Abstract
Cell transplantation is an attractive treatment strategy for a variety of brain disorders, as it promises to replenish lost functions and rejuvenate the brain. In particular, transplantation of astrocytes has come into light recently as a therapy for amyotrophic lateral sclerosis (ALS); moreover, grafting of astrocytes also showed positive results in models of other conditions ranging from neurodegenerative diseases of older age to traumatic injury and stroke. Despite clear differences in etiology, disorders such as ALS, Parkinson’s, Alzheimer’s, and Huntington’s diseases, as well as traumatic injury and stroke, converge on a number of underlying astrocytic abnormalities, which include inflammatory changes, mitochondrial damage, calcium signaling disturbance, hemichannel opening, and loss of glutamate transporters. In this review, we examine these convergent pathways leading to astrocyte dysfunction, and explore the existing evidence for a therapeutic potential of transplantation of healthy astrocytes in various models. Existing literature presents a wide variety of methods to generate astrocytes, or relevant precursor cells, for subsequent transplantation, while described outcomes of this type of treatment also differ between studies. We take technical differences between methodologies into account to understand the variability of therapeutic benefits, or lack thereof, at a deeper level. We conclude by discussing some key requirements of an astrocyte graft that would be most suitable for clinical applications.
Keywords: astrocyte, transplantation, neurodegeneration, injury, brain, spinal cord
Main Points
Astrocytic pathologies are found in many disorders, thus transplantation of healthy astrocytes can have therapeutic benefits. Regional and functional astrocyte heterogeneity has to be considered when choosing the optimal transplant source.
Introduction
Cell therapies constitute an emerging class of therapeutic approaches to promote regeneration of damaged tissues. In particular, the brain presents a lucrative target since many neurological disorders result from the death of specific cell types; replenishing the cells of the central nervous system (CNS) aims to restore lost abilities and modify the course of a disease.
For example, neurons represent a valuable therapeutic tool to replenish the loss of specific neuronal subtypes in neurological disorders, and some benefits can be seen upon neuronal engraftment in Parkinson’s disease (PD) patients and models1–5. However, neuronal axons can reach up to 1 m in length in the healthy CNS, and even if the cell bodies survive and integrate well into the local host network with their shorter dendrites, there is little evidence supporting the ability of transplanted neurons (or neuron-differentiated stem cells) to send long axonal projections to their correct targets. In line with this, several groups observed that the functional benefit derived from the neuronal transplantation therapy cannot be explained by restoration of correct axonal circuits, but rather may be due to the secretion of protective factors and integration in local cellular networks at the site of implantation where they encourage plasticity within existing cells6–10.
Astrocytic processes, on the other hand, are well-positioned to physiologically integrate into the local cellular networks to take place of lost or diseased host astrocytes. Their ability to interact with and regulate multiple aspects of functionality of other cell types, such as neurons, makes it plausible that astrocytic replacement would have an effect on the astrocytic networks and beyond.
Stem cells have also been widely used for grafting. While some positive indications were obtained in several early studies11,12, results remain variable, and safety concerns persist as the precise mechanisms of action of stem cell grafts are uncertain 13 . The exact differentiation paths of these grafts within the host tissue are unpredictable, and stem cells are more likely to give rise to unwanted cellular phenotypes under the hostile conditions of the diseased CNS than pre-differentiated cells. This may at least partially account for the inconsistency of functional outcomes. Moreover, transplanted stem cells can remain undifferentiated and migratory 14 , which does not preclude the possibility of tumor development longer term. Tumorigenicity after stem cell transplantation has been described in several models15,16, and the short life span of common model organisms does not allow for predictions of how such grafts may behave in, for instance, stroke or PD patients, who are able to survive for decades after their diagnosis.
A growing body of literature highlights the importance of astrocytes in the healthy brain. These cells represent a cell population that forms complex networks capable of cross-talking to neuronal nets17–19. Astrocytic networks appear to act as nexus points interacting with, and regulating key aspects of functions of neurons20,21, oligodendrocytes22,23, microglia 24 , and the neurovascular unit18,25; and the presence of human-specific astrocyte types26,27 points toward potentially novel roles of these cells to be considered in the case of patient-specific grafts. Astrocyte-lineage cells also act as stem cells with neurogenic potential that can replenish neuronal populations, especially under inflammatory conditions28,29.
Moreover, astrocytic abnormalities have been described in association with, and sometimes as a cause of, various developmental and degenerative disorders ranging from autism 30 and Down syndrome 31 to multiple sclerosis32,33 and psychiatric conditions34,35. These findings call for an assessment of the utility of astrocyte transplantation as a clinical strategy. Experimental depletion of these cells, either pharmacologic or genetic, has shown that the lack of functional astrocytes can lead to depressive symptoms36,37, cognitive 38 and motor39,40 impairment, seizures 41 , abnormal behaviours41,42, memory disturbance43,44, and neuronal death40,45 in the healthy animals, indicating that the healthy astrocytes are indispensable for the normal brain function. In the context of disease and inflammation, however, both positive and negative outcomes of astrocyte depletion have been reported45–47, suggesting that the roles of these cells are complex, and it is necessary to understand the protective pathways in greater depth.
Traditionally, neuronal and neural stem cell (NSC) replacement have been considered as the main avenue for the treatment of brain disorders since neuronal death is a prominent hallmark of many conditions of the brain, and a good number of detailed reviews on this topic exist48–51. At the same time, despite growing academic and commercial (e.g. AstranauTX, Astrocyte Pharmaceuticals Inc, and Kadimastem) interest in astrocyte-directed therapeutics, the literature on astrocyte transplantation is more limited compared to the neuronal counterpart, and opinions on its effectiveness diverge.
In the current review, we outline the disorders in which transplantation of astrocytes has been attempted so far. We describe examples of transplantation strategies employed in the following disorders, or the models of: amyotrophic lateral sclerosis (ALS), Parkinson’s, Alzheimer’s, and Huntington’s diseases, traumatic injury, and ischaemic stroke. We examine some common themes emerging across these pathologies—including immune milieu alternations, calcium signaling abnormalities, and mitochondrial dysfunction, and consider the mechanisms through which healthy astrocytes can therefore ameliorate these disorders.
A number of recent reviews discussed astrocyte-lineage cells in the context of transplantation as therapeutic and research modeling options. Transplantation of astrocytes and their progenitors has been reviewed in the context of ALS, and the importance of the location of the graft has been emphasised52,53. In PD, the dual beneficial versus harmful nature of astrocytic involvement has been considered, and some potentially protective genetic modifications of transplanted cells have been put forward 54 . Replacement of damaged cells, including astrocyte-lineage cells, after spinal cord injury (SCI), has been discussed, alongside the use of biomaterials to aid in cell guidance and integration into the host tissues55,56. Several reviews focused on the regenerative potential of stem cells and progenitors such as glial-restricted precursor cells (GRPs) capable of differentiating into astrocytes among other cell types as sources of cellular grafts57–64, or the ability of transplanted stem cells to interact with the host astrocytes and stimulate protective pathways in the latter65–68 in several conditions, including stroke, SCI, and Alzheimer’s disease (AD). In particular, protection of the blood-brain barrier (BBB) and the neurovascular unit by transplanted astrocytes has received attention in studies of neurodegenerative diseases 63 . Heterogeneity of astrocytic phenotypes (“neurotoxic” A1 and “neuroprotective” A2) and its influence on their ability to exert protective functions in disorders such as stroke have been considered 69 . Moreover, human chimeric mice, in which cell transplantation has been employed to model neurological diseases, allowed researchers to study disease mechanisms in highly translationally relevant models 70 .
The therapeutic benefits of astrocyte transplantation have been variable, and the reasons for these discrepancies have not yet been examined in depth, nor summarized in a single publication. We hypothesize that the discrepancies could be, at least in part, attributed to the astrocyte heterogeneity due to the technical differences in the astrocyte generation as well as the brain region (of cell origin and of the transplant location within the recipient). We take a closer look at the proposed roles of astrocytes in brain disorders and how these cells specifically become affected by various pathologies. Then, we outline the methodologies used to generate astrocytes, or precursor cells, for subsequent engraftment, in order to identify the most promising strategies that could be scaled up for future clinical applications. Overview of the full mechanisms of the listed pathologies beyond astrocytes (including neuronal pathology and an in-depth overview of the neurogenic potential of astrocytes) is outside of the scope of the current review, and interested readers are provided with a brief summary of each disorder supplemented with some useful references.
Amyotrophic Lateral Sclerosis
ALS is a disorder in which benefits of astrocytic grafting are most clearly established as demonstrated by the outcomes of the phase II clinical trial for ALS (10 patients split across two cohorts) released in December 2020. The treatment involved intrathecal injection of astrocytes into the spinal cord and was found to be well-tolerated; it also reduced the disease progression rate in a significant and clinically meaningful manner (Table 1).
Table 1.
Disease or disease model and age at transplantation | Type of astrocyte-lineage cells or precursors transplanted | Site of transplantation within the CNS | Endpoint/treatment duration | Outcome summary | References |
---|---|---|---|---|---|
ALS—human adults, 10 patients (5 patients per cohort) | Pre-differentiated astrocytes derived from human embryonic stem cells—AstroRx® (Produced by Kadimastem); 100 × 106 cells per patient in Cohort A, 250 × 106 cells per patient in Cohort B |
Intrathecal administration—spinal cord under local anesthesia | 6 months for Cohorts A and B. Repeated treatment schedule (every 2–3 months) planned for the upcoming Cohort C |
Phase I&IIa clinical trials, assessment of safety and preliminary efficiency. No treatment-related serious adverse effects (SAEs), no dose-limiting toxicities. Statistically significant reduction in the muscle function decline assessed by the average ALS Functional Rating Scale-Revised (ALSFRS-R) change for 3–4 months post-treatment compared to the pre-treatment period |
a |
ALS—SOD1G93A rats, 90 day-old | GRPs derived from healthy rodent (rat or mouse) embryonic spinal cords | Cervical spinal cord ventral horn levels C4,5, and 6 bilaterally | Until end-stage, up to approx. 3 months post-transplantation (end stage was defined by the inability of rats to right themselves within 30s when placed on the side) | GRPs differentiated preferentially into astrocytes; reduced microgliosis, slower decline in forelimb motor control and respiratory function, reduced motor neuron loss. GRPs deficient in GLT1 served as a negative control and did not show positive effects. | Lepore et al. 71 |
ALS—SOD1G93A mice, 50- to 60-day old | Human GRPs (Q-Cells®) derived from fetal forebrain | Cervical spinal cord ventral horn levels C4 and 5 bilaterally | Until end-stage, up to approx. 3 months post-transplantation (end stage was defined by the inability of mice to right themselves within 30s when placed on the side) | Human GRPs differentiated preferentially into astrocytes, up to 10% of cells continued to proliferate 3 months post-engraftment. No therapeutic benefit achieved. | Lepore et al. 72 |
ALS—SOD1G93A mice, 90 day-old | Human iPSCs line 201B7 clone selected for its low tumorigenicity, pre-differentiated into glial-rich neural progenitors (GRNPs) by BMP4+LIF treatment | Lumbar spinal cord ventral horn levels L3 and 4 bilaterally | Until end-stage, up to approx. 2.5 months post-transplantation | Modest lifespan prolongation, transient improvement of the lower limb function. | Kondo et al. 73 |
ALS—SOD1G93A mice, 67 ± 2 days-old, or 2 injections at 67 ± 2 and 97 ± 2 days of age | Human astrocytes derived from human ESCs (two stem cell lines: HADC100 and NCL-14) through astrocyte progenitor cell (APC) stage | Injection into the CSF through cisterna magna | Until end-stage, approx. 2 months post-first transplantation point | Trends toward longer survival, improved motor performance on the rotarod, significantly delayed disease onset; two injections are more efficient than one. | Izrael et al. 74 |
ALS—SOD1G93A rats, 2 injections at 50 ± 2 and 70 ± 2 days of age | Same as above | Intrathecal injection by lumbar puncture in the subarachnoid space between L5 and L6 (most relevant to the human administration method) | Until end-stage, up to 5.5 months post-first transplantation point | Trends toward longer survival, significantly delayed disease onset, treated rats maintained their body weight significantly longer, improved motor performance on the rotarod and in the grip strength test; no tumors formed. | Izrael et al. 74 |
ALS: amyotrophic lateral sclerosis; BMP4: bone morphogenic protein 4; CNS: central nervous system; GLT1: glutamate transporter 1; GRPs: glial-restricted precursors; GRNPs: glial-rich neural progenitors; CSF: cerebrospinal fluid; ESCs: embryonic stem cells; LIF: leukemia inhibitory factor; SOD1: superoxide dismutase 1.
ALS usually affects adults with an average age of diagnosis of 55 years and an incidence of about two per 100 000 people. Only 5% to 10% of all ALS cases run in families, as opposed to sporadic onset75,76. Superoxide dismutase 1 (SOD1) gene mutations are most commonly associated with ALS accounting for 20% of familial cases 77 . This mitochondrial enzyme acts by scavenging superoxide radicals to protect cells from excessive amounts of reactive oxygen species (ROS). Other genes such as a DNA-binding protein TDP-43/TARDPB and C9ORF72 have also been implicated in ALS pathogenesis, suggesting that multiple aetiological pathways have the capacity to lead to the eventual ALS manifestation—profound loss of upper and lower motor neurons in the spinal cord and brain leading to eventual paralysis75,78,79.
Interestingly, even though the neuronal pathology is well-described, profound changes in astrocytes accompany or even precede the disease. Causal effects of astrocytic aberrations in the ALS pathology are demonstrated by the motor dysfunction and motor neuron degeneration initiated after transplantation of the SODG93A harboring GRPs 80 or ALS patient-iPSC-derived astrocytes81,82 into the spinal cord of the wild-type mice. Following this line of evidence, healthy astrocyte transplantation showed a beneficial effect upon engraftment into the spinal cords of SOD1 mutant mice and rats71,73,74, prolonging survival time and diminishing the disease progression rate in these animals (Table 1). In accordance with these data, astrocyte-specific decrease in the mutant SOD1 load attenuated disease progression in mouse ALS models83,84.
In addition to the non-cell autonomous effects on motor neurons caused by SOD mutations in astrocytes, ALS-associated mutations induce drastic changes in astrocyte biology, at least in vivo, including reactive astrocytosis, also known as astrogliosis, as evidenced by the increased GFAP expression and process hypertrophy85–90, and ultimately astrocyte degeneration and apoptosis 91 . These can result from either, or a combination of, mutations within astrocytes themselves, or from altered functions of surrounding neurons and other cells such as microglia. Curiously, some of the proliferating GFAP-positive cells in ALS may not be true astrocytes, but aberrant glial cells with an astrocyte-like phenotype of microglial origin, which may not be sufficient to substitute for the loss of the true astrocytic functions while contributing to the pro-inflammatory milieu generation92,93.
As suggested by the fact that SOD is an enzyme involved in mitochondrial protection from excessive levels of ROS, oxidative stress alongside mitochondrial damage are well-established hallmarks of ALS94–96, which is also true in cases of ALS caused by other mutations97–99. Since astrocytes are a major source of the antioxidant glutathione (GSH) in the brain100,101, oxidative damage to astrocytes is likely to make the brain milieu more vulnerable to ROS. Mitochondrial abnormalities, elevated levels of inducible nitric oxide synthase (iNOS), and ultimately increased levels of ROS in astrocytes are associated with motor neuron degeneration102–104. Since mitochondrial functions are closely associated with calcium signalling105–107, it is not surprising that altered calcium homeostasis has been observed in astrocytes carrying ALS-linked mutations108–110. Abnormally elevated intracellular calcium levels in response to stimuli such as ATP 109 could result in caspase activation with subsequent astrocytosis 111 . Enhanced SNARE-dependent exocytosis of the vesicles containing microRNA, glutamate, and ATP may also contribute to the neuronal toxicity of the mutant astrocyte-conditioned medium109,112,113.
Several secreted factors can also be responsible for such neurotoxic effects, including upregulated IFNγ, IL6, prostaglandin D2, tumor necrosis factor α (TNFα), and TGFβ that are released in the extracellular milieu114–116. Moreover, elevated extracellular glutamate levels were also found in ALS models where oxidative stress and potentially other changes in SOD1 mutant astrocytes result in downregulation of glutamate transporters117,118, initiating excitotoxic cascades in the neighboring motor neurons. Activation of caspase-3, a marker of apoptosis, in astrocytes can also downregulate glutamate transporter EEAT2 expression 119 , thereby contributing to the increased extracellular glutamate accumulation and excitotoxicity to perpetuate the vicious cycle. On the other hand, healthy astrocytes harboring a “younger” phenotype, such as the hESC-derived cells used for grafting in a murine model of ALS, exhibit a more protective secretory profile by supplying GDNF, VEGF, osteopontin, and CXCl-16 chemokine, which stimulate neuronal survival and regeneration, as well as matrix metalloproteinase inhibitors TIMP-1 and 2, in order to preserve supportive extracellular matrix (ECM) composition and suppress immune cell infiltration 74 .
Considering that the ALS-associated mutations affect all cells in the brain, selective vulnerability of motor neurons, but not other neuron types, co-cultured with SOD1 mutant astrocytes is intriguing. Activation of LT-βR by theTNF superfamily member LIGHT triggers a motor neuron-selective death pathway 120 , and so does the activation of FasL121,122. Both of these pathways are potentiated by diseased astrocytes through secretion of IFNγ 120 or ATP 123 , respectively. Additionally, NGF, secreted by astrocytes under inflammatory conditions such as those found in ALS104,124, promotes cell survival in cells expressing both TrkA and p75NTR, 125 but if TrkA is absent, cell death results126,127. Although motor neurons are normally devoid of both, in ALS re-expression of p75NTR becomes evident128–130, which may contribute to this selective degeneration.
In this context, replacement of healthy astrocytes is a promising approach for the treatment of ALS. Wild-type astrocytes can help reduce excitotoxicity by removing excessive glutamate, provide lactate for surrounding neurons, reduce oxidative stress, and secrete anti-inflammatory factors that are beneficial for both diseased neurons and astrocytes in the host tissue. Effective healthy astrocyte replacement should be able to improve the muscle function and ultimately prolong the patient’s lifespan, with Kadimastem’s AstroRx offering a potential candidate for further scaling up of this approach.
Parkinson’s Disease
PD is a progressive neurodegenerative condition that shares some pathophysiological similarities with ALS, including abnormal protein aggregation and mitochondrial dysfunction131–133. Moreover, up to a third of all ALS patients experience symptoms of Parkinsonism 134 , and the co-occurrence of the two pathologies is especially obvious in Lytico-bodig disease and in the ALS-parkinsonism-dementia complex 135 . PD is the second most common neurodegenerative condition that typically affects older adults, with 96% of cases diagnosed after the age of 50, with an incidence of almost 2,000 per 100 000 people over the age of 80 136 , 137 .
PD is a clinically heterogenous disorder characterized by the loss of dopaminergic neurons 138 accompanied by reactive changes in astrocytes and microglia 139 in the nigrostriatal system. Loss of the nigrostriatal dopaminergic innervation results in persistent tremor, bradykinesia, rigidity, and postural instability140,141. However, it becomes apparent that many other systems are affected by PD including autonomic and cognitive dysfunctions 142 . It has been suggested that the earliest signs of PD may start within the gastrointestinal system where resident neurons143,144 and astrocyte-lineage-related cells (enteric glial cells [EGCs]) 145 become affected. Accumulation of α-synuclein protein aggregates known as Lewy bodies is a common neuropathological finding that tends to spread in a stereotypic pattern known as Braak stages—hindbrain structures such as the brain stem and midbrain develop a higher protein aggregate load earlier in disease while forebrain structures remain relatively unaffected until later disease stages146–148. Current treatments include dopamine replacement and deep brain stimulation (DBS), which only provide symptomatic relief, while serious side effects including hallucinations and drug-induced dyskinesias often limit the therapeutic benefits of these approaches 138 . Interestingly, the mechanism of action of DBS may include stimulation of chemical transmitter release from astrocytes 149 .
Human astrocyte transplantation has been trialed in rodent models of PD with promising results (Table 2).
Table 2.
Disease or disease model and age at transplantation | Type of astrocyte-lineage cells or precursors transplanted | Site of transplantation within the CNS | Endpoint/treatment duration | Outcome summary | References |
---|---|---|---|---|---|
PD—6-OHDA injected (left striatum) Sprague-Dawley rats, adult | Human mesenchymal stem cells (MSCs) pre-differentiated into astrocyte-like cells | Left striatum, 6 weeks post-lesion | 120 days (4 months) after grafting) | Reduced rotational behavior, enhanced density of dopaminergic fibers. | Bahat-Stroomza et al. 150 |
PD—6-OHDA injected (right striatum) Fisher 344 male rats, adult | GDAs, human or Fisher 344 hPAP transgenic rat, treated with either BMP4 or CNTF | Right striatum, 4 weeks post-lesion | 7 weeks post-lesion (3 weeks post-transplantation); 2 weeks post-treatment for behavior analysis | GDABMP4 (but not GDACNTF) transplantation benefits: improvement in left paw usage, reduced rotational behavior, increased TH labeling (dopaminergic neuron marker) in the right striatum and protection of parvalbumin-expressing GABAergic interneurons, increased synaptophysin expression. | Proschel et al. 151 |
PD—6-OHDA injected (right side of substantia nigra and the median forebrain bundle), Sprague-Dawley female rats, adult | NPCs, from mouse embryonic ventral midbrain) alone or in combination with cortical or ventral midbrain rodent postnatal day 5- to 7-exctracted and cultured astrocytes; or with ventral midbrain astrocytes transduced with Nurr1+Foxa2 transcription factors | Striatum bilaterally | 6 months post-transplantation | Co-grafting NPCs with ventral midbrain astrocytes showed the greatest amount of benefits in terms of reduction of rotational behavior to levels comparable with controls and number of NPCs differentiating into dopaminergic neurons. | Song et al. 152 |
PD: Parkinson’s disease; BMP4: bone morphogenic protein 4; CNS: central nervous system; CNTF: ciliary neurotrophic factor; GDAs: glial precursor derived astrocytes; hPAP: human placental alkaline phosphatase; NPCs: nNeural precursor cells; 6-OHDA: 6-hydroxydopamine; TH: tyrosine hydroxylase.
