Access to this full-text is provided by Springer Nature.
Content available from Journal of Neuroinflammation
This content is subject to copyright. Terms and conditions apply.
REVIEW Open Access
© The Author(s) 2025. Open Access This article is licensed under a Creative Commons Attribution 4.0 International License, which permits use,
sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and
the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this
article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included
in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will
need to obtain permission directly from the copyright holder. To view a copy of this licence, visit h t t p : / / c r e a t i v e c o m m o n s . o r g / l i c e n s e s / b y / 4 . 0 /.
Zeng et al. Journal of Neuroinammation (2025) 22:72
https://doi.org/10.1186/s12974-025-03410-w
Introduction
Astrocytes are a major class of glial cells found in the cen-
tral nervous system (CNS), representing 19 to 40% of the
glial population [1]. Under normal condition, astrocytes
are responsible for maintaining homeostasis including
regulation of ion and water balance [2], maintenance of
the blood-brain barrier (BBB) [3], and regulation of local
cerebral blood ow [2, 3] (Fig. 1A-C). Astrocytes also
help to provide support for neuronal metabolic functions
[4] and maintain synaptic homeostasis through modulat-
ing synaptic formation, maturation, and elimination [5,
6] (Fig.1D-E). Furthermore, astrocytes play key roles in
Journal of Neuroinammation
†Jialiu Zeng and Jonathan Indajang contributed equally to this work.
*Correspondence:
Jialiu Zeng
jzeng22@syr.edu
Chih Hung Lo
clo101@syr.edu
1Department of Biomedical and Chemical Engineering, Syracuse
University, Syracuse, NY 13244, USA
2Interdisciplinary Neuroscience Program, Syracuse University, Syracuse,
NY 13244, USA
3Meinig School of Biomedical Engineering, Cornell University, Ithaca,
NY 14853, USA
4Department of Neurology, Yale School of Medicine, New Haven,
CT 06511, USA
5Department of Biology, Syracuse University, Syracuse, NY 13244, USA
Abstract
Astrocytes are a major cell type in the central nervous system (CNS) that play a key role in regulating homeostatic
functions, responding to injuries, and maintaining the blood-brain barrier. Astrocytes also regulate neuronal
functions and survival by modulating myelination and degradation of pathological toxic protein aggregates.
Astrocytes have recently been proposed to possess both autophagic activity and active phagocytic capability
which largely depend on suciently acidied lysosomes for complete degradation of cellular cargos. Defective
lysosomal acidication in astrocytes impairs their autophagic and phagocytic functions, resulting in the
accumulation of cellular debris, excessive myelin and lipids, and toxic protein aggregates, which ultimately
contributes to the propagation of neuroinammation and neurodegenerative pathology. Restoration of lysosomal
acidication in impaired astrocytes represent new neuroprotective strategy and therapeutic direction. In this
review, we summarize pathogenic factors, including neuroinammatory signaling, metabolic stressors, myelin and
lipid mediated toxicity, and toxic protein aggregates, that contribute to lysosomal acidication impairment and
associated autophagic and phagocytic dysfunction in astrocytes. We discuss the role of lysosomal acidication
dysfunction in astrocyte-mediated neuroinammation primarily in the context of neurodegenerative diseases along
with other brain injuries. We then highlight re-acidication of impaired lysosomes as a therapeutic strategy to
restore autophagic and phagocytic functions as well as lysosomal degradative capacity in astrocytes. We conclude
by providing future perspectives on the role of astrocytes as phagocytes and their crosstalk with other CNS cells to
impart neurodegenerative or neuroprotective eects.
Keywords Lysosomal acidication, Lysosomal alkalization, Autophagy, Phagocytosis, Metabolic dysfunction, Acidic
nanoparticles, Glial crosstalk, Neurodegeneration, Neuroinammation, Neuroprotective
Lysosomal acidication impairment
in astrocyte-mediated neuroinammation
JialiuZeng1,2*†, JonathanIndajang3†, DavidPitt4 and Chih HungLo2,5*
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 2 of 15
Zeng et al. Journal of Neuroinammation (2025) 22:72
phagocytic uptake of cellular debris, myelin/lipids, and
toxic protein aggregates [7, 8] as well as modulation of
neuroimmune responses [9] (Fig. 1F-G). Upon expo-
sure to external stimuli, astrocytes become reactive and
exhibit cellular heterogeneity, including alterations in
cell morphology and functions, gene expression proles,
cytokine production levels, as well as their response to
injuries, which have implications towards brain inam-
mation and neuronal death [10–14].
Recent studies have highlighted that autophagic and
phagocytic processes play key regulatory roles in astro-
cytic degradation capability and impairments in these
functions could contribute to neuroinammation and
neurodegeneration [15–17]. In addition to the process-
ing of external cargo by phagocytosis [18], the processing
of internal cargo by autophagy is important in astrocyte
dierentiation and maturation as well as regulation of
mitochondrial dynamics, reactive oxygen species (ROS)
generation, neuroimmune response, and cell death [19].
In astrocyte autophagy and phagocytosis, fusion of
autophagosomes and phagosomes with suciently acidi-
ed lysosomes as maintained by the lysosomal vacuolar
(H+)-ATPase (V-ATPase) is essential for their degrada-
tive functions [20]. In the homeostatic state, astrocyte
autophagy and phagocytosis are functional in the pres-
ence of optimal lysosomal acidication and these pro-
cesses maintain cellular homeostasis, support axonal
health, and regulate myelination, contributing to neuro-
nal plasticity, functions, and survival [21, 22] (Fig.2A).
In their reactive states, it has been demonstrated that
an elevation of lysosomal pH or defective lysosomal
acidication decreases the eciency and eectiveness of
Fig. 1 The multifaceted roles of astrocytes in the brain. (A) Astrocytes control ion homeostasis and water exchange in the brain microenvironment by
regulating channel proteins including aquaporin-4 (AQP4) water channel as well as potassium, sodium, and calcium channels. (B) Astrocytes support the
formation of tight junctions (e.g., claudin, occludin, junctional adhesion molecules (JAM), and cadherin) and the maintenance of epithelial cells at the
blood-brain barrier (BBB). (C) Contraction and blood ow movement of the BBB is mediated by smooth muscle tissue that respond to Ca2+ ions released
and regulated by astrocytes. (D) Astrocytes mediate nutrient transport to neurons to regulate neuronal metabolism. (E) Astrocytes operate as phagocytes
to carry out synaptic pruning as well as remove cell debris, damaged organelles, and myelin. (F) Astrocytes participate in autophagic degradation of intra-
cellular toxic protein aggregates, myelin/lipids and cellular debris phagocytosed from damaged neurons. (G) Astrocytes release inammatory cytokines
that can recruit microglia to sites of brain injury and/or induce neuronal impairment and death. The gure was created with BioRender.com
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 3 of 15
Zeng et al. Journal of Neuroinammation (2025) 22:72
astrocytes to perform autophagic and phagocytic func-
tions [23, 24]. e resulting accumulation of damaged
organelles, myelin debris, and toxic protein aggregates as
well as the release of inhibitory factors and ROS further
propagate neuroinammation and drive neurodegenera-
tion [25, 26] (Fig.2B). Under aging or diseased condi-
tions, there are evidence of synaptic and autophagosomal
proteins as well as toxic protein aggregates accumulated
and colocalized with poorly acidied lysosomes in reac-
tive astrocytes [27–30]. However, the role of dierent
stimuli and their molecular mechanisms associated with
lysosomal acidication impairment in astrocytes are
unclear and remain to be claried. While the accumu-
lation of unwanted and toxic materials may be a conse-
quence of lysosomal dysfunction, it is important to note
that these materials could also be the initial triggers in
impairing lysosomal acidication.
In this review, we summarize the role of neuroinam-
matory signaling, metabolic stressors, myelin and lipid
mediated toxicity, and toxic protein aggregates in lyso-
somal acidication impairment and associated autopha-
gic and phagocytic dysfunction in astrocytes (Fig.3). We
discuss these pathogenic factors primarily in the context
of neurodegenerative diseases along with other brain
injuries that provide insights to the role of lysosomal
acidication dysfunction in astrocyte-mediated neuroin-
ammation. We provide some insight into the feedback
mechanisms between pathogenic factors and lysosomal
dysregulation, both regarding how astrocytic dysregula-
tion is initiated by pathogenic factors, and how eventual
dysregulation accelerates neural degeneration. We then
highlight current therapeutic strategies to re-acidify lyso-
somes and restore astrocyte autophagic and phagocytic
functions. We conclude by providing future perspectives
on the role of astrocytes as phagocytes and their coopera-
tive role with other CNS cells such as microglia and neu-
rons to mediate neurodegeneration and neuroprotection.
Fig. 2 Autophagic and phagocytic degradative functions in normal and reactive astrocytes. (A) In normal astrocytes, lysosomes maintain a suciently
acidic environment, enabling proper vesicle fusion and optimal autophagic/phagocytic activities, including the clearance of damaged mitochondria and
myelin debris, thereby maintaining neuronal health. In addition, astrocytes release regenerative factors which contribute to neuron remyelination. (B)
In reactive astrocytes under exposure to pro-inammatory cytokines, excessive lipids, and toxic protein aggregates, lysosomal acidication is impaired
(poorly acidied lysosomes), leading to inhibition of autophagic/phagocytic activities. As a result, there is reduced mitochondrial turnover and increased
accumulation of damaged mitochondria, as well as release of neurotoxic factors such as ROS. In addition, damaged astrocytes can release undegraded
toxic materials as well as inhibitory factors that further impair neuronal function. Re-acidication of impaired lysosomes by lysosome-targeting small
molecules and nanoparticles restores autophagic/phagocytic functions in astrocytes, allowing for eective clearance of neurotoxic factors to maintain
neuronal health. The gure was created with BioRender.com
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 4 of 15
Zeng et al. Journal of Neuroinammation (2025) 22:72
Neuroinammatory signaling
Inammatory reactive astrocytes with reduced homeo-
static functions can be neurotoxic, contributing to neu-
rodegenerative diseases including Alzheimer’s disease
(AD), Parkinson’s disease (PD), and multiple sclerosis
(MS). Reactive astrocytes subjected to microglia-derived
pro-inammatory cytokines such as a combination of
tumor necrosis factor (TNF), interleukin-1α (IL-1α), and
complement component 1q (C1q) have reduced abil-
ity to promote neuronal survival, synaptogenesis and
Fig. 3 Factors aecting lysosomal acidication dysfunction in astrocytes. (A) Exposure to dierent cytokines triggers neuroinammatory signaling that
induce diverse astrocyte inammatory phenotypes. Cytokines exposure leads to a reduction of lysosomal V-ATPase levels, leading to elevated lysosomal
pH and reduced lysosomal enzyme degradative capacity. Chronic autolysosomal buildup due to incomplete degradation leads to exocytosis of neu-
rotoxic factors which impair surrounding neurons. (B) Changes in nutrient levels induce metabolic stress which lead to mitochondrial dysfunction and
impaired lysosomal acidication. (C) In astrocytes with lipids or myelin accumulation, lysosomal size is increased along with elevated lysosomal pH. In
addition, high fat diet intake and metabolic disorders that aect the peripheral organs can also aect astrocyte function and reactivity. (D) Toxic proteins
aggregates taken up by astrocytes localized into lysosomes and impaired lysosomal acidication, resulting in cellular dysfunction and spreading of pa-
thology due to inecient degradation and increased release of the toxic materials. The gure was created with BioRender.com
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 5 of 15
Zeng et al. Journal of Neuroinammation (2025) 22:72
phagocytosis, and can induce neuronal death in human
AD, PD and MS tissues [31]. Dierent cytokine cocktails
may induce dierent astrocyte phenotypes which could
be neurotoxic or neuroprotective [31–33]. Defective lyso-
somal acidication has been shown to be associated with
neurotoxic reactive astrocytes, age-related inammation,
and consequentially contribute to neurodegenerative dis-
eases [33, 34] (Fig.3A).
