Astrocytes in Selective Vulnerability To Neurodege
Astrocytes in Selective Vulnerability To Neurodege
Review
Selective vulnerability of specific brain regions and cell populations is a hallmark Highlights
of neurodegenerative disorders. Mechanisms of selective vulnerability involve Selective vulnerability in neurode-
neuronal heterogeneity, functional specializations, and differential sensitivities generative disorders affects specific
neuronal populations and may involve
to stressors and pathogenic factors. In this review we discuss the growing body
non-neuronal cells, such as astrocytes,
of literature suggesting that, like neurons, astrocytes are heterogeneous and spe- which have multifaceted roles in brain
cialized, respond to and integrate diverse inputs, and induce selective effects on function.
brain function. In disease, astrocytes undergo specific, context-dependent
Astrocytes are phenotypically and func-
changes that promote different pathogenic trajectories and functional outcomes. tionally heterogeneous and responsive
We propose that astrocytes contribute to selective vulnerability through maladap- to diverse signals as a function of age,
tive transitions to context-divergent phenotypes that impair specific brain regions brain region, environmental conditions,
and functions. Further studies on the multifaceted roles of astrocytes in disease and other variables.
may provide new therapeutic approaches to enhance resilience against neurode- Astrocytic responses and contributions
generative disorders. to neural impairments and pathogenic
processes in neurodegenerative condi-
tions are emerging as selective and
context-dependent.
Selective vulnerability is a common feature of neurodegenerative disorders
Neurodegenerative disorders are age-related neurological illnesses that cause progressive Astrocytes may promote selective
decline in behavioral, cognitive, social, and emotional processes. Examples of these disorders vulnerability in neurodegenerative dis-
orders by inducing distinct effects
include Alzheimer’s disease (AD), frontotemporal dementia (FTD), amyotrophic lateral sclerosis across different contexts.
(ALS), Huntington’s disease (HD), and Parkinson’s disease (PD). Neurodegenerative disorders
are characterized by proteinopathy, brain atrophy, neuronal dysfunction and cell death, glial
alterations, neuroimmune responses, and neurovascular changes. The underlying neuropathogenic
molecular pathways are complex and include impairments in proteostasis, RNA processing, gene
expression, metabolic pathways, and intracellular signaling mechanisms [1].
A central feature of all neurodegenerative disorders is the vulnerability of select neuronal populations
and brain regions to progressive dysfunction and pathology, whereas other regions and cell popu-
lations are largely spared pathology or exhibit functional resilience despite the presence of pathology.
In AD, impairments typically begin in pyramidal neurons of the entorhinal cortex and hippocampal
CA1 region, as well as in noradrenergic neurons in the locus coeruleus and regions of the neocortex
1
[2]. In FTD, van Economo neurons and fork cells within the frontoinsular cortex and anterior cingulate Appel Alzheimer’s Disease Research
Institute, Weill Cornell Medicine, New
cortex are more susceptible [3], whereas in ALS spinal cord motor neurons are lost early [4]. In
York, NY, USA
HD, striatal medium spiny GABAergic neurons degenerate [5], whereas PD involves the loss of 2
Feil Family Brain and Mind Research
dopaminergic neurons of the substantia nigra [6]. As these disorders progress, secondary neuronal Institute, Weill Cornell Medicine, New
York, NY, USA
vulnerability, spread of proteinopathy, and various neuropathological cascades continue to spread 3
Neuroscience Graduate Program, Weill
selectively across specific brain regions and neuronal populations. Cornell Medicine, New York, NY, USA
In addition to regional differences, there are also differences in impairments and degeneration
across affected individuals with similar diagnoses and genetic risk factors. Why particular neural
cell populations, brain regions, and individuals are more susceptible to degenerative processes *Correspondence:
and functional decline while others are resilient remains enigmatic. Identifying the processes [email protected] (A.G. Orr).
underlying selective vulnerability is likely crucial for pinpointing early pathogenic mechanisms and
advancing the development of novel prognostic biomarkers, preventative strategies, and thera-
peutics to halt disease onset and progression.
In this review we discuss recent evidence implicating astrocytes in selective vulnerability associated
with neurodegenerative disorders. We first highlight key disease-related mechanisms and the
involvement of non-neuronal cells, and then review the fundamental roles of astrocytes as integra-
tors of diverse cues and dynamic modulators of brain function. Focusing on recent studies in
the context of neurodegenerative disorders and their mechanisms, we describe converging
evidence that astrocytes have context-specific changes and roles in disease that may promote
selective vulnerability. These discoveries have important implications for the understanding of
astrocyte pathobiology and therapeutic development for neurodegenerative disorders.
Specific brain regions and neuronal circuits may be more vulnerable to disease as a function of
their neurotransmitter systems, ion buffering capacity, and other intrinsic factors, which can
cause distinct stress-related responses culminating in excitotoxicity, synapse loss, and cell
death [18–21]. Neurons also depend on efficient energy generation, and recent studies in
mouse models and human neural cells point to mitochondrial dysfunction and changes in neuro-
nal metabolism in degeneration of select neuronal populations [22–24]. Notably, aging is a strong
risk factor for neurodegenerative disorders and modulates many of the mechanisms underlying
neuronal vulnerability to disease [25,26].
In this broader view of disease pathogenesis, astrocytes occupy a crucial niche at the interface of
neurons, other glial cells, the vasculature, and infiltrating immune cells and peripheral factors
[41,42]. Historically, astrocytes have been viewed as largely homogeneous support cells with
similar functions and responses to disease. However, emerging evidence is revealing a rich diver-
sity of astrocytic features and functions within and across brain regions (Figure 2), which is likely
due to their developmental lineage, positional identity, and sensitivity to various local inputs
and cues in their microenvironment [43]. Considering their multifunctional roles in neurovascular
Contributing variables Multicellular interactions Diverse molecular changes Nonlinear and selective pathogenesis
Proteinopathy
Synaptic
impairments Subpopulation A
Age Genetics DNA and RNA
Neurovascular Astrocytic-neuronal dysregulation
Pathogenesis
Trends in Neurosciences
Figure 1. Vulnerability to neurodegenerative disorders is multifactorial. Multiple intrinsic variables affect the onset and progression of neurodegenerative disease,
including age and genetics, but also environmental and lifestyle factors. These and other variables influence the complex multicellular interactions between different cell
types in the brain and their functions. Dynamic molecular changes, detrimental and protective, occur within multiple cell types and affect cell–cell interactions,
contributing to disease complexity. Together, these changes result in nonlinear pathogenesis and selective vulnerability within specific subpopulations of cells, brain
regions, and individuals. Figure created with BioRender.