A unique population of astrocytes matured from GRPs through the exposure to bone morphogenic protein 4 (GRPsBMP4) rescued motor symptoms, which was accompanied by an increase in striatal dopamine production and neuronal survival 151 . Other methods of human astrocyte lineage cell generation, such as differentiation from the adult bone marrow mesenchymal stromal cells, yielded similar behavioral improvements 150 . In another study, modified astrocyte-lineage cells destined for the striatal graft were engineered to deliver tyrosine hydroxylase (TH), which is normally expressed by neurons, enhanced local dopamine synthesis, and improved motor coordination in a rat model 153 . Finally, co-grafting of astrocytes alongside neural progenitor cells into the striatum also led to an enhanced behavioral recovery in a mouse model of PD that surpassed the therapeutic benefit of grafting these progenitors alone 152 .
Perhaps the most compelling evidence of astrocyte involvement in PD was shown by the selective astrocytic expression of mutant A53T α-synuclein, associated with a familial form of the disorder, which led to profound dopaminergic and motor neuron degeneration accompanied by microglial activation within the hindbrain 154 . Moreover, co-culture of healthy neurons with patient-derived astrocytes differentiated from iPSCs led to neurodegeneration and α-synuclein accumulation 155 , showing that PD-linked mutation in astrocytes alone is sufficient for profound pathology in astrocytes and neurons to develop. It is therefore evident that these cells play an important role in PD progression. It is noteworthy that, under healthy conditions, mesencephalic regions such as SNpc (which are more vulnerable to PD-inducing insults) may be less dense in astrocytes compared to neighboring regions 156 , pointing at the neuroprotective effects of astrocyte presence. Curiously, two drugs currently being explored in human PD clinical trials, zonisamide 157 and rotigotine 158 , have been shown to increase astrocytic proliferation and stimulate secretion of neuroprotective factors from these cells.
Despite α-synuclein being a predominantly neuronal protein, α-synuclein aggregates in other cell types including astrocytes have been reported 148 , and specific upregulation of α-synuclein was seen in astrocytes derived from patient iPSCs carrying LRRK2 mutation (kinase involved in autophagy and associated with an autosomal dominant form of PD) 155 . Astrocytes carrying LRRK2 mutations that were derived from patient iPSC lines also showed decreased astrocytic marker expression and complexity, and these cells produced higher levels of ROS 159 . In addition to the cell-autonomous changes in PD astrocytes, these cells are capable of taking up the misfolded α-synuclein from neurons through endocytosis160–163, which can result in pathological activation of the former 164 . α-synuclein aggregates taken up by astrocytes can be cleared via the lysosomal pathway165,166, but excessive protein build-up resulting from the increased α-synuclein release 167 , or insufficient clearance 165 , can lead to mitochondrial stress 168 , autophagy dysfunction155,166,169, ER-Golgi system stress 170 , and eventual astrocyte apoptosis169–172. Importance of the mitochondrial disbalance in PD pathogenesis is further emphasized by the fact that PINK1, an autophagy-related gene that is associated with familial PD forms and is predominantly active within astrocytes as opposed to neurons 173 , encodes a mitochondrial kinase. Mutations in this protein can lead to defective astrocytic proliferation and ATP levels, heightened ROS levels, and a decreased ability to uptake glucose as well as to lower growth factor receptor expression 174 . DJ-1 is another mitochondrial-stabilizing gene whose deletion causes a familial PD form, and its deletion negatively affected astrocytic mitochondrial function and the ability of these cells to protect astrocyte-neuron co-cultures against toxic insults 175 .
In response to cellular stress caused by α-synuclein 164 and parkinsonism-inducing neurotoxins176–180, astrocytes undergo reactive changes 181 resulting in neuroinflammation. Reactive astrocytosis does not only prevent these cells from secreting trophic factors like GDNF family of ligands, BDNF, NT3, and mesencephalic astrocyte-derived neurotrophic factor (MANF)152,182–185, but also stimulates release of pro-inflammatory cytokines such as IL1β 186 . Consistently, it has been noted that differentiated astrocytes used for transplantation express trophic factors that are known to be neuroprotective, which can at least partially account for the benefits of the transplantation151,152.
Transplanted astrocytes, therefore, can provide multiple benefits in PD by replenishing the resident astrocytic pool in place of the apoptotic cells, secreting neuroprotective factors, restoring potassium buffering, degrading α-synuclein through lysosomal pathways, and reducing oxidative stress by supplying ROS-scavenging enzymes. Artificial expression of transcription factors (Nurr1 and Foxa2) in astrocytes co-grafted with stem cells promoted a non-reactive phenotype of the astroglia 152 , suggesting that bioengineering approaches can ensure that the cells maintain the beneficial phenotype even in the presence of ROS and pro-inflammatory cytokines among other PD-associated stressors. Effective healthy astrocyte replacement in PD could be able to address motor and/or non-motor symptoms of this disorder. While most studies focused on the motor manifestations such as tremor and rigidity, up to 50% of people with PD report the non-motor symptoms, including memory issues, sleep disturbance, and depression, to be the major determinants of their quality of life 187 . Recalling that the experimental loss of astrocyte function in the cortical areas and the hippocampus result in memory disturbance43,44 and depressive symptoms36,37, inclusion of the non-motor symptoms of PD in the efficacy assessment of pre-clinical and clinical PD trials of astrocyte-centered therapies could prove fruitful and open new avenues complimentary to dopamine replacement approaches.
Alzheimer’s Disease
AD is the most prevalent neurodegenerative disorder that shares important similarities with the pathogenesis of PD. These similarities include a progressive, stereotypic pattern of misfolded protein accumulation. It starts within the entorhinal cortex and medial temporal structures and gradually spreads to the basal ganglia188,189. Amyloid-β plaques derived from the amyloid precursor protein (APP), and hyperphosphorylated tau-containing neurofibrillary tangles are hallmarks of AD found extra- and intracellularly, respectively 190 . A number of tauopathies including frontotemporal dementia (FTD) are also associated with the tau tangles and lead to certain symptoms similar to those of AD191,192. Interestingly, amyloid-β and α-synuclein pathologies co-exist in up to 50% of AD patients193–196, suggesting that common cellular abnormalities, including those found in astrocytes, are likely to be found both in AD and PD.
Similarly to PD, AD typically manifests later in life, affecting around 10% of over 65-year-olds with disease prevalence strongly correlating with age197–199. Memory loss is an early symptom of AD 200 , but as the disease progresses, other neurological functions become affected and symptoms such as speech impairment and lack of motor coordination become prominent. Heritability of this disorder is estimated to range from 60% to 80%, suggesting an important role for genetic factors200,201. Mutations in the APP gene can predispose the amyloid-β protein to misfold and are often found in association with the familial form of the disorder. PSEN1 and PSEN2 are also commonly associated with AD and encode for the proteins in the γ-secretase complex, which is necessary to cleave amyloid-β from its precursor APP 200 . The ε4 allele of apolipoprotein (APOE4), a major cholesterol carrier, represents the strongest risk factor for sporadic late-onset AD 202 , and it is noteworthy that astrocytes show the highest degree of apolipoprotein expression among all CNS cell types 203 . Current AD treatment strategies are purely symptomatic; these include cholinesterase inhibitors, which aim to increase acetylcholine levels, and NMDA receptor antagonist memantine, which counteracts excitotoxicity 204 .
An interesting approach to transplantation has been attempted in a rat model of AD generated by the infusion of a toxic form of amyloid-β (1-42 peptide), where autologous EGCs that are related to astrocytes were harvested for grafting from animals’ own appendices. EGCs were delivered into cerebral ventricles, from which they were observed to migrate toward the amyloid plaques within the brains. These cells were not only able to reduce the plaque load, but also the cytokine profile in the treated brains was shifted toward a more anti-inflammatory phenotype with a significant decrease in TNFα, PGE2, and IL6, and increase in NGF, BDNF, and GDNF. Moreover, memory and learning skills were improved by the treatment 205 . These data are corroborated by the study conducted in a mouse transgenic model of FTD harboring human P301S tau, in which transplantation of NPC-differentiated astrocytes into the cortical gray matter reversed cortical neuron loss 206 (Table 3).
Table 3.
Disease or disease model and age at transplantation | Type of astrocyte-lineage cells or precursors transplanted | Site of transplantation within the CNS | Endpoint/treatment duration | Outcome summary | References |
---|---|---|---|---|---|
AD—Sprague-Dawley male rats infused with amyloid-β42 through a mini-osmotic pump device into the right cerebral ventricle for 20 days, adult | EGCs isolated from appendices of the same rats (appendices isolated during cannula insertion into the cerebral ventricle) and cultured for 20 days | Right cerebral ventricle through the same cannula left after the amyloid-β42 infusion | 8 weeks | Autologous EGCs increased neurotrophin release and neurogenesis, reduced inflammatory cytokine secretion, induced amyloid-β plaque degradation in the cortex and hippocampus, and significantly ameliorated cognitive deficits as assessed by Morris water maze and object recognition tasks. | Esposito et al. 205 |
Tauopathy / FTD—P301S tau mice, 8 weeks-old | NPCs were isolated from the cortices of the eGFP-expressing neonatal mice; astrocytes were differentiated from NPCs by BMP4 treatment | Cortical gray matter, one hemisphere per animal | 4 or 12 weeks | Both NPCs (which preferentially differentiated into astrocytes) and pre-differentiated astrocytes significantly reduced neuronal loss. | Hampton et al. 206 |
AD: Alzheimer’s disease; BMP4: bone morphogenic protein 4; CNS: central nervous system; EGCs: enteric glial cells; eGFP: enhanced green fluorescent protein; FTD: frontotemporal dementia; NPCs: neural precursor cells.
Multiple pathological changes in astrocytes have been observed in AD and animal models of this disease 207 . Reactive astrogliosis and disruption of the astrocytic domain organization occur in an AD mouse model even before the appearance of the amyloid plaques208,209; similar evidence of astrocyte activation and degeneration was found in the brains of AD patients 210 . Indeed, predominance of the pro-inflammatory (e.g. IL1, TNFα) versus anti-inflammatory cytokines are well-documented hallmarks of AD 211 . Alterations in the surface-expressed receptors can take place in AD, such as in the case of EphB2 receptor upregulation on hippocampal astrocytes that can downregulate synaptic plasticity 212 . While reactive astrocytes surrounding amyloid-β plaques may be protective at initial disease stages, astrocytic activation increases linearly with cognitive decline 213 , likely contributing to the disease progression. Accordingly, suppression of astrocyte activation through inhibition of the JAK-STAT3 cascade was found to improve outcomes in a mouse model of AD 214 .
Interestingly, even though astrocytes surrounding plaques were found activated and hypertrophic in triple-transgenic (3xTg-AD) mice (harboring mutations in APP, presenilin, and tau), astrocytes distant from the plaques, or those analyzed at stages prior to plaque formation, were found to have dystrophic branches with reduced complexity208,209. Moreover, atrophic astrocytes were also found in the hippocampi of PDAPP mice exhibiting high levels of human APP expression 215 . Regional heterogeneity of astrocytic response has been observed with entorhinal cortex exhibiting less astrocytic activation compared to other regions such as hippocampus 208 , which may underlie selective vulnerability of certain brain regions to the AD-related degeneration.
APP, unlike tau, is expressed not only by neurons but also by astrocytes 216 ; at the same time, healthy astrocytes do not express β-secretase (BACE1), a key enzyme necessary to cleave amyloid-β from APP. However, inflammation, or chronic stress in the context of AD can induce astrocytic BACE1 expression in AD, thereby contributing to the amyloid-β load216,217. Release of amyloid-β can have cell-autonomous effects that compromise astrocytic viability 218 , and also activate microglia 219 . In addition, formation of amyloid-β oligomers is able to induce ROS release from astrocytes and trigger the loss of protective transcriptional activity of STAT3 in neurons 220 . Accordingly, increased oxidative stress was observed in astrocytes harboring tau mutations associated with FTD 221 . Considering the close connection between the ROS production and abnormal mitochondrial function, it is not surprising that multiple alterations in mitochondria-related genes were found in astrocytes from AD patients’ brains compared to the healthy elderly controls 222 .
In AD, astrocytes have been shown to be capable of clearing misfolded amyloid proteins through endocytic mechanisms and promote its elimination through the lysosomal pathway 223 or transcytosis and cerebrospinal fluid (CSF) clearance 218 . Such process is hampered by mutations in the APOE4 allele in astrocytes, which is associated with the excessive endosomal acidification, defective autophagy, and ultimately lack of sufficient amyloid-β clearance through the endosome-lysosome pathway 224 .
Hence, astrocyte grafting can aid in clearance of senile plaques, reduce pro-inflammatory cytokine concentration, supply antioxidants and enhance neurotrophic factor release, as a neuroprotective mechanism in response to the amyloid-β42 challenge 225 . It can also substitute for the atrophic and apoptotic cells in the astrocytic syncytium which could buffer abnormally elevated calcium levels in the astrocytic network. Effective healthy astrocyte replacement in AD could improve the cognitive function or reduce the rate of the functional and cognitive decline.
Huntington’s Disease
Huntington’s disease (HD) is caused by a single gene mutation—a polyglutamine (CAG repeat) expansion on the N-terminal region of huntingtin gene (HTT), in which the size of the expanded region inversely correlates with the age of disease onset 226 . Similar to other neurodegenerative proteinopathies discussed above, the pathological hallmark of HD is the aggregation of the mutant huntingtin protein (mtHtt), which can trigger the progressive neurodegeneration, particularly of the striatal GABAergic medium spiny neurons (MSNs), although other areas such as cortex also become affected 227 . No cure exists except palliative treatments that aim to alleviate the involuntary movements, or chorea, by reducing dopaminergic neurotransmission, and to suppress psychiatric manifestations228,229.
The huntingtin protein is naturally expressed in various cell types including cells outside of the brain230–233. Astrocytic pathology has been implicated in HD as shown by the study in which specific expression of mtHtt in this cell type in mice was sufficient to induce profound pathology, including motor abnormalities, body weight loss, and lower life expectancy 234 . In vitro astrocyte-neuron co-cultures demonstrated that astrocytes harboring mtHtt increased vulnerability of neurons, while wild-type astrocytes protected neighboring neurons from excitotoxicity 231 . Even more strikingly, while neurons differentiated from patient-derived iPSCs were phenotypically normal and survived in the adult mouse brain, astrocytes generated from the same cells showed cytoplasmic vacuolation under basal cell culture conditions, suggesting that HTT mutations can induce astrocytic aberrations in a cell-autonomous manner, and that astrocytes are highly sensitive to mtHtt accumulation 235 . Human glial progenitor cells (hGPCs) expressing mtHtt fail to differentiate into mature GFAP-expressing astrocytes in the rodent brain 236 . Accordingly, specific reduction of mtHtt load in astrocytes slows down disease progression in a mouse model 237 .
It is therefore not entirely surprising that transplantation of healthy astrocytes can alleviate HD disease phenotype in transgenic mice. Indeed, hGPCs comprised of astrocyte-biased precursors, when engrafted neonatally into the striatum of HD mice, successfully differentiated into astrocytes (or persisted as precursors) and delayed motor and cognitive deterioration. Importantly, striatal atrophy was also reduced by the astrocyte transplantation (Table 4). On the other hand, reverse engraftment of mtHtt-bearing precursors into healthy rodents resulted in the manifestations of HD with impaired motor coordination 238 .
Table 4.
Disease or disease model and age at transplantation | Type of astrocyte-lineage cells or precursors transplanted | Site of transplantation within the CNS | Endpoint/treatment duration | Outcome summary | References |
---|---|---|---|---|---|
HD—R6/2 (heterozygous transgenic for the 5’-end of the human HTT gene) x rag1-/- (immunodeficient) mice, postnatal day 1 (P1) | Astrocyte-biased human fetal glial precursor cells (GPCs) were isolated from the forebrain tissue (18–22 week-gestational age) | Striatum bilaterally | 8–18 weeks depending on the test | Integrated human cells did not express mHTT aggregates; treated mice survived significantly longer, showed less striatal volume loss, exhibited improved motor performance assessed by the rotarod and gait tests, and better cognitive performance analyzed by the T-maze test. Striatal neurons showed improved electrophysiological properties, and reduced striatal potassium levels were found in the presence of transplanted glia. | Benraiss et al. 238 |
HD: Huntington’s disease; CNS: central nervous system; HTT: huntingtin; mHTT: mutant huntingtin.
One documented astrocytic pathology associated with the HTT mutation and symptom onset in HD models is the reduction in inwardly rectifying potassium channel Kir4.1 expression, which alters the electrophysiological profile of astrocytes and makes surrounding neurons more prone to excitotoxic death.
Concomitant with the Kir4.1 downregulation, astrocytic and neuronal depolarization alongside elevated extracellular potassium levels were observed in a mouse HD model. Artificial AAV-mediated expression of Kir4.1-GFP in astrocytes in this model attenuated the characteristic MSN hyperexcitability associated with HD, as well as improved motor function of the animals and increased their life span 239 . To further exacerbate excitotoxicity caused by Kir4.1 downregulation, deficit in glutamate transporters, especially GLT-1, was observed in mouse models and cultured astrocytes harboring mtHtt, which can further contribute to MSN hyperexcitability by increasing glutamate concentrations within striatum231,240–244. Nuclear mtHtt inclusions are able to directly suppress GLT1 expression through interaction with and inhibition of its promoter 234 .
Post-mortem analysis of HD brains revealed morphological changes and increased astrocytosis, which correlated with disease progression 243 . Inflammation and astrocyte activation are likely to play a role in HD pathogenesis as higher levels of NFκB were found in astrocytes from HD patients and mouse models, and systemic inflammatory stimuli (e.g. LPS) elicited more prominent cytokine release from such cells. Accordingly, inhibition of the IκB kinase-NFκB pathway improved neuronal survival and ameliorated motor and cognitive deficits in the R6/2 mice 245 . JAK-STAT signaling represents another canonical pathway involved in astrocytic activation and subsequent microglial recruitment in HD. Surprisingly, blockade of this pathway increased mtHtt aggregation without affecting neuronal survival, suggesting that some aspects of astrocytic reactivity serve as a protective compensatory mechanism aimed at elimination of excessive misfolded protein 246 . Reactive astrocytes are also able to exert neuroprotective effects by promoting mtHtt degradation through upregulation of autophagy, lysosome, and proteasome-related genes 247 , which may also be a cell-autonomous, protective mechanism underlying lower mtHtt accumulation in this cell type 232 . However, this may only represent an initial compensatory response that subsides over time, as progressive ubiquitin-proteasome system decrease was shown in neurons and astrocytes of aging mice232,233.
As expected, neurons are not the only cells whose excitability is altered in HD. A mouse HD model has demonstrated altered spontaneous calcium signaling in striatal astrocytes that had, on average, diminished frequency, amplitude, and duration compared to wild-type animals. At the same time, astrocytes responded more robustly to the activity of cortical neurons due to excessive glutamate accumulation resulting from an insufficient glutamate uptake and deficiency in potassium channels240,248. To corroborate these data, specific reduction in calcium signaling in wild-type mice by transducing striatal astrocytes with a construct encoding a calcium pump PMCA2, which extrudes calcium from the cytosol, leads to excessive self-grooming behavior reminiscent of that observed in R6/2 mice 249 . Furthermore, diminished calcium signaling was found in R6/2 HD mouse model, which preceded later-stage severe motor dysfunction associated with the striatal tissue loss 42 .
In addition to abnormal cytosolic calcium signaling, mitochondrial calcium dynamics are likely to contribute to HD pathology. It is noteworthy that mitochondria of striatal astrocytes (and neurons) are less capable of calcium buffering compared to cortical cells, providing another insight into the selective vulnerability of this brain region 250 . Moreover, striatal mitochondria present with reduced mitochondrial respiratory capacity compared to their cortical counterparts, and antioxidant (N-acetylcysteine) treatment ameliorated some motor symptoms in an HD mouse model 251 .
Multiple other astrocytic genes were found to be altered in HD models, which results in suppressed BDNF secretion, perturbed calcium signaling pathways, and astrocyte activation among other pathological changes252,253. Since astrocytosis is known to be associated with neurotoxicity, pro-inflammatory cytokine profile, and even eventual astrocyte apoptosis, as discussed above for other known disorders, these factors could be expected to contribute to MSN death and other manifestations of HD. Effective healthy astrocyte replacement in HD could improve the cognitive function or reduce the rate of cognitive decline, reduce psychiatric symptoms, and/or help control the motor manifestations of this disorder, thus increasing the functional independence of patients.
Traumatic Injury to the CNS
SCI and traumatic brain injury (TBI) are among the leading causes of preventable disability in the younger population. Nearly half of the SCI incidents occur between ages 16 and 30, while only less than 1% of the affected patients make a full recovery254,255. Globally, TBI is about twice as prevalent as SCI with the numbers of cases of approximately 760 per 100 000 and 370 per 100 000, respectively 256 . Even though vehicle collisions, sport-related traumas, and physical assault are common causes of CNS injuries in younger patients, falls account for more injuries in the older population 255 , sometimes becoming a co-morbidity of another degenerative disorder affecting motor functions such as those described above.
Unlike neurodegenerative conditions where cellular dysfunctions are found in multiple brain areas, traumatic injury in cases such as spinal fracture-dislocation represents a localized area of damage and inflammation. This makes cell transplantation therapies more applicable, although many cases of injury span beyond a single localized point, either due to the diffuse injury during the traumatic event or secondary damage because of the widely reaching inflammatory changes in the whole CNS257,258.
A specific type of astrocyte lineage cells differentiated from GRPs using BMP4, but not CNTF, stimulated functional motor recovery and had a protective effect on axotomised neurons259–261 in rats upon transplantation after SCI, without enhancing pain fiber sprouting (which presented a concern with some cell transplantation therapies in SCI 262 ). Interestingly, the same type of astrocytes (GRP-derived astrocytes, GDABMP4) was also shown to be protective in a rat model of PD 151 (Table 2). On the other hand, GRPs differentiated in the presence of CNTF specifically promoted mechanical allodynia and thermal hyperalgesia, which correlated with the increased pain fiber outgrowth 260 , providing key evidence of astrocytic lineage heterogeneity and offering at least partial explanation for inconsistent success of astrocyte transplantation in other studies 262 (Tables 5 & 6).
Table 5.