CRISPR interference screens were conducted on
human-induced pluripotent stem cells (hiPSC)-derived
astrocytes cultured with microglia-derived pro neu-
roinammatory cytokines (TNF, IL-1α, and C1q) to
determine relevant pathways propagating inamma-
tory astrocyte reactivity [32]. IL-6 and interferon signal-
ing downstream of canonical NF-κB activation drove
two distinct inammatory reactive signatures, and both
are modulated by the signal transducer and activator of
transcription 3 (STAT3). ese signatures were vali-
dated in both mouse models and in human AD brains
[32]. In addition, genes involved in the mammalian tar-
get of rapamycin (mTOR) pathway were found to be sig-
nicantly changed [32]. In a follow-up study by the same
group, CRISPR interference screens were conducted on
human iPSC-derived astrocytes treated with pro-inam-
matory cytokines (e.g., TNF, IL-1α, and C1q) to identify
molecular targets that aects lysosomal acidication dys-
function and exocytosis [33]. First, signicant lysosomal
alkalization and autophagic dysfunction were observed
in neurotoxic reactive astrocytes. Lysosomal alkaliza-
tion has been attributed to multiple downregulated genes
including V-ATPase subunits and lysosomal hydrolases
[33]. Importantly, it was found that mTOR is a central
upstream regulator of this phenotype which is linked
to lysosomal acidication dysfunction in inammatory
reactive astrocytes [33]. Inhibition of mTOR restored
lysosomal acidication and rescued this phenotype
associated with neurodegenerative diseases [33]. More-
over, these reactive astrocytes have increased lysosome
exocytosis, leading to the secretion of toxic materials.
In inammatory reactive astrocytes, mTOR activation
remodels lysosomal functions and induces unconven-
tional secretion of IL-32 which is involved in the polar-
ization of astrocyte reactive states [35], while mTOR
inhibition reduces the intracellular levels and secretion
of lipocalin-2 [36]. However, the relationship between
whether the secreted materials directly mediate neuronal
toxicity or if the materials contribute to toxicity through
an indirect mechanism such as autocrine-paracrine sig-
naling, remains to be elucidated. Emerging evidence indi-
cates that mTOR activity is age-dependent, with aged
and senescent astrocytes exhibiting distinct transcrip-
tional proles compared to other astrocytic populations
[37, 38]. In aging astrocytes, reduced mTOR activity
disrupts autophagy and lysosomal function, leading to
protein tracking defects and impaired synapse regu-
lation. A subset of aging astrocytes, termed autophagy-
dysregulated astrocytes, displays lysosomal dysfunction,
abnormal autophagosome accumulation, and impaired
proteasome function, resulting in synapse loss and
reduced dendritic spines [37]. While senescent astrocytes
exhibit proinammatory proles partly driven by active
mTOR signaling and DNA-damage response pathways
[38], these ndings highlight the shifting roles of mTOR
in aging astrocytes and its potential as a therapeutic tar-
get in neurodegenerative diseases.
In other disease contexts, it was shown that the stim-
ulation of toll-like receptor 3 (TLR3) triggers lysosomal
alkalization and release of adenosine triphosphate (ATP)
and luminal contents from optic nerve head astrocytes
[39]. In MS patients and mice, a subset of astrocytes that
expresses the lysosomal protein LAMP1 and the TNF-
related apoptosis-inducing ligand (TRAIL) has been
identied [40], bridging the link between lysosomal dys-
function and astrocyte-mediated neuroinammation. In
a lysosomal storage disease model of astrocytes, there
is progressive neuroinammatory response and inhi-
bition of autophagic function [41]. In addition, in Gau-
cher disease patients derived induced astrocytes, there is
reduced glucocerebrosidase activity, cathepsin D activ-
ity, and increased inammatory response [42]. In corti-
cal astrocytes isolated from mice with multiple sulfatase
deciency, there is impaired lysosomal/autophagic dys-
function and accumulation of autophagic substrates [43].
ere are also examples of activation of autophagy func-
tions in astrocytes to attenuate inammasome activa-
tion or inammatory phenotypes in neurodegenerative
diseases [44, 45]. Hence, the crosstalk between autoly-
sosomal acidication dysfunction and neuroinamma-
tion, and their pathogenic roles in neurodegenerative and
neuroimmune disorders warrant further investigations. It
is important to design future studies around the goal of
examining the association between autolysosomal acidi-
cation dysfunction and neuroinammation in heterog-
enous cell cultures or in vivo.
Metabolic stressors
Astrocytes and neurons operate as a tightly coupled unit
for energy metabolism in the brain. As neurons expend
a considerable amount of ATP on neurotransmission,
astrocytic mitochondrial metabolism can release signal-
ing molecules like ATP and glutamate, allowing neurons
to allocate more cellular resources to sustain high activ-
ity rates during information processing [46, 47]. Dys-
functional astrocytic mitochondria can lead to impaired
glutamate clearance and increased levels of extracel-
lular glutamate, ultimately inducing glutamate toxic-
ity in neighboring neurons [48]. Furthermore, impaired
mitochondria in astrocytes can also lead to increased
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 6 of 15
Zeng et al. Journal of Neuroinammation (2025) 22:72
ROS production or mitochondrial-derived damage-
associated molecular patterns into the cytoplasm, which
can also result in neuronal death [48, 49]. Hence, main-
taining astrocytic mitochondrial function is essential to
protect against neurodegeneration. Importantly, optimal
lysosomal acidication maintains the turnover of dam-
aged mitochondria through autophagic clearance, closely
regulating mitochondrial metabolism in astrocytes [50,
51]. On the other hand, mitochondria regulate lysosomal
pH by supplying ATP to activate the V-ATPase to acidify
the lysosomal lumen [52]. erefore, understanding and
maintaining proper mitochondria-lysosome crosstalk is
crucial to regulating astrocytic function (Fig.3B).
Chronic exposure to diet derived metabolites like
homocysteine and glucose aect astrocyte function.
Exposure to homocysteine, a homologue of the amino
acid cysteine, in astrocytes leads to lysosomal and
autophagic impairments, including lysosomal acidica-
tion dysfunction due to downregulation of V-ATPase
[53]. is further leads to increased oxidative stress and
astrocytic cell death [53]. Chronic exposure to high levels
of glucose in astrocytes induce mitochondrial oxidative
stress through 5’AMP-activated protein kinase-indepen-
dent pathways and lead to inhibition of transcription fac-
tor EB (TFEB) [54]. is consequently leads to impaired
lysosomal acidication, reduced phagocytic function, and
accumulation of oligomeric Aβ in astrocytes [54]. Expo-
sure to high glucose has also been reported to promote
ferroptosis of astrocytes by disrupting iron metabolism,
which can be rescued by administration of gembro-
zil, a peroxisome proliferator-activated receptor alpha
(PPARα) agonist. Gembrozil prevented the accumula-
tion of lipid peroxidation products and ROS induced by
iron deposition in astrocytes and inhibited ferroptosis of
astrocytes [55]. Gembrozil has been shown to upregu-
late TFEB and enhance lysosomal biogenesis in astrocytes
via PPARα [55], suggesting that lysosomal acidication
may play an important role in alleviating high glucose
induced metabolic dysfunction in astrocytes.
In a brain ischemia mouse model, increased expression
of inammatory cytokines TNF and IL-1β was seen in
astrocytes. Using an oxygen-glucose deprivation/reoxy-
genation (OGD/R) model of primary mouse astrocytes to
study brain ischemia in vitro, lysosomal impairment and
autophagic dysfunction were observed, as determined by
decreased lysosome number, increased lysosomal size,
and accumulation of autophagosome associated pro-
teins [56]. In a OGD primary rat astrocytes model, it was
shown that there is an increase in lysosomal membrane
permeabilization (LMP) and cathepsin release from lyso-
somes into the cytoplasm of astrocytes, leading to cell
death [57]. is was also seen in another study using a
similar model [58], alongside with disrupted mitochon-
drial membrane potential, increased production of ROS
and inammatory cytokines TNF, IL-6 and FasL, as well
as apoptosis in astrocytes [58]. e knockdown of recep-
tor interacting protein 1, an essential molecule in medi-
ating TNF signaling, blocked OGD-induced increase in
LMP and astrocyte death, suggesting that TNF down-
stream pathways contribute to lysosomal dysfunction in
ischemic astrocytes [57]. In another instance, conditional
knockdown of LAMP-2A in ischemic astrocytes inhib-
ited their activation and prevented the translocation of
the pro-apoptotic proteins Bax and Bad to mitochon-
dria, thereby preventing neuronal death, suggesting that
elevated astrocytic LAMP-2 A contributes to ischemic
vulnerability.
Myelin and lipid induced toxicity and dysfunction
Astrocytes are actively involved in lipid metabolism in
the brain and can be aected by increased levels of sat-
urated fatty acids associated with dysfunctional lipid
metabolism, neuronal myelin damage, and obesity [59–
61]. Cultured brain astrocytes present higher capacity
to process lipids in their oxidative metabolism and have
a higher propensity to uptake fatty acids than other cell
types in the brain [62]. e sensitivity of astrocytes and
their reactive state transformation under lipid-induced
oxidative stress such as sphingosine-1-phosphate (S1P)
and apolipoprotein E (APOE) mutations, myelin debris
accumulation and chronic exposure to fatty acids could
lead to failure in their functions (Fig.3C). S1P is a bio-
active signaling lipid involved in several vital processes,
including cellular proliferation, survival, and migration.
Autosomal recessive mutations in sphingosine-1-phos-
phate lyase 1 which encodes for S1P lyase, leads to neu-
rodevelopmental disorders. An excess of S1P due to
mutations in S1P lyase led to increased activity of regu-
latory enzymes involved in the tricarboxylic acid cycle
and increased cellular ATP content, which subsequently
activated mTOR and reduced lysosomal-autophagosome
fusion as well as reduced autophagic function of astro-
cytes [63]. Phospholysine phosphohistidine inorganic
pyrophosphate phosphatase (LHPP) is another enzyme
that is highly expressed in the brain which catalyzes
pyrophosphate to orthophosphate. LHPP is primarily
expressed in the lysosomes of astrocytes and has opti-
mal enzymatic activity at an acidic pH [64]. Under stress
conditions, LHPP modulates lysosomal acidication
through pyrophosphate hydrolysis driven proton trans-
port through the V-ATPase, thereby averting the adverse
impact of chronic stress on adult hippocampal neurogen-
esis [64].