ATP
ROS and facilitate precise, dynamic, and
multifaceted responses by astrocytes to
Metabolome Metabolism various physiological and pathological
signals. Ultimately, astrocytic responses
have direct and indirect effects on
neuronal function and survival, and
thereby have major roles in the
Surface proteins Local connectivity Neuronal modulation pathogenesis of neurodegenerative
diseases. Abbreviations: ATP, adenosine
triphosphate; ROS, reactive oxygen
species. Figure created with BioRender.
Trends in Neurosciences
dynamics, brain metabolism, neuronal activities, neural stress responses, and neuroimmune cas-
cades, astrocytes are well positioned to affect many of the mechanisms linked to selective
vulnerability or resilience to disease. In the following section, we briefly summarize recent evi-
dence indicating that astrocytes have extensive functional heterogeneity, and respond to stimuli
in a versatile and context-dependent manner. Thereafter, we discuss the potential involvement of
astrocytes in selective pathogenesis linked to neurodegenerative disorders.
Astrocytes integrate numerous cues and have diverse features and functions
Astrocytes are prevalent glial cells that tile the brain and are estimated to contact hundreds of
thousands of synapses and interact with microglia, oligodendrocytes, and neurovascular ele-
ments [44]. Astrocytes are involved in a wide range of normal and pathogenic processes [45]
through diverse arrays of receptors, channels, adhesion molecules, and other surface factors
that sense neurotransmitters, cytokines, growth factors, and other types of molecules pro-
duced by surrounding cells, afferent synaptic inputs, and the periphery [46,47]. In response
to these various cues, astrocytes regulate synaptic and neural circuit functions, other glial
cells, vascular elements, and homeostatic processes. Historically, astrocytes were frequently
studied in isolation or in the context of injury or disease, which can alter their phenotype and
function. In addition, long-enduring dogmas asserting that astrocytes are nonexcitable and
slow to respond have perpetuated a notion that they are unable to effectively detect, store,
and transmit behaviorally relevant neural information. These challenges, together with the slow de-
velopment of tools to precisely target and manipulate astrocytes in vivo, have delayed our under-
standing of astrocyte biology. However, recent studies using more advanced methods suggest
that astrocytes are crucial integrators and conductors of brain function, including behavior, in
part due to their intimate associations with neurons and other cell types and their precise effects
on neural function.
Astrocytes, like neurons, are heterogeneous and have region-specific morphological and molecular
signatures, as well as region- and context-specific calcium dynamics and neurotransmitter release
properties [48–50]. Astrocytes regulate diverse neuronal functions, such as basal synaptic strength
in hippocampal CA1 neurons, inhibition in cortical circuits related to goal-directed behaviors, and
modulation of sensory transmission in arousal and sleep [51–53], as established primarily in rodent
models. Astrocytes exhibit distinct functional properties involving fast and slow signaling mecha-
nisms [53,54], and modulate neural network excitation and synchronization required for sensory
information processing, storage, and behavioral output [55,56]. Indeed, in mouse models, astrocytic
signaling has been shown to regulate cognitive function and behavior [57,58], including learning and
memory-linked processes [59–61]. Aberrant changes in the activation of astrocytic G-protein-
coupled signaling can lead to neural hyperactivity and specific behavioral deficits [59,62–64]. Studies
are also revealing astrocytic contributions to social and emotion-related behaviors and neuro-
psychiatric conditions, as recently reviewed [65].
Evidence in rodent and ferret models suggests that astrocyte engagement in neural circuit activities
is selective, dynamic, and dependent on context. Astrocytes in the striatum can discriminate and
differentially regulate neuronal activities in specific circuits despite being physically intermingled
with multiple circuits [66], and these astrocytes lack evoked calcium-based responses to neocor-
tical inputs, but gain aberrant evoked responsiveness in the context of HD-related pathology [67].
Somewhat similarly, astrocytes in the neocortex discriminate between cholinergic inputs from
neuronal populations in the nucleus basalis activated by visual stimuli [68], tune their responses
to distinct features of visual stimuli [69], and shift their responsiveness and calcium signaling
patterns based on the sleep–wake cycle and levels of norepinephrine [49,53,70,71]. In addition,
astrocytes in the CA1 region of the hippocampus can selectively modulate CA1 neurons projecting
to the anterior cingulate cortex but not those projecting to the nucleus accumbens, thereby
modulating remote memory [62]. Moreover, CA1 astrocytes participate in frequency-specific
dendritic integration [72]. Thus, in contrast to previous views that astrocytes are functionally homo-
geneous, emerging studies reveal a high degree of context-dependent functional precision and
adaptation in astrocytic–neuronal interactions across a range of circuits and domains [73].
general markers of astrocyte reactivity, such as increased glial fibrillary acidic protein (GFAP)
expression, have historically served as indicators of astrocytic alterations, the use of binary charac-
terization (e.g., normal or reactive) is now recognized as insufficient and potentially misleading due
to the diversity of astrocytic changes and their influence across different contexts [86,87]. For
example, in mouse models, astrocytes in the spinal cord and striatum adopt diverse and dynamic
reactive states across different pathological contexts through changes in the activities of multiple
transcriptional regulators, as assessed by chromatin accessibility measures, RNA sequencing,
and gene perturbation [88,89].
Aging and neuroinflammation are major risk factors for neurodegenerative disorders and facilitate
disease onset and progression [25,90]. The effects of aging and immune responses on glial cell
phenotypes are brain-region-specific and appear to be pronounced in the white matter [32,91].
Aging induces divergent transcriptional profiles in astrocytes within different brain regions and is
associated with diminished astrocyte-mediated synaptic plasticity in mice and humans [92–94].
It has not been firmly established, however, whether certain aging-induced astrocytic changes in
specific brain areas promote regional vulnerability or resilience to dementia-linked pathology, and
if so, how. In animal models, astrocytes also have regional and time-dependent responses in
lipopolysaccharide (LPS)-induced neuroinflammation, which induces distinct subpopulations of
astrocytes with differing transcriptional profiles [95,96]. Therefore, spatiotemporal differences in
astrocytic phenotypes consequent to aging and inflammation may contribute to differential brain
vulnerability to disease, possibly synergizing with specific neuronal alterations in susceptible
regions and giving rise to selective and localized neurodegenerative cascades. In the following
we highlight further evidence that astrocytes are differentially affected by dementia-associated
factors and pathological processes, and in turn influence neuronal functions and pathogenic trajec-
tories in a selective and context-dependent manner (Figure 3).
Proteinopathy
Astrocytes can differentially modulate proteinopathy and its effects on brain function. The uptake
and clearance of protein aggregates by astrocytes can become progressively impaired in patho-
logical conditions [97]. Astrocytes take up protein aggregates to either degrade them intracellu-
larly or deliver them to waste removal systems, although alternative pathways might also exist.