Disease or disease model and age at transplantation | Type of astrocyte-lineage cells or precursors transplanted | Site of transplantation within the CNS | Endpoint/treatment duration | Outcome summary | References |
---|---|---|---|---|---|
SCI—L5 dorsal root crush, albino rats, adult | Immature astrocytes from E16-18 fetal rat spinal cords, cultured for 2 days, plated on nitrocellulose Millipore inserts (‘pennants’) for further 1–2 days before insertion | Dorsal root entry zone at the CNS-PNS interface, implanted at the time of injury | Approx. 3 weeks | Enhanced but variable fiber regeneration into the spinal cord gray matter, attenuated inflammatory response, astrocytic coating of the implant prevented hemorrhage and cavitation. | Kliot et al. 263 |
SCI—C3 fasciculus gracilis (carries sensory input from caudal parts of the body) aspiration, male Sprague-Dawley rats, adult | Cultured astrocytes from E14 fetal rat spinal cords, or whole pieces of E14 fetal rat spinal cord | C3 fasciculus gracilis area, implanted at the time of injury | Up to 90 days | Whole fetal spinal cord grafts improved hindlimb function; astrocytes from such grafts migrated into the nucleus gracilis of the medulla and prevented neuronal atrophy. Pre-cultured astrocytes also migrated to the medulla, but provided no neuroprotection, and exacerbated hindlimb dysfunction. |
Bernstein and Goldberg 264 |
SCI—hemisection of the dorsal L3 spinal cord, female Sprague-Dawley rats, adult | Cultured (>2 weeks) astrocytes from newborn rat cerebral cortex, either in HBSS solution or grown in gelfoam for 3 days | Dorsal L3 spinal cord, implanted at the time of injury | 2 weeks to 2 months | Astrocytes in suspension and gelfoam reduced scarring and potentially increased neurofilament outgrowth; implanted astrocytes are capable of migrating. | Wang et al. 265 |
SCI—focal infarcts at dorsal L13, male PVG rats, adult | (1) Neonatal PVG rat forebrain astrocytes cultured for 7 days (A1 astrocytes); (2) Neonatal kitten forebrain astrocytes cultured for 7 days; (3) CG-4 (bipotential rat glial progenitor cell line) -differentiated astrocytes (A2 astrocytes) |
Dorsal L13 spinal cord, 3 days after the injury | 4 weeks | Cultured rat astrocytes were the most prone to clumping and only filled 25% of the post-injury cyst; feline astrocytes and CG-4-differentiated astrocytes filled the cyst with fine processes more uniformly. CG-4-differentiated astrocytes increased vascularization, promoted some axonal sparing and potentially stimulated ECM production. No significant effect on axonal regeneration. | Olby and Blakemore 266 |
SCI—bilateral dorsal hemisection of T8-9 spinal cord, female Wistar rats, young adult | Cultured cortical neonatal (P3) rat astrocytes (type 1) in collagen type I; cultured in collagen overnight prior to transplantation; 2mm wide section | Thoracic spinal cord, implanted at the time of injury | 28 days / 4 weeks | Significant increase in neuronal process ingrowth into the astrocyte-containing implant from both rostral and caudal areas of the graft compared to collagen graft only; temporary motor improvements as assessed by the open-field locomotion analysis and crossing a walkway / catwalk analysis. Little astrocyte migration outside of the implant. | Joosten et al. 267 |
SCI—complete section of T8 spinal cord, female Fischer 344 rats, adult | Cultured cortical astrocytes from adult female Fischer 344 rats from the same inbred as the host animals, GFP-labeled using a lentiviral vector | T11 spinal cord, 7 days after injury | 6 weeks | Transplanted astrocytes migrated and accumulated at the site of injury; transgene was downregulated from approx. 80 to 7% over 6 weeks—to consider for gene therapy development. | Pencalet et al. 268 |
SCI—dorsal column unilateral transection at C1-2, or rubrospinal tract unilateral transection at C3-4; female Sprague-Dawley or Fischer 344 rats, 3 months-old (adult) | (GRP, A2B5+)-GDAs: GRPs isolated from E13.5 Fischer 344 transgenic (expressing human placental alkaline phosphatase) rat embryos, differentiated by exposure to BMP4 (GDABMP, type 1 astrocytes); GFP-labeled adult mouse DRG neuron implantation caudal to the injury site to assess axonal outgrowth in some animal groups |
6 different areas of injection per lesion site, implanted at the time of injury | 4 days to 5 weeks, depending on the test | Robust axon growth across lesion sites bridged with GDAsBMP, suppressed red nucleus neuron atrophy, reduced astrogliosis, increased linear alignment of host astrocytes at the lesion margin, delayed expression of inhibitory proteoglycans, recovery of locomotor functions as assessed by the grid-walk test—all seen after transplantation of GDAsBMP, but not GRPs. Greater variation in lesion size and margin morphology in Fischer 344 rats. |
Davies et al. 259 |
SCI—as above, female Sprague-Dawley rats, 3 months-old (adult) | GDAs differentiated from GRPs, either as above by treatment with BMP4 (GDABMP), or by treatment with CNTF (GDACNTF); GFP-labeled adult mouse DRG neuron implantation caudal to the injury site to assess axonal outgrowth in some animal groups |
As above | As above | GDAsBMP, but not GDAsCNTF or non-differentiated GRPs, support axonal regeneration and behavioral recovery as described above; GDAsCNTF or non-differentiated GRPs, but not GDAsBMP, promote CGRP immunoreactive nociceptive c-fiber sprouting which results in mechanical allodynia and thermal hyperalgesia. |
Davies et al. 260 |
SCI—C3-4 unilateral transection, female Sprague-Dawley rats, 3 months-old (adult) | Human GDAsBMP and GDAsCNTF, differentiated from human spinal cord glial precursors (9 week-gestational age) | As above | 3 days to 5 weeks | GDAsBMP, but not GDAsCNTF, promoted axon ingrowth into the injury site and locomotor recovery, as assessed by the grid-walk test. These cells also expressed higher levels of GDNF, GLT1, and Connexin 43. |
Davies et al. 261 |
SCI—T9-10 moderate contusion injury, female Sprague-Dawley rats, 8 weeks-old (young adult) | GFP-labeled mouse iPSCs were differentiated into astrocytes through neurosphere and neural stem cell stages by treatment with FBS | T9-10, 3 or 7 days post-injury | Up to 8 weeks | Transplanted astrocytes extended processes longitudinally, but no motor improvement was observed; increased sensitivity to mechanical stimuli. | Hayashi et al. 269 |
SCI—T10 contusion, female athymic rats, adult | Human brain-derived GRPs (18-to 24-week-gestational age); or human GDAsBMP differentiated from GRPs |
In and around (2mm rostral and caudal) the injury site, 9 days post-injury | Up to 8 weeks | Majority of transplanted human GRPs differentiated into astrocytes with some oligodendrocyte differentiation. Both GRP and GDABMP grafts reduced cyst and scar formation. GRPs attenuated hyperactive bladder reflexes; GDABMP graft improved sensory function of the hindpaw. No allodynia and pain and no locomotor improvement were observed, modest decrease in motor performance (grid-walk) upon GDAsBMP transplantation. |
Jin et al. 270 |
SCI—C4-5 small excision of the right dorsal column, female Sprague-Dawley rats, adult | 1) Rat GRPs were generated from spinal cord and cortices via 3 different methods; 2) Human GRPs were derived from the brain tissue (18- to 24-week-gestational age); 3) Astrocytes were differentiated from GRPs through treatment with either FBS, BMP4, or CNTF |
C4-5, implanted at the time of injury | 3 weeks | Differentiated astrocytes remained phenotypically plastic. Majority of engrafted cells differentiated into astrocytes. All grafts supported axonal growth; sensory axons could grow into but not out of the graft area. |
Haas et al. 271 |
SCI—T9 moderate to severe contusion, Fischer 344 rats, adult | GRPs from E14 Fischer 344 rat spinal cord were differentiated into GDAs by exposure to BMP4; GDAs were transduced with D15A multi-neurotrophin using a lentiviral vector | In and around the injury site, 8 or 9 days post-injury | Up to 8 weeks | Most grafted GRPs differentiated into astrocytes with some differentiating into oligodendroglial lineage cells. D15A-GDAs, but not GRPs or GDAs, increased spared white matter and promoted motor recovery assessed by the open-field locomotor test. No allodynia was detected. |
Fan et al. 272 |
SCI—C4-5 unilateral excision severing the right dorsal column, female athymic rats, adult | Human GRPs were derived from the brain tissue (20- to 21-week-gestational age); GDAs were differentiated through exposure to BMP4 or CNTF |
C4-5, implanted at the time of injury | 5 weeks | Each of the grafts (GDABMP or GDACNTF) supported sensory axon outgrowth into, but not out of the graft. Pre-differentiated astrocytes retained morphologic and phenotypic plasticity. | Haas and Fischer 273 |
SCI—C4-5 unilateral excision severing the right dorsal column, female Sprague-Dawley rats, adult | Human GRPs were derived from the brain tissue (20- to 21-week-gestational age) from frozen stocks | As above | As above | See above | Haas and Fischer 273 |
SCI—T8 contusion, female Sprague-Dawley rats, adult | Rat GRPs were derived from E13.5 spinal cords, and GDAs were differentiated through exposure to BMP4; Injected alone or in combination with human recombinant decorin (hr decorin) |
Around the injury site, implanted at the time of injury | 4 weeks (28 days) | Both GDAsBMP and GDAsBMP + hr decorin reduced glial scar formation, filled the injury-induced cavity, enhanced linear alignment of astrocytic processes, increased motor and sensory axon regeneration, improved hindlimb motor function. GDABMP + hr decorin graft was the most efficient as inhibiting the inflammatory response. |
Wu et al. 274 |
SCI—C1-2 right dorsal column transection, female Sprague-Dawley rats, adult | Fischer 344 rat GRPs were derived from E13.5 spinal cords, and GDAs were differentiated through exposure to BMP4 | Around the injury site, implanted at the time of injury | 7 days | GDABMP graft promoted significant axonal growth into the lesion center. Knock-down of periostin (POSTN), a secreted protein, by shRNA diminished the axonal regeneration. |
Shih et al. 275 |
SCI—C2 hemisection, female Sprague-Dawley rats, adult | Fischer 344 rat GRPs were derived from E13.5 spinal cords | C2, implanted at the time of injury | 5 weeks (35 days) | GRPs efficiently differentiated into astrocytes, reduced the macrophage response, stimulated regeneration of ventral respiratory group axons and sprouting of spared fibers, and protected diaphragm function. Notably, re-growing axons did not exit the graft site, suggesting that axonal recovery was probably not responsible for the functional effect. | Goulão et al. 8 |
TBI—left frontal cortex defects were created with a scalpel, Sprague-Dawley rats, 12 weeks-old (adult) | hUC-MSCs co-grafted with astrocytes (region unspecified) from P1-2 Sprague-Dawley rats activated by 1ng/ml LPS 6h prior to use; transplanted in a peptide hydrogel R-B-SPH scaffold – a self-assembling peptide hydrogel enriched with a BDNF protein-derived sequence |
Into the injury site, implanted at the time of injury | Up to 8 weeks | Presence of activated astrocytes increased BDNF secretion and hUC-MSC proliferation within the graft, promoted neuronal differentiation of hUC-MSC, and enhanced neuronal network reconstitution. | Shi et al. 276 |
TBI—unilateral controlled cortical impact (CCI) that produced a moderate injury, male Sprague-Dawley rats, 3 months-old (adult) | GFP-expressing murine ESCs were differentiated into astrocytes in the presence of CNTF; GABAergic neuronal cells differentiated from the same ESCs and growth arrested stromal cells (bone marrow stromal fibroblasts) were used in parallel transplantations |
2 sites in the injured cortex around the injury, 7 days following injury | Up to 6 weeks | ESC-derived GABAergic neurons, but not ESC-derived GDAsCNTF nor stromal cells, improved sensorimotor recovery. | Becerra et al. 277 |
TBI—lesion of the right motor cortex with probe pre-cooled in liquid nitrogen, male Sprague-Dawley rats, 7 weeks-old (adult) | Astrocyte-enriched mixed glial cultures were prepared from cerebral cortices of newborn rats and grown in FBS; cortical neurons from E16 rat embryos were used in parallel transplantations | Into the injured cortex, 5 days following injury | Up to 6 weeks | Glial graft significantly increased body weight and promoted motor recovery (assessed via the rotarod test) compared to the neuronal graft; both glial and neuronal grafts reduced the injury area. | Quan et al. 278 |
SCI: spinal cord injury; TBI: traumatic brain injury; CNS: central nervous system; ECM: extracellular matrix; GRP: glial-restricted precursor; GDAs: Glial precursor derived astrocytes; CGRP: calcitonin-gene-related peptide; hUC-MSCs: Human umbilical cord mesenchymal stem cells; PNS: peripheral nervous system; BMP4: bone morphogenic protein 4; CNTF: ciliary neurotrophic factor; HBSS: Hanks’ balanced salt solution; FBS: fetal bovine serum; DRG: dorsal root ganglia; iPSCs: induced pluripotent stem cells; ESCs: embryonic stem cells; LPS: lipopolysaccharide; BDNF: brain-derived neurotrophic factor.
Table 6.
Cell type of origin | BMP4 concentration | Duration of BMP4 treatment | Condition treated by transplantation and main outcome | References |
---|---|---|---|---|
Human induced PSCs (hiPSCs) | 10 ng/ml (+ 10 ng/ml LIF) |
20 days | ALS model—modest lifespan and motor improvement | Kondo et al. 73 |
Rat (Fisher 344 hPAP transgenic) GRPs from the spinal cord of E13.5 embryos | 10 ng/ml (+ 1 ng/ml bFGF) |
7 days - ? | PD model—functional improvement, neuroprotection | Proschel et al. 151 |
Human GRPs from the rostral neural tube of 9- to 10-week-old embryos | 10 ng/ml | As above | As above | Proschel et al. 151 |
NPCs from ubiquitous eGFP-expressing mouse cerebral cortex | 10 ng/ml | 4 days (96 h) | FTD model—reduced neuronal loss similarly to NPCs | Hampton et al. 206 |
Rat (Fisher 344 hPAP transgenic) GRPs from the spinal cord of E13.5 embryos, A2B5-positive | 10 ng/ml | 7 days | SCI—functional improvement | Davies et al. 259 |
As above | 10 ng/ml | 7 days | SCI—functional improvement which was not seen with GRP-differentiated astrocytes (GDAs) differentiated in the presence of CNTF (GDACNTF) | Davies et al. 260 |
Human glial progenitor cells (hGPCs) from spinal cords of 9 weeks-old embryos, A2B5-positive | 20 ng/ml | 7 days | SCI—functional improvement which was not seen with astrocytes differentiated in the presence of CNTF | Davies et al. 261 |
Human GRPs from the brain tissue of 18- to 24-week-old embryos | 50 ng/ml | 5 days | SCI—improved sensory function, modestly decreased motor performance | Jin et al. 270 |
Rat GRPs from the spinal cord generated via 3 different methods; Or human GRPs from the brain tissue of 18- to 24-week-old embryos |
10 ng/ml, or 50 ng/ml in select experiments | 6 days | SCI—supported axonal growth, remained phenotypically plastic despite pre-differentiation | Haas et al. 271 |
Rat GRPs (Fischer 344) from E14 spinal cord, A2B5-positive | 20 ng/ml | 3 days | SCI—only GDAs that were transduced to express D15A neurotrophin showed positive effects on motor recovery and white matter sparing | Fan et al. 272 |
Human GRPs from the brain tissue of 20- to 21-week-old embryos | 10 ng/ml | 10 days | SCI—supported sensory axon outgrowth, remained phenotypically plastic despite pre-differentiation | Haas and Fischer 273 |
Rat GRPs (Sprague-Dawley) from E13.5 spinal cord | 10 ng/ml | 7 days | SCI—improved motor function | Wu et al. 274 |
Rat GRPs (Fisher 344 hPAP transgenic) from E13.5 spinal cord | 10 ng/ml (+1 ng/ml bFGF) |
6 days | SCI—axonal regeneration dependent on periostin secretion | Shih et al. 275 |
BMP4: bone morphogenic protein 4; ALS: amyotrophic lateral sclerosis; GRPs: glial-restricted precursors; PD: Parkinson’s disease; NPCs: Neural precursor cells; FTD: frontotemporal dementia; SCI: spinal cord injury; GDAs: Glial precursor derived astrocytes; LIF: leukemia inhibitory factor; hPAP: human placental alkaline phosphatase; bFGF: basic fibroblast growth factor; eGFP: enhanced green fluorescent protein; CNTF: ciliary neurotrophic factor.
One obvious way in which an astrocyte graft can alleviate the symptoms associated with trauma is by filling up the cavity and replenishing lost cells in the damaged area. Lesioned axons fail to send new projections through a fluid-filled cyst that can result post-trauma, while astrocytes, which can be engrafted in a gel biomaterial such as collagen 267 , contribute to restoring a more physiological ECM composition. Even though some earlier reports considered astrocytes to be the primary source of inhibitory ECM components that are detrimental to axonal regrowth, new evidence strongly suggests that astrocytic presence at the lesion site is crucial for healing279,280. Furthermore, healthy transplanted astrocytes were able to delay expression of inhibitory ECM molecules when engrafted during early post-lesion stages, suppress astrogliosis of the resident cell populations, and re-organize the injured tissue to potentially make the scar border more permissive to axonal growth259,274.
In addition to the primary damage to the tissue at the time of trauma 281 , secondary axonal injury leading to neuronal body atrophy and apoptosis282–285, also occurs. Astrocyte-mediated protection of myelin sheaths surrounding damaged areas can be prompted by a combination of connexin cell-to-cell contacts 22 and secreted factors such as thrombin protease inhibitors 23 , ATP, and LIF 286 . Additionally, astrocyte-derived CXCL10 can promote microglia-mediated phagocytosis of myelin debris 32 , which is essential for remyelination 287 . Astrocytes can also support surviving neurons and encourage new neurite outgrowth by providing neurotrophic cues, both secreted (such as BDNF and NT-3262,272) and cell surface bound (such as Eph receptors 288 ).
Purinergic 289 and glutamatergic 290 excitotoxicity due to excessive release of ATP or glutamate, respectively, represent another feature of traumatic CNS injury, and therefore blockade of these pathways ameliorates secondary cell death and function loss after trauma. Oxidative stress also contributes to the cell death following injury as rapid generation of reactive oxygen and nitrogen species exacerbate excitotoxicity and damage mitochondria 291 . The ability of astrocytic syncytium to buffer excessive ATP and glutamate as well as to supply glutathione and other antioxidants were reviewed above in the context of other conditions, and it is likely that similar canonical pathways could be tapped into in the context of SCI and TBI.
Another important aspect of the functional recovery from trauma is restoration of the BBB and vascular supply to the injured area, in which astrocytes play a crucial role. Indeed, transplanted human iPSC-derived astrocytes were found to interact closely with the blood vessels 292 , and were able to increase vascularization of the lesioned area266,263.
Inconsistency of the astrocyte transplantation success between different models of SCI is an important concern, and several reasons for this phenomenon were suggested 262 . First, the type of injury, location, and severity are likely to be important determinants of what extent of functional recovery can be reasonably expected. For example, some studies on SCI rodent models have demonstrated locomotor recovery 259 while others presented sensory recovery with no motor improvement 270 . It is possible that the cell numbers, density, and vehicle used for astrocyte delivery (e.g. collagen-based gel or liquid medium) can play a role in determining the outcome (Table 5).
Second, the time of transplantation post-injury is a key variable that needs to be assessed. It has been suggested that inflammation and reactive astrogliosis are beneficial at the early healing stages293,294 (e.g. due to recruiting microglia to promote debris clearance) and therefore not allowing the natural protective response of resident astrocytes to develop could be detrimental. In fact, delayed transplantation studies are more likely to be relevant to the real clinical cases where many patients would seek treatment days, weeks, or years after the initial injury.
Finally, it is obvious from the published SCI studies that the type of astrocytes to be transplanted (e.g. mature vs embryonic, A1 vs A2 type, BMP4- or CNTF-differentiated GDAs, plus regional differences) plays a crucial role in determining the likelihood of recovery. For instance, different subtypes of astrocytes exhibit differential tropism for encouraging support of specific neuronal fibers as demonstrated by the GDAsBMP4, which specifically promoted motor axon outgrowth, while GDACNTF mostly enhanced extension of nociceptive calcitonin-gene-related peptide (CGRP) c-fibers 260 . Several different methods used to differentiate rodent and human astrocytes using BMP4 are summarized in Table 6. The ability of transplanted GDAs to maintain their beneficial phenotype long-term in a hostile niche has been debated; however, since their effect on functional recovery may depend more on the initial positive effect on the host tissue than the continued presence of these cells 259 , cell pre-differentiation could still be a viable therapeutic approach. In an attempt to ensure that GDABMP4 retain their beneficial phenotype and continue providing the trophic factors even under unfavorable conditions, bioengineering approaches have been employed: retroviral transduction of these cells with D15A (an engineered neurotrophin that combines NT-3 and BDNF activities) enhanced the ability of the graft to improve locomotion in rats with SCI 272 . Effective healthy astrocyte replacement in traumatic CNS injury would help a patient to regain the lost neurological function fully or partially, and/or reduce the burden of neuropathic pain, thereby increasing their capacity for independent living.
Ischemia/Stroke
Cerebral ischemia, most commonly resulting from stroke, is one of the leading causes of disability and death worldwide, which is associated with devastating losses of various neurological functions and increased risk of dementia for the patients 295 . The incidence of stroke ranges from 95 to 290 new cases per 100 000 people per year, and 13% to 35% of cases lead to death within 1 month 296 . Stroke is more common in older patients with the mean age of occurrence around 70 years 297 , but up to 15% of stroke patients are young adults, for whom personal and economic implications of the life-long disability are much greater 298 . While some genetic factors such as history of familial hypertension predispose certain people to stroke, a combination of environmental factors and lifestyle has a large impact on the probability of development of this pathology, which include obesity, lack of exercise, psychological stress, and smoking 299 .
Ischaemic stroke is commonly caused by an acute blockade of a cerebral artery by a thrombus that results in the cessation of blood supply to particular brain areas, leading to cell death at the center of the infarction within minutes of the onset. As in the case of traumatic injury, specific location of the ischaemic event determines the extent and nature of the disability that is likely to result from it.
To rescue memory deficits in the aftermath of an ischaemic stroke in rats, co-transplantation of NSCs alongside astrocytes, brain microvascular endothelial cells (BMECs), or both, has been performed. While NSC graft alone did not affect rats’ cognitive performance, double (astrocytes+NSCs or BMECs+NSCs) or triple transplantation led to significant improvements 300 . Another approach to employ astrocytic transplantation to improve cognitive and motor function after ischemia in rats involved knocking down astrocytic CDK5 prior to engraftment301,302 (Table 7).
Table 7.