Apolipoprotein E (ApoE) plays a major role in choles-
terol and phospholipid regulation within the CNS [65]
and astrocytes are the primary source of ApoE in the
brain [66]. e E4 allele of APOE (APOE4) is the stron-
gest genetic risk factor for the development of late onset
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 7 of 15
Zeng et al. Journal of Neuroinammation (2025) 22:72
AD. APOE4 astrocytes accumulate high amounts of lipid
droplets and have decreased fatty acid uptake and oxi-
dation compared to APOE3 astrocytes [67]. is obser-
vation is also consistent with other reports related to
lysosomal dysfunctions associated with APOE4 astro-
cytes [68], with a specic study illustrating that lyso-
somes in APOE4 astrocytes have a higher lysosomal pH
than APOE3 astrocytes [69]. APOE4 also downregulates
the sodium-hydrogen exchanger 6, which create dys-
regulation of the endosomal pH in astrocytes, leading to
Aβ accumulation within the astrocytes [70]. e impair-
ments of endolysosomal and autophagic function in
APOE4 astrocytes has also been reported in other studies
[69, 71, 72]. In addition, APOE4 astrocytes induce cho-
lesterol accumulation that impairs lysosomal turnover
of damaged mitochondria and treatment of therapeutic
agents that remove cholesterol restores autophagic and
mitochondrial activity [73].
In the CNS, myelin uptake is thought to occur primar-
ily by microglia. However, recent studies have shown that
astrocytes also participate in this process [74, 75]. Astro-
cytes actively phagocytose myelin debris during demy-
elination [76, 77]. In a study using primary rat astrocytes
culture, the myelin debris was taken up by astrocytes
through receptor-mediated endocytosis and are trans-
ported to lysosomes for degradation. Exposure to exces-
sive myelin debris resulted in astroglial nuclear factor
kappa B (NF-κB) activation and secretion of inamma-
tory cytokines [76]. ese ndings were conrmed in
the context of MS, where myelin-positive astrocytes had
increased nuclear localization of NF-κB and cytokine
expression compared to astrocytes lacking myelin [76].
In a similar astrocyte culture model of spinal cord injury,
engulfed myelin debris are transported to lysosomes for
degradation and led to an increase in lysosomal size,
which consequently resulted in excessive glia scar forma-
tion [77]. Apart from myelin debris accumulation, accu-
mulation of cholesterol can also lead to impairment of
lysosomal acidication and increased lysosomal leakiness
in foamy phagocytes, which can eventually lead to cell
death [78].
Chronic exposure of fatty acid (e.g., palmitic acid) in
astrocytes have led to intracellular lipid accumulation
with an elevation of the pH in the lysosomes along with
autophagic dysfunction, and an increase in the mRNA
expression of pro-inammatory cytokines [79]. High-
fat diet (HFD) feeding induces peripheral obesity which
develops inammation and has been shown to aect
CNS function [80, 81]. In mice under HFD, there is
increased hypothalamic inammatory signaling, reactive
astrogliosis and microgliosis, along with neuronal injury
[82]. In a similar study, HFD feeding to mice reduced
mitochondrial number and increased mitochondrial size,
thereby contributing to reduced activity of hypothalamic
astrocytes [83]. In addition, there are other models of
liver related injury or inammation that has been found
to lead to neurodegeneration [80]. For instance, in alco-
holic liver disease mouse model, exposure to ethanol
impaired lysosomal acidication and function in astro-
cytes and reduced autophagic function [84]. In a hepatic
encephalopathy model where mice are exposed to high
levels of ammonia, astrocytes had decreased lysosomal
acidication and increased accumulation of ROS [85],
leading to neuronal toxicity. ROS has been shown to fur-
ther interact with lysosomal membranes through per-
oxidation, which destabilizes the membrane [86], leading
to decreased ecacy of proton pumps and pH “leaking”,
which would impair lysosomal acidication [86, 87].
Toxic protein aggregates
Astrocytes play a key role in the clearance of toxic protein
aggregates that are hallmarks of many neurodegenera-
tive diseases [88–91]. However, they are still vulnerable
to the toxic eects of protein aggregation in both familial
and sporadic pathologies, which have been implicated in
lysosomal dysfunction, autophagic inhibition, and propa-
gation of neurodegenerative pathology (Fig.3D). In AD,
accumulation of toxic aggregates such as amyloid beta
(Aβ) has been attributed to astrocytes dysfunction [92].
Presenilin-1 (PS1) mutation has been shown to impair
lysosomal vesicle tracking, leading to reduced degrada-
tion capacity, and accumulation of Aβ brils in astrocytes
[93]. APP/PS1 transgenic mice displayed a higher density
of astrocytes and increased accumulation of lysosomes
in cells, potentially due to a higher phagocytic activity
required to clear a higher burden of toxic materials in
the mice [94]. e stimulation of lysosomal biogenesis
with TFEB in astrocytes has been shown to reduce Aβ
plaque load in the hippocampus of APP/PS1 mice [95].
Another study has shown that a small molecule agonist
of angiotensin-(1–7) receptor (AVE 0991) could suppress
astrocyte neuroinammatory responses by enhancing
autophagy. Treatment of AVE 0991 reduced Aβ deposi-
tion as well as rescued neuronal death and cognitive de-
cits in APP/PS1 mice [96].
Human iPSC-derived astrocytes or primary astrocytes
exposed to Aβ brils or oligomers showed accumulation
of Aβ inclusions that were enclosed within LAMP1-pos-
itive lysosomes and sustained markers associated with
reactivity [97, 98]. Additionally, Aβ uptake and accumu-
lation in astrocytes resulted in endoplasmic reticulum
and mitochondrial swelling, autolysosomal dysregula-
tion, LMP, formation of pathological lipid structures, and
increased secretion of chemokines and cytokines [97,
99, 100]. Furthermore, increasing evidence have pointed
to the role of astrocytes in Aβ brils uptake and aggre-
gation, leading to indirect neurotoxic qualities [101,
102]. Specically, high level of glycoprotein YKL-40
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 8 of 15
Zeng et al. Journal of Neuroinammation (2025) 22:72
in astrocytes can promote neurotoxicity and YKL-40
knockout astrocytes exhibit enhanced lysosomal acidi-
cation as well as increased uptake and degradation of Aβ
peptides [103]. In a study using neuron-astrocyte co-cul-
tures, it was shown that incomplete astrocyte phagocyto-
sis of Aβ brils leads to increased astrocytic secretion of
toxic vesicles as well as accumulation of Aβ brils in neu-
rons and neuronal cell death [101]. Ineective clearance
in astrocytes is possibly attributed to increased levels of
Rab27a protein, which reduces lysosomal acidity through
Nox2 recruitment [101]. In addition, increasing levels of
autophagic ux in astrocytes via progesterone has been
shown to be eective in enhancing the neuroprotective
and anti-inammatory eect of astrocytes in models of
AD [104]. Furthermore, stimulation of autophagy with
Sirtuin-1 in primary rat astrocytes has also been shown
to improve lysosomal function through upregulation of
V-ATPase subunits and increase in lysosome number,
leading to more eective clearance of Aβ brils [105].
In primary human astrocytes exposed to preformed 4R
tau brils, the endocytosis of tau aggregates causes lyso-
somal swelling, permeabilization, and lysosomal deacidi-
cation [106]. In primary mouse astrocytes treated with
tau pre-formed brils, expression of TFEB enhanced
lysosomal activity, increased tau degradation and inhib-
ited tau transmission [107]. TFEB activation enhances
the phagocytic capacity of astrocytes, through increas-
ing the uptake of pre-formed brils, and increases the
incidence of phagocytosed pre-formed brils inside the
lysosome, indicating that TFEB enhances both uptake
and degradation of phagocytosed proteins [107]. In addi-
tion, astroglial TFEB overexpression reduced tau pathol-
ogy, spreading, and gliosis in the hippocampus of PS19
tauopathy mice [107]. Studies in another tauopathy
model using rTg4510 tau transgenic mouse have also
shown that TFEB expression enhanced lysosomal activity
and clearance of autophagic substrates and phosphory-
lated tau [108]. Another study from the same group high-
lights TFEB’s role in mediating the lysosomal exocytosis
of mutant truncated tau, both in vitro and in PS19 trans-
genic mice. is process, dependent on the lysosomal
target TRPML1, is positively correlated with tau clear-
ance. Loss of TFEB increases tau pathology and spread-
ing, suggesting that TFEB-mediated lysosomal exocytosis
of tau acts as a clearance mechanism to reduce intracel-
lular tau under pathological conditions [109].
In PD, α-synuclein is the major component of neuro-
nal cytoplasmic aggregates called Lewy bodies, which
are the main pathological hallmark of the disease. In
immortalized astrocyte cell lines, overexpression of
wild-type α-synuclein as well as A30P and A53T mutant
α-synuclein led to inhibition of autophagy, loss of mito-
chondrial membrane potential, and cell death [110]. Pri-
mary astrocytes with A53T α-synuclein overexpression
or treatment with α-synuclein aggregates had decreased
lysosomal acidication and reduced lysosomal enzyme
activity, thereby contributing to the release of more
extracellular vesicles which propagate PD pathology
[111]. Incubation of astrocytes with Lewy body extracts
from human PD patients or α-synuclein preformed brils
led to α-synuclein colocalization in lysosomes, indicating
aggregate buildup due to reduced lysosomal degradative
capacity [112, 113]. Additionally, there is increased mito-
chondrial driven cytotoxicity in astrocytes [112]. Dose
dependent treatment with lysosomal V-ATPase inhibitor
Balomycin A1 led to an increase in the accumulation
of α-synuclein brils in astrocytes, indicating that lyso-
somal acidication plays an important role in modulating
α-synuclein buildup in astrocytes [114]. Increasing levels
of autophagic ux in astrocytes via rapamycin have been
shown to be eective in enhancing the neuroprotective
and anti-inammatory eect of astrocytes in models
of PD [115]. In other types of familial PD, mutations in
LRRK2, ATP13A2, GBA1, and PARK7 impair lysosomal
function and degradative capacity of astrocytes [42,
116–118]. LRRK2 G2019S primary mouse astrocytes
have enlarged lysosomes and abnormal lysosomal pH,
which led to reduced lysosomal activity, and is regulated
by LRRK2 localization to lysosomes [116]. Inhibition of
LRRK2 kinase activity with PF-06447475 restored defects
in lysosomal morphology and function [116]. ATP13A2
mutations in astrocytes resulted in decreased lysosomal
proteolysis function and increased accumulation and
propagation of α-synuclein [117]. Patient derived induced
astrocytes with GBA mutations also exhibited impaired
lysosomal enzyme activity, leading to α-synuclein accu-
mulation [42]. In DJ1 knockout iPSC-derived midbrain
organoid models, impaired lysosomal proteolysis results
in increased α-synuclein phosphorylation, protein aggre-
gation, and the accumulation of advanced glycation end
products. Astrocytes play a role in these eects, as DJ1
loss diminishes their metabolic support capacity and
promotes a pro-inammatory phenotype. In co-culture
models, DJ1-expressing astrocytes have been shown to
rescue proteolysis decits [118].
