Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 2964 word(s) 2964 2022-03-01 05:10:05 |
2 format correction Meta information modification 2964 2022-03-14 03:04:10 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Milanese, M. Human iPSC-Derived Astrocytes in Neurological Disorders. Encyclopedia. Available online: https://encyclopedia.pub/entry/20501 (accessed on 08 July 2024).
Milanese M. Human iPSC-Derived Astrocytes in Neurological Disorders. Encyclopedia. Available at: https://encyclopedia.pub/entry/20501. Accessed July 08, 2024.
Milanese, Marco. "Human iPSC-Derived Astrocytes in Neurological Disorders" Encyclopedia, https://encyclopedia.pub/entry/20501 (accessed July 08, 2024).
Milanese, M. (2022, March 11). Human iPSC-Derived Astrocytes in Neurological Disorders. In Encyclopedia. https://encyclopedia.pub/entry/20501
Milanese, Marco. "Human iPSC-Derived Astrocytes in Neurological Disorders." Encyclopedia. Web. 11 March, 2022.
Human iPSC-Derived Astrocytes in Neurological Disorders
Edit

Astrocytes, the most-abundant non-neuronal cell population in the central nervous system, play a vital role in these processes. They are involved in various functions in the brain, such as the regulation of synapse formation, neuroinflammation, and lactate and glutamate levels. The development of human-induced pluripotent stem cells (iPSCs) reformed the research in neurodegenerative disorders allowing for the generation of disease-relevant neuronal and non-neuronal cell types that can help in disease modeling, drug screening, and, possibly, cell transplantation strategies.

human-induced pluripotent stem cells neurodegenerative disease iPSC-derived astrocytes disease modeling

1. Introduction

Neurodegenerative diseases involve chronic and progressive damage to neurons, resulting in various complications such as cognitive impairment, memory deficits, changes in perception and mood, and loss of sensitivity and motor abilities [1]. Most neurodegenerative diseases share common pathological pathways such as the over-accumulation of toxic-aggregated proteins, mitochondrial dysfunction, axonal transport defects, excitotoxicity, and chronic inflammation, which lead to neurodegeneration [2]. The increasing incidence of neurodegenerative disorders such as Alzheimer’s disease (AD) [3], amyotrophic lateral sclerosis (ALS) [4], multiple sclerosis (MS) [5], and Parkinson’s disease (PD) [6] represents a significant impact on global health because of the severity of their symptoms and the lack of effective therapies [7].
Due to the complexity of the pathological disease mechanisms and the challenge of accessing human samples, human modeling of neurodegenerative diseases has always been a difficult task. Animal models have commonly been used, due to the intrinsic features of in vivo preclinical experimentation and the possibility to establish ex vivo mechanistic studies. However, animal model studies usually require many animals and take a long time, thus limiting the number of experimental conditions tested [8]. The development of induced pluripotent stem cell (iPSC) allowed the creation of a new scenario, complementing animal disease models of neurodegenerative diseases.
In 2007, Yamanaka and colleagues paved the way for this achievement by describing iPSC technology implemented in mice [9]. They generate iPSCs from embryonic fibroblasts and adult tail-tip fibroblasts of mice using four transcription factors: Octamer-binding transcription factor 3/4 (Oct3/4), sex-determining region Y (Sox2), the master regulator of cell cycle entry and proliferative metabolism (c-Myc), and Krüppel-like factor 4 (Klf4). This protocol took almost 30 days to generate rodent iPSCs and a further 30 days to differentiate them [9]. Subsequent studies showed that human iPSCs could also differentiate into any cell type in the human body; thus, well-established models of human disease, including both developmental and adult-onset diseases, often use iPSCs from patients [10][11]. Due to the rapid growth of discoveries, it is now possible to differentiate iPSCs from patients and healthy donors in almost all neural cells, allowing human neurodegenerative disease modeling with intrinsic advantages over in-vivo models. Advances in iPSC technology are fundamental to adapt astrocyte research to translational studies since human astrocytes derived from fibroblasts of patients match phenotypes of various neurodegenerative diseases such as AD [12][13], ALS [14][15], HD [16], and fragile X syndrome [17]. Nevertheless, establishing iPSC-derived astrocytes as bona fide models of human astrocytes and individual heterogeneity of in vivo astrocytes should be approached with caution since these cells may modify the phenotype according to the culture conditions [18].