Disease or disease model and age at transplantation | Type of astrocyte-lineage cells or precursors transplanted | Site of transplantation within the CNS | Endpoint/treatment duration | Outcome summary | References |
---|---|---|---|---|---|
Ischemia—middle cerebral artery occlusion and reperfusion, male Sprague-Dawley rats, adult | Astrocytes were derived from neonatal Sprague-Dawley rat cortices and cultured in FBS-containing medium; NSCs and brain microvascular endothelial cells (BMECs) were also isolated from newborn rat cortices and used in parallel transplantations |
Hippocampal CA1, 12 days after injury | 8 weeks | Co-transplantation of all 3 cell types was the most efficient at memory improvement as assessed by the Morris water maze test. NSCs alone had no effect on memory. | Cai et al. 300 |
Global cerebral ischemia—permanent left carotid artery occlusion, temporary right carotid artery occlusion (20min) and reperfusion, male Wistar albino rats, 3 months-old (adult) | Astrocytes were obtained from the P1-2 rat cerebral cortices; CDK5 microRNA knock-down (CDK5-KD) was employed | Somatosensory cortex, immediately after the injury | 24 h to 15 days | CDK5-KD astrocytes were significantly more efficient at rescuing cognitive and motor impairment after ischemia as assessed by rotarod and neurological evaluation, prevented loss of neurons, increased an endothelial cell marker expression, and upregulated BDNF levels. | Becerra-Calixto and Cardona-Gómez 301 |
Global cerebral ischemia—as above | As above | As above | 4 months | CDK5-KD astrocytes prevented loss of host astrocyte and neurons post-injury, protected endothelial cells, enhanced BBB recovery, enhanced BDNF production by endogenous astrocytes, and improved neurological and motor (rotarod) performance. | Becerra-Calixto et al. 302 |
CNS: central nervous system; NSCs: neural stem cells; BBB: blood-brain barrier; FBS: fetal bovine serum; BDNF: brain-derived neurotrophic factor; CDK5: cyclin-dependent kinase 5.
CDK5 hyperactivation has been observed in several disorders including ischemia, AD, and ALS, which results from the calcium-dependent p35 to p25 cleavage303,304. It was therefore hypothesized that preventing CDK5 activation can render astrocytes in their non-pathologically activated state. Indeed, CDK5 knock-down increased the ability of transplanted astrocytes to prevent local cell loss, protect the BBB, and ultimately improve neurological scores as well as locomotor recovery301,302.
Consistent with the detrimental role of calcium-mediated hyperactivation of CDK5 in ischaemic stroke, abnormally increased levels of astrocytic calcium were seen in reactive astrocytes following the insult, which result from the extracellular calcium entry 305 or release of calcium from the intracellular stores 306 . Interestingly, there appears to be a level of heterogeneity in the roles played by astrocytic calcium responses, which may stem from the spatial (endfeet vs soma), or temporal (e.g. frequency of calcium oscillations 307 ), location of signaling after the ischaemic insult. For instance, release of calcium in the astrocytic endfeet, but not cell bodies, enhances vasodilation to improve brain microcirculation, which may be protective in ischemia 308 . At the same time, mGluR activation that promotes rhythmic calcium oscillations leads to the pathological swelling in astrocytes 309 , but also has cytoprotective effects on white matter astrocytes 310 . Decreasing CDK5 may specifically dampen the detrimental effects of calcium dyshomeostasis under ischaemic conditions while allowing the protective signals to remain.
Excitotoxicity is a common feature of many pathologies, and ischemia is no exception. Excessive calcium oscillations triggered in astrocytes by ischaemic insults were associated with CaMKII inhibition, which led to the reduced ability of astrocytes to buffer extracellular glutamate as well as an increase in ATP release 311 . This therefore perpetuates the excitotoxic cycle by acting on astrocytic glutamatergic receptors—the expression of which can be stimulated by hypoxic conditions 312 , and purinergic receptors, which further dysregulates intracellular calcium levels. Damage to astrocytic mitochondria was observed in other studies as well313,314, which further contributes to the BBB breakdown 315 and GLT1 glutamate transporter downregulation 316 . Certain regional populations of astrocytes, such as those found in the hippocampus, were found to be specifically vulnerable to ischemia-induced mitochondrial damage and oxidative stress, leading to the early loss of glutamate transporters 317 which further impacts neighboring hippocampal neurons. This may be the underlying reason of frequently observed memory deficits following stroke.
Inflammatory molecules such as interleukin-1β (IL-1β) are known to contribute to cell damage in stroke, and one of the inflammation-induced mechanisms of damage involves activation of p38/stress-activated protein kinase 2 (p38/SAPK2), leading to gap junctional closure. Accordingly, inhibition of p38/SAPK2 pathway was found to reduce the area of ischaemic lesion 318 , suggesting that the gap junctional communication is crucial in preventing the injury spread. Moreover, human marrow stromal cell (hMSC) transplantation aided neurological recovery in rats following stroke, and these cells were shown to be able to release soluble factors that upregulate gap junctional communication in astrocytes through activation of PI3K/Akt pathway 319 . At the same time, blockade of hemichannels composed of connexin 43, the opening of which is potentiated under inflammatory and hypoxic conditions, had beneficial effects through downregulation of inflammatory cytokine production and microglial activation alongside prevention of release of ROS, ATP, and glutamate320–322.
Even though neurons were thought to be more vulnerable to ischemia than astrocytes, it is becoming clearer that both cell types suffer from pathological changes that commonly result in cell death 323 . Transplanted astrocytes can therefore replenish the resident panglial network, support the BBB, and improve the blood flow to the brain, and supply neuro- and astro-protective factors such as erythropoietin309,324. Effective healthy astrocyte replacement in the ischaemic stroke could, similarly to the traumatic injury to the CNS, help restore the compromised functionality, the exact nature of which would depend on the affected region of the brain.
Overview
Astrocyte biology is an expanding topic of brain science research with the functions of these cells continually expanding from neuronal supporters to active partners at synapses to cellular hubs integrating signals and influencing functions of all major brain cell types. This review explores some prominent examples of astrocyte transplantation strategies with promising functional benefits in translational models of ALS, PD, AD, HD, ischemia, and traumatic CNS injury, and a pioneering clinical trial with human ALS patients points at a clear therapeutic interest of these studies.
Considering that many more brain and spinal cord disorders are found to be associated with astrocytic abnormalities, it is likely that the insights gained from these experimental transplantation strategies can be expanded to treatments of other diseases. For example, astrocytic abnormalities were found to precede demyelination in multiple sclerosis 325 , and astrocytes carrying mutations associated with a higher risk of autism were found to inflict damage on healthy neurons co-cultured with these mutant cells 30 . Therefore, replenishing healthy astrocyte pools can alleviate symptoms of more than one condition, especially considering that, despite different aetiologies, many disorders share key pathological alterations in astrocyte biology, demonstrated in this review. Transplantation of healthy cells and/or tapping into the canonical pathway onto which multiple pathologies converge (e.g. inflammation-induced astrocytosis, mitochondrial stress, or astrocytic syncytium-mediated calcium, ATP, and glutamate buffering) represents a robust and versatile approach.
Autologous cell sources are especially promising in human astrocyte transplantation therapies due to the lack of ethical concerns and minimized risk of rejection. Enteric glia have proven beneficial in an AD model upon engraftment in the brain 205 ; however, recent studies point toward early dysfunction of these cells in degenerative disorders such as AD and PD 326 . Moreover, in case a patient harbors a mutation in all somatic cells that predisposes one to a certain disorder, even reprogramming of patient own fibroblasts into seemingly healthy astrocyte-lineage cells may result in inherently diseased astrocytes devoid of beneficial properties 81 . Gene-editing technologies such as CRISPR could be employed to correct known mutations associated with familial forms of diseases prior to implantation of the cell graft. Increased expression of transcription factors and neurotrophins, or knock-down of factors known to be associated with pathologically reactive astrocyte states, which is likely to shift the astrocytic phenotype toward a less inflammatory state, could also be additionally employed152,272,301,302.
One interesting point to consider during cellular reprogramming is regional heterogeneity of vulnerability to particular diseases—it is becoming increasingly apparent that brain cells in different brain regions exhibit different levels of susceptibility to pathologies associated with PD, AD, ALS, or ischaemic challenge. At the same time, progress has been made in the field of regional patterning of reprogrammed astrocytes327–329. Thence, should the transplantation therapy aim to replace the cells that correspond to the most vulnerable and affected region since these cells are more likely to be lost or severely affected by the disease, or should it supply those cells exhibiting a more “robust” regional phenotype?
Astrocytes, once matured and integrated in the brain, are not migratory330,331, and they retain many aspects of their regional identity in vitro 332 ; hence, they can be expected to retain important aspects of such identity after transplantation as well. One study has shown that grafting midbrain, and not cortical, astrocytes were more beneficial in a PD model, arguing that the midbrain cells are more naturally adapted to supporting dopaminergic neurons 152 . One limitation of this study is that these astrocytes were transplanted into the rat striata, while no astrocytes from striatal origin were engrafted, and no transplantation into the midbrain was explored. Another good example of this point can be found in the field of ALS research. While cervical spinal cord-localized transplantation of rodent spinal cord-derived astrocyte precursors was beneficial in a rat model of ALS 71 , human forebrain-derived astrocyte precursors showed no protective effects in a mouse ALS model 72 . Even though both studies were conducted by the same group and used the same overall approach, the number of variables makes it difficult to compare these studies directly (e.g. mouse vs rat model, rat vs human cells); nevertheless, it is possible that the spinal cord astrocytes are specifically more apt to integrate into the spinal cord astrocytic and neuronal networks and restore relevant functions. Another corroboration of this idea comes from a study of the corticospinal tract (CST) axonal growth in neonatal rat spinal cords—when nitrocellulose inserts were placed in the cords to deflect the normal CST growth path, only spinal cord astrocytes grown on the inserts were able to support CST axon growth as opposed to cortical astrocytes 333 . On the other hand, enteric astrocytes have proven protective in an AD model when transplanted into the brain 205 , even though no comparison with other regional types was offered by that study. In addition, it has been suggested that astrocytes from the brain regions representing axonal targets, such as cortical astrocytes for hippocampal neurones, could promote axonal outgrowth more efficiently that astrocytes from the same brain regions as neurones, or unrelated regions of the brain. 334 This property could be useful in cases where prompt neurite extension is desirable, such as after acute SCI, but the long-term effects of non-native astrocyte subtypes in a particular region of the brain are not known. More research is needed to address this issue in greater depth.
Engraftment of neurons, as described above2–5, and other cell types such as brain microvascular endothelial cells (BMECs) 300 and oligodendrocytes 335 also provide valuable therapeutic strategies, and, in fact, olidogendroglial differentiation of GRPs (in addition to astrocytic differentiation) can be at least partially responsible for the success of the GRP engraftment in some of the studies discussed above8,270.
Astrocytic transplantation can be conducted in parallel with engraftment of other cell lineages, as explored in several studies152,300, in order to maximize the beneficial effects, since even in those CNS disorders where pathology is commonly found most prominently in one cell type, multiple cellular perturbations are expected, and the cellular culprit of the pathology is not always obvious. Examples of that include the non-cell autonomous neuronal death in rodent models of ALS bearing mutant astrocytes80–82 and myelination pathology in Alexander’s disease caused by mutations in GFAP 336 —an intermediate filament protein associated with the astrocytic lineage.
But does cell transplantation merely alleviate disease symptoms? A fascinating study demonstrated that human glial precursors, which abundantly differentiate into astrocytes, when grafted into the mouse forebrain could integrate into the local cellular circuits and maintain their highly branched morphology, and also increase the number of synaptic contacts. Most importantly, these human cells enhanced the memory retention ability of these animals 337 (Table 8).
Table 8.
Disease or disease model and age at transplantation | Type of astrocyte-lineage cells or precursors transplanted | Site of transplantation within the central nervous system | Endpoint/treatment duration | Outcome summary | Reference |
---|---|---|---|---|---|
Healthy neonatal immunodeficient (rag1-/- or rag2-/-) mice | Human fetal glial progenitors extracted from the forebrain ventricular and subventricular zones (17- to 22-week-gestational age); murine glial progenitor cells were prepared from newborn pups in a similar fashion and used in parallel transplantations |
Two locations within the forebrain, within a day of birth | 2 weeks to 20 months | Glial precursors differentiated largely into astrocytes and retained hominid features (larger size, threefold faster calcium wave propagation); chimaeric mice containing human cells exhibited enhanced long-term potentiation (LTP) and learning, as assessed by Barnes maze test, object-location memory, and fear conditioning. No enhancement of memory was observed in animals who received murine grafts. | Han et al. 337 |
Considering the accumulating data showing that attention-modifying and psycho-modulatory (e.g. antidepressants) drugs commonly used in humans, which used to be considered acting purely on neuronal networks, are increasingly recognized to also affect astrocyte biology 338 , and that glial precursors (abundantly differentiated into astrocytes) derived from schizophrenic patients were able to recapitulate certain key behavioral aspects of this pathology in rodents after transplantation 339 , the idea of treating more subtle psychological disorders and even enhancing cognitive abilities by modifying the phenotype and number of astrocytes in the human brain may become a realistic perspective. Perhaps attenuation of aging-related decline could represent a more immediately achievable goal as benefits of rejuvenation of the astrocytic niche have started being explored in aged animals 340 .
Summary and Conclusions
Grafting of healthy astrocytes can slow down the disease progression and/or improve functional outcome in translational models of ALS, PD, AD, HD, ischemia, and traumatic CNS injury, and a human phase IIa clinical trial has shown a significant patient benefit in the case of ALS.
There are some common pathological features that are typically found in the diseased native astrocytes in these conditions that can lead to astrocyte atrophy and death, damage to surrounding neurons, loss of myelin, and recruitment of neurotoxic microglia (Fig. 1). These include:
Pro-inflammatory cytokine release profile (e.g. IL-6, TNFα, IL-1β) and loss of ability to release protective factors (e.g. BDNS, NT-3, GDNF);
Mitochondrial damage and increased ROS production, decreased ATP production;
Calcium signaling disturbance;
Connexin dysregulation including abnormal hemichannel opening;
Loss of glutamate transporters leading to excitotoxicity;
Loss of potassium buffering ability further leading to increased excitability of surrounding neurons;
Autophagy and proteasome deficits;
Apoptosis.
Transplanted astrocytes may therefore be able to (among other, disease-specific benefits):
Replace the apoptotic/necrotic astrocytes;
Reduce astrocytosis and inflammation;
Supply protective soluble (e.g. anti-inflammatory cytokines) and membrane-bound (e.g. Eph receptors, connexins) factors;
Reduce accumulation of toxic compounds including misfolded proteins and excessive glutamate, potassium, and calcium through lysosomal and proteasomal degradation and “glymphatic unit” clearance;
Act as stem cell-like cells with neurogenic potential.
Considering the emerging role of astrocytes in many other disorders, astrocyte transplantation is likely to become a more widespread therapeutic approach, alone or in combination with transplantation of other cell types such as neurons or NSCs.
The source of the cells suitable for transplantation needs to be determined with caution. Scalability and reproducibility issues with the available cell material may be a limitation of this therapeutic approach when scaling up from the experimental settings into the clinical trials and wider administration to the patients. There are three main types of cell sources that could be used for this purpose that we have identified:
Astrocyte precursor from ethically derived from the human embryos. This source has been used e.g. for the derivation of stem cells in transplantation trials involving people with PD3,1;
Commercially available pre-differentiated cells such as AstroRx produced by Kadimastem for the treatment of ALS;
Autologous cell transplantation where fibroblasts or mesenchymal stem cells are harvested from each patient and differentiated into the astrocyte-lineage cells ready for transplantation.
While there is a certain advantage to using the latter type of cells that are patient-specific, and thereby minimize the potential rejection and the extent of immunosuppression needed, they present two considerable limitations. First, differentiation of fibroblasts or mesenchymal stem cells into astrocyte-lineage cells, especially with the addition of time to characterize the resulting cells, can take several months, therefore delaying the benefit to the patient. Second, the presence of the known and yet-unknown mutations and epigenetic modifications that contribute to the disease phenotype may persist in the reprogrammed cells. Related to this issue, the batch-to-batch variability would be high between the cell transplants influenced by the patient-specific characteristics, and faithful comparison of the treatment outcomes would be difficult.
The precursor cells derived from human embryos could be considered more reliable in terms of their true astrocytic phenotype when compared with the differentiated cells, the identity of which is typically determined by the expression of rather generic astrocyte markers including GFAP. At the same time, use of human embryonic material presents an obvious limitation in the quantity of cells that can be obtained and the ethical considerations of scaling the cell supply up. In addition, batch-to-batch variability related to the differences between the embryos remains.
With this in mind, commercially available pre-differentiated and pre-characterized cells such as AstroRx could prove the most practical for the clinical use, as corroborated by their success in the first astrocyte transplantation clinical trial. These cells can be expanded, pre-characterized, and cryopreserved—these processes are currently being standardized to make the cells more suitable for the clinic. Refining the astrocyte-generation technology by creating more brain-region specific astrocyte types can expand this approach to a more diverse range of brain conditions.
The following additional consideration could be taken into account before this treatment finds its way into clinic:
Any cell graft has a small risk of over-proliferation. Pre-differentiation of stem cells into committed astrocyte precursors can help reduce the risk of graft overgrowth;
Hostile microenvironment of the diseased/lesioned tissue can predispose initially healthy cells to adopt a reactive and detrimental phenotype. Engineering astrocytes to artificially express trophic factors such as D15A (a human-designed neurotrophin that combines NT-3 and BDNF activities 272 ), transcription factors (Nurr1 and Foxa2 152 ) that are known to promote a non-reactive phenotype, or suppressing factors such as CDK5 that enhance pathological reactivity301,302 can ensure long-term maintenance of a healthy phenotype even in the presence of unfavorable conditions. Additionally, other small molecule or gene therapy-based approaches can be employed alongside transplantation to reduce inflammatory responses. For instance, minocycline and COX-2 inhibitors suppress neuroinflammation and extend lifespan of ALS mice341,342; more specific astrocyte- and brain-targeted interventions can be developed in the near future.
Acknowledgments
We wish to thank our colleagues at the Department of Clinical Neurosciences, University of Cambridge, for many intellectually engaging discussions that inspired the current review. In particular, we thank Dr Saifur Rahman for insightful views on calcium disturbances in disease states, and Prof Roger Barker and his research group for emphasizing the importance of inflammatory changes in neurodegeneration.
Footnotes
Author Contributions: N.H. conceptualized the paper and its structure, performed the literature review, wrote the paper, and generated the figure and tables. W.L.K., A.O., and M.K. provided substantial additions and edits. All authors contributed to multiple parts of the paper as well as the overall content.
Ethical Approval: This study was approved by our institutional review board.
Statement of Human and Animal Rights: This article does not contain any studies with human or animal subjects.
Statement of Informed Consent: There are no human subjects in this article and informed consent is not applicable.
Declaration of Conflicting Interests: The author(s) declared no potential conflicts of interest with respect to the research, authorship, and/or publication of this article.
Funding: The author(s) disclosed receipt of the following financial support for the research, authorship, and/or publication of this article: NH is supported by the Addenbrooke’s Charitable Trust (ACT), Ferblanc Foundation, and the Rosetrees Trust (Seedcorn2020/100084); WLK is supported by the Medical Research Council (MR/S005528/1); AO is supported by Sight Research UK (SAC 041); MRNK is supported by the National Institute for Health Research (NIHR, CS-2015-15-023) the Brain Injury MedTech Co-operative based at Cambridge University Hospitals NHS Foundation Trust and University of Cambridge. The views expressed in this publication are those of the authors and not necessarily those of the NHS, the National Institute for Health Research, or the Department of Health and Social Care.
