 39(Web Server issue):W270–W277
P, Yue W-S, Ortwine DF, Man C-F, Baragi V, 23. Irwin JJ, Sterling T, Mysinger MM, Bolstad
Kilgore K, Dyer RD, Han H-K (2008) ES, Coleman RG (2012) ZINC: a free tool to
Quinazolinones and pyrido[3,4-d]pyrimidin- discover chemistry for biology. J Chem Inf
4-ones as orally active and specific matrix Model 52(7):1757–1768
metalloproteinase-13 inhibitors for the treat- 24. Irwin JJ, Shoichet BK (2005) ZINC—a free
ment of osteoarthritis. J Med Chem database of commercially available compounds
51(4):835–841 for virtual screening. J Chem Inf Model
14. Cai H, Agrawai AK, Putt DA, Hashim M, 45(1):177–182
Reddy A, Brodfuehrer J, Surendran N, Lash 25. Lipinski CA (2000) Drug-like properties and
LH (2009) Assessment of the renal toxicity of the causes of poor solubility and poor permea-
novel anti-inflammatory compounds using bility. J Pharmacol Toxicol Methods
cynomolgus monkey and human kidney cells. 44(1):235–249
Toxicology 258(1):56–63 26. Monovich LG, Tommasi RA, Fujimoto RA,
15. Gege C, Bao B, Bluhm H, Boer J, Gallagher Blancuzzi V, Clark K, Cornell WD, Doti R,
BM, Korniski B, Powers TS, Steeneck C, Doughty J, Fang J, Farley D, Fitt J, Ganu V,
Virtual High-Throughput Screening for Matrix Metalloproteinase Inhibitors 271
Goldberg R, Goldstein R, Lavoie S, Kulathila 30. Gall AL, Ruff M, Kannan R, Cuniasse P, Yiotakis
R, Macchia W, Parker DT, Melton R, O’Byrne A, Dive V, Rio MC, Basset P, Moras D (2001)
E, Pastor G, Pellas T, Quadros E, Reel N, Crystal structure of the stromelysin-3 (MMP-11)
Roland DM, Sakane Y, Singh H, Skiles J, catalytic domain complexed with a phosphinic
Somers J, Toscano K, Wigg A, Zhou S, Zhu L, inhibitor mimicking the transition-state. J Mol
Shieh W-C, Xue S, McQuire LW (2009) Biol 307(2):577–586
Discovery of potent, selective, and orally active 31. Browner MF, Smith WW, Castelhano AL
carboxylic acid based inhibitors of matrix (1995) Matrilysin-inhibitor complexes: com-
metalloproteinase-13. J Med Chem mon themes among metalloproteases.
52(11):3523–3538 Biochemistry 34(20):6602–6610
27. Singh J, Deng Z, Narale G, Chuaqui C (2006) 32. Schrödinger Release 2016-2: LigPrep, version
Structural interaction fingerprints: a new 3.8 (2016) Schrödinger, LLC, New York, NY
approach to organizing, mining, analyzing, 33. Sastry M, Lowrie JF, Dixon SL, Sherman W
and designing protein-small molecule com- (2010) Large-scale systematic analysis of 2D
plexes. Chem Biol Drug Des 67(1):5–12 fingerprint methods and parameters to improve
28. Deng Z, Chuaqui C, Singh J (2004) Structural virtual screening enrichments. J Chem Inf
interaction fingerprint (SIFt): a novel method Model 50(5):771–784
for analyzing three-dimensional protein-ligand 34. Berthold MR, Cebron N, Dill F, Gabriel TR,
binding interactions. J Med Chem Kötter T, Meinl T, Ohl P, Sieb C, Thiel K,
47(2):337–344 Wiswedel B (2007) Knime: the Konstanz
29. Hashimoto H, Takeuchi T, Komatsu K, information miner. In: Studies in classification,
Miyazaki K, Sato M, Higashi S (2011) data analysis, and knowledge organization.
Structural basis for matrix metalloprotein- Springer, New York
ase-2 (MMP-2)-selective inhibitory action of 35. Baell J, Walters MA (2014) Chemistry: chemi-
beta-amyloid precursor protein-derived inhib- cal con artists foil drug discovery. Nature
itor. J Biol Chem 286(38):33236–33243 513(7519):481–483
Chapter 15
Abstract
Matrix metalloproteinases (MMPs) are a family of zinc-containing enzymes required for homeostasis.
These enzymes are an important class of drug targets as their over expression is associated with many dis-
ease states. Most of the inhibitors reported against this class of proteins have failed in clinical trials due to
lack of specificity. In order to assist in drug design endeavors for MMP targets, a computationally tractable
pathway is presented, comprising, (1) docking of small molecule inhibitors against the target MMPs, (2)
derivation of quantum mechanical charges on the zinc ion in the active site and the amino acids coordinat-
ing with zinc including the inhibitor molecule, (3) molecular dynamics simulations on the docked ligand–
MMP complexes, and (4) evaluation of binding affinities of the ligand–MMP complexes via an accurate
scoring function for zinc containing metalloprotein–ligand complexes. The above pathway was applied to
study the interaction of the inhibitor Batimastat with MMPs, which resulted in a high correlation between
the predicted and experimental binding free energies, suggesting the potential applicability of the
pathway.
Key words Matrix metalloprotease, Computer-aided drug design, Docking and scoring, Molecular
dynamics simulations
1 Introduction
Charles A. Galea (ed.), Matrix Metalloproteases: Methods and Protocols, Methods in Molecular Biology, vol. 1579,
DOI 10.1007/978-1-4939-6863-3_15, © Springer Science+Business Media LLC 2017
273
274 Tanya Singh et al.
2 Methods
Table 1
Experimental IC50 values in nM reported for the binding of Batimastat
with MMP-1, MMP-2, MMP-3, MMP-8, MMP-9, and MMP-13 [1]
Fig. 2 Computational pathway showing the steps followed in docking and scoring study of Batimastat binding
to different MMPs
2.2 Molecular The target protein-ligand complex and the inhibitor molecule
Docking and Scoring are provided as input to Sanjeevini software suite for docking
and scoring studies. The docking module [76, 77] of Sanjeevini
docks the ligand molecule to the binding site and generates sev-
eral (i.e., 103) configurations via a six-dimensional rigid body
Monte Carlo methodology resulting in many ligand configura-
tions that are scored based on the scoring function built into
278 Tanya Singh et al.
2.5 Post facto Structures at equal intervals over the last 40 ns of the molecular
Analyses of Molecular dynamics simulation run trajectory are extracted (i.e., approxi-
Dynamics Simulations mately 100 structures in total) for each system (see Note 3). For
each structure the binding free energy is estimated using the
Bappl-Z scoring function [28]. For these calculations, the system
is parameterized within the additivity approximation where the net
free energy change is treated as a sum of contributions from indi-
vidual energy components. The equation for the estimation of the
free energy change upon binding is:
22
DG ° = a Eel + b Evdw + å s AD ALSA + l ( DSCR ) + d (1)
( A =1)
The Bappl-Z scoring function used for calculating binding free
energy estimates has been thoroughly validated earlier [28]. The
scoring function captures the theoretical rigor of the MMGBSA/
MMPBSA [91–97] methodologies as well as the rapidity of empiri-
cal/knowledge-based methods [28, 78, 79]. The individual terms
in Eq. 1 are described below.