2. Astrocytes Phenotypes and Functions

Astrocytes, the most abundant glial cell type in the central nervous system (CNS), play an essential role in neuronal development and function and are very specialized and heterogeneous throughout the CNS. They help maintain synaptogenesis, synaptic plasticity, the extracellular ion concentration, the blood–brain barrier (BBB), promote myelination in the white matter, and support neurons [19]. Apart from their fundamental physiological functions, it is generally agreed that astrocytes play a role in toxic/pathological events and that their dysfunction can generate neurological disorders such as neurodegenerative diseases, neurodevelopmental diseases, epilepsy, and astrogliomas [20]. In classical taxonomy, protoplasmic and fibrous astrocytes are two prominent gray and white matter classes, respectively [21]. Astrocytes form large homologous networks linked by gap junctions and heterologous networks with oligodendrocytes in different brain regions [22][23]. Recent studies showed that astrocytes change their morphology and function depending on age and their location in the brain [24][25][26][27].
Reactive gliosis is defined as changes in the neuroglia morphology, representing an actual reaction rather than a simple indicator of noxious influences on the CNS. Accordingly, the terms “reactive astrogliosis” and “astrocyte reactivity” define a pathological event that determines morphological, biochemical, and metabolic remodeling other than transcriptional regulation, which translates into the gain or loss of homeostatic functions [28]. Thus, morphology does not represent the only way to depict the role of astrocytes in neuropathologies since it does not correlate with functional phenotypes or their ability to impact the biology of other cells. Overall, the impact of reactive astrocytes varies and is complex and may depend on the pathological condition, which can be beneficial in one disease and detrimental in another [29]. The scenario is even more complex due to the presence of reactive microglia. Microglia can shift astrocyte signaling from a physiological to pathological status by increasing the production of factors such as tumor necrosis factor α (TNFα), which in turn has been reported to alter synaptic functions and behavior [30].
Specific molecular profiles and functions characterize the reactivity of astrocytes and their distinct impact on diseases and produce astrocyte phenotypes, representing the unique outcome of a given state. Notably, astrocyte reactivity characterizes pathological contexts while astrocyte activation represents physiological conditions [31]. Astrocyte reactivity is secondary to extrinsic signals and may evolve differently, exhibiting pathological modification and etiological features with a remarkable impact on disease progression [32].
In recent decades, scientists dramatically accelerated their research and deepened the roles of astrocytes, thanks to new interdisciplinary approaches combining omics with physiology and genetics. In this context, transcriptomics and proteomics have shown that in the healthy brain, following embryonic patterning programs or local neuronal cues, astrocytes are specialized to perform specific tasks in distinct CNS circuits [33]. The advent of genetic engineering opened a new research phase based on astrocyte-targeted manipulation, thus extending studies on detrimental astrocyte phenotypes. These studies identified other functional alterations in reactive astrocytes involving neurotransmitter and ion buffering, gap junctions, phagocytic activity, metabolic coupling, and most importantly, increased neuronal death due to the release of toxic factors and not merely the loss of trophic or antioxidant support from astrocytes [30].
This evolution generated the need to unify the nomenclature and refine the concepts that precisely define astrocyte heterogeneity by using a systematic approach to contextualize the contribution of astrocytes to CNS disorders. Recent studies have proposed that mouse astrocytes have two discrete pathological conditions: The A1 neurotoxic phenotype when exposed to specific cytokines secreted by activated microglia and the A2 neuroprotective phenotype when exposed to an experimental paradigm mimicking ischemic stroke [34][35]. However, this binary polarization should be approached cautiously since more recent studies recommend moving beyond the A1–A2 classification [30]. Indeed, only a subset of mixed A1 and A2 or pan-reactive transcripts are up-regulated in astrocytes of human patients or mouse models of chronic diseases or acute CNS injuries [36][37][38][39]. Moreover, advanced multidimensional data and the co-clustering statistical approach revealed that the A1 and A2 transcriptomes represent only two of the many potential astrocyte transcriptomes. As transcriptomic data are a fundamental tool used to characterize the functional diversity of reactive astrocytes, a multidimensional analysis should be applied to establish the uniqueness of astrocyte phenotypes [40].

3. Human iPSC-Derived Astrocytes in Neurological Disorders

The classical drug discovery and validation method has some drawbacks, such as time-consuming procedures, the need for many animals for experiments, and the testing of a limited number of compounds under limited experimental conditions. Recent advancements in the available techniques led to high-throughput screenings, which can test hundreds or even thousands of compounds in less time and with lower relative costs. Although these procedures are more efficient than previous ones, they have poor validation rates due to the rare biological relevance of the screening platforms when related to the disease to treat and the use of non-human derivatives, which may not be related to specific human biology. The great advantage of using iPSC technology in developing treatments for human disease is evident.