ORCID iD: Nataly Hastings https://orcid.org/0000-0002-5376-339X
References
- 1. Freed CR, Greene PE, Breeze RE, Tsai WY, DuMouchel W, Kao R, Dillon S, Winfield H, Culver S, Trojanowski JQ, Eidelberg D, et al. Transplantation of embryonic dopamine neurons for severe Parkinson’s disease. N Engl J Med. 2001;344(10):710–19. [DOI] [PubMed] [Google Scholar]
- 2. Wernig M, Zhao J-P, Pruszak J, Hedlund E, Fu D, Soldner F, Broccoli V, Constantine-Paton M, Isacson O, Jaenisch R. Neurons derived from reprogrammed fibroblasts functionally integrate into the fetal brain and improve symptoms of rats with Parkinson’s disease. Proc Natl Acad Sci. 2008;105(15): 5856–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3. Barker RA, Barrett J, Mason SL, Björklund A. Fetal dopaminergic transplantation trials and the future of neural grafting in Parkinson’s disease. Lancet Neurol. 2013;12(1):85–91. [DOI] [PubMed] [Google Scholar]
- 4. Evans JR, Mason SL, Barker RA. Current status of clinical trials of neural transplantation in Parkinson’s disease. Prog Brain Res. 2012;200:169–98. [DOI] [PubMed] [Google Scholar]
- 5. Barker R, Drouin-Ouellet J, Parmar M. Cell-based therapies for Parkinson disease-past insights and future potential. Nat Rev Neurol. 2015;11(9):492–503. [DOI] [PubMed] [Google Scholar]
- 6. Xu L, Yan J, Chen D, Welsh AM, Hazel T, Johe K, Hatfield G, Koliatsos VE. Human neural stem cell grafts ameliorate motor neuron disease in SOD-1 transgenic rats. Transplantation. 2006;82(7):865–75. [DOI] [PubMed] [Google Scholar]
- 7. Xu L, Ryugo DK, Pongstaporn T, Johe K, Koliatsos VE. Human neural stem cell grafts in the spinal cord of SOD1 transgenic rats: differentiation and structural integration into the segmental motor circuitry. J Comp Neurol. 2009;514(4):297–309. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 8. Goulão M, Ghosh B, Urban MW, Sahu M, Mercogliano C, Charsar BA, Komaravolu S, Block CG, Smith GM, Wright MC, Lepore AC. Astrocyte progenitor transplantation promotes regeneration of bulbospinal respiratory axons, recovery of diaphragm function, and a reduced macrophage response following cervical spinal cord injury. Glia. 2019;67(3):452–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9. Southwell DG, Froemke RC, Alvarez-Buylla A, Stryker MP, Gandhi SP. Cortical plasticity induced by inhibitory neuron transplantation. Science. 2010;327(5969):1145–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10. Davis MF, Figueroa Velez DX, Guevarra RP, Yang MC, Habeeb M, Carathedathu MC, Gandhi SP. Inhibitory neuron transplantation into adult visual cortex creates a new critical period that rescues impaired vision. Neuron. 2015;86(4):1055–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Curtis E, Martin JR, Gabel B, Sidhu N, Rzesiewicz TK, Mandeville R, Van Gorp S, Leerink M, Tadokoro T, Marsala S, Jamieson C, et al. A first-in-human, phase I study of neural stem cell transplantation for chronic spinal cord injury. Cell Stem Cell. 2018;22(6):941–50. [DOI] [PubMed] [Google Scholar]
- 12. Bhasin A, Srivastava MV, Mohanty S, Bhatia R, Kumaran SS, Bose S. Stem cell therapy: a clinical trial of stroke. Clin Neurol Neurosurg. 2013;115(7):1003–1008. [DOI] [PubMed] [Google Scholar]
- 13. Barker RA, Parmar M, Kirkeby A, Björklund A, Thompson L, Brundin P. Are stem cell-based therapies for Parkinson’s disease ready for the clinic in 2016? J Parkinsons Dis. 2016;6(1):57–63. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Gonzalez R, Garitaonandia I, Crain A, Poustovoitov M, Abramihina T, Noskov A, Jiang C, Morey R, Laurent LC, Elsworth JD, Snyder EY, et al. Proof of concept studies exploring the safety and functional activity of human parthenogenetic-derived neural stem cells for the treatment of Parkinson’s disease. Cell Transplant. 2015;24(4):681–90. [DOI] [PubMed] [Google Scholar]
- 15. Guan Y, Zou H, Chen X, Zhao C, Wang J, Cai Y, Chan P, Chen L, Zhang YA. Ischemia, immunosuppression, and SSEA-1-negative cells all contribute to tumors resulting from mouse embryonic stem cell-derived neural progenitor transplantation. J Neurosci Res. 2014;92(1):74–85. [DOI] [PubMed] [Google Scholar]
- 16. Pustavoitau A, Bhardwaj A, Stevens R. Neurological complications of transplantation. J Intensive Care Med. 2011;26(4): 209–22. [DOI] [PubMed] [Google Scholar]
- 17. Kuga N, Sasaki T, Takahara Y, Matsuki N, Ikegaya Y. Large-scale calcium waves traveling through astrocytic networks in vivo. J Neurosci. 2011;31(7):2607–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Winship IR, Plaa N, Murphy TH. Rapid astrocyte calcium signals correlate with neuronal activity and onset of the hemodynamic response in vivo. J Neurosci. 2007;27(23):6268–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19. Szabó Z, Héja L, Szalay G, Kékesi O, Füredi A, Szebényi K, Dobolyi Á, Orbán TI, Kolacsek O, Tompa T, Miskolczy Z, et al. Extensive astrocyte synchronization advances neuronal coupling in slow wave activity in vivo. Sci Rep. 2017;7(1): 6018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20. Henneberger C, Papouin T, Oliet SHR, Rusakov DA. Long-term potentiation depends on release of d-serine from astrocytes. Nature. 2010;463(7278):232–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21. Dallérac G, Chever O, Rouach N. How do astrocytes shape synaptic transmission? Insights from electrophysiology. Front Cell Neurosci. 2013;7:159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22. Lutz SE, Zhao Y, Gulinello M, Lee SC, Raine CS, Brosnan CF. Deletion of astrocyte connexins 43 and 30 leads to a dysmyelinating phenotype and hippocampal CA1 vacuolation. J Neurosci. 2009;29(24):7743–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23. Dutta DJ, Woo DH, Lee PR, Pajevic S, Bukalo O, Huffman WC, Wake H, Basser PJ, SheikhBahaei S, Lazarevic V, Smith JC, et al. Regulation of myelin structure and conduction velocity by perinodal astrocytes. Proc Natl Acad Sci. 2018; 115(46):11832–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Bohlen CJ, Bennett FC, Tucker AF, Collins HY, Mulinyawe SB, Barres BA. Diverse requirements for microglial survival, specification, and function revealed by defined-medium cultures. Neuron. 2017;94(4):759–773.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Bozoyan L, Khlghatyan J, Saghatelyan A. Astrocytes control the development of the migration-promoting vasculature scaffold in the postnatal brain via VEGF signaling. J Neurosci. 2012;32(5):1687–704. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Oberheim NA, Takano T, Han X, He W, Lin JHC, Wang F, Xu Q, Wyatt JD, Pilcher W, Ojemann JG, Ransom BR, et al. Uniquely hominid features of adult human astrocytes. J Neurosci. 2009;29(10):3276–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Oberheim NA, Wang X, Goldman S, Nedergaard M. Astrocytic complexity distinguishes the human brain. Trends Neurosci. 2006;29(10):547–53. [DOI] [PubMed] [Google Scholar]
- 28. Michelucci A, Bithell A, Burney MJ, Johnston CE, Wong KY, Teng SW, Desai J, Gumbleton N, Anderson G, Stanton LW, Williams BP, et al. The neurogenic potential of astrocytes is regulated by inflammatory signals. Mol Neurobiol. 2016;53(6):3724–39. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29. Zamboni M, Llorens-Bobadilla E, Magnusson JP, Frisén J. A Widespread neurogenic potential of neocortical astrocytes is induced by injury. Cell Stem Cell. 2020;27(4):605–17.e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. Russo FB, Freitas BC, Pignatari GC, Fernandes IR, Sebat J, Muotri AR, Beltrão-Braga PCB. Modeling the interplay between neurons and astrocytes in autism using human induced pluripotent stem cells. Biol Psychiatry. 2018;83(7):569–78. [DOI] [PubMed] [Google Scholar]
- 31. Mizuno GO, Wang Y, Shi G, Wang Y, Sun J, Papadopoulos S, Broussard GJ, Unger EK, Deng W, Weick J, Bhattacharyya A, et al. Aberrant calcium signaling in astrocytes inhibits neuronal excitability in a human down syndrome stem cell model. Cell Rep. 2018;24(2):355–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32. Skripuletz T, Hackstette D, Bauer K, Gudi V, Pul R, Voss E, Berger K, Kipp M, Baumgärtner W, Stangel M. Astrocytes regulate myelin clearance through recruitment of microglia during cuprizone-induced demyelination. Brain. 2013;136(1):147–67. [DOI] [PubMed] [Google Scholar]
- 33. Correale J, Farez MF. The role of astrocytes in multiple sclerosis progression. Front Neurol. 2015;6:180. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Williams MR, Hampton T, Pearce RKB, Hirsch SR, Ansorge O, Thom M, Maier M. Astrocyte decrease in the subgenual cingulate and callosal genu in schizophrenia. Eur Arch Psychiatry Clin Neurosci. 2013;263(1):41–52. [DOI] [PubMed] [Google Scholar]
- 35. Kim R, Healey KL, Sepulveda-Orengo MT, Reissner KJ. Astroglial correlates of neuropsychiatric disease: from astrocytopathy to astrogliosis. Prog Neuro-Psychopharmacology Biol Psychiatry. 2018;87(Pt A):126–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. David J, Gormley S, McIntosh AL, Kebede V, Thuery G, Varidaki A, Coffey ET, Harkin A. L-alpha-amino adipic acid provokes depression-like behaviour and a stress related increase in dendritic spine density in the pre-limbic cortex and hippocampus in rodents. Behav Brain Res. 2019;362:90–102. [DOI] [PubMed] [Google Scholar]
- 37. Etiévant A, Oosterhof C, Bétry C, Abrial E, Novo-Perez M, Rovera R, Scarna H, Devader C, Mazella J, Wegener G, Sánchez C, et al. Astroglial control of the antidepressant-like effects of prefrontal cortex deep brain stimulation. EBioMedicine. 2015;2(8):898–908. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Lima A, Sardinha VM, Oliveira AF, Reis M, Mota C, Silva MA, Marques F, Cerqueira JJ, Pinto L, Sousa N, Oliveira JF. Astrocyte pathology in the prefrontal cortex impairs the cognitive function of rats. Mol Psychiatry. 2014;19(7):834–41. [DOI] [PubMed] [Google Scholar]
- 39. Cui W, Allen ND, Skynner M, Gusterson B, Clark AJ. Inducible ablation of astrocytes shows that these cells are required for neuronal survival in the adult brain. Glia. 2001; 34(4):272–82. [DOI] [PubMed] [Google Scholar]
- 40. Schreiner B, Romanelli E, Liberski P, Ingold-Heppner B, Sobottka-Brillout B, Hartwig T, Chandrasekar V, Johannssen H, Zeilhofer HU, Aguzzi A, Heppner F, et al. Astrocyte depletion impairs redox homeostasis and triggers neuronal loss in the adult CNS. Cell Rep. 2015;12(9):1377–84. [DOI] [PubMed] [Google Scholar]
- 41. Chang YF, Cauley RK, Chang JD, Rao VV. L-α-aminoadipate inhibits kynurenate synthesis in rat brain hippocampus and tissue culture. Neurochem Res. 1997;22(7):825–29. [DOI] [PubMed] [Google Scholar]
- 42. Yu X, Taylor AMW, Nagai J, Golshani P, Evans CJ, Coppola G, Khakh BS. Reducing Astrocyte calcium signaling in vivo alters striatal microcircuits and causes repetitive behavior. Neuron. 2018;99(6):1170–87.e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43. Adamsky A, Kol A, Kreisel T, Doron A, Ozeri-Engelhard N, Melcer T, Refaeli R, Horn H, Regev L, Groysman M, London M, et al. Astrocytic activation generates de novo neuronal potentiation and memory enhancement. Cell. 2018;174:59–71.e14. [DOI] [PubMed] [Google Scholar]
- 44. Pereira MF, Amaral IM, Lopes C, Leitão C, Madeira D, Lopes JP, Gonçalves FQ, Canas PM, Cunha RA, Agostinho P. l-α-aminoadipate causes astrocyte pathology with negative impact on mouse hippocampal synaptic plasticity and memory. FASEB J. 2021;35(8):e21726. [DOI] [PubMed] [Google Scholar]
- 45. Jäkel S, Dimou L. Glial cells and their function in the adult brain: a journey through the history of their ablation. Front Cell Neurosci. 2017;11:24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46. O’Neill E, Chiara Goisis R, Haverty R, Harkin A. L-alpha-aminoadipic acid restricts dopaminergic neurodegeneration and motor deficits in an inflammatory model of Parkinson’s disease in male rats. J Neurosci Res. 2019;97(7):804–16. [DOI] [PubMed] [Google Scholar]
- 47. Madadi S, Pasbakhsh P, Tahmasebi F, Mortezaee K, Khanehzad M, Boroujeni FB, Noorzehi G, Kashani IR. Astrocyte ablation induced by La-aminoadipate (L-AAA) potentiates remyelination in a cuprizone demyelinating mouse model. Metab Brain Dis. 2019;34(2):593–603. [DOI] [PubMed] [Google Scholar]
- 48. Jang SE, Qiu L, Chan LL, Tan EK, Zeng L. Current status of stem cell-derived therapies for Parkinson’s disease: from cell assessment and imaging modalities to clinical trials. Front Neurosci. 2020;14:558532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Hayashi Y, Lin HT, Lee CC, Tsai KJ. Effects of neural stem cell transplantation in Alzheimer’s disease models. J Biomed Sci. 2020;27(1):1–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Bazarek S, Johnston BR, Sten M, Mandeville R, Eggan K, Wainger BJ, Brown JM. Spinal motor neuron transplantation to enhance nerve reconstruction strategies: towards a cell therapy. Exp Neurol. 2022;353:114054. [DOI] [PubMed] [Google Scholar]
- 51. Fischer I, Dulin JN, Lane MA. Transplanting neural progenitor cells to restore connectivity after spinal cord injury. Nat Rev Neurosci. 2020;21(7):366–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52. Baloh RH, Glass JD, Svendsen CN. Stem cell transplantation for amyotrophic lateral sclerosis. Curr Opin Neurol. 2018;31(5):655–61. [DOI] [PubMed] [Google Scholar]
- 53. Izrael M, Slutsky SG, Revel M. Rising stars: astrocytes as a therapeutic target for ALS disease. Front Neurosci. 2020;14:824. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Man JHK, Groenink L, Caiazzo M. Cell reprogramming approaches in gene- and cell-based therapies for Parkinson’s disease. J Control Release. 2018;286:114–24. [DOI] [PubMed] [Google Scholar]
- 55. Liu S, Schackel T, Weidner N, Puttagunta R. Biomaterial-supported cell transplantation treatments for spinal cord injury: challenges and perspectives. Front Cell Neurosci. 2018;11:430. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Xu AK, Gong Z, He YZ, Xia KS, Tao HM. Comprehensive therapeutics targeting the corticospinal tract following spinal cord injury. J Zhejiang Univ Sci B. 2019;20(3):205–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Grochowski C, Radzikowska E, Maciejewski R. Neural stem cell therapy—brief review. Clin Neurol Neurosurg. 2018;173:8–14. [DOI] [PubMed] [Google Scholar]
- 58. Ghosh HS. Adult neurogenesis and the promise of adult neural stem cells. J Exp Neurosci. 2019;13:1179069519856876. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Zhang GL, Zhu ZH, Wang YZ. Neural stem cell transplantation therapy for brain ischemic stroke: review and perspectives. World J Stem Cells. 2019;11(10):817–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60. Martins-Macedo J, Lepore AC, Domingues HS, Salgado AJ, Gomes ED, Pinto L. Glial restricted precursor cells in central nervous system disorders: current applications and future perspectives. Glia. 2021;69(3):513–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Goldman SA, Mariani JN, Madsen PM. Glial progenitor cell-based repair of the dysmyelinated brain: progression to the clinic. Semin Cell Dev Biol. 2021;116:62–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Serrenho I, Rosado M, Dinis A, Cardoso CM, Grãos M, Manadas B, Baltazar G. Stem cell therapy for neonatal hypoxic-ischemic encephalopathy: a systematic review of preclinical studies. Int J Mol Sci. 2021;22(6):3142. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63. Zhao Y, Wang S, Song X, Yuan J, Qi D, Gu X, Yin MY, Han Z, Zhu Y, Liu Z, Zhang Y, et al. Glial cell-based vascular mechanisms and transplantation therapies in brain vessel and neurodegenerative diseases. Front Cell Neurosci. 2021;15: 627682. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Albert K, Niskanen J, Kälvälä S, Lehtonen Š. Utilising induced pluripotent stem cells in neurodegenerative disease research: focus on glia. Int J Mol Sci. 2021;22(9):4334. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65. Baez-Jurado E, Hidalgo-Lanussa O, Barrera-Bailón B, Sahebkar A, Ashraf GM, Echeverria V, Barreto GE. Secretome of mesenchymal stem cells and its potential protective effects on brain pathologies. Mol Neurobiol. 2019;56(10):6902–27. [DOI] [PubMed] [Google Scholar]
- 66. Das M, Mayilsamy K, Mohapatra SS, Mohapatra S. Mesenchymal stem cell therapy for the treatment of traumatic brain injury: progress and prospects. Rev Neurosci. 2019;30(8):839–55. [DOI] [PubMed] [Google Scholar]
- 67. Zhang L, Dong Z-F, Zhang J-Y. Immunomodulatory role of mesenchymal stem cells in Alzheimer’s disease. Life Sci. 2020;246:117405. [DOI] [PubMed] [Google Scholar]
- 68. Lv B, Zhang X, Yuan J, Chen Y, Ding H, Cao X, Huang A. Biomaterial-supported MSC transplantation enhances cell-cell communication for spinal cord injury. Stem Cell Res Ther. 2021;12(1):36. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69. Hatakeyama M, Ninomiya I, Otsu Y, Omae K, Kimura Y, Onodera O, Fukushima M, Shimohata T, Kanazawa M. Cell therapies under clinical trials and polarized cell therapies in pre-clinical studies to treat ischemic stroke and neurological diseases: a literature review. Int J Mol Sci. 2020;21(17):1–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70. Mariani JN, Zou L, Goldman SA. Human glial chimeric mice to define the role of glial pathology in human disease. Methods Mol Biol. 2019;1936:311–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. Lepore AC, Rauck B, Dejea C, Pardo AC, Rao MS, Rothstein JD, Maragakis NJ. Focal transplantation-based astrocyte replacement is neuroprotective in a model of motor neuron disease. Nat Neurosci. 2008;11(11):1294–301. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Lepore AC, O’Donnell J, Kim AS, Williams T, Tuteja A, Rao MS, Kelley LL, Campanelli JT, Maragakis NJ. Human glial-restricted progenitor transplantation into cervical spinal cord of the SOD1 G93A mouse model of ALS. PLoS ONE. 2011;6(10):e25968. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. Kondo T, Funayama M, Tsukita K, Hotta A, Yasuda A, Nori S, Kaneko S, Nakamura M, Takahashi R, Okano H, Yamanaka S, et al. Focal transplantation of human iPSC-derived glial-rich neural progenitors improves lifespan of ALS mice. Stem Cell Rep. 2014;3(2):242–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Izrael M, Slutsky SG, Admoni T, Cohen L, Granit A, Hasson A, Itskovitz-Eldor J, Krush Paker L, Kuperstein G, Lavon N, Yehezkel Ionescu S, et al. Safety and efficacy of human embryonic stem cell-derived astrocytes following intrathecal transplantation in SOD1 G93A and NSG animal models. Stem Cell Res. Ther. 2018;9(1):152. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Renton AE, Chiò A, Traynor BJ. State of play in amyotrophic lateral sclerosis genetics. Nat Neurosci. 2014;17(1):17–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76. Mathis S, Goizet C, Soulages A, Vallat JM, Masson GL. Genetics of amyotrophic lateral sclerosis: a review. J Neurol Sci. 2019;399:217–26. [DOI] [PubMed] [Google Scholar]
- 77. Calvo AC, Manzano R, Mendonça DMF, Muñoz MJ, Zaragoza P, Osta R. Amyotrophic lateral sclerosis: a focus on disease progression. Biomed Res Int. 2014;2014:925101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Al-Chalabi A, Jones A, Troakes C, King A, Al-Sarraj S, Van Den Berg LH. The genetics and neuropathology of amyotrophic lateral sclerosis. Acta Neuropathol. 2012;124(3):339–52. [DOI] [PubMed] [Google Scholar]
- 79. Taylor JP, Brown RH, Cleveland DW. Decoding ALS: from genes to mechanism. Nature. 2016;539(7628):197–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Papadeas ST, Kraig SE, O’Banion C, Lepore AC, Maragakis NJ. Astrocytes carrying the superoxide dismutase 1 (SOD1G93A) mutation induce wild-type motor neuron degeneration in vivo. Proc Natl Acad Sci. 2011;108(43):17803–808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Qian K, Huang H, Peterson A, Hu B, Maragakis NJ, Ming G-L, Chen H, Zhang SC. Sporadic ALS astrocytes induce neuronal degeneration in vivo. Stem Cell Reports. 2017;8(4):843–55. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82. Chen H, Qian K, Chen W, Hu B, Blackbourn LW, Du Z, Ma L, Liu H, Knobel KM, Ayala M, Zhang SC. Human-derived neural progenitors functionally replace astrocytes in adult mice. J Clin Invest. 2015;125(3):1033–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Wang L, Gutmann DH, Roos RP. Astrocyte loss of mutant SOD1 delays ALS disease onset and progression in G85R transgenic mice. Hum Mol Genet. 2011;20(2):286–93. [DOI] [PubMed] [Google Scholar]
- 84. Dirren E, Aebischer J, Rochat C, Towne C, Schneider BL, Aebischer P. SOD1 silencing in motoneurons or glia rescues neuromuscular function in ALS mice. Ann Clin Transl Neurol. 2015;2(2):167–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85. Gong YH, Parsadanian AS, Andreeva A, Snider WD, Elliott JL. Restricted expression of G86R Cu/Zn superoxide dismutase in astrocytes results in astrocytosis but does not cause motoneuron degeneration. J Neurosci. 2000;20(2):660–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86. Stephenson DT, Stephenson DT, Wright S. Reactive astrogliosis is widespread in the subcortical white matter of amyotrophic lateral sclerosis brain. J Neuropathol Exp Neurol. 1991;50(3):263–77. [DOI] [PubMed] [Google Scholar]