Eel: Electrostatic interactions including interactions between
protein and ligand atoms and the zinc ion with rest of the complex.
These electrostatic interactions are calculated based on Coulomb’s
law with a sigmoidal dielectric function for solvent screening effects
[28]. To model the electrostatic interactions of zinc with the rest
of the complex, we have adopted the nonbonded model described
by Stote and Karplus [85].
Evdw: Direct van der Waals interactions between protein and
ligand atoms and the zinc ion with the rest of the complex. Van der
Waals interactions are modeled using the Lennard-Jones potential
[28] while interactions with the zinc ion are modeled on the lines
of Stote and Karplus [85].
280 Tanya Singh et al.
3 Notes
Acknowledgments
References
1. Skiles JW, Gonnella NC, Jeng AY (2004) The 5. Hu J, Van den Steen PE, Sang QX, Opdenakker
design, structure, and clinical update of small G (2007) Matrix metalloproteinase inhibitors
molecular weight matrix metalloproteinase as therapy for inflammatory and vascular dis-
inhibitors. Curr Med Chem 11:2911–2977 eases. Nat Rev Drug Discov 6:480
2. Cawston T (1998) Matrix metalloproteinases 6. Ray JM, Stetler-Stevenson WG (1995)
and TIMPs: properties and implications for Gelatinase A activity directly modulates mela-
the rheumatic diseases. Mol Med Today noma cell adhesion and spreading. EMBO
4:130–137 J 14:908–917
3. Blavier L, Henriet P, Imren S, Declerck YA 7. Chambers AF, Matrisian LM (1997) Changing
(1999) Tissue inhibitors of matrix metallopro- views of the role of matrix metalloproteinases
teinases in cancer. Ann N Y Acad Sci 878: in metastasis. J Natl Cancer Inst 89:
108–119 1260–1270
4. Chang C, Werb Z (2001) The many faces of 8. Kahari VM, Saarialho-Kere U (1993) Matrix
metalloproteases: cell growth, invasion, angio- metalloproteinases and their inhibitors in
genesis and metastasis. Trends Cell Biol tumour growth and invasion. Ann Med
11:S37–S43 31:34–45
282 Tanya Singh et al.
34. Onisto M, Riccio MP, Scannapieco P, Caenazzo 45. Shubayev VI, Angert M, Dolkas J, Campana
C, Griggio L, Spina M, Stetler-Stevenson WG, WM, Palenscar K, Myers RR (2006) TNF
Garbisa S (1995) Gelatinase A/TIMP-2 imbal- alpha-induced MMP-9 promotes macrophage
ance in lymph-node-positive breast carcino- recruitment into injured peripheral nerve. Mol
mas, as measured by RT-PCR. Int J Cancer Cell Neurosci 31:407–415
63:621–626 46. Chattopadhyay S, Myers RR, Janes J, Shubayev
35. Engel G, Heselmeyer K, Auer G, Bäckdahl M, V (2007) Cytokine regulation of MMP-9 in
Eriksson E, Linder S (1994) Correlation peripheral glia: implications for pathological
between stromelysin-3 mRNA level and out- processes and pain in injured nerve. Brain
come of human breast cancer. Int J Cancer Behav Immun 21:561–568
58:830–835 47. Liu LY, Zheng H, Xiao HL, She ZJ, Zhao SM,
36. Ahmad A, Hanby A, Dublin E, Poulsom R, Chen ZL, Zhou GM (2008) Comparison of
Smith P, Barnes D, Rubens R, Anglard P, Hart blood-nerve barrier disruption and matrix
I (1998) Stromelysin 3: an independent prog- metalloprotease-9 expression in injured central
nostic factor for relapse-free survival in node- and peripheral nerves in mice. Neurosci Lett
positive breast cancer and demonstration of 434:155–159
novel breast carcinoma cell expression. Am 48. Platt CI, Krekoski CA, Ward RV, Edwards DR,
J Pathol 152:721–728 Gavrilovic J (2003) Extracellular matrix and
37. Freije JM, Diez-Itza I, Balbin M, Sánchez LM, matrix metalloproteinases in sciatic nerve.
Blasco R, Tolivia J, López-Otín C (1994) J Neurosci Res 74:417–429
Molecular cloning and expression of collage- 49. Shubayev VI, Myers RR (2008) Upregulation
nase-3, a novel human matrix metalloprotein- and interaction of TNFalpha and gelatinases A
ase produced by breast carcinomas. J Biol and B in painful peripheral nerve injury. Brain
Chem 269:16766–16773 Res 855:83–89
38. Ueno H, Nakamura H, Inoue M, Imai K, 50. Ramer R, Hinz B (2008) Inhibition of cancer
Noguchi M, Sato H, Seiki M, Okada Y (1997) cell invasion by cannabinoids via increased
Expression and tissue localization of expression of tissue inhibitor of matrix metallo-
membrane-types 1, 2, and 3 matrix metallo- proteinases-1. J Natl Cancer Inst 2:59–69
proteinases in human invasive breast carcino- 51. Sommer C, Schmidt C, George A, Toyka KV
mas. Cancer Res 57:2055–2060 (1997) A metalloprotease-inhibitor reduces
39. Jones JL, Glynn P, Walker RA (1999) Expression pain associated behavior in mice with experi-
of MMP-2 and MMP-9, their inhibitors, and mental neuropathy. Neurosci Lett 237:45–48
the activator MT1-MMP in primary breast car- 52. Noble LJ, Donovan F, Igarash T, Goussev S,
cinomas. J Pathol 189:161–168 Werb Z (2002) Matrix metalloproteinases limit
40. Cai QW, Li J, Li X, Wang J, Huang Y (2012) functional recovery after spinal cord injury by
Expression of STAT3, MMP-1 and TIMP-1 in modulation of early vascular events. J Neurosci
gastric cancer and correlation with pathological 1:7526–7535
features. Mol Med Rep 5:1438–1442 53. Pannu R, Christie DK, Barbosa E, Singh I,
41. Murray GI, Duncan ME, Arbuckle E, Melvin Singh AK (2007) Post-trauma Lipitor treat-
WT, Fothergill JE (1998) Matrix metallopro- ment prevents endothelial dysfunction, facili-
teinases and their inhibitors in gastric cancer. tates neuroprotection, and promotes locomotor
Gut 43:791–797 recovery following spinal cord injury.