Restoration of lysosomal acidication as a
therapeutic target
Functional lysosomal V-ATPase and ion channels such as
two-pore channels (TPC) are crucial in maintaining lyso-
somal acidication and function of astrocytes [119]. In
rat astrocytes, activation of TPC by nicotinic acid adenine
dinucleotide phosphate (NAADP) increases autopha-
gosome and lysosome formation [119]. To enhance the
function of lysosomal V-ATPase, C381 is a small-mole-
cule activator of V-ATPase that has been applied to pro-
mote lysosomal acidication in microglia [120], although
its eect in astrocytes remains to be tested. OSI-027 and
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 9 of 15
Zeng et al. Journal of Neuroinammation (2025) 22:72
PP242 are two other small-molecular mTOR inhibitors
that have been identied by high-throughput screening
using a uorescent protein based lysosomal pH biosen-
sor [121]. OSI-027 and PP242 were identied as the top
lysosome-acidifying hits in human iPSC-derived astro-
cytes which demonstrated increased lysosomal cathepsin
activity and improved autophagic function [121]. In pri-
mary astrocytes under exposure to environmental toxins,
lysosomal acidication and autophagic ux are impaired
and can be restored by PP242 treatment [122]. Recent
developments using lysosomal-acidifying nanoparticles
to target and restore acidication of impaired lysosomes
have been demonstrated in astrocytes [23, 24]. e intro-
duction of lysosomal-acidifying nanoparticles to mouse
primary astrocytes has led to increased lysosomal acidi-
cation which increased the lysosomal cathepsin activity
and astrocytic phagocytosis of cell debris [23, 24]. Other
types of lysosomal-acidifying nanoparticles have also
been developed [123–125] to re-acidify impaired lyso-
somes and promote autophagic degradation, and this has
been reviewed elsewhere [126–128].
Another approach to promote lysosomal acidication
is through increasing cyclic adenosine monophosphate
(cAMP) levels [129]. Balomycin A1 treatment to astro-
cytes induced dysfunctional V-ATPase and lysosomal
alkalization, while increasing cAMP levels via activation
of PKA signaling pathway restored lysosomal acidica-
tion [130]. Treatment with cilostazol, a phosphodies-
terase inhibitor that inhibits the degradation of cAMP,
reacidies lysosomes in astrocytes, thereby increasing
Aβ degradation in astrocytes [131]. In addition, while
the acute treatment of cAMP activates the AKT survival
pathway in astrocytes, chronic exposure of cAMP has
been observed to activate the FoxO-mediated Bim/Bax
death pathway [132]. erefore, the reliance on cAMP
elevation to acidify lysosomes requires dosage optimiza-
tion, as high cAMP level is observed to exacerbate the
vulnerability of astrocytes to oxidative stress [132, 133].
Interestingly, pharmacological inhibitors and siRNAs
of H+/K+-ATPase elevated lysosomal pH in balomycin
A1 and cAMP co-treated astrocytes, suggesting that H+/
K+-ATPase may function as an alternative proton pump
for lysosomes when the V-ATPase function is impaired
[130]. Hence, pharmacological agents that target the H+/
K+-ATPase may be a new avenue for lysosome-acidifying
therapeutics. Other molecular targets to restore lyso-
somal pH and autophagic function have been explored.
An important therapeutic target is TFEB, where its
expression regulates lysosome biogenesis and expression
of V-ATPase, thereby maintaining lysosomal acidication
[107, 134]. Stereotaxic injection of adeno-associated viral
particles carrying TFEB driven by a glial brillary acidic
protein (GFAP) promoter was used to achieve astrocyte-
specic expression of the gene in the hippocampus of
APP/PS1 transgenic mice. Expression of TFEB in these
astrocytes enhanced lysosome function, resulting in
reduced Aβ plaques in the hippocampus [95]. Aspirin has
also been shown to upregulate TFEB and increases lyso-
somal biogenesis in mouse astrocytes through inducing
the activation of PPARα and stimulated the transcrip-
tion of TFEB [135]. Furthermore, progranulin may also
be a promising target as it mediates TFEB expression [95,
136]. However, careful regulation of TFEB expression is
essential, as excessive activation could lead to potential
side eects, including its role as an oncogenic regulatory
marker [137, 138].
Summary and future perspectives
Astrocytes play a critical role in maintaining energy
metabolism and neuronal health in the CNS [4, 139].
e eectiveness of autophagic and phagocytic func-
tions by astrocytes depends on the extent of lysosomal
acidication and degradation (Fig.4A). Pathogenic fac-
tors, including neuroinammatory signaling, metabolic
stressors, and the accumulation of lipids and toxic pro-
tein aggregates, contribute to the impairment of astro-
cytic lysosomal acidication, resulting in dysfunctional
autophagy and phagocytosis. However, it remains unclear
whether these pathogenic factors directly drive lysosomal
dysfunction in astrocytes or if the defects are secondary
eects of astrocyte reactivity induced by these stress-
ors, neither is mutually exclusive. Further studies are
required to disentangle these mechanisms and clarify the
causal relationships underlying astrocytic dysfunction.
Due to the crosstalk between mitochondria and lyso-
some [140], it is important to investigate how restoration
of mitochondria function, metabolic activity, and lipid
metabolism in astrocytes might oer potential avenues
to maintain optimal lysosomal acidication and eective
degradation.
e generation of the mRFP-eGFP-LC3 mouse model,
which is designed to monitor changes in autophagic ux
due to alterations in lysosomal acidication [141], with a
astrocyte promoter such as GFAP will enable the exami-
nation of the eects of lysosomal acidication impair-
ment in astrocytes in vivo. We also discussed molecular
targets and potential therapeutics that regulate lysosomal
pH, including small molecules and lysosome-targeting
nanoparticles. It is essential to further investigate how
these therapeutics modulate lysosomal acidication and
overall cellular function under dierent disease condi-
tions and reactivity states of astrocytes. Furthermore,
proling of astrocyte heterogeneity using omics charac-
terizations will reveal cellular subtypes that are directly
implicated by lysosomal acidication dysfunction and
enable the elucidation of new molecular targets for bet-
ter and more eective intervention [32, 142–145]. While
this review focuses on targeting lysosomal acidication
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 10 of 15
Zeng et al. Journal of Neuroinammation (2025) 22:72
in astrocytes, it is important to note that lysosomal dys-
function also occurs in other CNS cell types, such as
microglia and neurons, under pathological conditions
[127, 128, 146]. In addition to defects in lysosomal acidi-
cation, lysosomal enzyme deciencies play a critical role
in maintaining astrocytic function, as well as the function
of other CNS cell types [147–149]. In infantile neuronal
ceroid lipofuscinosis (CLN1 disease), primary cultures
of astrocytes, microglia, and neurons derived from
Ppt1-decient mice exhibit impaired cellular function.
Ppt1-decient astrocytes display dysregulated calcium
signaling, resulting in increased cell death. In co-culture
experiments, the presence of both Ppt1-decient astro-
cytes and microglia further disrupted the morphology
of both wild-type and Ppt1-decient neurons [150], sug-
gesting that the astrocytes be cross primed by impaired
microglia to become neurotoxic under disease condi-
tions [150, 151]. Given the interconnected roles of these
cells, a broader therapeutic approach targeting lysosomal
function across multiple CNS cell types may oer more
comprehensive benets, highlighting the need to assess
the extent and specic cellular contributions of lysosomal
impairments.
e supportive functions of astrocytes have recently
been extended to them taking on a more active role as
phagocytes similar to microglia. ese functions include
the phagocytosis of cellular debris, synapse elimination,
and the regulation of neuronal activity [8, 152]. Com-
parative studies suggest that astrocytes complement the
phagocytic activity of microglia, although their mecha-
nisms of action are distinct [8, 152]. Astrocytic activity-
dependent synaptic pruning requires the involvement
of phagocytic receptors multiple EGF-like-domains 10
(MEGF10) and MER Tyrosine Kinase (MERTK), both
of which are highly expressed in developing astrocytes
[153]. Subsequent studies have highlighted the critical
role of MEGF10 in synaptic pruning within adult mice,
where astrocytes eliminate excitatory synapses in the
hippocampus to maintain circuit homeostasis and sup-
port memory formation [154]. In contrast, microglia-
mediated synapse elimination involves activation of the
classical complement pathway [155–157]. Specically,
the complement cascade initiator, C1q, localizes to devel-
oping synapses, marking them for microglial phagocy-
tosis in a complement component 3-dependent manner
[155]. A recent study illustrates that the lysosomal pH
Fig. 4 Astrocyte-microglia interactions to promote clearance of unwanted and toxic materials in the cells. (A) Under normal conditions where the un-
wanted materials are within the clearance capacity of astrocytes, phagocytic/autophagic degradation of the accumulated materials can proceed. During
chronic exposure to these unwanted materials, lysosomal functions are impaired, and this reduces the capacity of astrocytes to degrade, leading to the
accumulation of toxic materials within the astrocytes and their subsequent dysfunction. (B) Astrocytes-microglia crosstalk through tunneling nanotubes
(TNTs). In astrocyte-microglia interaction, unwanted and toxic cellular materials such as impaired lysosomes, undegraded autophagosomes, and dam-
aged mitochondria can migrate to microglia, where the latter can assist in more eective degradation of these toxic products. The gure was created
with BioRender.com
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 11 of 15
Zeng et al. Journal of Neuroinammation (2025) 22:72
of astrocytes is lower than that of microglia and astro-
cytes are more resistant to alterations in lysosomal pH
compared to microglia [158]. Despite being more acidic,
another study has shown that astrocytes phagocytose
less Aβ than microglia in cell culture and rat brain slices
[159]. In the presence of dying cells, astrocytes appear
to degrade most cells at proximity without the need for
constant cell migration, while microglia are sparse and
require constant movement to detect and engage dying
cells. is suggests that astrocytic phagocytosis would
be more energetically favorable than microglia [160]. A
similar study in the degradation of toxic materials like
Aβ show a complementary feedback between microglia
and astrocytes to remove the aggregate [161]. Further-
more, there is evidence to suggest that astrocytes can
potentially exchange materials with microglia through
tunneling nanotubes (Fig.4B), which may promote more
ecient phagocytosis, although the exact mechanism
remains to be investigated [162–164].
An important future direction of study would be to
document crosstalk between microglia and astrocytes,
which would provide more detailed understanding and
insights into the overall phagocytic processes [165,
166]. is new appreciation for the phagocytic function
of astrocytes complements the basal autophagy func-
tions of astrocytes to contribute to neurodegenerative
and neuroprotective mechanisms in the CNS. is shifts
the current treatment paradigm to consider restoration
of lysosomal acidication and degradative functions in
astrocytes as a therapeutic target for neurodegenera-
tive diseases [24, 95]. Astrocytes and microglia may have
cooperative or opposing interactions during phagocyto-
sis as well as further interactions with neurons [167, 168].
e extent of which they target specic synapses or toxic
proteins and how they work together in dierent circum-
stances requires further investigation. ere is also inter-
action between astrocytes and endothelial cells through
microRNA that targets V-ATPase and modulates lyso-
somal acidication which can determine the level of
endothelial adhesion molecules and the extent of neutro-
phil migration through the BBB [169]. It is important to
comprehend these interactions and their eects on brain
homeostasis under both healthy and diseased conditions,
as this knowledge is crucial for developing treatments for
neurological disorders.
Acknowledgements
The authors thank the funding sources for supporting this work.
Author contributions
C.H.L. conceived the review topic. J.Z., J.I., and C.H.L. wrote the manuscript
and prepared the gures. D.P. provided critical comments and edited the
manuscript.