3.1. Alzheimer’s Disease

AD is an age-related neurodegenerative disease that mainly affects memory and executive functions, which induce behavioral and neuropsychiatric changes. AD is characterized by the accumulation of amyloid-beta (Aβ) plaques and TAU-laden neurofibrillary tangles [41]. Almost 40 million people worldwide have dementia, most of them older than 60 years. This number is rapidly increasing and is estimated to double by at least 2050 [42], overtaking cancer as the second leading cause of death after cardiovascular diseases [43]. There is no treatment to stop the disease progression; therefore, identifying the exact cause and mechanism is most important.
Astrocytes play an essential role in the pathology of AD. They are reported to be involved in various brain functions and are known to become reactive in different disease pathologies, changing their gene expression profile and metabolism [44]. Astrocytes maintain neuronal excitability and synaptic transmission by regulating ion concentrations. Astrocytes also constitute a significant source of cholesterol and other lipids critical for many cellular functions, as well as lipoproteins such as APOE, which are essential regulators of brain Aβ clearance and degradation [45]. Astrocytes of AD patients have shown increased GFAP expression and gamma-aminobutyric acid (GABA) production and release [46]. Furthermore, Aβ accumulates in AD astrocytes.

3.2. Parkinson’s Disease

PD affects 7 to 10 million people worldwide, making it the second-most prevalent neurodegenerative disease after AD. Most PD cases are sporadic (85%), and only 15% of patients show familial mutations [47]. There are several hypotheses behind PD pathogenesis, including neuroinflammation, mitochondrial dysfunction, dysfunctional protein degradation, and alpha-synuclein (α-synuclein) pathology, but the exact cause is still unknown [48][49][50]. PD is mainly characterized by the loss of ventral midbrain dopaminergic neurons in the substantia nigra pars compacta and the presence of Lewy bodies in the brain. These pathological changes are considered responsible for the typical motor symptoms (bradykinesia, rigidity, rest tremor, and postural instability) seen in PD [51][52]. There is no proper cure for PD, and most interventions are aimed at relieving the motor symptoms with either dopamine replacement therapy or surgery [53].
In the early 1980s, researchers used fetal DA neural cells of human origin in a PD rat model for therapeutic purposes. They demonstrated that PD rats successfully restored functional activity after transplantation [54]. Ten years later, another group found a way to translate this approach to patients. They successfully transplanted human fetal DA neurons in a severe PD case, which engrafted and improved motor functions [55]. With the precedence of previous research, transplantation of iPSC-derived DA neurons is becoming a therapeutic opportunity for PD patients [56]. Some preclinical and clinical studies have used iPSC-derived DA neurons in PD rats and human models, demonstrating that neurons successfully improved motor functions and survival rate for at least two years [57][58]. In 2018, the so-called “Kyoto clinical trial” was started. The authors efficiently induced dopaminergic neurons from human iPSCs and purified dopaminergic progenitor cells by sorting. After in vivo preclinical studies in rats and monkeys, cell-based therapy for PD was started in humans [59]. At present, seven patients have been recruited, and the recruitment is now closed.
Apart from DA neurons, the contribution of glial cells to the pathogenesis of PD has been considered. A study compared the functionality of iPSC-derived astrocytes and dopaminergic neurons obtained from PD patients with the p.Gly2019Ser mutation and healthy controls. They found that astrocytes from PD patients secreted α-synuclein, which exerted a neurotoxic function on surrounding dopaminergic neurons, resulting in neuronal dysfunction. They also found dysfunctional chaperone-mediated autophagy and progressive α-synuclein accumulation in the PD iPSC-derived astrocytes [53].
Transplantation is a growing field in PD involving iPSC-derived dopaminergic neurons. Further, the knowledge of the role of astrocytes is increasing. This occurrence can be essential in identifying pathological astrocytic phenotypes in human stem cell models of familial and sporadic PD.