42. Kemik O, Kemik AS, Sümer A, Dulger AC, J Neurochem 101:182–200
Adas M, Begenik H, Hasirci I, Yilmaz O, 54. Fleming JC, Norenberg MD, Ramsay DA,
Purisa S, Kisli E, Tuzun S, Kotan C (2011) Dekaban GA, Marcillo AE, Saenz AD, Pasqale-
Levels of matrix metalloproteinase-1 and tissue Styles M, Dietrich WD, Weaver LC (2008) The
inhibitors of metalloproteinase-1 in gastric can- cellular inflammatory response in human spinal
cer. World J Gastroenterol 17:2109–2112 cords after injury. Brain 129:3249–3269
43. Fujimoto D, Hirono Y, Goi T, Katayama K, 55. Amantea D, Corasaniti MT, Mercuri NB,
Vamaguchi A (2008) Prognostic value of Bernardi G, Bagetta G (2008) Brain regional
Protease-activated Receptor-1 (PAR-1) and and cellular localization of gelatinase activity in
Matrix Metalloproteinase-1 (MMP-1) in gas- rat that have undergone transient middle cere-
tric cancer. Anticancer Res 28:847–854 bral artery occlusion. Neuroscience 152:8–17
44. Kawasaki Y, Xu ZZ, Wang X, Park JY, Zhuang 56. Nagel S, Su Y, Horstmann S, Heiland S,
ZY, Tan PH, Gao YJ, Roy K, Corfas G, Lo EH, Gardner H, Koziol J, Martinez-Torres FJ,
Ji RR (2008) Distinct roles of matrix metallopro- Wagner S (2008) Minocycline and hypother-
teases in the early- and late-phase development of mia for reperfusion injury after focal cerebral
neuropathic pain. Nat Med 14:331–336 ischemia in the rat: effects on BBB breakdown
284 Tanya Singh et al.
and MMP expression in the acute and subacute minogen activator, plasminogen activator
phase. Brain Res 1188:198–206 inhibitor and gelatinase-B in chronic wound
57. Ding YH, Li J, Rafols JA, Ding Y (2004) fluid switches from a chronic to acute wound
Reduced brain edema and matrix metallopro- profile with progression to healing. Wound
teinase (MMP) expression by pre-reperfusion Repair Regen 7:154–165
infusion into ischemic territory in rat. Neurosci 68. Warner RL, Bhagavathula N, Nerusu KC,
Lett 372:35–39 Lateef H, Younkin E, Johnson KJ, Varani
58. Fujimoto M, Takagi Y, Aoki T, Hayase M, J (2004) Matrix metalloproteinases in acute
Marumo T, Gomi M, Nishimura M, Kataoka H, inflammation: induction of MMP-3 and
Hashimoto N, Nozaki K (2008) Tissue inhibi- MMP-9 in fibroblasts and epithelial cells fol-
tor of metalloproteinases protect blood- brain lowing exposure to pro-inflammatory media-
barrier disruption in focal cerebral ischemia. tors in vitro. Exp Mol Pathol 76:189–195
J Cereb Blood Flow Metab 28:1674–1685 69. Sagel, S.D., Kapsner, R.K, and Osberg, I.
59. Vilalta A, Sahuquillo J, Rosell A, Poca MA, (2005) Induced sputum matrix metallopro-
Riveiro M, Montaner J (2008) Moderate and teinase-9 correlates with lung function and air-
severe traumatic brain injury induce early over- way inflammation in children with cystic
expression of systemic and brain gelatinases. fibrosis. Pediatr Pulmonol 39, 224–232.
Intensive Care Med 34:1384–1392 70. Yang H, Dai Y, Dong H, Zang D, Liu Q, Duan
60. Jiang X, Namura S, Nagata I (2001) Matrix H, Niu Y, Bin P, Zheng Y (2011)
metalloproteinase inhibitor KB-R7785 attenu- Trichloroethanol up-regulates matrix metallo-
ates brain damage resulting from permanent proteinase-9 and tissue inhibitor of metallopro-
focal cerebral ischemia in mice. Neurosci Lett teinase-1 in HaCaT cells. Toxicol In Vitro
305:41–44 25:1638–1643
61. Lee JE, Yoon YJ, Moseley ME, Yenari MA 71. Shibataa N, Ohnumaa T, Higashia S, Usuia
(2005) Reduction in levels of matrix metallo- C, Ohkuboa T, Kitajimaa A, Uekib A, Nagaoc
proteinases and increased expression of tissue M, Araia H (2005) Genetic association
inhibitor of metalloproteinase-2 in response to between matrix metalloproteinase MMP-9
mild hypothermia therapy in experimental and MMP-3 polymorphisms and Japanese
stroke. J Neurosurg 103:289–297 sporadic Alzheimer’s disease. Neurobiol
62. Yang Y, Estrada EY, Thompson JF, Liu W, Aging 26:1011–1014
Rosenberg GA (2007) Matrix metalloproteinase- 72. Kofla-Dlubacz A, Matusiewicz M, Krzystek-
mediated disruption of tight junction proteins in Korpacka M, Iwanczak B (2012) Correlation
cerebral vessels is reversed by synthetic matrix of MMP-3 and MMP-9 with Crohn’s disease
metalloproteinase inhibitor in focal ischemia in activity in children. Dig Dis Sci 57:706–712
rat. J Cereb Blood Flow Metab 4:697–709 73. Kumar MS, Vamsi G, Sripriya R, Sehgal PK
63. Wang Y, Deng XL, Xiao XH, Yuan BX (2007) A (2006) Expression of matrix metalloprotein-
non-steroidal anti-inflammatory agent provides ases (MMP-8 and -9) in chronic periodontitis
significant protection during focal ischemic patients with and without diabetes mellitus.
stroke with decreased expression of matrix metal- J Periodontol 77:1803–1808
loproteinases. Curr Neurovasc Res 3:176–183 74. Jayaram B, Singh T, Mukherjee G, Mathur A,
64. Truettner JS, Alonso OF, Dalton DW (2005) Shekhar S, Shekhar V (2012) Sanjeevini: a
Influence of therapeutic hypothermia on matrix freely accessible web-server for target directed
metalloproteinase activity after traumatic brain lead molecule discovery. BMC Bioinformatics
injury in rats. J Cereb Blood Flow Metab 13:S7
11:1505–1516 75. Mukherjee G, Jayaram B (2013) A rapid iden-
65. Sunami E, Tsuno N, Osada T, Saito S, Kitayama tification of hit molecules for target proteins via
J, Tomozawa S, Tsuruo T, Shibata Y, Muto T, physico-chemical descriptors. Phys Chem
Nagawa H (2000) MMP-1 is a prognostic Chem Phys 15:9107–9116
marker for hematogenous metastasis of 76. Gupta A, Gandhimathi A, Sharma P, Jayaram B
colorectal cancer. Oncologist 5:108–114 (2007) ParDOCK: an all atom energy-based
66. Rodrigues WF, Madeira MF, da Silva TA, Monte Carlo docking protocol for protein-ligand
Clemente-Napimoga JT, Miguel CB, Dias-da- complexes. Protein Pept Lett 14:632–646
Silva VJ, Barbosa-Neto O, Lopes AH, Napimoga 77. Singh T, Biswas D, Jayaram B (2011) AADS—
MH (2012) Low dose of propranolol down- An automated active site identification, dock-
modulates bone resorption by inhibiting inflam- ing and scoring protocol for protein targets
mation and osteoclast differentiation. Br based on physico-chemical descriptors. J Chem
J Pharmacol 165:2140–2151 Inf Model 51:2515–2527
67. Wysocki AB, Kusakabe AO, Chang S, Tuan TL 78. Jain T, Jayaram B (2005) An all atom energy-
(1997) Temporal expression of urokinase plas- based computational protocol for predicting
In Silico Design of MMP Inhibitors 285
binding affinities of protein-ligand complexes. 87. Kalra P, Reddy TV, Jayaram B (2011) Free
FEBS Lett 579:6659–6666 energy component analysis for drug design: a
79. Shaikh SA, Jayaram B (2007) A Swift all-atom case study of HIV-1 protease-inhibitor bind-
energy-based computational protocol to pre- ing. J Med Chem 44:4325–4338
dict DNA ligand binding affinity and ΔTm. 88. Horn HW, Swope WC, Pitera JW, Madura JD,
J Med Chem 50:2240–2244 Dick TJ, Hura GL, Head-Gordon T (2004)
80. Giangreco I, Lattanzi G, Nicolotti O, Catto M, Development of an improved four-site water
Laghezza A, Leonetti F, Stefanachi A, Carotti model for biomolecular simulations:
A (2011) Insights into the complex formed by TIP4P-Ew. J Chem Phys 120:9665–9678
matrix metalloproteinase-2 and alloxan inhibi- 89. Berendsen HJC, Postma JPM, van Gunsteren
tors: molecular dynamics simulations and free WF, DiNola A, Haak JR (1984) Molecular
energy calculations. PLoS One 6:1 dynamics with coupling to an external bath.