Funding
C.H.L. is supported by a start-up grant from the Department of Biology at
Syracuse University. J.Z. is supported by a start-up grant from the Department
of Biomedical and Chemical Engineering at Syracuse University.
Data availability
No datasets were generated or analysed during the current study.
Declarations
Ethics approval and consent to participate
Not applicable.
Consent for publication
All authors have approved the nal version of the manuscript.
Competing interests
The authors declare no competing interests.
Received: 2 May 2024 / Accepted: 5 March 2025
References
1. von Bartheld CS, Bahney J, Herculano-Houzel S. The search for true numbers
of neurons and glial cells in the human brain: A review of 150 years of cell
counting. J Comp Neurol. 2016. pp. 3865–95.
2. Valles SL, Singh SK, Campos-Campos J, Colmena C, Campo-Palacio I, Alvarez-
Gamez K et al. Functions of astrocytes under normal conditions and after a
brain disease. Int J Mol Sci. 2023.
3. Pociūtė A, Pivoriūnas A, Verkhratsky A. Astrocytes dynamically regulate the
blood-brain barrier in the healthy brain. Neural Regeneration Res. 2024;19.
4. Chen Z, Yuan Z, Yang S, Zhu Y, Xue M, Zhang J, et al. Brain energy metabolism:
astrocytes in neurodegenerative diseases. CNS Neurosci Ther. 2023;29:24–36.
5. Park J, Chung W-S. Astrocyte-dependent circuit remodeling by synapse
phagocytosis. Curr Opin Neurobiol. 2023;81:102732.
6. Ponath G, Park C, Pitt D. The role of astrocytes in multiple sclerosis. Front
Immunol. 2018;9:217.
7. Xu T, Liu C, Deng S, Gan L, Zhang Z, Yang G-Y, et al. The roles of microglia and
astrocytes in Myelin phagocytosis in the central nervous system. J Cereb
Blood Flow Metabolism. 2022;43:325–40.
8. Konishi H, Koizumi S, Kiyama H. Phagocytic astrocytes: emerging from the
shadows of microglia. Glia. 2022;70:1009–26.
9. Giovannoni F, Quintana FJ. The role of astrocytes in CNS inammation. Trends
Immunol. 2020;41:805–19.
10. Hasel P, Aisenberg WH, Bennett FC, Liddelow SA. Molecular and metabolic
heterogeneity of astrocytes and microglia. Cell Metabol. 2023;35:555–70.
11. Lo CH, Skarica M, Mansoor M, Bhandarkar S, Toro S, Pitt D. Astrocyte heteroge-
neity in multiple sclerosis: current Understanding and technical challenges.
Front Cell Neurosci. 2021;15:726479.
12. Matias I, Morgado J, Gomes FCA. Astrocyte heterogeneity: impact to brain
aging and disease. Front Aging Neurosci. 2019.
13. Moulson AJ, Squair JW, Franklin RJM, Tetzla W, Assinck P. Diversity of Reactive
Astrogliosis in CNS Pathology: Heterogeneity or Plasticity? Frontiers in Cellular
Neuroscience. 2021.
14. Brandebura AN, Paumier A, Onur TS, Allen NJ. Astrocyte contribution to
dysfunction, risk and progression in neurodegenerative disorders. Nat Rev
Neurosci. 2023;24:23–39.
15. Kreher C, Favret J, Maulik M, Shin D. Lysosomal functions in glia associated
with neurodegeneration. Biomolecules. 2021;11.
16. Sung K, Jimenez-Sanchez M. Autophagy in astrocytes and its implications in
neurodegeneration. J Mol Biol. 2020;432:2605–21.
17. Zhou Z, Zhou J, Liao J, Chen Z, Zheng Y. The emerging role of astrocytic
autophagy in central nervous system disorders. Neurochem Res. 2022.
18. Galloway DA, Phillips AEMM, Owen DRJJ, Moore CS. Phagocytosis in the
brain: homeostasis and disease. Front Immunol. 2019;10:790.
19. Wang J-L, Xu C-J. Astrocytes autophagy in aging and neurodegenerative
disorders. Biomed pharmacotherapy = Biomedecine Pharmacotherapie.
2020;122:109691.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 12 of 15
Zeng et al. Journal of Neuroinammation (2025) 22:72
20. Kawai A, Uchiyama H, Takano S, Nakamura N, Ohkuma S. Autophagosome-
lysosome fusion depends on the pH in acidic compartments in CHO cells.
Autophagy. 2007;3:154–7.
21. Papouin T, Dunphy J, Tolman M, Foley JC, Haydon PG. Astrocytic control of
synaptic function. Philosophical Trans Royal Soc Lond Ser B Biol Sci. 2017;372.
22. Kıray H, Lindsay SL, Hosseinzadeh S, Barnett SC. The multifaceted role of
astrocytes in regulating myelination. Exp Neurol. 2016;283:541–9.
23. Lööv C, Mitchell CH, Simonsson M, Erlandsson A. Slow degradation in
phagocytic astrocytes can be enhanced by lysosomal acidication. Glia.
2015;63:1997–2009.
24. Lööv C, Erlandsson A. Lysosomal acidication in cultured astrocytes using
nanoparticles. Methods in molecular biology. (Clifton NJ). 2017;1594:165–77.
25. Cox GM, Kithcart AP, Pitt D, Guan Z, Alexander J, Williams JL, et al. Macro-
phage migration inhibitory factor potentiates Autoimmune-Mediated
neuroinammation. J Immunol. 2013;191:1043–54.
26. Quan L, Uyeda A, Muramatsu R. Central nervous system regeneration: the
roles of glial cells in the potential molecular mechanism underlying remyelin-
ation. Inamm Regeneration. 2022;42:7.
27. Kallergi E, Siva Sankar D, Matera A, Kolaxi A, Paolicelli RC, Dengjel J, et al. Pro-
ling of puried autophagic vesicle degradome in the maturing and aging
brain. Neuron. 2023;111:2329–e23477.
28. Litwiniuk A, Juszczak GR, Stankiewicz AM, Urbańska K. The role of glial
autophagy in Alzheimer’s disease. Mol Psychiatry. 2023;28:4528–39.
29. Mitsui S, Yamaguchi J, Suzuki C, Uchiyama Y, Tanida I. TUNEL-positive struc-
tures in activated microglia and SQSTM1/p62-positive structures in activated
astrocytes in the neurodegenerative brain of a CLN10 mouse model. Glia.
2023;71:2753–69.
30. Wang T, Sun Y, Dettmer U. Astrocytes in Parkinson’s Disease: From Role to
Possible Intervention. Cells. 2023.
31. Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L, et
al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature.
2017;541:481–7.
32. Leng K, Rose IVL, Kim H, Xia W, Romero-Fernandez W, Rooney B, et al. CRISPRi
screens in human iPSC-derived astrocytes elucidate regulators of distinct
inammatory reactive States. Nat Neurosci. 2022;25:1528–42.
33. Rooney B, Leng K, Mccarthy F, Rose IVL, Herrington KA, Bax S et al. mTOR
controls neurotoxic lysosome exocytosis in inammatory reactive astrocytes.
2021;1–32.
34. Lo CH, Zeng J, Loi GWZ, Saipuljumri EN, O’Connor LM, Indajang J et al.
Defective lysosomal acidication contributes to TNFR1 mediated neuronal
necroptosis in Alzheimer’s disease. bioRxiv. 2023;2023.10.12.562041.
35. Leng K, Rooney B, McCarthy F, Xia W, Rose IVL, Bax S, et al. mTOR activation
induces endolysosomal remodeling and nonclassical secretion of IL-32 via
exosomes in inammatory reactive astrocytes. J Neuroinamm. 2024;21:198.
36. Jung B-K, Park Y, Yoon B, Bae J-S, Han S-W, Heo J-E, et al. Reduced secre-
tion of LCN2 (lipocalin 2) from reactive astrocytes through autophagic and
proteasomal regulation alleviates inammatory stress and neuronal damage.
Autophagy. 2023;19:2296–317.
37. Lee E, Jung Y-J, Park YR, Lim S, Choi Y-J, Lee SY, et al. A distinct astrocyte
subtype in the aging mouse brain characterized by impaired protein homeo-
stasis. Nat Aging. 2022;2:726–41.
38. Simmnacher K, Krach F, Schneider Y, Alecu JE, Mautner L, Klein P, et al. Unique
signatures of stress-induced senescent human astrocytes. Exp Neurol.
2020;334:113466.
39. Beckel JM, Gómez NM, Lu W, Campagno KE, Nabet B, Albalawi F et al. Stimula-
tion of TLR3 triggers release of lysosomal ATP in astrocytes and epithelial cells
that requires TRPML1 channels. Sci Rep. 2018;8.
40. Sanmarco LM, Wheeler MA, Gutiérrez-Vázquez C, Polonio CM, Lin-
nerbauer M, Pinho-Ribeiro FA, et al. Gut-licensed IFNγ + NK cells drive
LAMP1 + TRAIL + anti-inammatory astrocytes. Nature. 2021;590:473–9.
41. Di Malta C, Fryer JD, Settembre C, Ballabio A. Astrocyte dysfunction triggers
neurodegeneration in a lysosomal storage disorder. Proc Natl Acad Sci USA.
2012;109:E2334–42.
42. Aaki E, Stubbleeld BK, McGlinchey RP, McMahon B, Ory DS, Sidransky E. A
characterization of gaucher iPS-derived astrocytes: potential implications for
Parkinson’s disease. Neurobiol Dis. 2020;134:104647.
43. Di Malta C, Fryer JD, Settembre C, Ballabio A. Autophagy in astrocytes: a novel
culprit in lysosomal storage disorders. Autophagy. 2012;8:1871–2.
44. Dong A, Yang Y, Jiang S, Yao X, Qi D, Mao C, et al. Pramipexole inhibits astro-
cytic NLRP3 inammasome activation via Drd3-dependent autophagy in a
mouse model of Parkinson’s disease. Acta Pharmacol Sin. 2023;44:32–43.
45. Cheng X, Wei Y, Qian Z, Han L. Autophagy balances neuroinammation in
Alzheimer’s disease. Cell Mol Neurobiol. 2023;43:1537–49.
46. Ioannou MS, Jackson J, Sheu S-H, Chang C-L, Weigel AV, Liu H, et al. Neuron-
Astrocyte metabolic coupling protects against Activity-Induced fatty acid
toxicity. Cell. 2019;177:1522–e153514.
47. Volterra A, Meldolesi J. Astrocytes, from brain glue to communication ele-
ments: the revolution continues. Nat Rev Neurosci. 2005;6:626–40.
48. Bantle CM, Hirst WD, Weihofen A, Shlevkov E. Mitochondrial dysfunction in
astrocytes: A role in Parkinson’s disease?? Front Cell Dev Biology. 2021;8.
49. Lin M, Liu N, Qin Z, Wang Y. Mitochondrial-derived damage-associated
molecular patterns amplify neuroinammation in neurodegenerative dis-
eases. Acta Pharmacol Sin. 2022;43:2439–47.
50. Deus CM, Yambire KF, Oliveira PJ, Raimundo N. Mitochondria-Lysosome cross-
talk: from physiology to neurodegeneration. Trends Mol Med. 2020;26:71–88.