3.3. Amyotrophic Lateral Sclerosis

Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease characterized by the loss of cortical and spinal cord motor neurons, causing muscle weakness, atrophy, and paralysis, with the death of patients by respiratory failure within 3–5 years after diagnosis [60]. ALS is a rare disease, affecting about 2–3 in 100,000 individuals, and approximately 90% of ALS cases are sporadic (sALS) due to multiple genetic, epigenetic, and environmental factors, while about 10% are familial (fALS), which is clinically indistinguishable from sALS [61]. Mutations in more than 20 genes are associated with fALS, the most important being mutations in the chromosome 9 open reading frame 72 protein (C9orf72), superoxide dismutase type 1 enzyme (SOD1), 43 kDa transactive response-DNA binding protein (TARDBP, TDP43), and fused in sarcoma/translocated in liposarcoma protein (FUS/TLS) [62][63]. ALS is a complex disease due to its multiple causes, also involving non-neuronal cells [64][65]. In particular, astrocytes have a detrimental role in ALS progression [66][67] with several mechanisms, including excitotoxicity, altered astrocyte metabolism, inflammation, and oxidative stress [68][69][70][71].
ALS is a multi-factorial and multi-cellular disease where many genes play a decisive role, thus highlighting the limitations of animal models, each characterized by a specific gene mutation. Having access to samples from patients with different genetic signatures or sporadic is a significant advantage for the knowledge of pathology and stratifying the results based on individual patients.

3.4. Huntington’s Disease

HD is a hereditary, neurodegenerative disease caused by cytosine–adenine–guanine (CAG) repeat expansion in the huntingtin (HTT) gene, which codes for glutamine [72][73]. CAG repeats result in a polyglutamine tract, leading to progressive involuntary motor movements, cognitive disturbances, and dementia [74][75]. HD affects about 12 per 100,000 individuals in the European population [76]. The disease can occur from childhood to old age, with a mean age at onset of 45 years [77], and CAG extension strongly correlates with disease onset [78]. The HTT gene has critical roles in cellular homeostasis processes such as transcription, protein-protein interactions, transport, mitochondrial functions, cellular stress responses, and vesicular trafficking [79]. However, in pathological conditions, a mutation in the HTT gene by CAG expansion in exon 1 results in the expansion of polyglutamine residue at the N-terminus of the HTT protein [72]. A previous study also reported the formation of small oligomeric fragments and protein accumulation in the nucleus, leading to the death of medium spiny neurons (MSNs) in the striatum and other regions [79].
While neuron degeneration plays a critical role in the pathology of HD, glial cells are also involved. Dysfunctional astrocytes in the striatum lead to alteration of astrocytes structure and abnormalities of electrical properties, thereby maintaining ion homeostasis and neuronal signaling through the release of glutamate and adenosine triphosphate [80] and increasing pro-inflammatory and reducing anti-inflammatory cytokine production, as well as the brain-derived neurotrophic factor (BDNF) and Ccl5/RANTES chemokine [81].
From the studies in the literature, it appears that patient-derived astrocytes present apparent pathological features of the disease. Thus, iPSCs-derived astrocytes show promise as a valuable tool for HD modeling and emerge with great potential in approaches to cell replacement therapy.

3.5. Multiple Sclerosis

MS, a chronic, auto-immune, demyelinating CNS disease, is the leading cause of neurological disability in young adults [82][83]. The symptoms start with acute episodes of neurological dysfunction, followed by a relapsing-remitting course (RRMS) [84]. Later, approximately 80% of patients develop secondary progressive MS leading to permanent disability, including limb weakness, sensory loss, vision disturbances, pain, and muscle spasms. The remaining patients directly reach the progressive phase, referred to as primary progressive MS [84][85]. The pathology of MS remains mostly unexplained; however, shreds of evidence suggest that activated immune cells from the periphery migrate to the CNS, forming lesions characterized by primary demyelination and relative preservation of axons [82]. Currently, available disease-modifying treatments are proven to suppress the inflammatory components in RRMS; however, these drugs are practically ineffective in progressive forms [83].
Reactive astrocytes are instrumental in forming MS plaques from the early stages of the disease, recruiting lymphocytes and damaging tissue, confining inflammation, and promoting lesion repair [82][86]. Reactive astrocytes contribute to the various neuroinflammatory responses, including the production of and reactivity to soluble mediators (e.g., cytokines and chemokines), but they also regulate oxidative stress and maintain BBB integrity and function [87]. Moreover, various studies suggest that reactive astrocytes are associated with a detrimental effect by exacerbating inflammation and inhibiting regeneration; meanwhile, they can also contribute to neuroprotection [82][86][87][88]. These results highlight the complex and dual role of astrocyte-mediated regulation during disease progression.
iPSC-derived astrocytes represent an attractive approach to enhancing the knowledge of astrocyte dysfunction contributing to MS etiology [18][82][89]. However, at present, only two studies have been conducted on iPSC-derived astrocytes assessing the risk variants leading to MS susceptibility, with contradictory conclusions [18][89].
Astrocytes can have different roles in MS pathology depending on the pathological stage, the damage mechanisms considered, and the patient. Studies in the literature have not yet elucidated the mechanisms that give astrocytes this dual role. Therefore, the iPSC-derived astrocyte model is a robust platform that can help evaluate these aspects and highligh therapeutic targets capable of modifying the harmful elements of astrocytes in MS or enhancing the positive ones.