81. Jakalian A, Bush BL, Jack DB, Bayly CI (2004) J Chem Phys 81:3684–3690
Fast, efficient generation of high-quality atomic 90. Lavery R, Zakrzewska K, Beveridge DL,
charges. AM1-BCC model: I. Method. Bishop TC, Case DA, Cheatham T III, Dixit S,
J Comput Chem 21:132–146 Jayaram B, Lankas F, Laughton C, Maddocks
82. Wang J, Wolf RM, Caldwell JW, Kollman PA, JH, Michon A, Osman R, Orozco M, Perez A,
Case DA (2004) Development and testing of a Singh T, Spackova N, Sponer J (2009) A sys-
general amber force field. J Comput Chem tematic molecular dynamics study of nearest
25:1157–1174 neighbor effects on base pair and base pair step
83. Cornell WD, Cieplak P, Bayly CI, Gould IR, conformations and fluctuations in
Merz KM (1995) A second generation force B-DNA. Nucleic Acids Res 38:299–313
field for the simulation of proteins, nucleic 91. Rizzo RC, Toba S, Kuntz ID (2004) A molec-
acids, and organic molecules. J Am Chem Soc ular basis for the selectivity of thiadiazole urea
117:5179–5197 inhibitors with stromelysin-1 and gelatinase-A
84. Frisch MJ, Trucks GW, Schlegel HB, Scuseria from generalized born molecular dynamics
GE, Robb MA, Cheeseman JR, Scalmani G, simulations. J Med Chem 47:3065–3074
Barone V, Mennucci B, Petersson GA, 92. Chong S, Ham S (2013) Assessing the influ-
Nakatsuji H, Caricato M, Li X, Hratchian HP, ence of solvation models on structural
Izmaylov AF, Bloino J, Zheng G, Sonnenberg characteristics of intrinsically disordered pro-
JL, Hada M, Ehara M, Toyota K, Fukuda R, tein. Comput Theor Chem 1017:194–199
Hasegawa J, Ishida M, Nakajima T, Honda Y, 93. Shaikh SA, Ahmed SR, Jayaram B (2004) A
Kitao O, Nakai H, Vreven T, Montgomery JA molecular thermodynamic view of DNA-drug
Jr, Peralta JE, Ogliaro F, Bearpark M, Heyd JJ, interaction: a case study of 25 minor groove
Brothers E, Kudin KN, Staroverov VN, binders. Arch Biochem Biophys 429:81–99
Kobayashi R, Normand J, Raghavachari K, 94. Jayaram B, Mcconnell K, Dixit SB, Beveridge
Rendell A, Burant JC, Iyengar SS, Tomasi J, DL (2002) Free energy component analysis of
Cossi M, Rega N, Millam NJ, Klene M, Knox 40 protein-DNA complexes: a consensus view
JE, Cross JB, Bakken V, Adamo C, Jaramillo J, on the thermodynamics of binding at the
Gomperts R, Stratmann RE, Yazyev O, Austin molecular level. J Comput Chem 23:1–14
AJ, Cammi R, Pomelli C, Ochterski JW, Martin 95. Jayaram B, McConnell KJ, Dixit SB, Beveridge
RL, Morokuma K, Zakrzewski VG, Voth GA, DL (1999) Free energy analysis of protein-
Salvador P, Dannenberg JJ, Dapprich S, DNA binding: the EcoRI endonuclease—DNA
Daniels AD, Farkas Ö, Foresman JB, Ortiz JV, complex. J Comput Phys 151:333–357
Cioslowski J, Fox DJ (2009) Gaussian, Inc.,
Wallingford, CT 96. Fenley MO, Harris R, Jayaram B, Boschitsch
AH (2010) Revisiting the association of cationic
85. Stote RH, Karplus M (1995) Zinc binding in groove-binding drugs to DNA using a Poisson-
proteins and solution: a simple but accurate Boltzmann approach. Biophys J 99:879–886
nonbonded representation. Proteins Struct
Funct Genet 23:12–31 97. Wong S, Amaro RE, McCammon JA (2009)
MM-PBSA captures key role of intercalating
86. Pearlman DA, Case DA, Caldwell JW, Ross water molecules at a protein−protein interface.
WS, Cheathem JE III, DeBolt S, Ferguson D, J Chem Theory Comput 5:422–449
Seibel G, Kollman P (1995) AMBER, a pack-
age of computer programs for applying molec- 98. Singh T, Adekoya OA, Jayaram B (2015)
ular mechanics, normal mode analysis, Understanding the binding of inhibitors of
molecular dynamics and free energy calcula- matrix metalloproteinases by molecular docking,
tions to simulate the structural and energetic quantum mechanical calculations, molecular
properties of molecules. Comput Phys dynamics simulations, and a MMGBSA/
Commun 91:1–41 MMBappl study. Mol Biosyst 11:1041–1051
Chapter 16
Abstract
The blood-brain barrier is a multicellular and basement membrane unit that regulates molecular transport
between the blood and central nervous system. Many cerebral pathologies, such as acute stroke and
chronic vascular dementia, result in a disrupted blood-brain barrier, increasing its permeability and allow-
ing the entry of potentially neurotoxic molecules. The activation of matrix metalloproteinases mediates
further blood-brain barrier damage. The inhibition of matrix metalloproteinases is a potential strategy for
stroke therapy. As inhibitors are developed, efficient context-specific screening methods will be required.
Models of the blood-brain barrier have been extensively used to study neuropathologies and the effect of
various treatment options.
Herein, we describe a co-culture model of the blood-brain barrier composed of brain microvascular
endothelial cells and astrocytes grown on an artificial basement membrane-coated membrane insert. Our
cell model forms a barrier and is a simple first approximation of blood-brain barrier integrity. As currently
developed, the model may be applied to testing the effect of matrix metalloproteinases and matrix metal-
loproteinase inhibitors on blood-brain barrier physiology and pathophysiology. The model is a quick and
effective evaluation tool for generating nonclinical data in a living cell system before proceeding to animal
models.
1 Introduction
Charles A. Galea (ed.), Matrix Metalloproteases: Methods and Protocols, Methods in Molecular Biology, vol. 1579,
DOI 10.1007/978-1-4939-6863-3_16, © Springer Science+Business Media LLC 2017
287
288 Jennifer S. Myers et al.
between the cell layer, building on the examples of Culot et al. and
Megard et al. [15, 16]. We selected bBMVEC for relative ease of
culture and included astrocytes (see Note 1) because of their sig-
nificant contribution to barrier properties in endothelial cells such
as BBB formation and integrity [1, 17, 18]. The economic benefit
of our model is twofold. First, the permeability of cells can be sim-
ply quantified by the use of Lucifer yellow and a fluorescence plate
reader, a piece of equipment that is readily available and thus avoids
the use of equipment dedicated to measurement of transendothe-
lial electric resistance. Second, our choice of using primary cell
lines instead of freshly harvested cells avoids continual animal sac-
rifice for each experiment and the attendant difficulties and costs.