Brain clearance of protein aggregates depends in part on bulk flow through perivascular spaces
[98], and water movement through astrocytic aquaporin 4 (AQP4) channels is hypothesized to
facilitate Aβ removal [99–101]. Notably, AQP4 levels are reduced in astrocytic endfeet in mouse
models of AD pathology and in individuals with AD or cerebral amyloid angiopathy [102,159],
which is characterized by aberrant Aβ accumulation in perivascular spaces. Reductions in clear-
ance are associated with increased Aβ load in humans [103], implicating changes in astrocytic
clearance mechanisms in AD pathogenesis. AQP4 levels differ among astrocyte subpopulations
and subcompartments, with AQP4 enrichment in astrocytic endfeet [104,105]. Differences in AQP4
function may promote local variations in Aβ clearance and affect the capacity of brain regions to
cope with protein accumulation, thereby correlating with pathogenesis and neurocognitive changes.
In contrast to AQP4 reductions in AD, AQP4 levels are enhanced in FTD linked to progranulin
mutations [106], highlighting potential disease-specific changes in clearance mechanisms.
(A) Differential protein clearance Physiology Pathology (B) Differential metabolic load Physiology Pathology
Cortex Tau
Cortex
10mV
50ms
Aβ
α-syn Resilient
AQP4 Hyperexcitability
Other
Low metabolic load
Vulnerable
Hyperexcitability
(C) Differential neurotransmission effects FTD Injury (D) Differential synapse maintenance HD
Impaired
GPCRs
10mV
50ms
10mV
mV
V
50
0ms
10m
mV
mV
50
50
0ms synapse growth
α β/γ Altered
Glutamate GABA Striatum Gi/o signaling
Thalamus
Gene
1 0m V
10mV
expression
50ms
50m
5 0m
0 m
AD / FTD AD Aging
Aβ Impaired pruning
and growth
TDP-43
Chemokines Hippocampus
Hippocampus GABA Reduced phagocytosis
and synaptogenic factors
Disrupted
Presynaptic changes Tonic inhibition proteosomes
Aberrant excitation and lysosomes
Trends in Neurosciences
Figure 3. Astrocytes may contribute to selective vulnerability in disease through divergent, context-dependent phenotypes that affect specific brain
regions and functions. (A) Aberrant proteins implicated in disease are cleared from the brain through various mechanisms, including aquaporin-4 (AQP4)-dependent
protein removal. Astrocytes in basal brain regions may have a higher clearance burden and, in conjunction with changes in AQP4 localization and expression, might
promote selective vulnerability in the basal regions. (B) Cortical astrocytes contact more blood vessels and fewer synapses per cell than hippocampal astrocytes. The
resulting higher metabolic load carried by hippocampal astrocytes may predispose hippocampal synapses and other neural elements to metabolic vulnerability and the
detrimental effects of neural hyperexcitability. (C) Astrocytes regulate neurotransmission in a state-dependent and region-specific manner. In frontotemporal dementia
(FTD), thalamic astrocytes may remove less glutamate via surface transporters and promote synaptic loss and, in brain injury, remove less GABA and induce
hyperexcitability. Aberrant TDP-43 accumulation in hippocampal astrocytes promotes region-specific expression of chemokines that induce presynaptic changes and
aberrant excitatory transmission. Hippocampal astrocytes in Alzheimer’s disease (AD) overproduce GABA and impair neuronal function through increases in tonic
inhibition. (D) Astrocytes can differentially modulate synaptic formation and loss. Striatal astrocytes in the context of Huntington’s disease (HD)-related pathology have
altered Gi/o-coupled signaling leading to gene expression changes that impair synaptogenesis. In aging, subpopulations of hippocampal astrocytes have disrupted
proteosomes and lysosomes, which impairs synaptic pruning and secretion of synaptogenic factors. Abbreviations: α-syn, α-synuclein; Aβ, amyloid-β; Gi/o, Gi/o-coupled
receptor signaling; GPCRs, G-protein-coupled receptors; TDP-43, transactive response DNA-binding protein of 43 kDa. Figure created with BioRender.
phagocytic and AQP4-dependent clearance mechanisms may synergize to hasten the accumula-
tion and aggregation of aberrant proteins. Most cerebrospinal fluid (CSF) is hypothesized to drain at
the base of the brain via lymphatic vessels running in parallel to cranial nerves [111]. Accordingly,
despite generally similar levels of astrocytic AQP4 throughout the brain, there is likely a high local
burden on astrocytic clearance mechanisms in basal brain regions. Interestingly, these regions
are also among the earliest to display signs of Aβ, tau, and α-synuclein accumulation in humans,
including basal brain regions and midbrain, suggesting that astrocytes in those regions could be
affected by aging and other risk factors and promote regional vulnerability to altered proteostasis
and onset of pathogenic cascades that subsequently spread to other brain areas.
homeostasis, and that genetic deletion of mutant SOD1 selectively in mouse astrocytes slows
disease progression [112]. Moreover, in mouse models involving targeted viral vector manipula-
tions, tau accumulation in hilar astrocytes of the hippocampus was sufficient to cause bioener-
getic failure, reductions in neurogenesis, and selective impairments in inhibitory neurons [113].
Similarly, TDP-43 accumulation induced in hippocampal astrocytes of adult mice causes
progressive memory loss through alterations in astrocytic antiviral pathways and selective impair-
ments in presynaptic function [114]. Of note, learning, motor function, and innate behaviors in
mice were not affected by astrocytic TDP-43 mislocalization, even when occurring throughout
the brain [114], suggesting that TDP-43 pathology in astrocytes has region-selective effects on
brain function. Mutant huntingtin and tau also disturb astrocytic cholesterogenesis, which is
essential for neuronal function [115–117]. Therefore, heterogeneous proteinopathy in astrocytes,
along with local variations in BBB-linked clearance mechanisms, could result in selective
vulnerabilities in protein handling and specific types of neuronal and neurocognitive impair-
ments. In support, correlational analyses of Aβ, tau, and astrocyte pathology, as assessed
by plasma GFAP levels, suggest that astrocytic alterations in humans may reflect an early
pathogenic transition in preclinical AD, predicting tau pathology and thereby linking Aβ
accumulation to clinical symptoms [118].
Metabolic pathways
Neurodegenerative disorders also involve changes in astrocytic metabolic pathways [119–122].
Astrocytes supply essential metabolites to neurons and other cells, including lactate for energy,
cholesterol for membrane homeostasis, glutamine as a precursor for neurotransmitters, and
factors to maintain redox homeostasis [123,124]. Astrocytes also regulate cerebral blood flow
to sustain neuronal energy and potentially integrate peripheral metabolic cues [47,125].