- 87. Schiffer D, Cordera S, Cavalla P, Migheli A. Reactive astrogliosis of the spinal cord in amyotrophic lateral sclerosis. J Neurol Sci. 1996;139:27–33. [DOI] [PubMed] [Google Scholar]
- 88. Vargas MR, Johnson JA. Astrogliosis in amyotrophic lateral sclerosis: role and therapeutic potential of astrocytes. Neurotherapeutics. 2010;7(4):471–781. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89. Schiffer D, Fiano V. Astrogliosis in ALS: possible interpretations according to pathogenetic hypotheses. Amyotroph Lateral Scler Other Mot. Neuron Disord. 2004;5(1):22–25. [DOI] [PubMed] [Google Scholar]
- 90. Wegorzewska I, Bell S, Cairns NJ, Miller TM, Baloh RH. TDP-43 mutant transgenic mice develop features of ALS and frontotemporal lobar degeneration. Proc Natl Acad Sci U S A. 2009;106(44):18809–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91. Rossi D, Brambilla L, Valori CF, Roncoroni C, Crugnola A, Yokota T, Bredesen DE, Volterra A. Focal degeneration of astrocytes in amyotrophic lateral sclerosis. Cell Death Differ. 2008;15(11):1691–700. [DOI] [PubMed] [Google Scholar]
- 92. Trias E, Díaz-Amarilla P, Olivera-Bravo S, Isasi E, Drechsel DA, Lopez N, Bradford CS, Ireton KE, Beckman JS, Barbeito L. Phenotypic transition of microglia into astrocyte-like cells associated with disease onset in a model of inherited ALS. Front Cell Neurosci. 2013;7:274. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Trias E, Ibarburu S, Barreto-Núñez R, Barbeito L. Significance of aberrant glial cell phenotypes in pathophysiology of amyotrophic lateral sclerosis. Neurosci Lett. 2017;636:27–31. [DOI] [PubMed] [Google Scholar]
- 94. Almer G, Vukosavic S, Romero N, Przedborski S. Inducible nitric oxide synthase up-regulation in a transgenic mouse model of familial amyotrophic lateral sclerosis. J Neurochem. 1999;72(6):2415–25. [DOI] [PubMed] [Google Scholar]
- 95. Martin LJ, Liu Z, Chen K, Price AC, Yan P, Swaby JA, Golden WC. Motor neuron degeneration in amyotrophic lateral sclerosis mutant superoxide dismutase-1 transgenic mice: mechanisms of mitochondriopathy and cell death. J Comp Neurol. 2007;500(1):20–46. [DOI] [PubMed] [Google Scholar]
- 96. Hensley K, Mhatre M, Mou S, Pye QN, Stewart C, West M, Williamson KS. On the relation of oxidative stress to neuroinflammation: lessons learned from the G93A-SOD1 mouse model of amyotrophic lateral sclerosis. Antioxidants Redox Signal. 2006;8(11–12):2075–87. [DOI] [PubMed] [Google Scholar]
- 97. Birger A, Ben-Dor I, Ottolenghi M, Turetsky T, Gil Y, Sweetat S, Perez L, Belzer V, Casden N, Steiner D, Izrael M, et al. Human iPSC-derived astrocytes from ALS patients with mutated C9ORF72 show increased oxidative stress and neurotoxicity. EBioMedicine. 2019;50:274–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98. Anneser JMH, Cookson MR, Ince PG, Shaw PJ, Borasio GD. Glial cells of the spinal cord and subcortical white matter up-regulate neuronal nitric oxide synthase in sporadic amyotrophic lateral sclerosis. Exp Neurol. 2001;171(2): 418–21. [DOI] [PubMed] [Google Scholar]
- 99. Rojas F, Cortes N, Abarzua S, Dyrda A, van Zundert B. Astrocytes expressing mutant SOD1 and TDP43 trigger motoneuron death that is mediated via sodium channels and nitroxidative stress. Front Cell Neurosci. 2014;8:24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100. Dringen R, Hirrlinger J. Glutathione pathways in the brain. Biol Chem. 2003;384(4):505–16. [DOI] [PubMed] [Google Scholar]
- 101. Chen Y, Vartiainen NE, Ying W, Chan PH, Koistinaho J, Swanson RA. Astrocytes protect neurons from nitric oxide toxicity by a glutathione-dependent mechanism. J Neurochem. 2001;77(6):1601–10. [DOI] [PubMed] [Google Scholar]
- 102. Cassina P, Cassina A, Pehar M, Castellanos R, Gandelman M, De León A, Robinson KM, Mason RP, Beckman JS, Barbeito L, Radi R. Mitochondrial dysfunction in SOD1G93A-bearing astrocytes promotes motor neuron degeneration: prevention by mitochondrial-targeted antioxidants. J Neurosci. 2008; 28(16):4115–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Harlan BA, Pehar M, Sharma DR, Beeson G, Beeson CC, Vargas MR. Enhancing NAD+ salvage pathway reverts the toxicity of primary astrocytes expressing amyotrophic lateral sclerosis-linked mutant superoxide dismutase 1 (SOD1). J Biol Chem. 2016;291(20):10836–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104. Pehar M, Cassina P, Vargas MR, Castellanos R, Viera L, Beckman JS, Estévez AG, Barbeito L. Astrocytic production of nerve growth factor in motor neuron apoptosis: implications for amyotrophic lateral sclerosis. J Neurochem. 2004;89(2):464–73. [DOI] [PubMed] [Google Scholar]
- 105. Lin DT, Wu J, Holstein D, Upadhyay G, Rourk W, Muller E, Lechleiter JD. Ca2+ signaling, mitochondria and sensitivity to oxidative stress in aging astrocytes. Neurobiol Aging. 2007;28(1):99–111. [DOI] [PubMed] [Google Scholar]
- 106. Boitier E, Rea R, Duchen MR. Mitochondria exert a negative feedback on the propagation of intracellular Ca2+ waves in rat cortical astrocytes. J Cell Biol. 1999;145(4):795–808. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Decuypere JP, Monaco G, Missiaen L, De Smedt H, Parys JB, Bultynck G. IP(3) receptors, mitochondria, and Ca2+ signaling: implications for aging. J Aging Res. 2011;2011:920178. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108. Agarwal A, Wu PH, Hughes EG, Fukaya M, Tischfield MA, Langseth AJ, Wirtz D, Bergles DE. Transient opening of the mitochondrial permeability transition pore induces microdomain calcium transients in astrocyte processes. Neuron. 2017;93(3):587–605.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Kawamata H, Ng SK, Diaz N, Burstein S, Morel L, Osgood A, Sider B, Higashimori H, Haydon PG, Manfredi G, Yang Y. Abnormal intracellular calcium signaling and SNARE dependent exocytosis contributes to SOD1G93A astrocyte- mediated toxicity in amyotrophic lateral sclerosis. J Neurosci. 2014;34(6):2331–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Norante RP, Peggion C, Rossi D, Martorana F, De Mario A, Lia A, Massimino ML, Bertoli A. ALS-associated SOD1(G93A) decreases SERCA pump levels and increases store-operated Ca2+ entry in primary spinal cord astrocytes from a transgenic mouse model. Int J Mol Sci. 2019;20(20):5151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111. Aras R, Barron AM, Pike CJ. Caspase activation contributes to astrogliosis. Brain Res. 2012;1450:102–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Varcianna A, Myszczynska MA, Castelli LM, O’Neill B, Kim Y, Talbot J, Nyberg S, Nyamali I, Heath PR, Stopford MJ, Hautbergue GM, et al. Micro-RNAs secreted through astrocyte-derived extracellular vesicles cause neuronal network degeneration in C9orf72 ALS. EBioMedicine. 2019;40:626–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113. Bezzi P, Gundersen V, Galbete JL, Seifert G, Steinhäuser C, Pilati E, Volterra A. Astrocytes contain a vesicular compartment that is competent for regulated exocytosis of glutamate. Nat Neurosci. 2004;7(6):613–20. [DOI] [PubMed] [Google Scholar]
- 114. Hensley K, Abdel-Moaty H, Hunter J, Mhatre M, Mhou S, Nguyen K, Potapova T, Pye QN, Qi M, Rice H, Stewart C, et al. Primary glia expressing the G93A-SOD1 mutation present a neuroinflammatory phenotype and provide a cellular system for studies of glial inflammation. J Neuroinflammation. 2006;3:2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Haidet-Phillips AM, Hester ME, Miranda CJ, Meyer K, Braun L, Frakes A, Song S, Likhite S, Murtha MJ, Foust KD, Rao M, et al. Astrocytes from familial and sporadic ALS patients are toxic to motor neurons. Nat Biotechnol. 2011;29(9):824–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 116. Tripathi P, Rodriguez-Muela N, Klim JR, de Boer AS, Agrawal S, Sandoe J, Lopes CS, Ogliari KS, Williams LA, Shear M, Rubin LL, et al. Reactive astrocytes promote ALS-like degeneration and intracellular protein aggregation in human motor neurons by disrupting autophagy through TGF-β1. Stem Cell Rep. 2017;9(2):667–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117. Howland DS, Liu J, She Y, Goad B, Maragakis NJ, Kim B, Erickson J, Kulik J, DeVito L, Psaltis G, DeGennaro LJ, et al. Focal loss of the glutamate transporter EAAT2 in a transgenic rat model of SOD1 mutant-mediated amyotrophic lateral sclerosis (ALS). Proc Natl Acad Sci U S A. 2002;99(3):1604–609. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118. Tong J, Huang C, Bi F, Wu Q, Huang B, Liu X, Li F, Zhou H, Xia XG. Expression of ALS-linked TDP-43 mutant in astrocytes causes non-cell-autonomous motor neuron death in rats. EMBO J. 2013;32(13):1917–26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119. Rosenblum LT, Shamamandri-Markandaiah S, Ghosh B, Foran E, Lepore AC, Pasinelli P, Trotti D. Mutation of the caspase-3 cleavage site in the astroglial glutamate transporter EAAT2 delays disease progression and extends lifespan in the SOD1-G93A mouse model of ALS. Exp Neurol. 2017;292:145–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120. Aebischer J, Cassina P, Otsmane B, Moumen A, Seilhean D, Meininger V, Barbeito L, Pettmann B, Raoul C. IFNγ triggers a LIGHT-dependent selective death of motoneurons contributing to the non-cell-autonomous effects of mutant SOD1. Cell Death Differ. 2011;18(5):754–68. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Raoul C, Estévez AG, Nishimune H, Cleveland DW, DeLapeyrière O, Henderson CE, Haase G, Pettmann B. Motoneuron death triggered by a specific pathway downstream of Fas: potentiation by ALS-linked SOD1 mutations. Neuron. 2002;35(6):1067–83. [DOI] [PubMed] [Google Scholar]
- 122. Petri S, Kiaei M, Wille E, Calingasan NY, Flint Beal M. Loss of Fas ligand-function improves survival in G93A-transgenic ALS mice. J Neurol Sci. 2006;251(1–2):44–49. [DOI] [PubMed] [Google Scholar]
- 123. Gandelman M, Levy M, Cassina P, Barbeito L, Beckman JS. P2X7 receptor-induced death of motor neurons by a peroxynitrite/FAS-dependent pathway. J Neurochem. 2013;126(3): 382–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124. Vargas MR, Pehar M, Cassina P, Estévez AG, Beckman JS, Barbeito L. Stimulation of nerve growth factor expression in astrocytes by peroxynitrite. In Vivo (Brooklyn). 2004;18(3): 269–74. [PubMed] [Google Scholar]
- 125. Deinhardt K, Chao MV. Trk receptors. Handb Exp Pharmacol. 2014;220:103–19. [DOI] [PubMed] [Google Scholar]
- 126. Miller FD, Kaplan DR. Life and death decisions: a biological role for the p75 neurotrophin receptor. Cell Death Differ. 1998;5(5):343–45. [DOI] [PubMed] [Google Scholar]
- 127. Kraemer BR, Yoon SO, Carter BD. The biological functions and signaling mechanisms of the p75 neurotrophin receptor. Handb Exp Pharmacol. 2014;220:121–64. [DOI] [PubMed] [Google Scholar]
- 128. Lowry KS, Murray SS, McLean CA, Talman P, Mathers S, Lopes EC, Cheema SS. A potential role for the p75 low-affinity neurotrophin receptor in spinal motor neuron degeneration in murine and human amyotrophic lateral sclerosis. Amyotroph Lateral Scler Other Mot Neuron Disord. 2001;2(3):127–34. [DOI] [PubMed] [Google Scholar]
- 129. Copray JCVM, Jaarsma D, Küst BM, Bruggeman RWG, Mantingh I, Brouwer N, Boddeke HWGM. Expression of the low affinity neurotrophin receptor p75 in spinal motoneurons in a transgenic mouse model for amyotrophic lateral sclerosis. Neuroscience. 2003;116(3):685–94. [DOI] [PubMed] [Google Scholar]
- 130. Smith KS, Rush RA, Rogers ML. Characterization and changes in neurotrophin receptor p75N-expressing motor neurons in SOD1(G93A)G1H mice. J Comp Neurol. 2015;523(16):2476. [DOI] [PubMed] [Google Scholar]
- 131. Schapira a H, Cooper JM, Dexter D, Jenner P, Clark JB, Marsden CD. Mitochondrial complex I deficiency in Parkinson’s disease. Lancet. 1989;1(8649):1269. [DOI] [PubMed] [Google Scholar]
- 132. Parihar MS, Parihar A, Fujita M, Hashimoto M, Ghafourifar P. Mitochondrial association of alpha-synuclein causes oxidative stress. Cell Mol Life Sci. 2008;65(7–8):1272–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133. Schapira AH. Mitochondria in the aetiology and pathogenesis of Parkinson’s disease. Lancet Neurol. 2008;7(1):97–109. [DOI] [PubMed] [Google Scholar]
- 134. Calvo A, Chiò A, Pagani M, Cammarosano S, Dematteis F, Moglia C, Solero L, Manera U, Martone T, Brunetti M, Balma M, et al. Parkinsonian traits in amyotrophic lateral sclerosis (ALS): a prospective population-based study. J Neurol. 2019;266(7):1633–42. [DOI] [PubMed] [Google Scholar]
- 135. Kuzuhara S, Kokubo Y. Atypical Parkinsonism of Japan: amyotrophic lateral sclerosis-parkinsonism-dementia complex of the Kii peninsula of Japan (Muro disease): an update. Mov Disord. 2005;20(Suppl 12): S108–13. [DOI] [PubMed] [Google Scholar]
- 136. de Lau LM, Breteler MM. Epidemiology of Parkinson’s disease. Lancet Neurol. 2006;5(6):525–35. [DOI] [PubMed] [Google Scholar]
- 137. Tysnes OB, Storstein A. Epidemiology of Parkinson’s disease. J Neural Transm. 2017;124(8):901–905. [DOI] [PubMed] [Google Scholar]
- 138. Obeso JA, Stamelou M, Goetz CG, Poewe W, Lang AE, Weintraub D, Burn D, Halliday GM, Bezard E, Przedborski S, Lehericy S, et al. Past, present, and future of Parkinson’s disease: a special essay on the 200th Anniversary of the Shaking Palsy. Mov Disord. 2017;32(9):1264–310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139. Brück D, Wenning GK, Stefanova N, Fellner L. Glia and alpha-synuclein in neurodegeneration: a complex interaction. Neurobiol Dis. 2016;85:262–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. Azizi SA. Parkinson’s Disease. In: Koob GF, Le Moal M, Thompson RF, eds. Encyclopedia of Behavioral Neuroscience. 1st ed. Elsevier Science; 2010:24–28. [Google Scholar]
- 141. Kalia LV, Lang AE. Parkinson’s disease. Lancet. 2015;386(9996):896–912. [DOI] [PubMed] [Google Scholar]
- 142. Martinez-Martin P, Rodriguez-Blazquez C, Kurtis MM, Chaudhuri KR. The impact of non-motor symptoms on health-related quality of life of patients with Parkinson’s disease. Mov Disord. 2011;26(3):399–406. [DOI] [PubMed] [Google Scholar]
- 143. Lebouvier T, Chaumette T, Paillusson S, Duyckaerts C, Bruley Des Varannes S, Neunlist M, Derkinderen P. The second brain and Parkinson’s disease. Eur J Neurosci. 2009;30(5):735–41. [DOI] [PubMed] [Google Scholar]
- 144. Devos D, Lebouvier T, Lardeux B, Biraud M, Rouaud T, Pouclet H, Coron E, Bruley des Varannes S, Naveilhan P, Nguyen JM, Neunlist M, et al. Colonic inflammation in Parkinson’s disease. Neurobiol Dis. 2013;50:42–48. [DOI] [PubMed] [Google Scholar]
- 145. Clairembault T, Leclair-Visonneau L, Neunlist M, Derkinderen P. Enteric glial cells: new players in Parkinson’s disease? Mov Disord. 2015;30(4):494–98. [DOI] [PubMed] [Google Scholar]
- 146. Braak H, Braak E. Pathoanatomy of Parkinson’s disease. J Neurol. 2000;247(Suppl 2): II3–10. [DOI] [PubMed] [Google Scholar]
- 147. Braak H, Del Tredici K, Rüb U, De Vos RAI, Jansen Steur ENH, Braak E. Staging of brain pathology related to sporadic Parkinson’s disease. Neurobiol Aging. 2003;24(2):197–211. [DOI] [PubMed] [Google Scholar]
- 148. Braak H, Sastre M, Del Tredici K. Development of α-synuclein immunoreactive astrocytes in the forebrain parallels stages of intraneuronal pathology in sporadic Parkinson’s disease. Acta Neuropathol. 2007;114(3):231–41. [DOI] [PubMed] [Google Scholar]
- 149. Tawfik VL, Chang SY, Hitti FL, Roberts DW, Leiter JC, Jovanovic S, Lee KH. Deep brain stimulation results in local glutamate and adenosine release: investigation into the role of astrocytes. Neurosurgery. 2010;67(2):367–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150. Bahat-Stroomza M, Barhum Y, Levy YS, Karpov O, Bulvik S, Melamed E, Offen D. Induction of adult human bone marrow mesenchymal stromal cells into functional astrocyte-like cells: potential for restorative treatment in Parkinson’s disease. J Mol Neurosci. 2009;39(1–2):199–210. [DOI] [PubMed] [Google Scholar]
- 151. Proschel C, Stripay JL, Shih CH, Munger JC, Noble MD. Delayed transplantation of precursor cell-derived astrocytes provides multiple benefits in a rat model of Parkinsons. EMBO Mol Med. 2014;6(4):504–18. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152. Song JJ, Oh SM, Kwon OC, Wulansari N, Lee HS, Chang MY, Lee E, Sun W, Lee SE, Chang S, An H, et al. Cografting astrocytes improves cell therapeutic outcomes in a Parkinson’s disease model. J Clin Invest. 2018;128(1):463–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153. Tornatore C, Baker-Cairns B, Yadid G, Hamilton R, Meyers K, Atwood W, Cummins A, Tanner V, Major E. Expression of tyrosine hydroxylase in an immortalized human fetal astrocyte cell line; in vitro characterization and engraftment into the rodent striatum. Cell Transplant. 1996;5(2):145–63. [DOI] [PubMed] [Google Scholar]
- 154. Gu XL, Long CX, Sun L, Xie C, Lin X, Cai H. Astrocytic expression of Parkinson’s disease-related A53T alpha-synuclein causes neurodegeneration in mice. Mol Brain. 2010;3:12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155. di Domenico A, Carola G, Calatayud C, Pons-Espinal M, Muñoz JP, Richaud-Patin Y, Fernandez-Carasa I, Gut M, Faella A, Parameswaran J, Soriano J, et al. Patient-specific iPSC-derived astrocytes contribute to non-cell-autonomous neurodegeneration in Parkinson’s disease. Stem Cell Rep. 2019;12(2):213–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Ji KA, Eu MY, Kang SH, Gwag BJ, Jou I, Joe EH. Differential neutrophil infiltration contributes to regional differences in brain inflammation in the substantia nigra pars compacta and cortex. Glia. 2008;56(10):1039–47. [DOI] [PubMed] [Google Scholar]
- 157. Asanuma M, Miyazaki I, Diaz-Corrales FJ, Kimoto N, Kikkawa Y, Takeshima M, Miyoshi K, Murata M. Neuroprotective effects of zonisamide target astrocyte. Ann Neurol. 2010;67(2):239–49. [DOI] [PubMed] [Google Scholar]
- 158. Isooka N, Miyazaki I, Kikuoka R, Wada K, Nakayama E, Shin K, Yamamoto D, Kitamura Y, Asanuma M. Dopaminergic neuroprotective effects of rotigotine via 5-HT1A receptors: possibly involvement of metallothionein expression in astrocytes. Neurochem Int. 2020;132:104608. [DOI] [PubMed] [Google Scholar]
- 159. Ramos-Gonzalez P, Mato S, Chara JC, Verkhratsky A, Matute C, Cavaliere F. Astrocytic atrophy as a pathological feature of Parkinson’s disease with LRRK2 mutation. NPJ Park Dis. 2021;7(1):31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160. Lee HJ, Kim C, Lee SJ. Alpha-synuclein stimulation of astrocytes: potential role for neuroinflammation and neuroprotection. Oxid Med Cell Longev. 2010;3(4):283–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Lee HJ, Suk JE, Patrick C, Bae EJ, Cho JH, Rho S, Hwang D, Masliah E, Lee SJ. Direct transfer of α-synuclein from neuron to astroglia causes inflammatory responses in synucleinopathies. J Biol Chem. 2010;285(12):9262–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162. Sorrentino ZA, Giasson BI, Chakrabarty P. α-Synuclein and astrocytes: tracing the pathways from homeostasis to neurodegeneration in Lewy body disease. Acta Neuropathol. 2019; 138(1):1–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 163. Cavaliere F, Cerf L, Dehay B, Ramos-Gonzalez P, De Giorgi F, Bourdenx M, Bessede A, Obeso JA, Matute C, Ichas F, Bezard E. 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. [DOI] [PubMed] [Google Scholar]
- 164. Fellner L, Irschick R, Schanda K, Reindl M, Klimaschewski L, Poewe W, Wenning GK, Stefanova N. Toll-like receptor 4 is required for α-synuclein dependent activation of microglia and astroglia. Glia. 2013;61(3):349–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165. Aflaki E, Stubblefield 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. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166. Gan L, Vargas MR, Johnson DA, Johnson JA. Astrocyte-specific overexpression of Nrf2 delays motor pathology and synuclein aggregation throughout the CNS in the alpha-synuclein mutant (A53T) mouse model. J Neurosci. 2012;32(49): 17775–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167. Alvarez-Erviti L, Seow Y, Schapira AH, Gardiner C, Sargent IL, Wood MJA, Cooper JM. Lysosomal dysfunction increases exosome-mediated alpha-synuclein release and transmission. Neurobiol Dis. 2011;42(3):360–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168. Braidy N, Gai WP, Xu YH, Sachdev P, Guillemin GJ, Jiang XM, Ballard JWO, Horan MP, Fang ZM, Chong BH, Chan DKY. Uptake and mitochondrial dysfunction of alpha-synuclein in human astrocytes, cortical neurons and fibroblasts. Transl Neurodegener. 2013;2(1):20. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169. Erustes AG, Stefani FY, Terashima JY, Stilhano RS, Monteforte PT, da Silva Pereira GJ, Han SW, Calgarotto AK, Hsu Y, Te Ureshino RP, Bincoletto C, et al. Overexpression of α-synuclein in an astrocyte cell line promotes autophagy inhibition and apoptosis. J Neurosci Res. 2018;96(1):160–71. [DOI] [PubMed] [Google Scholar]