51. Yazdankhah M, Ghosh S, Liu H, Hose S, Zigler JS, Sinha D. Mitophagy in Astro-
cytes Is Required for the Health of Optic Nerve. Cells. 2023.
52. Demers-Lamarche J, Guillebaud G, Tlili M, Todkar K, Bélanger N, Grondin
M, et al. Loss of mitochondrial function impairs lysosomes. J Biol Chem.
2016;291:10263–76.
53. Tripathi M, Zhang CW, Singh BK, Sinha RA, Moe KT, DeSilva DA, et al. Hyper-
homocysteinemia causes ER stress and impaired autophagy that is reversed
by vitamin B supplementation. Cell Death Dis. 2016;7:e2513–2513.
54. Huang YC, Hsu SM, Shie FS, Shiao YJ, Chao L-JJ, Chen HW et al. Reduced
mitochondria membrane potential and lysosomal acidication are associated
with decreased oligomeric Aβ degradation induced by hyperglycemia: A
study of mixed glia cultures. PLoS ONE. 2022;17.
55. Wang N, Zhao Y, Wu M, Li N, Yan C, Guo H et al. Gembrozil alleviates cogni-
tive impairment by inhibiting ferroptosis of astrocytes via restoring the Iron
metabolism and promoting antioxidant capacity in type 2 diabetes. Mol
Neurobiol. 2023.
56. Luo D, Ye W, Chen L, Yuan X, Zhang Y, Chen C et al. PPARa Inhibits Astrocyte
Inammation Activation by Restoring Autophagic Flux after Transient Brain
Ischemia. Biomedicines. 2023.
57. Du H, Guo Y, Zhu Y, Gao D, Lin B, Liu Y, et al. RIPK1 Inhibition contributes to
lysosomal membrane stabilization in ischemic astrocytes via a lysosomal
Hsp70.1B-dependent mechanism. Acta Pharmacol Sin. 2023;44:1549–63.
58. Liu J, Yang L, Tian H, Ma Q. Cathepsin D is involved in the oxygen and glucose
deprivation/reperfusion-induced apoptosis of astrocytes. Int J Mol Med.
2016;38:1257–63.
59. Zhang X, Chen C, Liu Y. Navigating the metabolic maze: anomalies in fatty
acid and cholesterol processes in Alzheimer’s astrocytes. Alzheimer’s Res
Therapy. 2024;16:63.
60. Zeng J, Lo CH, Editorial. Lipid metabolism dysregulation in Obesity-Related
diseases and neurodegeneration. Front Endocrinol. 2025;16.
61. Yang D, Wang X, Zhang L, Fang Y, Zheng Q, Liu X, et al. Lipid metabolism
and storage in neuroglia: role in brain development and neurodegenerative
diseases. Cell and Bioscience. BioMed Central Ltd; 2022.
62. Ortiz-Rodriguez A, Arevalo M-A. The contribution of astrocyte autophagy to
systemic metabolism. Int J Mol Sci. 2020.
63. Alam S, Afsar SY, Van Echten-Deckert G. S1P released by SGPL1-Decient
astrocytes enhances astrocytic ATP production via S1PR2,4, thus keeping
autophagy in check: potential consequences for brain health. Int J Mol Sci.
2023.
64. Sha L, Li J, Shen H, Wang Q, Meng P, Zhang X, et al. LHPP-mediated inorganic
pyrophosphate hydrolysis-driven lysosomal acidication in astrocytes regu-
lates adult neurogenesis. Cell Rep. 2023;42:112975.
65. Zhao N, Liu C-C, Qiao W, Bu G, Apolipoprotein E. Receptors, and modulation
of Alzheimer’s disease. Biol Psychiatry. 2018;83:347–57.
66. Huang Y, Mahley RW, Apolipoprotein E, Pt. 3–12.
67. Farmer BC, Kluemper J, Johnson LA. Apolipoprotein E4 alters astrocyte fatty
acid metabolism and lipid droplet formation. Cells. 2019;8.
68. Schmukler E, Solomon S, Simonovitch S, Goldshmit Y, Wolfson E, Michaelson
DM, et al. Altered mitochondrial dynamics and function in APOE4-expressing
astrocytes. Cell Death Dis. 2020;11:578.
69. Larramona-Arcas R, González-Arias C, Perea G, Gutiérrez A, Vitorica J, García-
Barrera T, et al. Sex-dependent calcium hyperactivity due to lysosomal-
related dysfunction in astrocytes from APOE4 versus APOE3 gene targeted
replacement mice. Mol Neurodegeneration. 2020;15:35.
70. Prasad H, Rao R. Amyloid clearance defect in ApoE4 astrocytes is reversed
by epigenetic correction of endosomal pH. Proc Natl Acad Sci USA.
2018;115:E6640–9.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 13 of 15
Zeng et al. Journal of Neuroinammation (2025) 22:72
71. Nuriel T, Peng KY, Ashok A, Dillman AA, Figueroa HY, Apuzzo J et al. The
endosomal-lysosomal pathway is dysregulated by APOE4 expression in vivo.
Front NeuroSci. 2017;11.
72. Persson T, Lattanzio F, Calvo-Garrido J, Rimondini R, Rubio-Rodrigo M,
Sundström E, et al. Apolipoprotein E4 elicits lysosomal cathepsin D release,
decreased Thioredoxin-1 levels, and apoptosis. J Alzheimer’s Disease.
2017;56:601–17.
73. Lee H, Cho S, Kim M-J, Park YJ, Cho E, Jo YS et al. ApoE4-dependent lysosomal
cholesterol accumulation impairs mitochondrial homeostasis and oxidative
phosphorylation in human astrocytes. Cell Rep. 2023;42.
74. Hammel G, Zivkovic S, Ayazi M, Ren Y. Consequences and mechanisms of
Myelin debris uptake and processing by cells in the central nervous system.
Cell Immunol. 2022;380:104591.
75. Chen K, Garcia Padilla C, Kiselyov K, Kozai TDY. Cell-specic alterations in
autophagy-lysosomal activity near the chronically implanted microelec-
trodes. Biomaterials. 2023;302:122316.
76. Ponath G, Ramanan S, Mubarak M, Housley W, Lee S, Sahinkaya FR, et al.
Myelin phagocytosis by astrocytes after Myelin damage promotes lesion
pathology. Brain. 2017;140:399–413.
77. Wang S, Deng J, Fu H, Guo Z, Zhang L, Tang P. Astrocytes directly clear Myelin
debris through endocytosis pathways and followed by excessive gliosis after
spinal cord injury. Biochem Biophys Res Commun. 2020;525:20–6.
78. Grajchen E, Hendriks JJA, Bogie JFJ. The physiology of foamy phagocytes in
multiple sclerosis. Acta Neuropathol Commun. 2018;6:124.
79. Ortiz-Rodriguez A, Acaz-Fonseca E, Boya P, Arevalo MA, Garcia-Segura LM.
Lipotoxic eects of palmitic acid on astrocytes are associated with autophagy
impairment. Mol Neurobiol. 2019;56:1665–80.
80. Asimakidou E, Saipuljumri EN, Lo CH, Zeng J. Role of metabolic dysfunction
and inammation along the liver–brain axis in animal models with obesity-
induced neurodegeneration. Neural Regeneration Res. 2025;20.
81. Zeng J, Lo CH, Cheong LYT. Therapeutic targeting of obesity-induced neuro-
inammation and neurodegeneration. Front Endocrinol. 2025;15:1456948.
82. Thaler JP, Yi C-X, Schur EA, Guyenet SJ, Hwang BH, Dietrich MO, et al. Obesity
is associated with hypothalamic injury in rodents and humans. J Clin Investig.
2012;122:153–62.
83. Varela L, Kim JG, Fernández-Tussy P, Aryal B, Liu ZW, Fernández-Hernando C,
et al. Astrocytic lipid metabolism determines susceptibility to diet-induced
obesity. Sci Adv. 2023;7:eabj2814.
84. Pla A, Pascual M, Guerri C. Autophagy constitutes a protective mechanism
against ethanol toxicity in mouse astrocytes and neurons. PLoS ONE. 2016;11.
85. Lu K, Zimmermann M, Görg B, Bidmon HJ, Biermann B, Klöcker N, et al.
Hepatic encephalopathy is linked to alterations of autophagic ux in astro-
cytes. EBioMedicine. 2019;48:539–53.
86. Pascua-Maestro R, Diez-Hermano S, Lillo C, Ganfornina MD, Sanchez D.
Protecting cells by protecting their vulnerable lysosomes: identication of a
new mechanism for preserving lysosomal functional integrity upon oxidative
stress. PLoS Genet. 2017;13:e1006603.
87. Wang F, Gómez-Sintes R, Boya P. Lysosomal membrane permeabilization and
cell death. Trac. 2018;19:918–31.
88. Giusti V, Kaur G, Giusto E, Civiero L. Brain clearance of protein aggregates: a
close-up on astrocytes. Mol Neurodegeneration. 2024;19:5.
89. Smethurst P, Risse E, Tyzack GE, Mitchell JS, Taha DM, Chen Y-R, et al. Distinct
responses of neurons and astrocytes to TDP-43 proteinopathy in amyo-
trophic lateral sclerosis. Brain. 2020;143:430–40.
90. Yang Y, Song J-J, Choi YR, Kim S, Seok M-J, Wulansari N et al. Therapeutic
functions of astrocytes to treat α-synuclein pathology in Parkinson’s disease.
Proceedings of the National Academy of Sciences. 2022;119:e2110746119.
91. Kim S, Chun H, Kim Y, Kim Y, Park U, Chu J, et al. Astrocytic autophagy plastic-
ity modulates Aβ clearance and cognitive function in Alzheimer’s disease.
Mol Neurodegeneration. 2024;19:55.
92. Chow SK, Yu D, MacDonald CL, Buibas M, Silva GA. Amyloid β-peptide directly
induces spontaneous calcium transients, delayed intercellular calcium waves
and gliosis in rat cortical astrocytes. ASN Neuro. 2010;2:15–23.
93. Stenovec M, Trkov S, Lasič E, Terzieva S, Kreft M, Rodríguez Arellano JJ, et al.
Expression of Familial alzheimer disease presenilin 1 gene attenuates vesicle
trac and reduces peptide secretion in cultured astrocytes devoid of patho-
logic tissue environment. GLIA. 2016;64:317–29.
94. St-Pierre M-K, Carrier M, González Ibáñez F, Khakpour M, Wallman M-J, Parent
M, et al. Astrocytes display ultrastructural alterations and heterogeneity in
the hippocampus of aged APP-PS1 mice and human post-mortem brain
samples. J Neuroinamm. 2023;20:73.
95. Xiao Q, Yan P, Ma X, Liu H, Perez R, Zhu A, et al. Enhancing astrocytic lysosome
biogenesis facilitates Aβ clearance and attenuates amyloid plaque pathogen-
esis. J Neuroscience: Ocial J Soc Neurosci. 2014;34:9607–20.
96. Deng Y, Wang S-Y, Wang Q-G, Xu Z-H, Peng Q, Chen S-Y, et al. AVE 0991 sup-
presses Astrocyte-Mediated neuroinammation of Alzheimer’s disease by
enhancing autophagy. J Inamm Res. 2023;16:391–406.