Moreover, astrocyte subpopulations have been identified with varying expression of genes
involved in energy-producing processes, including glycolysis and oxidative phosphorylation,
functions that might be compromised in aging, according to transcriptomic studies in mice
[32,93], and these differential bioenergetic states could promote impairments in select circuits
or brain regions.
Astrocytes are also involved in maintaining synaptic structure and density through the release of
synaptogenic factors and active elimination of synapses. These functions can vary across regions
and types of synapses. In aging mice, a specific population of hippocampal astrocytes has
impaired autophagy and synaptogenic mechanisms for regulating synapses [109]. In a mouse
model of HD, dysfunctional Gi/o-coupled receptor signaling in striatal astrocytes can selectively
dysregulate synaptogenesis and cause impairments in attention-associated behaviors [88]. In
mouse models and human cases of ALS, a selective loss of tripartite synapses may occur,
pointing to greater vulnerability of synapses containing astrocytic processes as compared with
bipartite synapses [146]. Human and mouse astrocytes are also implicated in increased AD
pathology-associated synapse elimination [147]. Of note, astrocytic synapse elimination in a
mouse model of tauopathy preferentially targets excitatory synapses over inhibitory synapses,
which may be engulfed more by microglia [148]. Astrocytes can also modulate microglia-
mediated synapse elimination [149] and could influence microglial roles in brain resilience to
tauopathy [31], but these potential astrocytic–microglial interactions require further investigation.
In addition to enhanced synapse removal, there is also evidence that astrocytes in transgenic
mice with amyloid pathology are impaired in the clearance of dystrophic synapses [150]. In
summary, astrocytes are known to influence synaptic structure and function, and recent studies
suggest that these effects are likely region-specific and selective, mediated by distinct astrocytic
subpopulations, differentially affected by pathogenic cascades, and potentially intertwined with
microglial activities.
Trends in Neurosciences
Figure 4. Astrocytes promote context-dependent effects in health and disease. (A) Astrocytes are sensitive to various cues in their microenvironment and have
diverse molecular, structural, and functional states as a result of the specific combination of cues received by individual astrocytes. These combinatorial inputs instruct
astrocytes to assume various context-integrated states that enable normal brain function. (B) In disease and other conditions, aberrant inputs and perturbations affect
individual astrocytes differently and may shift subsets or subcompartments of astrocytes into context-divergent states that selectively enhance or disrupt specific
aspects of brain function. Figure created with BioRender.
Further investigation of the exact molecular and cellular basis for selective vulnerability holds great Outstanding questions
potential for elucidating disease pathogenesis and developing more effective treatment options, The mechanisms by which astrocytes
with the possibility for earlier and more precise interventions. With regard to the contributions of regulate various types of disease
processes and neural impairments
astrocytes to disease progression, recent studies argue against the notion that disease-related
are incompletely understood. When
impairments are triggered largely by dysfunction in neuronal or other glial populations with and how do astrocytes engage
secondary or ancillary responses in astrocytes. Instead, studies are converging on a theory in different pathological cascades,
that dysfunctions in astrocytes are operating in concert with neuronal predispositions and poten- synaptic alterations, and neural circuit
dysfunctions?
tial abnormalities in other cells to trigger selective neurodegenerative cascades and progressive
behavioral alterations. It remains unclear whether astrocytes
are a diverse population with
Considering this emerging view, effective therapeutic strategies may require targeting of astrocytic developmentally defined and constant
states, whether they exhibit dynamic
functions that are linked to specific neuronal circuits most susceptible to impairment in a particular
shifts in function that generate
type of disorder and patient population. Whether targeting a discrete disease-associated astrocyte apparent heterogeneity, or whether
subcluster or a broader astrocyte network within a given brain region is required for therapeutic both scenarios are occurring in vivo.
efficacy remains an important open question. Further investigations are required to unravel exactly What are the exact triggers and
regulatory mechanisms that promote
how different astrocytic subpopulations, compartments, and functional states are organized and regional astrocyte heterogeneity in
dynamically tuned to different brain activities, and how these astrocytic states are selectively health and disease?
compromised and could be therapeutically targeted in neurodegenerative conditions.
What are the effects of disease-
linked astrocytic changes on specific
Concluding remarks and future perspectives neuronal subpopulations that are
Mounting evidence of astrocytic heterogeneity and functional selectivity suggests that astrocytes known to be most vulnerable to dis-
could promote selective vulnerability in disease. Significant progress has been made in broadly ease? What is the nature of astrocyte
contributions to cognitive and behavioral
profiling molecular changes and uncovering novel mechanisms that mediate astrocytic–neuronal
deficits across disorders and what
crosstalk and effects on pathogenic processes. However, an integrated understanding of are their specific pathophysiological
astrocytic and neuronal functions and their interconnected changes in disease is still lacking. mechanisms?
Additional studies are needed that address how astrocytes differentially regulate and contribute
Individual astrocytes have structural
to brain function and pathology in distinct biological and environmental contexts (see and functional subcompartments,
Outstanding questions). In elucidating these roles, it is important to be mindful of the limitations including endfeet and leaflets. What
of current models of neurodegenerative disorders and to explore neuropathological evidence are the disease-linked changes in
and multiomic datasets from human samples and use human iPSC-derived systems when these subcellular compartments,
and how do these changes affect
possible. Human astrocytes can differ from mouse astrocytes in certain morphological features, presynaptic and postsynaptic ele-
metabolic changes, and inflammatory pathways, emphasizing the importance of cross-species ments, neurovascular cells, and other
validation [157,158]. Future studies should investigate the roles of circuit-specific and molecularly glia?
defined subpopulations of astrocytes, take advantage of advanced genetically encoded indica-
Astrocytes are the primary cell type
tors and actuators, and rigorously compare astrocytic features and functions across different bi- involved in the synthesis of brain
ological and pathogenic contexts. lipoproteins and lipids, including ApoE
and cholesterol, which are essential
for brain function and implicated in
Acknowledgments
neurodegenerative disorders. How is
This work was supported by the National Institutes of Health: R01AG068091 (A.G.O.) and RF1NS118569 (A.G.O.),
astrocytic lipid metabolism and lipid-
BrightFocus Foundation: BFA2023008F (T.S.Z.), and the Alzheimer’s Association: 23AARF-1029892 (T.S.Z.). based crosstalk with other cell types
regulated by neuronal and neurovascular
Declaration of interests activities and altered by pathological
The authors declare no competing interests in relation to this work. processes?