3.6. Spinal Muscular Atrophy

Spinal muscular atrophy (SMA) is an autonomous hereditary disease characterized by a loss of motor neurons leading to muscle atrophy, respiratory failure, and death [90][91][92]. SMA is a rare disorder with a frequency of 1:11,000 individuals; however, it is the leading cause of infant death [90][91][92]. SMA is mainly caused by deletion or mutations of the survival motor neuron 1 (SMN1) gene on chromosome 5q, which codes for the SMN protein, found in neuronal and non-neuronal cells, including astrocytes [92][93]. This protein is essential for normal development and functional homeostasis in all species. Humans carry a second, closely related gene, called SMN2, an SMN1 homolog that can also produce the SMN protein. The greater the number of copies of this gene, the milder the SMA [94]. Currently, there are only four approved drugs for SMA: Nusinersen, onasemnogene, abeparvovec, and risdiplam; however, their long-term benefits remain to be assessed [95].
Astrocytes are crucial components contributing to MN damage and loss in SMA [90][92][96]. Restoring SMN selectively in astrocytes using scAAV-SMNgfap viral delivery, Rindt and colleagues [92] observed improved neuromuscular circuitry and an increase in the number and lifespan of neuromuscular junctions. They also hypothesized that astrocyte functions are disrupted in SMA, measured by the increased number of GFAP-positive cells and elevated production of pro-inflammatory cytokines. This increased expression of inflammatory cytokines was partially normalized in treated mice, suggesting that astrocytes contribute to the pathogenesis of SMA [92].