- 170. Liu M, Qin L, Wang L, Tan J, Zhang H, Tang J, Shen X, Tan L, Wang C. Alpha-synuclein induces apoptosis of astrocytes by causing dysfunction of the endoplasmic reticulum-Golgi compartment. Mol Med Rep. 2018;18(1):322–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171. Stefanova N, Klimaschewski L, Poewe W, Wenning GK, Reindl M. Glial cell death induced by overexpression of alpha-synuclein. J Neurosci Res. 2001;65(5):432–38. [DOI] [PubMed] [Google Scholar]
- 172. Díaz EF, Labra VC, Alvear TF, Mellado LA, Inostroza CA, Oyarzún JE, Salgado N, Quintanilla RA, Orellana JA. Connexin 43 hemichannels and pannexin-1 channels contribute to the α-synuclein-induced dysfunction and death of astrocytes. Glia. 2019;67(8):1598–619. [DOI] [PubMed] [Google Scholar]
- 173. Barodia SK, McMeekin LJ, Creed RB, Quinones EK, Cowell RM, Goldberg MS. PINK1 phosphorylates ubiquitin predominantly in astrocytes. NPJ Park Dis. 2019;5:29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174. Choi I, Kim J, Jeong H-K, Kim B, Jou I, Park M, Chen L, Kang U-J, Zhuang X, Joe E-H. PINK1 deficiency attenuates astrocyte proliferation through mitochondrial dysfunction, reduced AKT and increased p38 MAPK activation, and downregulation of EGFR. Glia. 2013;61(5):800–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175. Larsen NJ, Ambrosi G, Mullett SJ, Berman SB, Hinkle DA. DJ-1 knock-down impairs astrocyte mitochondrial function. Neuroscience. 2011;196:251–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176. Liu Y, Hu J, Wu J, Zhu C, Hui Y, Han Y, Huang Z, Ellsworth K, Fan W. α7 nicotinic acetylcholine receptor-mediated neuroprotection against dopaminergic neuron loss in an MPTP mouse model via inhibition of astrocyte activation. J Neuroinflammation. 2012;9:98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177. Episcopo F, Tirolo C, Testa N, Caniglia S, Morale M, Marchetti B. Reactive astrocytes are key players in nigrostriatal dopaminergic neurorepair in the MPTP mouse model of Parkinson’s disease: focus on endogenous neurorestoration. Curr Aging Sci. 2013;6(1):45–55. [DOI] [PubMed] [Google Scholar]
- 178. Goswami P, Gupta S, Joshi N, Sharma S, Singh S. Astrocyte activation and neurotoxicity: a study in different rat brain regions and in rat C6 astroglial cells. Environ Toxicol Pharmacol. 2015;40(1):122–39. [DOI] [PubMed] [Google Scholar]
- 179. Liu X, Sullivan KA, Madl JE, Legare M, Tjalkens RB. Manganese-induced neurotoxicity: the role of astroglial-derived nitric oxide in striatal interneuron degeneration. Toxicol Sci. 2006;91(2):521–31. [DOI] [PubMed] [Google Scholar]
- 180. Exil V, Ping L, Yu Y, Chakraborty S, Caito SW, Wells KS, Karki P, Lee E, Aschner M. Activation of MAPK and FoxO by Manganese (Mn) in rat neonatal primary astrocyte cultures. PLoS ONE. 2014;9(5):e94753. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181. Charron G, Doudnikoff E, Canron MH, Li Q, Véga C, Marais S, Baufreton J, Vital A, Oliet SHR, Bezard E. Astrocytosis in Parkinsonism: considering tripartite striatal synapses in physiopathology? Front Aging Neurosci. 2014;6:258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182. Liu Y, Zeng X, Hui Y, Zhu C, Wu J, Taylor DH, Ji J, Fan W, Huang Z, Hu J. Activation of α7 nicotinic acetylcholine receptors protects astrocytes against oxidative stress-induced apoptosis: implications for Parkinson’s disease. Neuropharmacology. 2015;91:87–96. [DOI] [PubMed] [Google Scholar]
- 183. Lindholm D, Mäkelä J, Di Liberto V, Mudò G, Belluardo N, Eriksson O, Saarma M. Current disease modifying approaches to treat Parkinson’s disease. Cell Mol Life Sci. 2016;73(7):1365–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 184. Ishii T, Warabi E, Mann GE. Circadian control of BDNF-mediated Nrf2 activation in astrocytes protects dopaminergic neurons from ferroptosis. Free Radic Biol Med. 2019;133: 169–78. [DOI] [PubMed] [Google Scholar]
- 185. Voutilainen MH, Bäck S, Pörsti E, Toppinen L, Lindgren L, Lindholm P, Peränen J, Saarma M, Tuominen RK. Mesencephalic astrocyte-derived neurotrophic factor is neurorestorative in rat model of Parkinson’s disease. J Neurosci. 2009;29(30):9651–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 186. Booth HDE, Hirst WD, Wade-Martins R. The role of astrocyte dysfunction in Parkinson’s disease pathogenesis. Trends Neurosci. 2017;40(6):358–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 187. Andrejack J, Mathur S. What people with Parkinson’s disease want. J Parkinsons Dis. 2020;10:S5–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188. Braak H, Braak E. Staging of Alzheimer’s disease-related neurofibrillary changes. Neurobiol Aging. 1995;16(3):271–78. [DOI] [PubMed] [Google Scholar]
- 189. Braak H, Braak E. Evolution of the neuropathology of Alzheimer’s disease. Acta Neurol Scand. 1996;165:3–12. [DOI] [PubMed] [Google Scholar]
- 190. Kumar A, Singh A, Ekavali. A review on Alzheimer’s disease pathophysiology and its management: an update. Pharmacol Rep. 2015;67(2):195–203. [DOI] [PubMed] [Google Scholar]
- 191. Neary D, Snowden J, Mann D. Frontotemporal dementia. Lancet Neurol. 2005;4(11):771–80. [DOI] [PubMed] [Google Scholar]
- 192. Spillantini MG, Goedert M. Tau pathology and neurodegeneration. Lancet Neurol. 2013;12(6):609–22. [DOI] [PubMed] [Google Scholar]
- 193. Irwin DJ, Hurtig HI. The contribution of tau, amyloid-beta and alpha-synuclein pathology to dementia in Lewy body disorders. J Alzheimer’s Dis Park. 2018;8(4):444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194. Compta Y, Parkkinen L, O’Sullivan SS, Vandrovcova J, Holton JL, Collins C, Lashley T, Kallis C, Williams DR, De Silva R, Lees AJ, et al. Lewy- and Alzheimer-type pathologies in Parkinson’s disease dementia: which is more important? Brain. 2011;134(Pt 5):1493–505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195. Clinton LK, Blurton-Jones M, Myczek K, Trojanowski JQ, LaFerla FM. Synergistic interactions between Aβ, tau, and α-synuclein: acceleration of neuropathology and cognitive decline. J Neurosci. 2010;30(21):7281–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196. Marsh SE, Blurton-Jones M. Examining the mechanisms that link β-amyloid and α-synuclein pathologies. Alzheimer’s Res Ther. 2012;4(2):11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 197. Ballard C, Gauthier S, Corbett A, Brayne C, Aarsland D, Jones E. Alzheimer’s disease. Lancet. 2011;377(9770):1019–31. [DOI] [PubMed] [Google Scholar]
- 198. Qiu C, Kivipelto M, Von Strauss E. Epidemiology of Alzheimer’s disease: occurrence, determinants, and strategies toward intervention. Dialogues Clin Neurosci. 2009;11(2): 111–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199. Masters CL, Bateman R, Blennow K, Rowe CC, Sperling RA, Cummings JL. Alzheimer’s disease. Nat Rev Dis Prim. 2015;1:15056. [DOI] [PubMed] [Google Scholar]
- 200. Van Cauwenberghe C, Van Broeckhoven C, Sleegers K. The genetic landscape of Alzheimer disease: clinical implications and perspectives. Genet Med. 2016;18(5):421–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 201. Gatz M, Reynolds CA, Fratiglioni L, Johansson B, Mortimer JA, Berg S, Fiske A, Pedersen NL. Role of genes and environments for explaining Alzheimer disease. Arch Gen Psychiatry. 2006;63(2):168–74. [DOI] [PubMed] [Google Scholar]
- 202. Riedel BC, Thompson PM, Brinton RD. Age, APOE and sex: triad of risk of Alzheimer’s disease. J Steroid Biochem Mol Biol. 2016;160:134–47. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203. Sun Y, Wu S, Bu G, Onifade MK, Patel SN, LaDu MJ, Fagan AM, Holtzman DM. Glial fibrillary acidic protein-apolipoprotein E (apoE) transgenic mice: astrocyte-specific expression and differing biological effects of astrocyte- secreted apoE3 and apoE4 lipoproteins. J Neurosci. 1998;18(9):3261–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 204. Yiannopoulou KG, Papageorgiou SG. Current and future treatments for Alzheimer’s disease. Ther Adv Neurol Disord. 2013;6(1):19–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 205. Esposito G, Sarnelli G, Capoccia E, Cirillo C, Pesce M, Lu J, Calì G, Cuomo R, Steardo L. Autologous transplantation of intestine-isolated glia cells improves neuropathology and restores cognitive deficits in β amyloid-induced neurodegeneration. Sci Rep. 2016;6:22605. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 206. Hampton DW, Webber DJ, Bilican B, Goedert M, Spillantini MG, Chandran S. Cell-mediated neuroprotection in a mouse model of human tauopathy. J Neurosci. 2010;30(30):9973–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 207. Osborn LM, Kamphuis W, Wadman WJ, Hol EM. Astrogliosis: an integral player in the pathogenesis of Alzheimer’s disease. Prog Neurobiol. 2016;144:121–41. [DOI] [PubMed] [Google Scholar]
- 208. Yeh CY, Vadhwana B, Verkhratsky A, Rodríguez JJ. Early astrocytic atrophy in the entorhinal cortex of a triple transgenic animal model of Alzheimer’s disease. ASN Neuro. 2011;3(5):271–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209. Olabarria M, Noristani HN, Verkhratsky A, Rodríguez JJ. Concomitant astroglial atrophy and astrogliosis in a triple transgenic animal model of Alzheimer’s disease. Glia. 2010;58(7):831–38. [DOI] [PubMed] [Google Scholar]
- 210. Hsu ET, Gangolli M, Su S, Holleran L, Stein TD, Alvarez VE, McKee AC, Schmidt RE, Brody DL. Astrocytic degeneration in chronic traumatic encephalopathy. Acta Neuropathol. 2018;136(6):955–72. [DOI] [PubMed] [Google Scholar]
- 211. Orre M, Kamphuis W, Osborn LM, Jansen AHP, Kooijman L, Bossers K, Hol EM. Isolation of glia from Alzheimer’s mice reveals inflammation and dysfunction. Neurobiol Aging. 2014;35(12):2746–60. [DOI] [PubMed] [Google Scholar]
- 212. Qi L, Cui EH, Ji CM, Zhang XB, Wang ZA, Sun YZ, Xu JC, Zhai XF, Chen ZJ, Li J, Zheng JY, et al. Specific knockdown of hippocampal astroglial EphB2 improves synaptic function via inhibition of D-serine secretion in APP/PS1 mice. Am J Transl Res. 2019;11(2):1073–83. [PMC free article] [PubMed] [Google Scholar]
- 213. Serrano-Pozo A, Mielke ML, Gómez-Isla T, Betensky RA, Growdon JH, Frosch MP, Hyman BT. Reactive glia not only associates with plaques but also parallels tangles in Alzheimer’s disease. Am J Pathol. 2011;179(3):1373–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214. Ceyzériat K, Ben Haim L, Denizot A, Pommier D, Matos M, Guillemaud O, Palomares MA, Abjean L, Petit F, Gipchtein P, Gaillard MC, et al. Modulation of astrocyte reactivity improves functional deficits in mouse models of Alzheimer’s disease. Acta Neuropathol Commun. 2018;6(1):104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 215. Beauquis J, Vinuesa A, Pomilio C, Pavía P, Galván V, Saravia F. Neuronal and glial alterations, increased anxiety, and cognitive impairment before hippocampal amyloid deposition in PDAPP mice, model of Alzheimer’s disease. Hippocampus. 2014;24(3):257–69. [DOI] [PubMed] [Google Scholar]
- 216. Zhao J, O’Connor T, Vassar R. The contribution of activated astrocytes to Aβ production: implications for Alzheimer’s disease pathogenesis. J Neuroinflammation. 2011;8:150. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217. Roßner S, Lange-Dohna C, Zeitschel U, Perez-Polo JR. Alzheimer’s disease β-secretase BACE1 is not a neuron-specific enzyme. J Neurochem. 2005;92(2):226–34. [DOI] [PubMed] [Google Scholar]
- 218. 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 Dis. 2018;65(4):1109–24. [DOI] [PubMed] [Google Scholar]
- 219. Zhong L, Wang Z, Wang D, Wang Z, Martens YA, Wu L, Xu Y, Wang K, Li J, Huang R, Can D, et al. Amyloid-beta modulates microglial responses by binding to the triggering receptor expressed on myeloid cells 2 (TREM2). Mol Neurodegener. 2018;13(1):15. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220. Muñoz Y, Paula-Lima AC, Núñez MT. Reactive oxygen species released from astrocytes treated with amyloid beta oligomers elicit neuronal calcium signals that decrease phospho-Ser727-STAT3 nuclear content. Free Radic Biol Med. 2018;117:132–44. [DOI] [PubMed] [Google Scholar]
- 221. Hallmann AL, Araúzo-Bravo MJ, Mavrommatis L, Ehrlich M, Röpke A, Brockhaus J, Missler M, Sterneckert J, Schöler HR, Kuhlmann T, Zaehres H, et al. Astrocyte pathology in a human neural stem cell model of frontotemporal dementia caused by mutant TAU protein. Sci Rep. 2017;7:42991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222. Sekar S, McDonald J, Cuyugan L, Aldrich J, Kurdoglu A, Adkins J, Serrano G, Beach TG, Craig DW, Valla J, Reiman EM, et al. Alzheimer’s disease is associated with altered expression of genes involved in immune response and mitochondrial processes in astrocytes. Neurobiol Aging. 2015;36(2):583–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223. Li MZ, Zheng LJ, Shen J, Li XY, Zhang Q, Bai X, Wang QS, Ji JG. SIRT1 facilitates amyloid beta peptide degradation by upregulating lysosome number in primary astrocytes. Neural Regen Res. 2018;13(11):2005–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224. Prasad H, Rao R. Amyloid clearance defect in ApoE4 astrocytes is reversed by epigenetic correction of endosomal pH. Proc Natl Acad Sci U S A. 2018;115(28):E6640–49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225. Kimura N, Takahashi M, Tashiro T, Terao K. Amyloid β up-regulates brain-derived neurotrophic factor production from astrocytes: rescue from amyloid β-related neuritic degeneration. J Neurosci Res. 2006;84(4):782–89. [DOI] [PubMed] [Google Scholar]
- 226. Kuan WL, Kasis A, Yuan Y, Mason SL, Lazar AS, Barker RA, Goncalves J. Modelling the natural history of Huntington’s disease progression. J Neurol Neurosurg Psychiatry. 2015;86(10):1143–49. [DOI] [PubMed] [Google Scholar]
- 227. Waldvogel HJ, Kim EH, Tippett LJ, Vonsattel JPG, Faull RLM. The neuropathology of Huntington’s disease. Curr Top Behav Neurosci. 2015;22:33–80. [DOI] [PubMed] [Google Scholar]
- 228. Harris KL, Kuan WL, Mason SL, Barker RA. Antidopaminergic treatment is associated with reduced chorea and irritability but impaired cognition in Huntington’s disease (Enroll-HD). J Neurol Neurosurg Psychiatry. 2020;91:622–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229. Wyant KJ, Ridder AJ, Dayalu P. Huntington’s disease—update on treatments. Curr Neurol Neurosci Rep. 2017;17(4):33. [DOI] [PubMed] [Google Scholar]
- 230. Carroll JB, Bates GP, Steffan J, Saft C, Tabrizi SJ. Treating the whole body in Huntington’s disease. Lancet Neurol. 2015;14(11):1135–42. [DOI] [PubMed] [Google Scholar]
- 231. Shin JY, Fang ZH, Yu ZX, Wang CE, Li SH, Li XJ. Expression of mutant huntingtin in glial cells contributes to neuronal excitotoxicity. J Cell Biol. 2005;171(6):1001–12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232. Tydlacka S, Wang CE, Wang X, Li S, Li XJ. Differential activities of the ubiquitin-proteasome system in neurons versus glia may account for the preferential accumulation of misfolded proteins in neurons. J Neurosci. 2008;28(49):13285–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233. Wang CE, Tydlacka S, Orr AL, Yang SH, Graham RK, Hayden MR, Li S, Chan AWS, Li XJ. Accumulation of N-terminal mutant huntingtin in mouse and monkey models implicated as a pathogenic mechanism in Huntington’s disease. Hum Mol Genet. 2008;17(17):2738–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234. Bradford J, Shin JY, Roberts M, Wang CE, Li XJ, Li S. Expression of mutant huntingtin in mouse brain astrocytes causes age-dependent neurological symptoms. Proc Natl Acad Sci U S A. 2009;106(52):22480–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 235. Juopperi TA, Kim WR, Chiang CH, Yu H, Margolis RL, Ross CA, Ming GL, Song H. Astrocytes generated from patient induced pluripotent stem cells recapitulate features of Huntington’s disease patient cells. Mol Brain. 2012;5:17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236. Osipovitch M, Asenjo Martinez A, Mariani JN, Cornwell A, Dhaliwal S, Zou L, Chandler-Militello D, Wang S, Li X, Benraiss SJ, Agate R, et al. Human ESC-derived chimeric mouse models of Huntington’s disease reveal cell-intrinsic defects in glial progenitor cell differentiation. Cell Stem Cell. 2019;24(1):107–122.e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 237. Wood TE, Barry J, Yang Z, Cepeda C, Levine MS, Gray M. Mutant huntingtin reduction in astrocytes slows disease progression in the BACHD conditional Huntington’s disease mouse model. Hum Mol Genet. 2019;28(3):487–500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238. Benraiss A, Wang S, Herrlinger S, Li X, Chandler-Militello D, Mauceri J, Burm HB, Toner M, Osipovitch M, Jim Xu Q, Ding F, et al. Human glia can both induce and rescue aspects of disease phenotype in Huntington disease. Nat Commun. 2016;7:11758. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239. Tong X, Ao Y, Faas GC, Nwaobi SE, Xu J, Haustein MD, Anderson MA, Mody I, Olsen ML, Sofroniew MV, Khakh BS. Astrocyte Kir4.1 ion channel deficits contribute to neuronal dysfunction in Huntington’s disease model mice. Nat Neurosci. 2014;17(5):694–703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240. Jiang R, Diaz-Castro B, Looger LL, Khakh BS. Dysfunctional calcium and glutamate signaling in striatal astrocytes from Huntington’s disease model mice. J Neurosci. 2016;36(12): 3453–70. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241. Estrada-Sánchez AM, Montiel T, Segovia J, Massieu L. Glutamate toxicity in the striatum of the R6/2 Huntington’s disease transgenic mice is age-dependent and correlates with decreased levels of glutamate transporters. Neurobiol Dis. 2009;34(1):78–86. [DOI] [PubMed] [Google Scholar]
- 242. Liévens JC, Woodman B, Mahal A, Spasic-Boscovic O, Samuel D, Kerkerian-Le Goff L, Bates GP. Impaired glutamate uptake in the R6 Huntington’s disease transgenic mice. Neurobiol Dis. 2001;8(5):807–21. [DOI] [PubMed] [Google Scholar]
- 243. Faideau M, Kim J, Cormier K, Gilmore R, Welch M, Auregan G, Dufour N, Guillermier M, Brouillet E, Hantraye P, DéGlon N, et al. In vivo expression of polyglutamine-expanded huntingtin by mouse striatal astrocytes impairs glutamate transport: a correlation with Huntington’s disease subjects. Hum Mol Genet. 2010;19(15):3053–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244. Chen LL, Wu JC, Wang LH, Wang J, Qin ZH, Difiglia M, Lin F. Rapamycin prevents the mutant huntingtin-suppressed GLT-1 expression in cultured astrocytes. Acta Pharmacol Sin. 2012;33(3):385–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245. Hsiao HY, Chen YC, Chen HM, Tu PH, Chern Y. A critical role of astrocyte-mediated nuclear factor-κB-dependent inflammation in Huntington’s disease. Hum Mol Genet. 2013;22(9):1826–42. [DOI] [PubMed] [Google Scholar]
- 246. Haim L, Ben Ceyzériat K, Sauvage MAC, de Aubry F, Auregan G, Guillermier M, Ruiz M, Petit F, Houitte D, Faivre E, Vandesquille M, et al. The JAK/STAT3 pathway is a common inducer of astrocyte reactivity in Alzheimer’s and Huntington’s diseases. J Neurosci. 2015;35(6):2817–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247. Abjean L, Haim L, Ben Sauvage MAC, Hérard A-S, Derbois C, Petit F, Gipstein P, Guillermier M, Gaudin M, Bernier S, Dufour N, et al. A42 Reactive astrocytes promote proteostasis in huntington’s disease. J Neurol Neurosurg Psychiatry. 2018;89:A15. [Google Scholar]
- 248. Khakh BS, Beaumont V, Cachope R, Munoz-Sanjuan I, Goldman SA, Grantyn R. Unravelling and exploiting astrocyte dysfunction in Huntington’s disease. Trends Neurosci. 2017;40(7):422–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249. Steele AD, Jackson WS, King OD, Lindquist S. The power of automated high-resolution behavior analysis revealed by its application to mouse models of Huntington’s and prion diseases. Proc Natl Acad Sci U S A. 2007;104(6):1983–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250. Oliveira JMA. Mitochondrial bioenergetics and dynamics in Huntington’s disease: tripartite synapses and selective striatal degeneration. J Bioenerg Biomembr. 2010;42(3):227–34. [DOI] [PubMed] [Google Scholar]
- 251. Wright DJ, Renoir T, Smith ZM, Frazier AE, Francis PS, Thorburn DR, McGee SL, Hannan AJ, Gray LJ. N-Acetylcysteine improves mitochondrial function and ameliorates behavioral deficits in the R6/1 mouse model of Huntington’s disease. Transl Psychiatry. 2015;5(1):e492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 252. Diaz-Castro B, Gangwani MR, Yu X, Coppola G, Khakh BS. Astrocyte molecular signatures in Huntington’s disease. Sci Transl Med. 2019;11(514):eaaw8546. [DOI] [PubMed] [Google Scholar]
- 253. Al-Dalahmah O, Sosunov AA, Shaik A, Ofori K, Liu Y, Vonsattel JP, Adorjan I, Menon V, Goldman JE. Single-nucleus RNA-seq identifies Huntington disease astrocyte states. Acta Neuropathol Commun. 2020;8(1):19. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254. Wyndaele M, Wyndaele JJ. Incidence, prevalence and epidemiology of spinal cord injury: what learns a worldwide literature survey? Spinal Cord. 2006;44(9):523–29. [DOI] [PubMed] [Google Scholar]
- 255. Singh A, Tetreault L, Kalsi-Ryan S, Nouri A, Fehlings MG. Global prevalence and incidence of traumatic spinal cord injury. Clin Epidemiol. 2014;6:309–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 256. Badhiwala JH, Wilson JR, Fehlings MG. Global burden of traumatic brain and spinal cord injury. Lancet Neurol. 2019;18(1):24–25. [DOI] [PubMed] [Google Scholar]