2 Materials
3 Methods
3.1 Cell Culture All experiments were performed in a biological safety cabinet at
room temperature unless otherwise stated. Inserts are easily punc-
tured by pipet tips; extreme care must be taken to avoid contact
with the membrane at each handling.
1. Prepare a stock solution of Type 1 rat tail collagen dissolved in
0.02 N acetic acid. Using this stock makes a dilution of suffi-
cient concentration to provide 15 μg collagen/cm2 of flask
surface. Add the collagen solution to the culture flask and
incubate at 37°C for 1 h (see Note 3). Remove the collagen
solution and rinse the flask with TBS and then with warm
medium.
2. Expand bBMVEC culture by plating at 5,000 cells/cm2 in
collagen-treated flask. Culture cells until 75–80% confluence
and then harvest and freeze using method of choice. This will
be a stock for future experiments. Freeze in multiple small
batches such that each aliquot provides enough cells for a sin-
gle experiment (see Note 4).
3. Expand NHA culture by plating between 5,000 cells/cm2 in
an untreated flask. Culture cells until 75–80% confluence and
then harvest and freeze using method of choice. This will be a
stock for future experiments. Freeze in multiple small batches
such that each aliquot provides enough cells for a single experi-
ment (see Note 4).
4. Three to four days prior to starting the co-culture, thaw an
aliquot of bBMVEC into a collagen-coated culture flask and
allow to recover with regular medium changes.
5. One day prior to starting the co-culture, prepare a dilution of
Type 1 rat tail collagen dissolved in sterile H2O to a sufficient
concentration to deliver 15 μg collagen/cm2 of membrane
insert surface. Apply collagen solution to the apical surface
inside the well of the membrane insert and allow to dry uncov-
ered overnight (see Note 5). After drying, rinse with TBS (see
Note 6).
Cellular BBB Model for MMP Inhibitor Screening 291
Fig. 2 Experimental workflow for testing the effect of MMP inhibition on monolayer permeability. The integrity
of the monolayer formed in the co-culture was assessed by its permeability to Lucifer yellow fluorescent
tracer. The effect of MMP inhibitors of the integrity of the monolayer was assessed after disrupting the mono-
layer with tissue plasminogen activator (tPA). First, cells were incubated with a treatment solution (tPA, tPA +
inhibitor, inhibitor, etc.) for 6 h. Then, the treatment solution was replaced with a solution of Lucifer yellow. After
the introduction of the tracer, the membrane insert and the well below were sampled at various time points up
to 1 h. The passage of Lucifer yellow between the cells was determined by fluorescence spectroscopy
that ensures that the liquid level inside and outside the insert is
roughly equal (see Note 15).
7. Immediately remove 100 μL aliquots from the well and the
membrane insert and place in the 96-well assay plate. Then
add 100 μL of fresh transport buffer to the membrane
compartment. Repeat sampling at desired time points, keep-
ing the culture in the incubator between time points (see
Note 16).
8. Measure the fluorescence of the samples using a microtiter
plate reader. Excite at 485 nm and record emission at 538 nm.
Calculate the concentration of Lucifer yellow in the sample
using a calibration curve.
294 Jennifer S. Myers et al.
4 Notes
Acknowledgments
References
1. Abbott NJ, Ronnback L, Hansson E (2006) 10. Naik P, Cucullo L (2012) In vitro blood-brain
Astrocyte-endothelial interactions at the blood- barrier models: current and perspective tech-
brain barrier. Nat Rev Neurosci 7(1):41–53 nologies. J Pharm Sci 101(4):1337–1354
2. Roycik MD, Myers JS, Newcomer RG, et al. 11. He Y, Yao Y, Tsirka SE, et al. (2014) Cell-
(2013) Matrix metalloproteinase inhibition in culture models of the blood-brain barrier.
atherosclerosis and stroke. Curr Mol Med Stroke 45(8):2514–2526
13(8):1299–1313 12. Nakagawa S, Deli MA, Kawaguchi H, et al.
3. Niego B, Medcalf RL (2014) Plasmin- (2009) A new blood-brain barrier model using
dependent modulation of the blood-brain bar- primary rat brain endothelial cells, pericytes and
rier: a major consideration during tPA-induced astrocytes. Neurochem Int 54(3–4):253–263
thrombolysis? J Cereb Blood Flow Metab 13. Hatherell K, Couraud P, Romero IA, et al.
34(8):1283–1296 (2011) Development of a three-dimensional,
4. Gasche Y, Soccal PM, Kanemitsu M, et al. all-human in vitro model of the blood–brain
(2006) Matrix metalloproteinases and diseases barrier using mono-, co-, and tri-cultivation
of the central nervous system with a special Transwell models. J Neurosci Methods
emphasis on ischemic brain. Front Biosci 199(2):223–229
11:1289–1301 14. Xue Q, Liu Y, Qi H, et al. (2013) A novel brain
5. Copin J, Bengualid DJ, Da Silva RF, et al. neurovascular unit model with neurons, astro-
(2011) Recombinant tissue plasminogen cytes and microvascular endothelial cells of rat.
activator induces blood–brain barrier break- Int J Biol Sci 9(2):174–189
down by a matrix metalloproteinase-9-inde- 15. Megard I, Garrigues A, Orlowski S, et al.
pendent pathway after transient focal cerebral (2002) A co-culture-based model of human
ischemia in mouse. Eur J Neurosci 34(7): blood-brain barrier: application to active trans-
1085–1092 port of indinavir and in vivo-in vitro correla-
6. Candelario-Jalil E, Yang Y, Rosenberg GA tion. Brain Res 927(2):153–167
(2009) Diverse roles of matrix metalloprotein- 16. Culot M, Lundquist S, Vanuxeem D, et al.
ases and tissue inhibitors of metalloproteinases (2008) An in vitro blood-brain barrier model
in neuroinflammation and cerebral ischemia. for high throughput (HTS) toxicological
Neuroscience 158(3):983–994 screening. Toxicol In Vitro 22(3):799–811
7. Cecchelli R, Berezowski V, Lundquist S, et al. 17. Siddharthan V, Kim YV, Liu S, et al. (2007)
(2007) Modelling of the blood-brain barrier in Human astrocytes/astrocyte-conditioned
drug discovery and development. Nat Rev medium and shear stress enhance the barrier
Drug Discov 6(8):650–661 properties of human brain microvascular endo-
8. Cucullo L, Hossain M, Rapp E, et al. (2007) thelial cells. Brain Res 1147:39–50
Development of a humanized in vitro blood- 18. Kuo YC, Lu CH (2011) Effect of human astro-
brain barrier model to screen for brain penetra- cytes on the characteristics of human brain-
tion of antiepileptic drugs. Epilepsia microvascular endothelial cells in the
48(3):505–516 blood-brain barrier. Colloids Surf B
9. Deli MA, Abraham CS, Kataoka Y, et al. (2005) Biointerfaces 86(1):225–231
Permeability studies on in vitro blood-brain 19. Cecchelli R, Dehouck B, Descamps L, et al.
barrier models: physiology, pathology, and (1999) In vitro model for evaluating drug
pharmacology. Cell Mol Neurobiol transport across the blood-brain barrier. Adv
25(1):59–127 Drug Deliv Rev 36(2–3):165–178
Part VII
Abstract
Matrix metalloprotease play a vital role in many cellular processes. Dysfunction in activity of these enzymes
has been implicated in the pathogenesis of a number of diseases. Factors that affect the balanced interac-
tion between MMPs and their inhibitors, such as genetic mutations of extracellular matrix components or
dysregulation of MMP expression, can lead to various diseases. Due to their essential role in ECM remod-
eling, MMPs have become targets of interest as biomarkers for the diagnosis and prognosis of diseases
associated with alterations of the ECM.