References

  1. Dugger, B.N.; Dickson, D.W. Pathology of neurodegenerative diseases. Cold Spring Harb. Perspect. Biol. 2017, 9, a028035.
  2. Dawson, T.M.; Golde, T.E.; Lagier-Tourenne, C. Animal models of neurodegenerative diseases. Nat. Neurosci. 2018, 21, 1370–1379.
  3. Winblad, B.; Amouyel, P.; Andrieu, S.; Ballard, C.; Brayne, C.; Brodaty, H.; Cedazo-Minguez, A.; Dubois, B.; Edvardsson, D.; Feldman, H.; et al. Defeating Alzheimer′s disease and other dementias: A priority for European science and society. Lancet Neurol. 2016, 15, 455–532.
  4. Arthur, K.C.; Calvo, A.; Price, T.R.; Geiger, J.T.; Chio, A.; Traynor, B.J. Projected increase in amyotrophic lateral sclerosis from 2015 to 2040. Nat. Commun. 2016, 7, 1–6.
  5. Benito-León, J. Are the prevalence and incidence of multiple sclerosis changing? Neuroepidemiology 2011, 36, 148.
  6. Dorsey, E.R.; Bloem, B.R. The Parkinson pandemic—A call to action. JAMA Neurol. 2018, 75, 9–10.
  7. Patrícia, B.; Pereira, A. Impact and prevention of neurodegenerative diseases in society: Alzheimer and Parkinson. In Neurodegenerative Diseases; SM Group Open Access eBooks: Dover, DE, USA, 2016.
  8. Gitler, A.D.; Dhillon, P.; Shorter, J. Neurodegenerative disease: Models, mechanisms, and a new hope. Dis. Models Mech. 2017, 10, 499–502.
  9. Takahashi, K.; Okita, K.; Nakagawa, M.; Yamanaka, S. Induction of pluripotent stem cells from fibroblast cultures. Nat. Protoc. 2007, 2, 3081.
  10. Shi, Y.; Inoue, H.; Wu, J.C.; Yamanaka, S. Induced pluripotent stem cell technology: A decade of progress. Nat. Rev. Drug Discov. 2017, 16, 115–130.
  11. Takahashi, K.; Tanabe, K.; Ohnuki, M.; Narita, M.; Ichisaka, T.; Tomoda, K.; Yamanaka, S. Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell 2007, 131, 861–872.
  12. Israel, M.A.; Yuan, S.H.; Bardy, C.; Reyna, S.M.; Mu, Y.; Herrera, C.; Hefferan, M.P.; Van Gorp, S.; Nazor, K.L.; Boscolo, F.S.; et al. Probing sporadic and familial Alzheimer’s disease using induced pluripotent stem cells. Nature 2012, 482, 216–220.
  13. Poon, A.; Schmid, B.; Pires, C.; Nielsen, T.T.; Hjermind, L.E.; Nielsen, J.E.; Holst, B.; Hyttel, P.; Freude, K.K. Generation of a gene-corrected isogenic control hiPSC line derived from a familial Alzheimer′s disease patient carrying a L150P mutation in presenilin 1. Stem Cell Res. J. 2016, 17, 466–469.
  14. Dimos, J.T.; Rodolfa, K.T.; Niakan, K.K.; Weisenthal, L.M.; Mitsumoto, H.; Chung, W.; Croft, G.F.; Saphier, G.; Leibel, R.; Goland, R.; et al. Induced pluripotent stem cells generated from patients with ALS can be differentiated into motor neurons. Science. 2008, 321, 1218–1221.
  15. Burkhardt, M.F.; Martinez, F.J.; Wright, S.; Ramos, C.; Volfson, D.; Mason, M.; Garnes, J.; Dang, V.; Lievers, J.; Shoukat-Mumtaz, U.; et al. A cellular model for sporadic ALS using patient-derived induced pluripotent stem cells. Mol. Cell. Neurosci. 2013, 56, 355–364.
  16. Park, I.H.; Arora, N.; Huo, H.; Maherali, N.; Ahfeldt, T.; Shimamura, A.; Lensch, M.W.; Cowan, C.; Hochedlinger, K.; Daley, G.Q. Disease-specific induced pluripotent stem cells. Cell 2008, 134, 877–886.
  17. Kaufmann, M.; Schuffenhauer, A.; Fruh, I.; Klein, J.; Thiemeyer, A.; Rigo, P.; Gomez-Mancilla, B.; Heidinger-Millot, V.; Bouwmeester, T.; Schopfer, U.; et al. High-throughput screening using iPSC-derived neuronal progenitors to identify compounds counteracting epigenetic gene silencing in fragile X syndrome. J. Biomol. Screen. 2015, 20, 1101–1111.
  18. Perriot, S.; Mathias, A.; Perriard, G.; Canales, M.; Jonkmans, N.; Merienne, N.; Meunier, C.; El Kassr, L.; Perrier, A.L.; Laplaud, D.A.; et al. Human induced pluripotent stem cell-derived astrocytes are differentially activated by multiple sclerosis-associated cytokines. Stem Cell Rep. 2018, 11, 1199–1210.