97. Konstantinidis E, Dakhel A, Beretta C, Erlandsson A. Long-term eects of
amyloid-beta deposits in human iPSC-derived astrocytes. Mol Cell Neurosci.
2023;125:103839.
98. Chen X, Gao R, Song Y, Xu T, Jin L, Zhang W, et al. Astrocytic AT1R deciency
ameliorates Aβ-induced cognitive decits and synaptotoxicity through
β-arrestin2 signaling. Prog Neurobiol. 2023;228:102489.
99. Wirth S, Schlößer A, Beiersdorfer A, Schweizer M, Woo MS, Friese MA et al.
Astrocytic uptake of posttranslationally modied amyloid-β leads to endoly-
sosomal system disruption and induction of pro-inammatory signaling. Glia.
2024;n/a.
100. Gong C, Bonli L, Zheng Y, Cecarini V, Cuccioloni M, Angeletti M, et al. Immor-
talized Alzheimer’s disease astrocytes: characterization of their proteolytic
systems. Mol Neurobiol. 2023;60:2787–800.
101. Söllvander S, Nikitidou E, Brolin R, Söderberg L, Sehlin D, Lannfelt L, et al.
Accumulation of amyloid-β by astrocytes result in enlarged endosomes
and microvesicle-induced apoptosis of neurons. Mol Neurodegeneration.
2016;11:38.
102. Domínguez-Prieto M, Velasco A, Tabernero A, Medina JM. Endocytosis and
transcytosis of Amyloid-β peptides by astrocytes: A possible mechanism
for Amyloid-β clearance in Alzheimer’s disease. J Alzheimer’s Disease.
2018;65:1109–24.
103. Zeng X, Cheung SKK, Shi M, Or PMY, Li Z, Liu JYH, et al. Astrocyte-specic
knockout of YKL-40/Chi3l1 reduces Aβ burden and restores memory func-
tions in 5xFAD mice. J Neuroinamm. 2023;20:290.
104. Hong Y, Liu Y, Zhang G, Wu H, Hou Y. Progesterone suppresses Aβ(42)-
induced neuroinammation by enhancing autophagy in astrocytes. Int
Immunopharmacol. 2018;54:336–43.
105. Li M-Z, Zheng L-J, Shen J, Li X-Y, Zhang Q, Bai X, et al. SIRT1 facilitates amyloid
beta peptide degradation by upregulating lysosome number in primary
astrocytes. Neural Regeneration Res. 2018;13:2005–13.
106. Rose K, Jepson T, Shukla S, Maya-Romero A, Kampmann M, Xu K et al.
Tau brils induce nanoscale membrane damage and nucleate cytosolic
tau at lysosomes. Proceedings of the National Academy of Sciences.
2024;121:e2315690121.
107. Martini-Stoica H, Cole AL, Swartzlander DB, Chen F, Wan Y-W, Bajaj L, et al.
TFEB enhances astroglial uptake of extracellular Tau species and reduces Tau
spreading. J Exp Med. 2018;215:2355–77.
108. Polito VA, Li H, Martini-Stoica H, Wang B, Yang L, Xu Y, et al. Selective clearance
of aberrant Tau proteins and rescue of neurotoxicity by transcription factor
EB. EMBO Mol Med. 2014;6:1142–60.
109. Xu Y, Du S, Marsh JA, Horie K, Sato C, Ballabio A, et al. TFEB regulates lyso-
somal exocytosis of Tau and its loss of function exacerbates Tau pathology
and spreading. Mol Psychiatry. 2021;26:5925–39.
110. Erustes AG, Stefani FY, Terashima JY, Stilhano RS, Monteforte PT, da Silva
Pereira GJ, et al. Overexpression of α-synuclein in an astrocyte cell line pro-
motes autophagy Inhibition and apoptosis. J Neurosci Res. 2018;96:160–71.
111. Wang P, Lan G, Xu B, Yu Z, Tian C, Lei X, et al. α-Synuclein-carrying astrocytic
extracellular vesicles in Parkinson pathogenesis and diagnosis. Translational
Neurodegeneration. 2023;12:40.
112. Cavaliere F, Cerf L, Dehay B, Ramos-Gonzalez P, De Giorgi F, Bourdenx M, et
al. In vitro α-synuclein neurotoxicity and spreading among neurons and
astrocytes using lewy body extracts from Parkinson disease brains. Neurobiol
Dis. 2017;103:101–12.
113. Hong B, Ohtake Y, Itokazu T, Yamashita T. Glial senescence enhances
α-synuclein pathology owing to its insucient clearance caused by
autophagy dysfunction. Cell Death Discovery. 2024;10:50.
114. Lee H-J, Suk J-E, Patrick C, Bae E-J, Cho J-H, Rho S, et al. Direct transfer of
α-Synuclein from neuron to astroglia causes inammatory responses in
synucleinopathies. J Biol Chem. 2010;285:9262–72.
115. Zhang Y, He X, Wu X, Lei M, Wei Z, Zhang X, et al. Rapamycin upregulates
glutamate transporter and IL-6 expression in astrocytes in a mouse model of
Parkinson’s disease. Cell Death Dis. 2017;8:e2611.
116. Henry AG, Aghamohammadzadeh S, Samaroo H, Chen Y, Mou K, Needle E, et
al. Pathogenic LRRK2 mutations, through increased kinase activity, produce
enlarged lysosomes with reduced degradative capacity and increase
ATP13A2 expression. Hum Mol Genet. 2015;24:6013–28.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 14 of 15
Zeng et al. Journal of Neuroinammation (2025) 22:72
117. Tsunemi T, Ishiguro Y, Yoroisaka A, Valdez C, Miyamoto K, Ishikawa K, et
al. Astrocytes protect human dopaminergic neurons from α-Synuclein
accumulation and propagation. J Neuroscience: Ocial J Soc Neurosci.
2020;40:8618–28.
118. Morrone Partt G, Coccia E, Goldman C, Whitney K, Reyes R, Sarrafha L, et al.
Disruption of lysosomal proteolysis in astrocytes facilitates midbrain organoid
proteostasis failure in an early-onset Parkinson’s disease model. Nat Commun.
2024;15:447.
119. Pereira GJS, Hirata H, Fimia GM, do Carmo LG, Bincoletto C, Han SW, et al. Nic-
otinic acid adenine dinucleotide phosphate (NAADP) regulates autophagy in
cultured astrocytes. J Biol Chem. 2011;286:27875–81.
120. Vest RT, Chou C-C, Zhang H, Haney MS, Li L, Laqtom NN, et al. Small
molecule C381 targets the lysosome to reduce inammation and ame-
liorate disease in models of neurodegeneration. Proc Natl Acad Sci USA.
2022;119:e2121609119.
121. Chin MY, Ang K-H, Davies J, Alquezar C, Garda VG, Rooney B, et al. Phenotypic
screening using High-Content imaging to identify lysosomal pH modulators
in a neuronal cell model. ACS Chem Neurosci. 2022;13:1505–16.
122. Li B, Liu T, shen Y, Qin J, Chang X, Wu M et al. TFEB/LAMP2 contributes to
PM0.2-induced autophagy-lysosome dysfunction and alpha-synuclein
dysregulation in astrocytes. J Environ Sci. 2023.
123. Zeng J, Acin-Perez R, Assali EA, Martin A, Brownstein AJ, Petcherski A, et al.
Restoration of lysosomal acidication rescues autophagy and metabolic
dysfunction in non-alcoholic fatty liver disease. Nat Commun. 2023;14:2573.
124. Lo CH, O’Connor LM, Loi GWZ, Saipuljumri EN, Indajang J, Lopes KM, et al.
Acidic nanoparticles restore lysosomal acidication and rescue metabolic
dysfunction in pancreatic β-Cells under lipotoxic conditions. ACS Nano.
2024;18:15452–67.
125. Trudeau KM, Colby AH, Zeng J, Las G, Feng JH, Grinsta MW, et al. Lysosome
acidication by photoactivated nanoparticles restores autophagy under
lipotoxicity. J Cell Biol. 2016;214:25–34.
126. Dai D, He L, Chen Y, Zhang C. Astrocyte responses to nanomaterials:
functional changes, pathological changes and potential applications. Acta
Biomater. 2021;122:66–81.
127. Lo CH, Zeng J. Defective lysosomal acidication: a new prognostic marker
and therapeutic target for neurodegenerative diseases. Translational Neuro-
degeneration. 2023;12:29.
128. Quick JD, Silva C, Wong JH, Lim KL, Reynolds R, Barron AM, et al. Lysosomal
acidication dysfunction in microglia: an emerging pathogenic mechanism
of neuroinammation and neurodegeneration. J Neuroinamm. 2023;20:185.
129. Koh J-Y, Kim HN, Hwang JJ, Kim Y-H, Park SE. Lysosomal dysfunction in pro-
teinopathic neurodegenerative disorders: possible therapeutic roles of cAMP
and zinc. Mol Brain. 2019;12:18.
130. Lee H, Koh JY. Roles for H+/K+-ATPase and zinc transporter 3 in cAMP-
mediated lysosomal acidication in Balomycin A1-treated astrocytes. GLIA.
2021;69:1110–25.
131. Kim HN, Seo BR, Kim H, Koh JY. Cilostazol restores autophagy ux in Balomy-
cin A1-treated, cultured cortical astrocytes through lysosomal reacidication:
roles of PKA, zinc and Metallothionein 3. Sci Rep. 2020;10:9175.
132. Shim MS, Kim KY, Bu JH, Nam HS, Jeong SW, Park TL et al. Elevated intracel-
lular cAMP exacerbates vulnerability to oxidative stress in optic nerve head
astrocytes Article. Cell Death Dis. 2018;9.
133. Ogura M, Taniura H, Nakamichi N, Yoneda Y. Upregulation of the glutamine
transporter through transactivation mediated by cAMP/protein kinase A
signals toward exacerbation of vulnerability to oxidative stress in rat neocorti-
cal astrocytes. J Cell Physiol. 2007;212:375–85.
134. Song W, Wang F, Savini M, Ake A, di ronza A, Sardiello M, et al. TFEB regulates
lysosomal proteostasis. Hum Mol Genet. 2013;22:1994–2009.
135. Chandra S, Jana M, Pahan K. Aspirin induces lysosomal biogenesis and
attenuates amyloid plaque pathology in a mouse model of Alzheimer’s
disease via PPARα. J Neuroscience: Ocial J Soc Neurosci. 2018;38:6682–99.
136. Tanaka Y, Suzuki G, Matsuwaki T, Hosokawa M, Serrano G, Beach TG, et al. Pro-
granulin regulates lysosomal function and biogenesis through acidication
of lysosomes. Hum Mol Genet. 2017;26:969–88.
137. Caliò A, Segala D, Munari E, Brunelli M, Martignoni G. MiT family translocation
renal cell carcinoma: from the early descriptions to the current knowledge.
Cancers. 2019;11.
138. Perera RM, Stoykova S, Nicolay BN, Ross KN, Fitamant J, Boukhali M, et al. Tran-
scriptional control of autophagy-lysosome function drives pancreatic cancer
metabolism. Nature. 2015;524:361–5.
139. Beard E, Lengacher S, Dias S, Magistretti PJ, Finsterwald C. Astrocytes as key
regulators of brain energy metabolism: new therapeutic perspectives. Front
Physiol. 2021;12:825816.