7. Fu, H. et al. (2018) Selective vulnerability in neurodegenerative 34. Goutman, S.A. et al. (2022) Emerging insights into the complex potential differences between sexes.
diseases. Nat. Neurosci. 21, 1350–1358 genetics and pathophysiology of amyotrophic lateral sclerosis. Are astrocytic effects on cognition and
8. Roussarie, J.P. et al. (2020) Selective neuronal vulnerability in Lancet Neurol. 21, 465–479
disease pathogenesis sex-dependent?
Alzheimer's disease: a network-based analysis. Neuron 107, 35. Simons, M. et al. (2023) Tipping points in neurodegeneration.
821–835 e812 Neuron 111, 2954–2968
9. Silva, M.C. et al. (2020) Prolonged tau clearance and stress 36. Murdock, M.H. and Tsai, L.H. (2023) Insights into Alzheimer's
vulnerability rescue by pharmacological activation of autophagy disease from single-cell genomic approaches. Nat. Neurosci.
in tauopathy neurons. Nat. Commun. 11, 3258 26, 181–195
10. Bourdenx, M. et al. (2021) Chaperone-mediated autophagy 37. Chatterjee, P. et al. (2021) Plasma glial fibrillary acidic protein is
prevents collapse of the neuronal metastable proteome. Cell elevated in cognitively normal older adults at risk of Alzheimer's
184, 2696–2714 e2625 disease. Transl. Psychiatry 11, 27
11. Nana, A.L. et al. (2019) Neurons selectively targeted in fronto- 38. Barker, S.J. et al. (2021) MEF2 is a key regulator of cognitive
temporal dementia reveal early stage TDP-43 pathobiology. potential and confers resilience to neurodegeneration. Sci.
Acta Neuropathol. 137, 27–46 Transl. Med. 13, eabd7695
12. Lin, L.C. et al. (2019) Preferential tau aggregation in von 39. Faraco, G. et al. (2019) Dietary salt promotes cognitive impair-
Economo neurons and fork cells in frontotemporal lobar ment through tau phosphorylation. Nature 574, 686–690
degeneration with specific MAPT variants. Acta Neuropathol. 40. Choi, S.H. et al. (2018) Combined adult neurogenesis and
Commun. 7, 159 BDNF mimic exercise effects on cognition in an Alzheimer's
13. Lau, A. et al. (2020) alpha-Synuclein strains target distinct brain mouse model. Science 361, aan8821
regions and cell types. Nat. Neurosci. 23, 21–31 41. Sanmarco, L.M. et al. (2021) Functional immune cell–astrocyte
14. Vogel, J.W. et al. (2023) Connectome-based modelling of interactions. J. Exp. Med. 218, e20202715
neurodegenerative diseases: towards precision medicine and 42. Brandebura, A.N. et al. (2023) Astrocyte contribution to dys-
mechanistic insight. Nat. Rev. Neurosci. 24, 620–639 function, risk and progression in neurodegenerative disorders.
15. Vaquer-Alicea, J. and Diamond, M.I. (2019) Propagation of protein Nat. Rev. Neurosci. 24, 23–39
aggregation in neurodegenerative diseases. Annu. Rev. Biochem. 43. Khakh, B.S. and Deneen, B. (2019) The emerging nature of
88, 785–810 astrocyte diversity. Annu. Rev. Neurosci. 42, 187–207
16. Uemura, N. et al. (2020) Cell-to-cell transmission of tau and 44. Torres-Ceja, B. and Olsen, M.L. (2022) A closer look at astro-
alpha-synuclein. Trends Mol. Med. 26, 936–952 cyte morphology: development, heterogeneity, and plasticity
17. Arboleda-Velasquez, J.F. et al. (2019) Resistance to autosomal at astrocyte leaflets. Curr. Opin. Neurobiol. 74, 102550
dominant Alzheimer's disease in an APOE3 Christchurch ho- 45. Blanco-Suarez, E. et al. (2017) Role of astrocyte–synapse inter-
mozygote: a case report. Nat. Med. 25, 1680–1683 actions in CNS disorders. J. Physiol. 595, 1903–1916
18. Selvaraj, B.T. et al. (2018) C9ORF72 repeat expansion causes 46. Sofroniew, M.V. (2020) Astrocyte reactivity: subtypes, states,
vulnerability of motor neurons to Ca(2+)-permeable AMPA and functions in CNS innate immunity. Trends Immunol. 41,
receptor-mediated excitotoxicity. Nat. Commun. 9, 347 758–770
19. Zhang, F. et al. (2020) β-Amyloid redirects norepinephrine 47. Nampoothiri, S. et al. (2022) Glial cells as integrators of peripheral
signaling to activate the pathogenic GSK3beta/tau cascade. and central signals in the regulation of energy homeostasis. Nat.
Sci. Transl. Med. 12, eaay6931 Metab. 4, 813–825
20. Leng, K. et al. (2021) Molecular characterization of selectively 48. Lanjakornsiripan, D. et al. (2018) Layer-specific morphological
vulnerable neurons in Alzheimer's disease. Nat. Neurosci. 24, and molecular differences in neocortical astrocytes and their
276–287 dependence on neuronal layers. Nat. Commun. 9, 1623
21. Zhao, R. et al. (2022) Activity disruption causes degeneration of 49. Tsunematsu, T. et al. (2021) Region-specific and state-
entorhinal neurons in a mouse model of Alzheimer’s circuit dys- dependent astrocyte Ca(2+) dynamics during the sleep–wake
function. eLife 11, e83813 cycle in mice. J. Neurosci. 41, 5440–5452
22. Traxler, L. et al. (2022) Warburg-like metabolic transformation 50. de Ceglia, R. et al. (2023) Specialized astrocytes mediate gluta-
underlies neuronal degeneration in sporadic Alzheimer's disease. matergic gliotransmission in the CNS. Nature 622, 120–129
Cell Metab. 34, 1248–1263 e1246 51. Chipman, P.H. et al. (2021) Astrocyte GluN2C NMDA receptors
23. Moya, M.V. et al. (2022) Unique molecular features and cellu- control basal synaptic strengths of hippocampal CA1 pyramidal
lar responses differentiate two populations of motor cortical neurons in the stratum radiatum. eLife 10, e70818
layer 5b neurons in a preclinical model of ALS. Cell Rep. 38, 52. Mederos, S. et al. (2021) GABAergic signaling to astrocytes in
110556 the prefrontal cortex sustains goal-directed behaviors. Nat.