  19. Kim, Y.; Park, J.; Choi, Y.K. The role of astrocytes in the central nervous system focused on BK channel and heme oxygenase metabolites: A review. Antioxidants 2019, 8, 121.
  20. Suga, M.; Kondo, T.; Inoue, H. Modeling neurological disorders with human pluripotent stem cell-derived astrocytes. Int. J. Mol. Sci. 2019, 20, 3862.
  21. Miller, R.H.; Raff, M.C. Fibrous and protoplasmic astrocytes are biochemically and developmentally distinct. J. Neurosci. 1984, 4, 585–592.
  22. Griemsmann, S.; Höft, S.P.; Bedner, P.; Zhang, J.; Von Staden, E.; Beinhauer, A.; Degen, J.; Dublin, P.; Cope, D.W.; Richter, N.; et al. Characterization of panglial gap junction networks in the thalamus, neocortex, and hippocampus reveals a unique population of glial cells. Cereb. Cortex. 2015, 25, 3420–3433.
  23. Beiersdorfer, A.; Scheller, A.; Kirchhoff, F.; Lohr, C. Panglial gap junctions between astrocytes and olfactory ensheathing cells mediate transmission of Ca2+ transients and neurovascular coupling. Glia 2019, 67, 1385–1400.
  24. Liddelow, S.A.; Barres, B.A. Reactive astrocytes: Production, function, and therapeutic potential. Immunity 2017, 6, 957–967.
  25. Zhang, Y.; Chen, K.; Sloan, S.A.; Bennett, M.L.; Scholze, A.R.; O’Keeffe, S.; Phatnani, H.P.; Guarnieri, P.; Caneda, C.; Ruderisch, N.; et al. An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J. Neurosci. 2014, 34, 11929–11947.
  26. Boisvert, M.M.; Erikson, G.A.; Shokhirev, M.N.; Allen, N.J. The aging astrocyte transcriptome from multiple regions of the mouse brain. Cell Rep. 2018, 22, 269–285.
  27. Emsley, J.G.; Macklis, J.D. Astroglial heterogeneity closely reflects the neuronal-defined anatomy of the adult murine CNS. Neuron Glia Biol. 2006, 2, 175.
  28. Pekny, M.; Pekna, M. Astrocyte reactivity and reactive astrogliosis: Costs and benefits. Physiol. Rev. 2014, 94, 1077–1098.
  29. Escartin, C.; Guillemaud, O.; Carrillo-de Sauvage, M.A. Questions and (some) answers on reactive astrocytes. Glia 2019, 67, 2221–2247.
  30. Habbas, S.; Santello, M.; Becker, D.; Stubbe, H.; Zappia, G.; Liaudet, N.; Klaus, F.R.; Kollias, G.; Fontana, A.; Pryce, C.R.; et al. Neuroinflammatory TNFα impairs memory via astrocyte signaling. Cell 2015, 163, 1730–1741.
  31. Escartin, C.; Galea, E.; Lakatos, A.; O’Callaghan, J.P.; Petzold, G.C.; Serrano-Pozo, A.; Steinhäuser, C.; Volterra, A.; Carmignoto, G.; Agarwal, A.; et al. Reactive astrocyte nomenclature, definitions, and future directions. Nat. Neurosci. 2021, 24, 312–325.
  32. Verkhratsky, A.; Zorec, R.; Parpura, V. Stratification of astrocytes in healthy and diseased brain. Brain Pathol. 2017, 27, 629–644.
  33. Chai, H.; Diaz-Castro, B.; Shigetomi, E.; Monte, E.; Octeau, J.C.; Yu, X.; Cohn, W.; Rajendran, P.S.; Vondriska, T.M.; Whitelegge, J.P.; et al. Neural circuit-specialized astrocytes: Transcriptomic, proteomic, morphological, and functional evidence. Neuron. 2017, 95, 531–549.
  34. Zamanian, J.L.; Xu, L.; Foo, L.C.; Nouri, N.; Zhou, L.; Giffard, R.G.; Barres, B.A. Genomic analysis of reactive astrogliosis. J. Neurosci. 2012, 32, 6391–6410.
  35. Liddelow, S.A.; Guttenplan, K.A.; Clarke, L.E.; Bennett, F.C.; Bohlen, C.J.; Schirmer, L.; Bennett, M.L.; Münch, A.E.; Chung, W.S.; Peterson, T.C.; et al. Neurotoxic reactive astrocytes are induced by activated microglia. Nature 2017, 541, 481–487.
  36. Ceyzériat, K.; Haim, L.B.; Denizot, A.; Pommier, D.; Matos, M.; Guillemaud, O.; Palomares, M.A.; Abjean, L.; Petit, F.; Gipchtein, P.; et al. Modulation of astrocyte reactivity improves functional deficits in mouse models of Alzheimer’s disease. Acta Neuropathol. Commun. 2018, 6, 1–23.
  37. Diaz-Castro, B.; Gangwani, M.R.; Yu, X.; Coppola, G.; Khakh, B.S. Astrocyte molecular signatures in Huntington’s disease. Sci. Transl. Med. 2019, 11, eaaw8546.