140. Cisneros J, Belton TB, Shum GC, Molakal CG, Wong YC. Mitochondria-
lysosome contact site dynamics and misregulation in neurodegenerative
diseases. Trends Neurosci. 2022;45:312–22.
141. Lee J-H, Yang D-S, Goulbourne CN, Im E, Stavrides P, Pensalni A, et al. Faulty
autolysosome acidication in Alzheimer’s disease mouse models induces
autophagic build-up of Aβ in neurons, yielding senile plaques. Nat Neurosci.
2022;25:688–701.
142. Sun W, Liu Z, Jiang X, Chen MB, Dong H, Liu J et al. Spatial and single-cell
transcriptomics reveal neuron-astrocyte interplay in long-term memory.
BioRxiv. 2023;2023.03.20.533566.
143. Kodam P, Sai Swaroop R, Pradhan SS, Sivaramakrishnan V, Vadrevu R.
Integrated multi-omics analysis of Alzheimer’s disease shows molecular
signatures associated with disease progression and potential therapeutic
targets. Sci Rep. 2023;13:3695.
144. O’Connor LM, O’Connor BA, Zeng J, Lo CH. Data Mining of Microarray Datas-
ets in Translational Neuroscience. Brain Sciences. 2023.
145. O’Connor LM, O’Connor BA, Lim S, Bin, Zeng J, Lo CH. Integrative multi-omics
and systems bioinformatics in translational neuroscience: A data mining
perspective. J Pharm Anal. 2023;13:836–50.
146. Lo CH. Towards clinical translation of biomarkers and therapies targeting
autolysosomal acidication dysfunction in neuroinammation and neurode-
generation—an interview with prof. David Rubinsztein. Aging Pathobiology
Ther. 2024;6:138–40.
147. Rama Rao KV, Kielian T. Astrocytes and lysosomal storage diseases. Neurosci-
ence. 2016;323:195–206. Available from: h t t p s : / / w w w . s c i e n c e d i r e c t . c o m / s c i e
n c e / a r t i c l e / p i i / S 0 3 0 6 4 5 2 2 1 5 0 0 5 0 3 5
148. Bosch ME, Kielian T. Astrocytes in juvenile neuronal ceroid lipofuscinosis
(CLN3) display metabolic and calcium signaling abnormalities. J Neurochem.
2019;148:612–24. Available from: h t t p s : / / d o i . o r g / 1 0 . 1 1 1 1 / j n c . 1 4 5 4 5
149. Platt FM, d’Azzo A, Davidson BL, Neufeld EF, Tit CJ. Lysosomal storage
diseases. Nat Rev Dis Primers. 2018;4:27. Available from: h t t p s : / / d o i . o r g / 1 0 . 1 0
3 8 / s 4 1 5 7 2 - 0 1 8 - 0 0 2 5 - 4
150. Lange J, Haslett LJ, Lloyd-Evans E, Pocock JM, Sands MS, Williams BP et al.
Compromised astrocyte function and survival negatively impact neurons
in infantile neuronal ceroid lipofuscinosis. Acta Neuropathol Commun.
2018;6:74. Available from: h t t p s : / / d o i . o r g / 1 0 . 1 1 8 6 / s 4 0 4 7 8 - 0 1 8 - 0 5 7 5 - 4
151. Liddelow SA, Guttenplan KA, Clarke LE, Bennett FC, Bohlen CJ, Schirmer L et
al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature.
2017;541:481–7. Available from: h t t p s : / / d o i . o r g / 1 0 . 1 0 3 8 / n a t u r e 2 1 0 2 9
152. Konishi H, Okamoto T, Hara Y, Komine O, Tamada H, Maeda M, et al. Astrocytic
phagocytosis is a compensatory mechanism for microglial dysfunction.
EMBO J. 2020;39:e104464.
153. Chung W-S, Clarke LE, Wang GX, Staord BK, Sher A, Chakraborty C, et al.
Astrocytes mediate synapse elimination through MEGF10 and MERTK path-
ways. Nature. 2013;504:394–400.
154. Lee J-H, Kim J-Y, Noh S, Lee H, Lee SY, Mun JY, et al. Astrocytes phagocytose
adult hippocampal synapses for circuit homeostasis. Nature. 2021;590:612–7.
155. Stevens B, Allen NJ, Vazquez LE, Howell GR, Christopherson KS, Nouri N, et al.
The classical complement cascade mediates CNS synapse elimination. Cell.
2007;131:1164–78.
156. Chu Y, Jin X, Parada I, Pesic A, Stevens B, Barres B et al. Enhanced synaptic
connectivity and epilepsy in C1q knockout mice. Proceedings of the National
Academy of Sciences. 2010;107:7975–80.
157. Schafer DP, Lehrman EK, Kautzman AG, Koyama R, Mardinly AR, Yamasaki R, et
al. Microglia sculpt postnatal neural circuits in an activity and Complement-
Dependent manner. Neuron. 2012;74:691–705.
158. Deng D, Wang B, Guan Y, Zheng H, Mutlu AS, Wang MC. Quantitative proling
of lysosomal pH heterogeneity using uorescence lifetime imaging micros-
copy. BioRxiv. 2023;2023.09.25.559395.
159. Prakash P, Jethava KP, Korte N, Izquierdo P, Favuzzi E, Rose IVLL, et al. Monitor-
ing phagocytic uptake of amyloid Β into glial cell lysosomes in real time.
Chem Sci. 2021;12:10901–18.
160. Damisah EC, Hill RA, Rai A, Chen F, Rothlin CV, Ghosh S, et al. Astrocytes and
microglia play orchestrated roles and respect phagocytic territories during
neuronal corpse removal in vivo. Sci Adv. 2020;6:eaba3239.
161. Lian H, Litvinchuk A, Chiang AC-A, Aithmitti N, Jankowsky JL, Zheng H.
Astrocyte-Microglia Cross Talk through Complement Activation Modulates
Amyloid Pathology in Mouse Models of Alzheimer’s Disease. The Journal of
Neuroscience. 2016;36:577 LP – 589.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
Page 15 of 15
Zeng et al. Journal of Neuroinammation (2025) 22:72
162. Rostami J, Holmqvist S, Lindström V, Sigvardson J, Westermark GT, Ingelsson
M et al. Human Astrocytes Transfer Aggregated Alpha-Synuclein via Tunnel-
ing Nanotubes. The Journal of Neuroscience. 2017;37:11835 LP – 11853.
163. Scheiblich H, Dansokho C, Mercan D, Schmidt SV, Bousset L, Wischhof L, et
al. Microglia jointly degrade brillar alpha-synuclein cargo by distribution
through tunneling nanotubes. Cell. 2021;184:5089–e510621.
164. Wiersma VI, Polymenidou M. Sharing is caring: the benets of distributing
protein aggregates among microglial networks. Neuron. 2021;109:3228–30.
165. Gotoh M, Miyamoto Y, Ikeshima-Kataoka H. Astrocytic neuroimmunological
roles interacting with microglial cells in neurodegenerative diseases. Int J Mol
Sci. 2023.
166. Jha MK, Jo M, Kim JH, Suk K. Microglia-Astrocyte Crosstalk: An Intimate
Molecular Conversation. Neuroscientist. 2019. pp. 227–40.
167. Chakraborty R, Nonaka T, Hasegawa M, Zurzolo C. Tunnelling nanotubes
between neuronal and microglial cells allow bi-directional transfer of
α-Synuclein and mitochondria. Cell Death Dis. 2023;14:329.
168. Stanca S, Rossetti M, Bongioanni P. Astrocytes as Neuroimmunocytes in
Alzheimer’s Disease: A Biochemical Tool in the Neuron–Glia Crosstalk along
the Pathogenetic Pathways. International Journal of Molecular Sciences.
2023.
169. Wu X, Liu H, Hu Q, Wang J, Zhang S, Cui W, et al. Astrocyte-Derived extracel-
lular vesicular miR-143-3p dampens autophagic degradation of endothelial
adhesion molecules and promotes neutrophil transendothelial migration
after acute brain injury. Adv Sci. 2024;11:2305339.
Publisher’s note
Springer Nature remains neutral with regard to jurisdictional claims in
published maps and institutional aliations.
Content courtesy of Springer Nature, terms of use apply. Rights reserved.
1.
2.
3.
4.
5.
6.
Terms and Conditions
Springer Nature journal content, brought to you courtesy of Springer Nature Customer Service Center GmbH (“Springer Nature”).
Springer Nature supports a reasonable amount of sharing of research papers by authors, subscribers and authorised users (“Users”), for small-
scale personal, non-commercial use provided that all copyright, trade and service marks and other proprietary notices are maintained. By
accessing, sharing, receiving or otherwise using the Springer Nature journal content you agree to these terms of use (“Terms”). For these
purposes, Springer Nature considers academic use (by researchers and students) to be non-commercial.
These Terms are supplementary and will apply in addition to any applicable website terms and conditions, a relevant site licence or a personal
subscription. These Terms will prevail over any conflict or ambiguity with regards to the relevant terms, a site licence or a personal subscription
(to the extent of the conflict or ambiguity only). For Creative Commons-licensed articles, the terms of the Creative Commons license used will
apply.
We collect and use personal data to provide access to the Springer Nature journal content. We may also use these personal data internally within
ResearchGate and Springer Nature and as agreed share it, in an anonymised way, for purposes of tracking, analysis and reporting. We will not
otherwise disclose your personal data outside the ResearchGate or the Springer Nature group of companies unless we have your permission as
detailed in the Privacy Policy.
While Users may use the Springer Nature journal content for small scale, personal non-commercial use, it is important to note that Users may
not:
use such content for the purpose of providing other users with access on a regular or large scale basis or as a means to circumvent access
control;
use such content where to do so would be considered a criminal or statutory offence in any jurisdiction, or gives rise to civil liability, or is
otherwise unlawful;
falsely or misleadingly imply or suggest endorsement, approval , sponsorship, or association unless explicitly agreed to by Springer Nature in
writing;
use bots or other automated methods to access the content or redirect messages
override any security feature or exclusionary protocol; or
share the content in order to create substitute for Springer Nature products or services or a systematic database of Springer Nature journal
content.
In line with the restriction against commercial use, Springer Nature does not permit the creation of a product or service that creates revenue,
royalties, rent or income from our content or its inclusion as part of a paid for service or for other commercial gain. Springer Nature journal
content cannot be used for inter-library loans and librarians may not upload Springer Nature journal content on a large scale into their, or any
other, institutional repository.
These terms of use are reviewed regularly and may be amended at any time. Springer Nature is not obligated to publish any information or
content on this website and may remove it or features or functionality at our sole discretion, at any time with or without notice. Springer Nature
may revoke this licence to you at any time and remove access to any copies of the Springer Nature journal content which have been saved.
To the fullest extent permitted by law, Springer Nature makes no warranties, representations or guarantees to Users, either express or implied
with respect to the Springer nature journal content and all parties disclaim and waive any implied warranties or warranties imposed by law,
including merchantability or fitness for any particular purpose.
Please note that these rights do not automatically extend to content, data or other material published by Springer Nature that may be licensed
from third parties.
If you would like to use or distribute our Springer Nature journal content to a wider audience or on a regular basis or in any other manner not
expressly permitted by these Terms, please contact Springer Nature at
onlineservice@springernature.com