24. Gonzalez-Rodriguez, P. et al. (2021) Disruption of mitochondrial Neurosci. 24, 82–92
complex I induces progressive parkinsonism. Nature 599, 650–656 53. Wang, F. et al. (2023) Distinct astrocytic modulatory roles in
25. Hou, Y. et al. (2019) Ageing as a risk factor for neurodegenerative sensory transmission during sleep, wakefulness, and arousal
disease. Nat. Rev. Neurol. 15, 565–581 states in freely moving mice. Nat. Commun. 14, 2186
26. Wegmann, S. et al. (2019) Experimental evidence for the age depen- 54. Stobart, J.L. et al. (2018) Cortical circuit activity evokes rapid
dence of tau protein spread in the brain. Sci. Adv. 5, eaaw6404 astrocyte calcium signals on a similar timescale to neurons.
27. Sun, N. et al. (2023) Single-nucleus multiregion transcriptomic Neuron 98, 726–735 e724
analysis of brain vasculature in Alzheimer's disease. Nat. 55. Lines, J. et al. (2020) Astrocytes modulate sensory-evoked
Neurosci. 26, 970–982 neuronal network activity. Nat. Commun. 11, 3689
28. Gleichman, A.J. and Carmichael, S.T. (2020) Glia in neurode- 56. Reitman, M.E. et al. (2023) Norepinephrine links astrocytic
generation: drivers of disease or along for the ride? Neurobiol. activity to regulation of cortical state. Nat. Neurosci. 26, 579–593
Dis. 142, 104957 57. Santello, M. et al. (2019) Astrocyte function from information
29. Sweeney, M.D. et al. (2018) The role of brain vasculature in processing to cognition and cognitive impairment. Nat. Neurosci.
neurodegenerative disorders. Nat. Neurosci. 21, 1318–1331 22, 154–166
30. Park, L. et al. (2020) Tau induces PSD95-neuronal NOS uncoupling 58. Akther, S. and Hirase, H. (2022) Assessment of astrocytes as a
and neurovascular dysfunction independent of neurodegeneration. mediator of memory and learning in rodents. Glia 70,
Nat. Neurosci. 23, 1079–1089 1484–1505
31. Udeochu, J.C. et al. (2023) Tau activation of microglial cGAS-IFN 59. Orr, A.G. et al. (2015) Astrocytic adenosine receptor A2A and
reduces MEF2C-mediated cognitive resilience. Nat. Neurosci. 26, Gs-coupled signaling regulate memory. Nat. Neurosci. 18,
737–750 423–434
32. Hahn, O. et al. (2023) Atlas of the aging mouse brain reveals 60. Robin, L.M. et al. (2018) Astroglial CB(1) receptors determine
white matter as vulnerable foci. Cell 186, 4117–4133 e4122 synaptic D-serine availability to enable recognition memory.
33. Blanchard, J.W. et al. (2022) APOE4 impairs myelination via Neuron 98, 935–944 e935
cholesterol dysregulation in oligodendrocytes. Nature 611, 61. Doron, A. et al. (2022) Hippocampal astrocytes encode reward
769–779 location. Nature 609, 772–778
62. Kol, A. et al. (2020) Astrocytes contribute to remote memory 88. Yu, X. et al. (2020) Context-specific striatal astrocyte molecu-
formation by modulating hippocampal–cortical communication lar responses are phenotypically exploitable. Neuron 108,
during learning. Nat. Neurosci. 23, 1229–1239 1146–1162 e1110
63. Nagai, J. et al. (2021) Specific and behaviorally consequential 89. Burda, J.E. et al. (2022) Divergent transcriptional regulation of
astrocyte G(q) GPCR signaling attenuation in vivo with astrocyte reactivity across disorders. Nature 606, 557–564
ibetaARK. Neuron 109, 2256–2274 e2259 90. Chen, X. and Holtzman, D.M. (2022) Emerging roles of innate
64. Delepine, C. et al. (2023) Differential effects of astrocyte manipula- and adaptive immunity in Alzheimer's disease. Immunity 55,
tions on learned motor behavior and neuronal ensembles in the 2236–2254
motor cortex. J. Neurosci. 43, 2696–2713 91. Allen, W.E. et al. (2023) Molecular and spatial signatures of mouse
65. Shigetomi, E. and Koizumi, S. (2023) The role of astrocytes in brain aging at single-cell resolution. Cell 186, 194–208 e118
behaviors related to emotion and motivation. Neurosci. Res. 92. Soreq, L. et al. (2017) Major shifts in glial regional identity are a
187, 21–39 transcriptional hallmark of human brain aging. Cell Rep. 18,
66. Martin, R. et al. (2015) Circuit-specific signaling in astrocyte– 557–570
neuron networks in basal ganglia pathways. Science 349, 93. Boisvert, M.M. et al. (2018) The aging astrocyte transcriptome
730–734 from multiple regions of the mouse brain. Cell Rep. 22,
67. Jiang, R. et al. (2016) Dysfunctional calcium and glutamate 269–285
signaling in striatal astrocytes from Huntington's disease 94. Csemer, A. et al. (2023) Astrocyte- and NMDA receptor-
model mice. J. Neurosci. 36, 3453–3470 dependent slow inward currents differently contribute to synaptic
68. Chen, N. et al. (2012) Nucleus basalis-enabled stimulus-specific plasticity in an age-dependent manner in mouse and human
plasticity in the visual cortex is mediated by astrocytes. Proc. neocortex. Aging Cell 22, e13939
Natl. Acad. Sci. USA 109, E2832–E2841 95. Diaz-Castro, B. et al. (2021) Molecular and functional properties
69. Schummers, J. et al. (2008) Tuned responses of astrocytes of cortical astrocytes during peripherally induced neuroinflam-
and their influence on hemodynamic signals in the visual cortex. mation. Cell Rep. 36, 109508
Science 320, 1638–1643 96. Hasel, P. et al. (2021) Neuroinflammatory astrocyte subtypes in
70. Paukert, M. et al. (2014) Norepinephrine controls the mouse brain. Nat. Neurosci. 24, 1475–1487
astroglial responsiveness to local circuit activity. Neuron 82, 97. Smethurst, P. et al. (2022) The role of astrocytes in prion-like
1263–1270 mechanisms of neurodegeneration. Brain 145, 17–26
71. Bojarskaite, L. et al. (2020) Astrocytic Ca(2+) signaling is 98. Rasmussen, M.K. et al. (2022) Fluid transport in the brain. Physiol.