1 Introduction
1.1 Definition, The space between the cells that form all tissues and organs is filled
Classification, with a series of compounds and macromolecules that constitute the
and Roles of MMPs extracellular matrix (ECM). Therefore, the ECM represents the
noncellular components of all tissues and organs. The ECM has tis-
sue-specific characteristics; however, it is basically composed of water
and relatively stable macromolecules, which are not only produced,
exported, and bound by cells but also responsible for the modula-
tion of their structure, physiology, biochemical interactions, and the
biomechanics of tissues. Such molecules, essential for morphogene-
sis, differentiation, and tissue homeostasis, are classified into three
categories: glycosaminoglycan polysaccharide chains, fibrous pro-
teins with structural roles (collagens and elastins), and non-fibrous
proteins with adhesion properties (laminins and fibronectins) [1].
Extracellular matrix remodeling is a dynamic and continuous
process in which the cell deposits new components and degrades
old ones by proteolytic action [2]. ECM degradation is essential
not only for processes like embryonic development, morphogene-
sis, reproduction and tissue reabsorption and remodeling [3] but
Charles A. Galea (ed.), Matrix Metalloproteases: Methods and Protocols, Methods in Molecular Biology, vol. 1579,
DOI 10.1007/978-1-4939-6863-3_17, © Springer Science+Business Media LLC 2017
299
300 Fernando Luiz Affonso Fonseca et al.
Table 1
Substrates cleaved by various MMPs and their chromosomal location
3.2 Sepsis Some studies show a direct correlation between increased levels of
MMPs and low survival rates of patients with sepsis [31]. Aerts
et al. [32] have recently demonstrated that a hydroxypyrone-based
inhibitor selective for MMP-12 displays anti-inflammatory activity
in mice.
306 Fernando Luiz Affonso Fonseca et al.
3.4 Conjuncti Despite the fact the causes of CCh are still uncertain, it is very
vochalasis (CCh) likely that the accumulation of MMPs in the tears of these patients
may contribute to the progression of the disease [34]. In addition,
the effectiveness of surgery to treat CCh has been shown to be
associated with a postoperative decrease in MMP-9 levels in tears.
In fact, many authors suggest that the quantification of MMP-9
could be important for the evaluation of many eye diseases [34].
3.5 Coronary Heart The inflammatory process seems to play a role in the pathogenesis
Disease (CHD) of vascular diseases. Markers of inflammation like C-reactive pro-
tein, fibrinogen, and interleukin-6 are associated with the risk of
developing CHD [35]. However, other markers, such as MMP-9,
also need to be accessed [36, 37].
3.6 Inflammatory Ulcerative colitis (UC) and Crohn’s disease (CD) are autoimmune
Bowel Disease (IBD) diseases characterized by inflammation of the gastrointestinal sys-
tem. Although MMPs are present in IBD, their role is not clear
and further studies are required [38]. The colon of patients with
IBD exhibit high levels of MMP-1, -3, -7, -9, -10, and -12 [39],
and intestinal biopsies of patients with UC demonstrate that vascu-
lar endothelial cells and leukocytic infiltrate produce MMP-7 and
-13 [40]. MMP-9 serum levels are correlated to both CD and UC
disease, with higher levels detected in the latter. This difference in
levels between these MMPs could help to differentiate between
UC disease and CD [41].
Future studies should elucidate the roles of MMPs and tissue
inhibitors of metalloproteinases (TIMPs), thus contributing to the
diagnosis and treatment of inflammatory diseases [42, 43].
4.1 Breast Cancer Overexpression of MMPs in the serum of breast cancer patients is
associated with a poor prognosis. For example, overexpression of
MMP-9 and MMP-2 correlates with a higher risk and poor prog-
nosis for this type of cancer [45].
MMP-13 has also been studied and characterized with tumor
progression. Overexpression of MMP-13 has been correlated with
the expression of HER2/neu, more aggressive tumor phenotypes
and decreased life expectancy [46]. A recent study examining the
relationship between MMP and TIMP expression in tumors (i.e.,
tumor center or tumor invasive front) and distant metastasis in
mononuclear inflammatory cells (MICs) and cancer associated
fibroblasts (CAFs) of patients with primary breast cancer, showed
that the expression of MMP-11 by MICs and TIMP-2 by CAFs in
the tumor center or tumor invasive front were the most potent
independent prognostic factors for predicting clinical outcome for
patients [47].
4.2 Prostate Cancer It has recently been shown that genotypic variations of MMP-3
and TIMP-3 (polymorphisms) may affect the balance between
these proteins, thus increasing the risk of developing prostate can-
cer [48].
The relationship between testosterone and MMP-13 levels in
prostate cancer patients has been previously reported [8]. Low lev-
els of testosterone have been linked to a poor prognosis [49]. This
relationship suggests an important biochemical interaction between
this hormone and these endopeptidases. Immunohistochemical
analysis of MMP-3, MMP-13 and TIMP-3, MMP-9, and TIMP-1
levels in prostate cancer also suggests that an imbalance between
MMP-9 and TIMP-1 may explain the invasive potential of pros-
tatic adenocarcinoma [50].
4.3 Lung Cancer MMP-13 is not only associated with breast and prostate cancers
but may also be the most important target among the MMPs in
lung cancer. In a recent study, MMP-2, -3, -9, and -13 were
308 Fernando Luiz Affonso Fonseca et al.
4.5 Bladder Cancer Studies have investigated the role of MMPs in bladder cancer by
identifying metalloproteinase polymorphisms and gene expression
levels as well as measuring MMP levels in urine. These analyses of
neoplasia have looked at various biological samples, including
urine, and the results establish MMPs as potential diagnostic bio-
markers for this neoplasia.
Both MMP-3 and MMP-9 have recently been proposed as
possible early markers for bladder cancer. Using ELISA-based
assays the authors were able to show that levels of both MMPs
were elevated in the urine of patients with bladder cancer when
compared with a control group. Furthermore, MMP-3 levels were
evaluated in urine samples from patients with early stage schisto-
somal and non-schistosomal bladder cancer. Remarkably, MMP-3
levels remained elevated at advanced stages of these two histologi-
cal types of cancer. On the other hand, MMP-9 levels were elevated
only at advanced stages of schistosomal and non-schistosomal
Matrix Metalloproteases as Biomarkers of Disease 309
References
1. Frantz C, Stewart KM, Weaver VM (2010) 9. Chaudhary KA, Pandya S, Ghosh K, Nadkarni
The extracellular matrix at a glance. J Cell Sci A (2013) Matrix metalloproteinase and its