- 257. Wu J, Stoica BA, Luo T, Sabirzhanov B, Zhao Z, Guanciale K, Nayar SK, Foss CA, Pomper MG, Faden AI. Isolated spinal cord contusion in rats induces chronic brain neuroinflammation, neurodegeneration, and cognitive impairment: involvement of cell cycle activation. Cell Cycle. 2014;13(15): 2446–58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258. Wu J, Zhao Z, Sabirzhanov B, Stoica BA, Kumar A, Luo T, Skovira J, Faden AI. Spinal cord injury causes brain inflammation associated with cognitive and affective changes: role of cell cycle pathways. J Neurosci. 2014;34(33):10989–1006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259. Davies JE, Huang C, Proschel C, Noble M, Mayer-Proschel M, Davies SJA. Astrocytes derived from glial-restricted precursors promote spinal cord repair. J Biol. 2006;5(3):7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260. Davies JE, Pröschel C, Zhang N, Noble M, Mayer-Pröschel M, Davies SJA. Transplanted astrocytes derived from BMP- or CNTF-treated glial-restricted precursors have opposite effects on recovery and allodynia after spinal cord injury. J Biol. 2008;19(7):24. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261. Davies SJA, Shih C-HH, Noble M, Mayer-Proschel M, Davies JE, Proschel C. Transplantation of Specific human astrocytes promotes functional recovery after spinal cord injury. PLoS ONE. 2011;6(3):e17328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262. Chu T, Zhou H, Li F, Wang T, Lu L, Feng S. Astrocyte transplantation for spinal cord injury: current status and perspective. Brain Res Bull. 2014;107:18–30. [DOI] [PubMed] [Google Scholar]
- 263. Kliot M, Smith GM, Siegal JD, Silver J. Astrocyte-polymer implants promote regeneration of dorsal root fibers into the adult mammalian spinal cord. Exp Neurol. 1990;109(1):57–69. [DOI] [PubMed] [Google Scholar]
- 264. Bernstein JJ, Goldberg WJ. Grafted fetal astrocyte migration can prevent host neuronal atrophy: comparison of astrocytes from cultures and whole piece donors. Restor Neurol Neurosci. 1991;2(4):261–70. [DOI] [PubMed] [Google Scholar]
- 265. Wang JJ, Chuah MI, Yew DTW, Leung PC, Tsang DSC. Effects of astrocyte implantation into the hemisected adult rat spinal cord. Neuroscience. 1995;65(4):973–81. [DOI] [PubMed] [Google Scholar]
- 266. Olby NJ, Blakemore WF. Reconstruction of the glial environment of a photochemically induced lesion in the rat spinal cord by transplantation of mixed glial cells. J Neurocytol. 1996;25(8):481–98. [DOI] [PubMed] [Google Scholar]
- 267. Joosten EAJ, Veldhuis WB, Hamers FPT. Collagen containing neonatal astrocytes stimulates regrowth of injured fibers and promotes modest locomotor recovery after spinal cord injury. J Neurosci Res. 2004;77(1):127–42. [DOI] [PubMed] [Google Scholar]
- 268. Pencalet P, Serguera C, Corti O, Privat A, Mallet J, Giménez Y, Ribotta M. Integration of genetically modified adult astrocytes into the lesioned rat spinal cord. J Neurosci Res. 2006;83(1):61–67. [DOI] [PubMed] [Google Scholar]
- 269. Hayashi K, Hashimoto M, Koda M, Naito AT, Murata A, Okawa A, Takahashi K, Yamazaki M. Increase of sensitivity to mechanical stimulus after transplantation of murine induced pluripotent stem cell-derived astrocytes in a rat spinal cord injury model: laboratory investigation. J Neurosurg Spine. 2011;15(6):582–93. [DOI] [PubMed] [Google Scholar]
- 270. Jin Y, Neuhuber B, Singh A, Bouyer J, Lepore A, Bonner J, Himes T, Campanelli JT, Fischer I. Transplantation of human glial restricted progenitors and derived astrocytes into a contusion model of spinal cord injury. J Neurotrauma. 2011;28(4):579–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271. Haas C, Neuhuber B, Yamagami T, Rao M, Fischer I. Phenotypic analysis of astrocytes derived from glial restricted precursors and their impact on axon regeneration. Exp Neurol. 2012;233(2):717–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272. Fan C, Zheng Y, Cheng X, Qi X, Bu P, Luo X, Kim DH, Cao Q. Transplantation of D15A-expressing glial-restricted-precursor-derived astrocytes improves anatomical and locomotor recovery after spinal cord injury. Int J Biol Sci. 2013;9(1):78–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273. Haas C, Fischer I. Human astrocytes derived from glial restricted progenitors support regeneration of the injured spinal cord. J Neurotrauma. 2013;30(12):1035–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274. Wu L, Li JJ, Chen L, Zhang H, Yuan L, Davies SJA. Combined transplantation of GDAs(BMP) and hr-decorin in spinal cord contusion repair. Neural Regen Res. 2013;8(24):2236–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275. Shih CH, Lacagnina M, Leuer-Bisciotti K, Pröschel C. Astroglial-derived periostin promotes axonal regeneration after spinal cord injury. J Neurosci. 2014;34(7):2438–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276. Shi W, Huang CJ, Xu XD, Jin GH, Huang RQ, Huang JF, Chen YN, Ju SQ, Wang Y, Shi YW, Qin JB, et al. Transplantation of RADA16-BDNF peptide scaffold with human umbilical cord mesenchymal stem cells forced with CXCR4 and activated astrocytes for repair of traumatic brain injury. Acta Biomater. 2016;45:247–61. [DOI] [PubMed] [Google Scholar]
- 277. Becerra GD, Tatko LM, Pak ES, Murashov AK, Hoane MR. Transplantation of GABAergic neurons but not astrocytes induces recovery of sensorimotor function in the traumatically injured brain. Behav Brain Res. 2007;179(1):118–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278. Quan FS, Chen J, Zhong Y, Ren WZ. Comparative effect of immature neuronal or glial cell transplantation on motor functional recovery following experimental traumatic brain injury in rats. Exp Ther Med. 2016;12(3):1671–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279. Anderson MA, Burda JE, Ren Y, Ao Y, O’Shea TM, Kawaguchi R, Coppola G, Khakh BS, Deming TJ, Sofroniew MV. Astrocyte scar formation aids central nervous system axon regeneration. Nature. 2016;532(7598):195–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 280. White RE, Jakeman LB. Don’t fence me in: harnessing the beneficial roles of astrocytes for spinal cord repair. Restor Neurol Neurosci. 2008;26(2–3):197–214. [PMC free article] [PubMed] [Google Scholar]
- 281. Kerschensteiner M, Schwab ME, Lichtman JW, Misgeld T. In vivo imaging of axonal degeneration and regeneration in the injured spinal cord. Nat Med. 2005;11(5):572–77. [DOI] [PubMed] [Google Scholar]
- 282. Becerra JL, Puckett WR, Hiester ED, Quencer RM, Marcillo AE, Post MJD, Bunge RP. MR-pathologic comparisons of wallerian degeneration in spinal cord injury. Am J Neuroradiol. 1995;16(1):125–33. [PMC free article] [PubMed] [Google Scholar]
- 283. Coleman MP, Perry VH. Axon pathology in neurological disease: a neglected therapeutic target. Trends Neurosci. 2002;25(10):532–37. [DOI] [PubMed] [Google Scholar]
- 284. Hu J, Zhang G, Rodemer W, Jin LQ, Shifman M, Selzer ME. The role of RhoA in retrograde neuronal death and axon regeneration after spinal cord injury. Neurobiol Dis. 2017;98:25–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 285. Rodemer W, Selzer ME. Role of axon resealing in retrograde neuronal death and regeneration after spinal cord injury. Neural Regen Res. 2019;14(3):399–404. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 286. Ishibashi T, Dakin KA, Stevens B, Lee PR, Kozlov SV, Stewart CL, Fields RD. Astrocytes promote myelination in response to electrical impulses. Neuron. 2006;49(6):823–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 287. Kotter MR, Li W-W, Zhao C, Franklin RJM. Myelin impairs CNS remyelination by inhibiting oligodendrocyte precursor cell differentiation. J Neurosci. 2006;26(1):328–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288. Goldshmit Y, McLenachan S, Turnley A. Roles of Eph receptors and ephrins in the normal and damaged adult CNS. Brain Res Rev. 2006;52(2):327–45. [DOI] [PubMed] [Google Scholar]
- 289. Peng W, Cotrina ML, Han X, Yu H, Bekar L, Blum L, Takano T, Tian GF, Goldman SA, Nedergaard M. Systemic administration of an antagonist of the ATP-sensitive receptor P2X7 improves recovery after spinal cord injury. Proc Natl Acad Sci U S A. 2009;106(30):12489–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 290. Park E, Velumian AA, Fehlings MG. The role of excitotoxicity in secondary mechanisms of spinal cord injury: a review with an emphasis on the implications for white matter degeneration. J Neurotrauma. 2004;21(6):754–74. [DOI] [PubMed] [Google Scholar]
- 291. Bains M, Hall ED. Antioxidant therapies in traumatic brain and spinal cord injury. Biochim Biophys Acta. 2012;1822(5): 675–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292. Krencik R, Weick JP, Liu Y, Zhang ZJ, Zhang SC. Specification of transplantable astroglial subtypes from human pluripotent stem cells. Nat Biotechnol. 2011;29(6):528–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 293. Rolls A, Shechter R, London A, Segev Y, Jacob-Hirsch J, Amariglio N, Rechavi G, Schwartz M. Two faces of chondroitin sulfate proteoglycan in spinal cord repair: a role in microglia/macrophage activation. PLoS Med. 2008;5(8):e171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294. Rolls A, Shechter R, Schwartz M. The bright side of the glial scar in CNS repair. Nat Rev Neurosci. 2009;10(3):235–41. [DOI] [PubMed] [Google Scholar]
- 295. Flynn RWV, MacWalter RSM, Doney ASF. The cost of cerebral ischaemia. Neuropharmacology. 2008;55(3):250–56. [DOI] [PubMed] [Google Scholar]
- 296. Béjot Y, Bailly H, Durier J, Giroud M. Epidemiology of stroke in Europe and trends for the 21st century. Press Med. 2016;45(12, Pt 2):e391–98. [DOI] [PubMed] [Google Scholar]
- 297. Appelros P, Stegmayr B, Terént A. Sex differences in stroke epidemiology: a systematic review. Stroke. 2009;40(4):1082–90. [DOI] [PubMed] [Google Scholar]
- 298. Smajlović D. Strokes in young adults: epidemiology and prevention. Vasc Health Risk Manag. 2015;11:157–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 299. O’Donnell MJ, Denis X, Liu L, Zhang H, Chin SL, Rao-Melacini P, Rangarajan S, Islam S, Pais P, McQueen MJ, Mondo C, et al. Risk factors for ischaemic and intracerebral haemorrhagic stroke in 22 countries (the INTERSTROKE study): a case-control study. Lancet. 2010;376(9735):112–23. [DOI] [PubMed] [Google Scholar]
- 300. Cai Q, Chen Z, Song P, Wu L, Wang L, Deng G, Liu B, Chen Q. Co-transplantation of hippocampal neural stem cells and astrocytes and microvascular endothelial cells improve the memory in ischemic stroke rat. Int J Clin Exp Med. 2015;8(8):13109–17. [PMC free article] [PubMed] [Google Scholar]
- 301. Becerra-Calixto A, Cardona-Gómez GP. Neuroprotection induced by transplanted CDK5 knockdown astrocytes in global cerebral ischemic rats. Mol Neurobiol. 2017;54(9):6681–96. [DOI] [PubMed] [Google Scholar]
- 302. Becerra-Calixto A, Posada-Duque R, Cardona-Gómez GP. Recovery of neurovascular unit integrity by CDK5-KD astrocyte transplantation in a global cerebral ischemia model. Mol Neurobiol. 2018;55(11):8563–85. [DOI] [PubMed] [Google Scholar]
- 303. Su SC, Tsai L-H. Cyclin-dependent kinases in brain development and disease. Annu Rev Cell Dev Biol. 2011;27:465–91. [DOI] [PubMed] [Google Scholar]
- 304. Meyer DA, Torres-Altoro MI, Tan Z, Tozzi A, Filippo M, Di DiNapoli V, Plattner F, Kansy JW, Benkovic SA, Huber JD, Miller DB, et al. Ischemic stroke injury is mediated by aberrant Cdk5. J Neurosci. 2014;34(24):8259–67. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 305. Butenko O, Dzamba D, Benesova J, Honsa P, Benfenati V, Rusnakova V, Ferroni S, Anderova M. The increased activity of TRPV4 channel in the astrocytes of the adult rat hippocampus after cerebral hypoxia/ischemia. PLoS ONE. 2012;7(6):e39959. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 306. Zanotti S, Charles A. Extracellular calcium sensing by glial cells: low extracellular calcium induces intracellular calcium release and intercellular signaling. J Neurochem. 1997;69(2):594–602. [DOI] [PubMed] [Google Scholar]
- 307. Tanaka M, Kawahara K, Kosugi T, Yamada T, Mioka T. Changes in the spontaneous calcium oscillations for the development of the preconditioning-induced ischemic tolerance in neuron/astrocyte co-culture. Neurochem Res. 2007;32(6):988–1001. [DOI] [PubMed] [Google Scholar]
- 308. Chen Y, Mancuso J, Zhao Z, Li X, Cheng J, Roman G, Wong STC. Vasodilation by in vivo activation of astrocyte endfeet via two-photon calcium uncaging as a strategy to prevent brain ischemia. J Biomed Opt. 2013;18(12):126012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 309. Gunnarson E, Song Y, Kowalewski JM, Brismar H, Brines M, Cerami A, Andersson U, Zelenina M, Aperia A. Erythropoietin modulation of astrocyte water permeability as a component of neuroprotection. Proc Natl Acad Sci U S A. 2009;106(5):1602–607. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 310. Vanzulli I, Butt AM. mGluR5 protect astrocytes from ischemic damage in postnatal CNS white matter. Cell Calcium. 2015;58(5):423–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 311. Ashpole NM, Chawla AR, Martin MP, Brustovetskys T, Brustovetskys N, Hudmon A. Loss of calcium/calmodulin-dependent protein kinase II activity in cortical astrocytes decreases glutamate uptake and induces neurotoxic release of ATP. J Biol Chem. 2013;288(20):14599–611. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 312. Krebs C, Fernandes HB, Sheldon C, Raymond LA, Baimbridge KG. Functional NMDA receptor subtype 2B is expressed in astrocytes after ischemia in vivo and anoxia in vitro. J Neurosci. 2003;23(8):3364–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 313. Dugan LL, Kim-Han JS. Astrocyte mitochondria in in vitro models of ischemia. J Bioenerg Biomembr. 2004;36(4):317–21. [DOI] [PubMed] [Google Scholar]
- 314. Bambrick L, Kristian T, Fiskum G. Astrocyte mitochondrial mechanisms of ischemic brain injury and neuroprotection. Neurochem Res. 2004;29(3):601–608. [DOI] [PubMed] [Google Scholar]
- 315. Nahirney PC, Reeson P, Brown CE. Ultrastructural analysis of blood-brain barrier breakdown in the peri-infarct zone in young adult and aged mice. J Cereb Blood Flow Metab. 2016;36(2):413–25. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 316. Ouyang Y-B, Xu L, Liu S, Giffard RG. Role of astrocytes in delayed neuronal death: GLT-1 and its novel regulation by MicroRNAs. Adv Neurobiol. 2014;11:171–88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 317. Ouyang Y-B, Voloboueva LA, Xu L, Giffard RG. Selective dysfunction of hippocampal CA1 astrocytes contributes to delayed neuronal damage after transient forebrain ischemia. J Neurosci. 2007;27(16):4253–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 318. Zvalova D, Cordier J, Mesnil M, Junier MP, Chneiweiss H. p38/SAPK2 controls gap junction closure in astrocytes. Glia. 2004;46(3):323–33. [DOI] [PubMed] [Google Scholar]
- 319. Gao Q, Katakowski M, Chen X, Li Y, Chopp M. Human marrow stromal cells enhance connexin43 gap junction intercellular communication in cultured astrocytes. Cell Transplant. 2005;14(2–3):109–17. [DOI] [PubMed] [Google Scholar]
- 320. Chen Y, Wang L, Zhang L, Chen B, Yang L, Li X, Li Y, Yu H. Inhibition of connexin 43 hemichannels alleviates cerebral ischemia/reperfusion injury via the TLR4 signaling pathway. Front Cell Neurosci. 2018;12:372. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 321. Ma D, Feng L, Cheng Y, Xin M, You J, Yin X, Hao Y, Cui L, Feng J. Astrocytic gap junction inhibition by carbenoxolone enhances the protective effects of ischemic preconditioning following cerebral ischemia. J Neuroinflammation. 2018;15(1):198. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 322. Yin X, Feng L, Ma D, Yin P, Wang X, Hou S, Hao Y, Zhang J, Xin M, Feng J. Roles of astrocytic connexin-43, hemichannels, and gap junctions in oxygen-glucose deprivation/reperfusion injury induced neuroinflammation and the possible regulatory mechanisms of salvianolic acid B and carbenoxolone. J Neuroinflammation. 2018;15(1):97. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 323. Giffard RG, Swanson RA. Ischemia-induced programmed cell death in astrocytes. Glia. 2005;50(4):299–306. [DOI] [PubMed] [Google Scholar]
- 324. Swanson R, Ying W, Kauppinen T. Astrocyte influences on ischemic neuronal death. Curr Mol Med. 2004;4(2):193–205. [DOI] [PubMed] [Google Scholar]
- 325. Matsushita T, Masaki K, Suzuki S, Matsuoka T, Yonekawa T, Wu XM, Tabira T, Iwaki T, Kira JI. Astrocytopathy in neuromyelitis optica, multiple sclerosis and Baló’s disease. Clin Neurol. 2011;51(11):898–900. [DOI] [PubMed] [Google Scholar]
- 326. Chalazonitis A, Rao M. Enteric nervous system manifestations of neurodegenerative disease. Brain Res. 2018;1693(Pt B): 207–13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 327. Batiuk MY, Martirosyan A, Wahis J, de Vin F, Marneffe C, Kusserow C, Koeppen J, Viana JF, Oliveira JF, Voet T, Ponting CP, et al. Identification of region-specific astrocyte subtypes at single cell resolution. Nat Commun. 2020;11(1):1220. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 328. Morel L, Chiang MSR, Higashimori H, Shoneye T, Iyer LK, Yelick J, Tai A, Yang Y. Molecular and functional properties of regional astrocytes in the adult brain. J Neurosci. 2017; 37(36):8706–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 329. Bradley RA, Shireman J, McFalls C, Choi J, Canfield SG, Dong Y, Liu K, Lisota B, Jones JR, Petersen A, Bhattacharyya A, et al. Regionally specified human pluripotent stem cell-derived astrocytes exhibit different molecular signatures and functional properties. Development. 2019;146(13):dev170910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 330. Molofsky AV, Kelley KW, Tsai H-H, Redmond SA, Chang SM, Madireddy L, Chan JR, Baranzini SE, Ullian EM, Rowitch DH. Astrocyte-encoded positional cues maintain sensorimotor circuit integrity. Nature. 2014;509(7499):189–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 331. Tsai HH, Li H, Fuentealba LC, Molofsky AV, Taveira-Marques R, Zhuang H, Tenney A, Murnen AT, Fancy SP, Merkle F, Kessaris N, Alvarez-Buylla A, et al. Regional astrocyte allocation regulates CNS synaptogenesis and repair. Science. 2012;337(6092):358–62. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 332. Lee SH, Kim WT, Cornell-Bell AH, Sontheimer H. Astrocytes exhibit regional specificity in gap-junction coupling. Glia. 1994;11(4):315–25. [DOI] [PubMed] [Google Scholar]
- 333. Schreyer DJ, Jones EG. Growth of corticospinal axons on prosthetic substrates introduced into the spinal cord of neonatal rats. Brain Res. 1987;432(2):291–99. [DOI] [PubMed] [Google Scholar]
- 334. Qian J, Bull MS, Levitt P. Target-derived astroglia regulate axonal outgrowth in a region-specific manner. Dev Biol. 1992;149(2):278–294. [DOI] [PubMed] [Google Scholar]
- 335. Keirstead HS, Nistor G, Bernal G, Totoiu M, Cloutier F, Sharp K, Steward O. Human embryonic stem cell-derived oligodendrocyte progenitor cell transplants remyelinate and restore locomotion after spinal cord injury. J Neurosci. 2005;25(19):4694–705. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 336. Namekawa M, Takiyama Y, Aoki Y, Takayashiki N, Sakoe K, Shimazaki H, Taguchi T, Tanaka Y, Nishizawa M, Saito K, Matsubara Y, et al. Identification of GFAP gene mutation in hereditary adult-onset Alexander’s disease. Ann Neurol. 2002;52(6):779–85. [DOI] [PubMed] [Google Scholar]
- 337. Han X, Chen M, Wang F, Windrem M, Wang S, Shanz S, Xu Q, Oberheim NA, Bekar L, Betstadt S, Silva AJ, et al. Forebrain engraftment by human glial progenitor cells enhances synaptic plasticity and learning in adult mice. Cell Stem Cell. 2013;12(3):342–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 338. Jeanson T, Pondaven A, Ezan P, Mouthon F, Charvériat M, Giaume C. Antidepressants impact connexin 43 channel functions in astrocytes. Front Cell Neurosci. 2016;9:495. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 339. Windrem MS, Osipovitch M, Liu Z, Bates J, Chandler-Militello D, Zou L, Munir J, Schanz S, McCoy K, Miller RH, Wang S, et al. Human iPSC glial mouse chimeras reveal glial contributions to schizophrenia. Cell Stem Cell. 2017;21(2):195–208.e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 340. Das MM, Avalos P, Suezaki P, Godoy M, Garcia L, Chang CD, Vit JP, Shelley B, Gowing G, Svendsen CN. Human neural progenitors differentiate into astrocytes and protect motor neurons in aging rats. Exp Neurol. 2016;280:41–49. [DOI] [PubMed] [Google Scholar]
- 341. Kriz J, Nguyen MD, Julien JP. Minocycline slows disease progression in a mouse model of amyotrophic lateral sclerosis. Neurobiol Dis. 2002;10(3):268–78. [DOI] [PubMed] [Google Scholar]
- 342. Drachman DB, Frank K, Dykes-Hoberg M, Teismann P, Almer G, Przedborski S, Rothstein JD. Cyclooxygenase 2 inhibition protects motor neurons and prolongs survival in a transgenic mouse model of ALS. Ann Neurol. 2002;52(6):771–78. [DOI] [PubMed] [Google Scholar]