reduced during sleep and is involved in the regulation of slow Rev. 102, 1025–1151
wave sleep. Nat. Commun. 11, 3240 99. Sapkota, D. et al. (2022) Aqp4 stop codon readthrough
72. Bohmbach, K. et al. (2022) An astrocytic signaling loop for facilitates amyloid-beta clearance from the brain. Brain 145,
frequency-dependent control of dendritic integration and 2982–2990
spatial learning. Nat. Commun. 13, 7932 100. Simon, M. et al. (2022) Loss of perivascular aquaporin-4 localiza-
73. Murphy-Royal, C. et al. (2023) A conceptual framework for tion impairs glymphatic exchange and promotes amyloid beta
astrocyte function. Nat. Neurosci. 26, 1848–1856 plaque formation in mice. Alzheimers Res. Ther. 14, 59
74. Farmer, W.T. et al. (2016) Neurons diversify astrocytes in the 101. Gomolka, R.S. et al. (2023) Loss of aquaporin-4 results in
adult brain through sonic hedgehog signaling. Science 351, glymphatic system dysfunction via brain-wide interstitial fluid
849–854 stagnation. eLife 12, e82232
75. Hasel, P. et al. (2017) Neurons and neuronal activity control 102. Greenberg, S.M. et al. (2020) Cerebral amyloid angiopathy and
gene expression in astrocytes to regulate their development Alzheimer disease – one peptide, two pathways. Nat. Rev.
and metabolism. Nat. Commun. 8, 15132 Neurol. 16, 30–42
76. Farhy-Tselnicker, I. et al. (2021) Activity-dependent modula- 103. de Leon, M.J. et al. (2017) Cerebrospinal fluid clearance in
tion of synapse-regulating genes in astrocytes. eLife 10, Alzheimer disease measured with dynamic PET. J. Nucl. Med.
e70514 58, 1471–1476
77. Chai, H. et al. (2017) Neural circuit-specialized astrocytes: 104. Boulay, A.C. et al. (2017) Translation in astrocyte distal
transcriptomic, proteomic, morphological, and functional processes sets molecular heterogeneity at the gliovascular
evidence. Neuron 95, 531–549 e539 interface. Cell Discov. 3, 17005
78. Endo, F. et al. (2022) Molecular basis of astrocyte diversity and 105. Batiuk, M.Y. et al. (2020) Identification of region-specific astro-
morphology across the CNS in health and disease. Science cyte subtypes at single cell resolution. Nat. Commun. 11, 1220
378, eadc9020 106. Marsan, E. et al. (2023) Astroglial toxicity promotes
79. Bayraktar, O.A. et al. (2020) Astrocyte layers in the mammalian synaptic degeneration in the thalamocortical circuit in fronto-
cerebral cortex revealed by a single-cell in situ transcriptomic temporal dementia with GRN mutations. J. Clin. Invest. 133,
map. Nat. Neurosci. 23, 500–509 JCI164919
80. Tan, C.X. et al. (2023) delta-Catenin controls astrocyte morpho- 107. Szebenyi, K. et al. (2021) Human ALS/FTD brain organoid slice
genesis via layer-specific astrocyte-neuron cadherin interactions. cultures display distinct early astrocyte and targetable neuronal
J. Cell Biol. 222, e202303138 pathology. Nat. Neurosci. 24, 1542–1554
81. Karpf, J. et al. (2022) Dentate gyrus astrocytes exhibit layer- 108. Jiwaji, Z. et al. (2022) Reactive astrocytes acquire neuroprotective
specific molecular, morphological and physiological features. as well as deleterious signatures in response to Tau and Abeta
Nat. Neurosci. 25, 1626–1638 pathology. Nat. Commun. 13, 135
82. Viana, J.F. et al. (2023) Astrocyte structural heterogeneity in the 109. Lee, E. et al. (2022) A distinct astrocyte subtype in the aging
mouse hippocampus. Glia 71, 1667–1682 mouse brain characterized by impaired protein homeostasis.
83. Soto, J.S. et al. (2023) Astrocyte–neuron subproteomes and Nat. Aging 2, 726–741
obsessive–compulsive disorder mechanisms. Nature 616, 110. Chandrasekaran, A. et al. (2021) Astrocytic reactivity triggered
764–773 by defective autophagy and metabolic failure causes neurotox-
84. Bindocci, E. et al. (2017) Three-dimensional Ca(2+) imaging icity in frontotemporal dementia type 3. Stem Cell Reports 16,
advances understanding of astrocyte biology. Science 356, 2736–2751
aai8185 111. Ma, Q. et al. (2017) Outflow of cerebrospinal fluid is predomi-
85. Salmon, C.K. et al. (2023) Organizing principles of astrocytic nantly through lymphatic vessels and is reduced in aged mice.
nanoarchitecture in the mouse cerebral cortex. Curr. Biol. 33, Nat. Commun. 8, 1434
957–972 e955 112. Yamanaka, K. et al. (2008) Astrocytes as determinants of
86. Escartin, C. et al. (2021) Reactive astrocyte nomenclature, disease progression in inherited amyotrophic lateral sclerosis.
definitions, and future directions. Nat. Neurosci. 24, 312–325 Nat. Neurosci. 11, 251–253
87. Patani, R. et al. (2023) Functional roles of reactive astrocytes in 113. Richetin, K. et al. (2020) Tau accumulation in astrocytes of the
neuroinflammation and neurodegeneration. Nat. Rev. Neurol. dentate gyrus induces neuronal dysfunction and memory def-
19, 395–409 icits in Alzheimer's disease. Nat. Neurosci. 23, 1567–1579
114. Licht-Murava, A. et al. (2023) Astrocytic TDP-43 dysregulation 137. Kaplan, L. et al. (2020) Neuronal regulation of the blood–brain barrier
impairs memory by modulating antiviral pathways and interferon- and neurovascular coupling. Nat. Rev. Neurosci. 21, 416–432
inducible chemokines. Sci. Adv. 9, eade1282 138. Hosli, L. et al. (2022) Direct vascular contact is a hallmark of
115. Benraiss, A. et al. (2021) Cell-intrinsic glial pathology is cerebral astrocytes. Cell Rep. 39, 110599
conserved across human and murine models of Huntington's 139. Palop, J.J. and Mucke, L. (2016) Network abnormalities and
disease. Cell Rep. 36, 109308 interneuron dysfunction in Alzheimer disease. Nat. Rev. Neurosci.
116. Glasauer, S.M.K. et al. (2022) Human tau mutations in cerebral 17, 777–792
organoids induce a progressive dyshomeostasis of cholesterol. 140. Mathys, H. et al. (2019) Single-cell transcriptomic analysis of
Stem Cell Reports 17, 2127–2140 Alzheimer's disease. Nature 570, 332–337
117. Gangwani, M.R. et al. (2023) Neuronal and astrocytic contribu- 141. Cooper, M.L. et al. (2020) Redistribution of metabolic resources
tions to Huntington's disease dissected with zinc finger protein through astrocyte networks mitigates neurodegenerative
transcriptional repressors. Cell Rep. 42, 111953 stress. Proc. Natl. Acad. Sci. USA 117, 18810–18821
118. Bellaver, B. et al. (2023) Astrocyte reactivity influences amyloid- 142. Semyanov, A. and Verkhratsky, A. (2021) Astrocytic processes:
beta effects on tau pathology in preclinical Alzheimer's disease. from tripartite synapses to the active milieu. Trends Neurosci.