123:4195–4200 drug targets therapy in solid and hematologi-
2. Harjanto D, Zaman MH (2013) Modeling cal malignances: an overview. Mutat Res
extracellular matrix reorganization in 3D envi- 753:7–23
ronments. PLoS One 8:e52509 10. Klein G, Schmal O, Aicher KW (2015) Matrix
3. Tallant C, Marrero A, Gomis-Rüth FX (2010) metalloproteinases in stem cell mobilization.
Matrix metalloproteinases: fold and function of Matrix Biol 44–46:175–183
their catalytic domains. Biochim Biophys Acta 11. MacColl E, Khalil RA (2015) Matrix metallo-
1803:20–28 proteinases as regulators of vein structure and
4. Bonnans C, Chou J, Werb Z (2014) function: implications in chronic venous dis-
Remodelling the extracellular matrix in devel- ease. J Pharmacol Exp Ther 355:410–428
opment and disease. Nat Rev Mol Cell Biol 12. Szmigielska-Kaplon A, Szemraj J, Hamara K,
15:786–801 et al. (2014) Polymorphism of CD44 influences
5. Theocharis AD, Skandalis SS, Gialeli C, the efficacy of CD34(+) cells mobilization in
Karamanos NK (2015) Extracellular matrix patients with hematological malignancies. Biol
structure. Adv Drug Deliv Rev 97:4–27 Blood Marrow Transplant 20:986–991
6. Giannandrea M, Parks WC (2014) Diverse 13. Kunisaki Y, Bruns I, Scheiermann C (2013)
functions of matrix metalloproteinases during Arteriolar niches maintain haematopoietic stem
fibrosis. Dis Model Mech 7:193–203 cell quiescence. Nature 502:637–643
7. Tenório PP, Duque MAA, Araújo MM, Pontes 14. Deryugina IE, Quigley PJ (2015) Tumor
Filho NT, Melo MR Jr (2012) The role of angiogenesis: MMP-mediated induction of
matrix metalloproteinases in the development intravasation- and metastasis-sustaining neo-
of aortic aneurysm. Avaliable at: http://files. vasculature. Matrix Biol 44–46:94–112
bvs.br/upload/S/0101-5907/2012/v26n4/ 15. Thushara MR, Hemshekhar M, Kemparaju B,
a3488.pdf [Portuguese] et al. (2015) Biologicals, platelet apoptosis and
8. Bonaldi CM, Azzalis LA, Junqueira VBC, human diseases: an outlook. Crit Rev Oncol
Oliveira CGB, Vilas Boas VA, Gáscon TM, Hematol 93:149–158
Gehrke FS, Kuniyoshi RK, Alves BCA, Fonseca 16. Poyer F, Coquerel B, Pegahi R, et al. (2009)
FLA (2015) Plasmatic levels of E-Cadherin Secretion of MMP-2 and MMP-9 induced by
and MMP-13 in prostate cancer patients: cor- VEGF autocrine loop correlates with clinical
relation with PSA, testosterone and pathologi- features in childhood acute lymphoblastic
cal parameters. Tumori 101:185–188 leukemia. Leuk Res 33:407–417
310 Fernando Luiz Affonso Fonseca et al.
in inflammation and immunity. Nat Rev Roels O, Sabourin JC, Thiberville L, Clapper
Immunol 13:649–665 ML (2015) MMP-13 in-vivo molecular imag-
43. Vandenbroucke RE, Libert C (2014) Is there ing reveals early expression in lung adenocarci-
new hope for therapeutic matrix metallopro- noma. PLoS One 10:e0132960
teinase inhibition? Nat Rev Drug Discov 52. Butkiewicz D, Krześniak M, Drosik A, Giglok
13:904–927 M, Gdowicz-Kłosok A, Kosarewicz A, Rusin M,
44. Wieczorek E, Jablonska E, Wasowicz W, Masłyk B, Gawkowska-Suwińska M, Suwiński R
Reszka E (2015) Matrix metalloproteinases (2015) The VEGFR2, COX-2 and MMP-2
and genetic mouse models in cancer research: a polymorphisms are associated with clinical out-
mini-review. Tumour Biol 36:163–175 come of patients with inoperable non-small cell
45. Ren F, Tang R, Zhang X, Madushi WM, Luo lung cancer. Int J Cancer 137:2332–2342
D, Dang Y, Li Z, Wei K, Chen G (2015) 53. Georgescu EF, Mogoantă SŞ, Costache A,
Overexpression of MMP family members func- Pârvănescu V, Totolici BD, Pătraşcu Ş,
tions as prognostic biomarker for breast cancer Stănescu C (2015) The assessment of matrix
patients: a systematic review and meta-analysis. metalloproteinase-9 expression and angiogen-
PLoS One 10(8):e0135544 esis in colorectal cancer. Rom J Morphol
46. Rosseti C, Reis B, Delgado PO, et al. (2015) Embryol 56:1137–1144
Adhesion molecules in breast carcinoma: a 54. Zheng CG, Chen R, Xie JB, Liu CB, Jin Z, Jin
challenge to the pathologist. Rev Assoc Med C (2015) Immunohistochemical expression of
Bras 61:81–85 Notch1, Jagged1, NF-κB and MMP-9 in
47. Eiró N, Fernandez-Garcia B, Vázquez J, Del Casar colorectal cancer patients and the relationship
JM, González LO, Vizoso FJ (2015) A phenotype to clinicopathological parameters. Cancer
from tumor stroma based on the expression of Biomark 15:889–897
metalloproteases and their inhibitors, associated 55. Chen H, Hu Y, Xiang W, Cai Y, Wang Z, Xiao
with prognosis in breast cancer. Oncoimmunology Q, Liu Y, Li Q, Ding K (2015) Prognostic sig-
4(7):e992222 eCollection 2015 nificance of matrix metalloproteinase 7 immu-
48. Srivastava P, Kapoor R, Mittal RD (2013) nohistochemical expression in colorectal
Impact of MMP-3 and TIMP-3 gene polymor- cancer: a meta-analysis. Int J Clin Exp Med
phisms on prostate cancer susceptibility in 8:3281–3290
North Indian cohort. Gene 530:273–277 56. Yan Q, Yuan Y, Yankui L, Jingjie F, Linfang
49. García-Cruz E, Piqueras M, Huguet J, Peri L, J, Yong P, Dong H, Xiaowei Q (2015) The
Izquierdo L, Musquera M, Franco A, Alvarez- expression and significance of CXCR5 and
Vijande R, Ribal MJ, Alcaraz A (2012) Low MMP-13 in colorectal cancer. Cell Biochem
testosterone levels are related to poor progno- Biophys 73:253–259
sis factors in men with prostate cancer prior to 57. El-Sharkawi F, El Sabah M, Hassan Z, Khaled
treatment. BJU Int 110(11 Pt B):E541–E546 H (2014) The biochemical value of urinary
50. Babichenko II, Andriukhin MI, Pulbere S, metalloproteinases 3 and 9 in diagnosis and
Loktev A (2014) Immunohistochemical prognosis of bladder cancer in Egypt. J Biomed
expression of matrix metalloproteinase-9 and Sci 21:72
inhibitor of matrix metalloproteinase-1 in pros- 58. Tao L, Li Z, Lin L, Lei Y, Hongyuan Y,
tate adenocarcinoma. Int J Clin Exp Pathol Hongwei J, Yang L, Chuize K (2015) MMP1,
7:9090–9098 2, 3, 7, and 9 gene polymorphisms and urinary
51. Salaün M, Peng J, Hensley HH, Roder N, cancer risk: a meta-analysis. Genet Test Mol
Flieder DB, Houlle-Crépin S, Abramovici- Biomarkers 19:548–555
Index
A F
Fluorescence imaging�����������������������������������������������245–247
Activated imaging probes����������������������������������������� 246, 248
Fluorogenic substrates�������������������������������� 51, 138–139, 179
Affinity chromatography�����������������������������10, 22, 28, 29, 71
Förster resonance energy transfer (FRET)
substrates���������������������������������������� 58, 138, 140, 167
B
Free energy calculations����������������������������������������������������119
BBB. See Blood-brain barrier (BBB)
bBMVEC. See Bovine brain microvascular endothelial cells H
(bBMVEC) High-throughput sequencing������������210, 211, 220–222, 224
Blood-brain barrier (BBB)���������������������������������������285–293
co-culture model���������������������������������������������������������286 I