Nat. Med. 29, 1775–1781 44, 781–792
119. Le Douce, J. et al. (2020) Impairment of glycolysis-derived l-serine 143. Koh, W. et al. (2023) GABA tone regulation and its cognitive
production in astrocytes contributes to cognitive deficits in functions in the brain. Nat. Rev. Neurosci. 24, 523–539
Alzheimer's disease. Cell Metab. 31, 503–517 e508 144. Cho, F.S. et al. (2022) Enhancing GAT-3 in thalamic astrocytes
120. Qi, G. et al. (2021) ApoE4 impairs neuron–astrocyte coupling of promotes resilience to brain injury in rodents. Sci. Transl. Med.
fatty acid metabolism. Cell Rep. 34, 108572 14, eabj4310
121. Tcw, J. et al. (2022) Cholesterol and matrisome pathways dysreg- 145. Romanos, J. et al. (2019) Differences in glutamate uptake
ulated in astrocytes and microglia. Cell 185, 2213–2233 e2225 between cortical regions impact neuronal NMDA receptor acti-
122. Mi, Y. et al. (2023) Loss of fatty acid degradation by astrocytic vation. Commun. Biol. 2, 127
mitochondria triggers neuroinflammation and neurodegenera- 146. Broadhead, M.J. et al. (2022) Selective vulnerability of tripartite
tion. Nat. Metab. 5, 445–465 synapses in amyotrophic lateral sclerosis. Acta Neuropathol.
123. Bonvento, G. and Bolanos, J.P. (2021) Astrocyte–neuron 143, 471–486
metabolic cooperation shapes brain activity. Cell Metab. 33, 147. Tzioras, M. et al. (2023) Human astrocytes and microglia show
1546–1564 augmented ingestion of synapses in Alzheimer's disease via
124. Andersen, J.V. et al. (2022) Astrocyte energy and neurotransmitter MFG-E8. Cell Rep. Med. 4, 101175
metabolism in Alzheimer's disease: integration of the glutamate/ 148. Dejanovic, B. et al. (2022) Complement C1q-dependent excitatory
GABA-glutamine cycle. Prog. Neurobiol. 217, 102331 and inhibitory synapse elimination by astrocytes and microglia in
125. Institoris, A. et al. (2022) Astrocytes amplify neurovascular Alzheimer's disease mouse models. Nat. Aging 2, 837–850
coupling to sustained activation of neocortex in awake mice. 149. Vainchtein, I.D. et al. (2018) Astrocyte-derived interleukin-33
Nat. Commun. 13, 7872 promotes microglial synapse engulfment and neural circuit
126. Ju, Y.H. et al. (2022) Astrocytic urea cycle detoxifies Abeta- development. Science 359, 1269–1273
derived ammonia while impairing memory in Alzheimer's disease. 150. Sanchez-Mico, M.V. et al. (2021) Amyloid-beta impairs the phago-
Cell Metab. 34, 1104–1120 e1108 cytosis of dystrophic synapses by astrocytes in Alzheimer's
127. Fecher, C. et al. (2019) Cell-type-specific profiling of brain mito- disease. Glia 69, 997–1011
chondria reveals functional and molecular diversity. Nat. 151. Lattke, M. et al. (2021) Extensive transcriptional and chromatin
Neurosci. 22, 1731–1742 changes underlie astrocyte maturation in vivo and in culture.
128. Ioannou, M.S. et al. (2019) Neuron–astrocyte metabolic Nat. Commun. 12, 4335
coupling protects against activity-induced fatty acid toxicity. 152. Baier, M.P. et al. (2022) Selective ablation of Sod2 in astrocytes
Cell 177, 1522–1535 e1514 induces sex-specific effects on cognitive function, D-serine
129. Polyzos, A.A. et al. (2019) Metabolic reprogramming in astro- availability, and astrogliosis. J. Neurosci. 42, 5992–6006
cytes distinguishes region-specific neuronal susceptibility in 153. Luengo-Mateos, M. et al. (2023) Hypothalamic astrocytic-
Huntington mice. Cell Metab. 29, 1258–1273 e1211 BMAL1 regulates energy homeostasis in a sex-dependent
130. Guttenplan, K.A. et al. (2021) Neurotoxic reactive astrocytes manner. Cell Rep. 42, 112949
induce cell death via saturated lipids. Nature 599, 102–107 154. Arnaud, L. et al. (2022) APOE4 drives inflammation in human
131. Chen, Z.P. et al. (2023) Lipid-accumulated reactive astrocytes astrocytes via TAGLN3 repression and NF-kappaB activation.
promote disease progression in epilepsy. Nat. Neurosci. 26, Cell Rep. 40, 111200
542–554 155. Jimenez-Blasco, D. et al. (2020) Glucose metabolism links
132. Fleeman, R.M. et al. (2023) Apolipoprotein E epsilon4 modu- astroglial mitochondria to cannabinoid effects. Nature 583,
lates astrocyte neuronal support functions in the presence of 603–608
amyloid-beta. J. Neurochem. 165, 536–549 156. Rasmussen, R.N. et al. (2023) Astrocytes: integrators of arousal
133. Lee, S.I. et al. (2021) APOE4-carrying human astrocytes over- state and sensory context. Trends Neurosci. 46, 418–425
supply cholesterol to promote neuronal lipid raft expansion 157. Li, J. et al. (2021) Conservation and divergence of vulnerability
and Abeta generation. Stem Cell Reports 16, 2128–2137 and responses to stressors between human and mouse astro-
134. Descalzi, G. et al. (2019) Lactate from astrocytes fuels learning- cytes. Nat. Commun. 12, 3958
induced mRNA translation in excitatory and inhibitory neurons. 158. Preman, P. et al. (2021) Human iPSC-derived astrocytes
Commun. Biol. 2, 247 transplanted into the mouse brain undergo morphological changes
135. Chen, M.B. et al. (2020) Persistent transcriptional programmes in response to amyloid-beta plaques. Mol. Neurodegener.
are associated with remote memory. Nature 587, 437–442 16, 68
136. Morant-Ferrando, B. et al. (2023) Fatty acid oxidation organizes 159. Zeppenfeld, D.M. et al. (2017) Association of perivascular localiza-
mitochondrial supercomplexes to sustain astrocytic ROS and tion of aquaporin-4 with cognition and Alzheimer disease in aging
cognition. Nat. Metab. 5, 1290–1302 brains. JAMA Neurol. 74, 91–99