permeability��������������������������������������������������������285–287
Imaging�������������������������������������������������� 64, 78–79, 245–254
Bovine brain microvascular endothelial cells
Inclusion bodies (IBs)���������������� 4, 6–8, 10, 13, 50, 53, 67–68
(bBMVEC)������������������������������������������ 286–290, 292
Inflammatory disease������������������������������������������������ 303, 304
C Isotopically labeled������������������������������������������� 20–25, 67–68
iTRAQ-TAILS���������������������������������������� 186–189, 191–192
Cancer���������������������������������3, 4, 17, 18, 36, 49, 87, 111, 231,
246, 260, 261, 263, 272, 273, 300, 304–307 L
Cell culture membrane inserts���������������������������������� 289, 292
Ligand binding����������������������������������������� 114, 131, 166, 272
Cleavage��������������������������� 4, 9–12, 15, 28, 31, 38, 53, 62, 137,
140, 166, 169, 173, 178, 185–188, 193–196, 199–206, M
219, 220, 223, 224, 226, 231, 260, 271, 301, 304
Collagen����������������3, 18, 36, 88, 90, 111, 113, 116, 122–124, Matrix metalloprotease (MMP)�������������������� 3–15, 17–31,
139–168, 239, 260, 262, 287, 288, 292, 297–299, 302 35, 36, 49–59, 61, 62, 64–68, 75, 79, 82, 87–106,
Colonoscopy���������������������������������������������������������������������245 111–132, 137–179, 185–197, 199, 210, 231–243,
Colorectal cancer������������������������������������������������������ 273, 306 245–254, 259–269, 271–279, 285–293, 297–307
Computer-aided drug design������������������������������������271–279 inhibitors������������������������������������111, 113, 115, 126, 260,
Crystallization���������������������������������50, 51, 53–55, 58, 88, 89 261, 263, 272–274, 286, 288, 290, 291
membrane-anchored����������������������������������� 18–20, 23, 50
D MMP2���������������������������������������������������������������� 4–15, 18
MMP-14����������������������������� 49, 68, 91, 93, 105, 114, 269
Degradome�����������������������������������������������������������������������186
MD. See Molecular dynamics (MD)
Detection�������������������������������� 14, 27, 79, 169, 226, 231–243,
Meprin����������������������������������������������������������������� 35–44, 298
245, 246, 250, 273, 303, 305
Metalloprotease��������������������������������������3–15, 17–31, 35, 36,
Diffraction�������������������������������������������������������������� 49–59, 90
49–59, 61–82, 87–106, 111–132, 137–179, 185–197,
Disease����������������������������������� 17, 36, 87, 224, 225, 231, 246,
209–227, 231–243, 245–254, 259–269, 271–279,
263, 273, 297–307
285–293, 297–307
Docking������������������������������������������65, 66, 76, 121–125, 260,
Metalloproteinase. See Metalloprotease
263–267, 272–275, 279
Metzincin������������������������������������������������������������������� 35, 298
and scoring���������������������������������������������������������273–276
Mice��������������������������������������������������������� 247, 248, 253, 303
Min mice�������������������������������������������������� 246–248, 252, 253
E
Molecular dynamics (MD)��������������������������������� 76, 111–132
Early cancer detection������������������������������������������������������246 simulations����������������������������������113–116, 119–129, 132,
Endoprotease����������������������������������������������������������������������35 272, 274, 276–279
Charles A. Galea (ed.), Matrix Metalloproteases: Methods and Protocols, Methods in Molecular Biology, vol. 1579,
DOI 10.1007/978-1-4939-6863-3, © Springer Science+Business Media LLC 2017
313
Matrix Metalloproteases: Methods and Protocols
314 Index
Molecular modeling������������������������������������������������� 115, 116 Protein–lipid interactions���������������������������������������������������74
MT-LOOP-dependent localization of membrane type I Proteomics������������������������������������������������ 185, 189, 195, 199
matrix metalloproteinase (MT1-MMP)������������18–20,
40, 50, 52–58, 144, 148, 149, 160–163, 165–167, 170 R
Multi-domain������������������������������������������������������������� 90, 112 Refolding����������������������������4–9, 13, 14, 50, 51, 53, 54, 58, 68
Multidomain enzymes�������������������������������������� 112, 113, 119
Mutagenesis������������������������������������������������� 62, 81, 209–227 S
N Semiquantitative���������������������������������������������������������������242
Stroke����������������������������������������������������������������������� 286, 300
Near-infrared imaging������������������������������������������������������248
Structural interaction fingerprints (SIFt)����������������� 266, 267
Normal human astrocytes
Structure-based prediction����������������������������������� 62, 65, 124
Nuclear magnetic resonance (NMR) spectroscopy�������������20,
Structure-based virtual screening (SBVS)�������� 260, 263, 265
23, 26–29, 31, 62–65, 67–77, 79–81, 88–90, 92, 101,
Structure-function relationship������������������������������������������89
102, 106, 112
Substrate��������������������������������������� 4, 9–12, 15, 36, 37, 39, 40, 43,
O 53, 88, 112, 113, 116, 120–124, 128, 130, 132, 137–179,
185–197, 199, 200, 209–227, 231–233, 239, 242, 243,
Oligomer����������������������������������������������������������������������������36 265, 269, 271, 298, 299
discovery����������������������������������������������������� 185, 186, 196
P
Paramagnetic relaxation enhancement (PRE)������������� 62, 68, T
71, 75 Time-resolved degradomics���������������������������������������������186
Peptide hydrolysis����������������������������������������������������� 131, 132 Tissue plasminogen activator (tPA)������������������������� 286, 288,
Peripheral membrane protein���������������������������������������61–82 290, 291, 293
Phage display��������������������������������������������137, 209–213, 215, Transmembrane domain��������������������������������������� 17–31, 112
217–221, 224 Triple-helical substrates���������������������116, 139–166, 168, 172
Pichia pastoris����������������������������������������������������������������� 35–44
Protease������������������������������������ 19, 20, 22, 25, 27, 28, 30, 31, Y
35–44, 64, 71, 89, 91, 185–188, 190, 191, 194, 196,
Yeast������������������������������������������������ 5, 20, 36, 37, 50, 80, 214
199–206, 209, 211, 215, 217, 219, 220, 224, 226, 232,
234, 242, 260 Z
Protein
dynamics�����������������������������������������������������������������������88 Zn-chelating inhibitor����������������������������������������������263–265
expression���������������������� 8, 13, 24, 29, 30, 63–64, 67, 210 Zymogram���������������������������������������������������������� 14, 232–234
flexibility���������������������������������������������������������������������122 Zymography������������������������������������6, 9–11, 14, 15, 231–243