  38. Zhou, Y.; Song, W.M.; Andhey, P.S.; Swain, A.; Levy, T.; Miller, K.R.; Poliani, P.L.; Cominelli, M.; Grover, S.; Gilfillan, S.; et al. Human and mouse single-nucleus transcriptomics reveal TREM2-dependent and TREM2-independent cellular responses in Alzheimer’s disease. Nat. Med. 2020, 26, 131–142.
  39. Das, S.; Li, Z.; Noori, A.; Hyman, B.T.; Serrano-Pozo, A. Meta-analysis of mouse transcriptomic studies supports a context-dependent astrocyte reaction in acute CNS injury versus neurodegeneration. J. Neuroinflamm. 2020, 17, 1–7.
  40. Heiland, D.H.; Ravi, V.M.; Behringer, S.P.; Frenking, J.H.; Wurm, J.; Joseph, K.; Garrelfs, N.W.; Strähle, J.; Heynckes, S.; Grauvogel, J.; et al. Tumor-associated reactive astrocytes aid the evolution of immunosuppressive environment in glioblastoma. Nat. Commun. 2019, 10, 1–2.
  41. Polanco, J.C.; Li, C.; Bodea, L.G.; Martinez-Marmol, R.; Meunier, F.A.; Götz, J. Amyloid-β and tau complexity—Towards improved biomarkers and targeted therapies. Nat. Rev. Neurol. 2018, 14, 22.
  42. Prince, M.; Bryce, R.; Albanese, E.; Wimo, A.; Ribeiro, W.; Ferri, C.P. The global prevalence of dementia: A systematic review and metaanalysis. Alzheimers Dement. 2013, 9, 63–75.
  43. Gammon, K. Neurodegenerative disease: Brain windfall. Nature 2014, 515, 299–300.
  44. Dossi, E.; Vasile, F.; Rouach, N. Human astrocytes in the diseased brain. Brain Res. Bull. 2018, 136, 139–156.
  45. Mahley, R.W. Central nervous system lipoproteins: ApoE and regulation of cholesterol metabolism. Arterioscler Thromb. Vasc. Biol. 2016, 36, 1305–1315.
  46. Kobayashi, E.; Nakano, M.; Kubota, K.; Himuro, N.; Mizoguchi, S.; Chikenji, T.; Otani, M.; Mizue, Y.; Nagaishi, K.; Fujimiya, M. Activated forms of astrocytes with higher GLT-1 expression are associated with cognitive normal subjects with Alzheimer pathology in human brain. Sci. Rep. 2018, 8, 1–2.
  47. Chu, Y.T.; Tai, C.H.; Lin, C.H.; Wu, R.M. Updates on the genetics of Parkinson′s disease: Clinical implications and future treatment. Acta Neurol. Taiwan 2021, 30, 83–93.
  48. Olanow, C.W.; Tatton, W.G. Etiology and pathogenesis of Parkinson′s disease. Ann. Rev. Neurosci. 1999, 22, 123–144.
  49. Schapira, A.H.; Jenner, P. Etiology and pathogenesis of Parkinson′s disease. Mov Disord. 2011, 26, 1049–1055.
  50. MacMahon Copas, A.N.; McComish, S.F.; Fletcher, J.M.; Caldwell, M.A. The pathogenesis of Parkinson′s disease: A complex interplay between astrocytes, microglia, and T lymphocytes? Front. Neurol. 2021, 12, 666737.
  51. Jankovic, J. Parkinson′s disease: Clinical features and diagnosis. J. Neurol. Neurosurg Psychiatr. 2008, 79, 368–376.
  52. Lee, S.H.; Park, S.M.; Yeo, S.S.; Kwon, O.; Lee, M.K.; Yoo, H.; Ahn, E.K.; Jang, J.Y.; Jang, J.H. Parkinson′s disease subtyping using clinical features and biomarkers: Literature review and preliminary study of subtype clustering. Diagnostics 2022, 12, 112.
  53. Di Domenico, A.; Carola, G.; Calatayud, C.; Pons-Espinal, M.; Muñoz, J.P.; Richaud-Patin, Y.; Fernandez-Carasa, I.; Gut, M.; Faella, A.; Parameswaran, J.; et al. Patient-specific iPSC-derived astrocytes contribute to non-cell-autonomous neurodegeneration in Parkinson′s disease. Stem Cell Rep. 2019, 12, 213–229.
  54. Perlow, M.J.; Freed, W.J.; Hoffer, B.J.; Seiger, A.; Olson, L.; Wyatt, R.J. Brain grafts reduce motor abnormalities produced by destruction of nigrostriatal dopamine system. Science 1979, 204, 643–647.
  55. Hallett, P.J.; Deleidi, M.; Astradsson, A.; Smith, G.A.; Cooper, O.; Osborn, T.M.; Sundberg, M.; Moore, M.A.; Perez-Torres, E.; Brownell, A.L.; et al. Successful function of autologous iPSC-derived dopamine neurons following transplantation in a non-human primate model of Parkinson’s disease. Cell Stem Cell 2015, 16, 269–274.
  56. Li, H.; Jiang, H.; Zhang, B.; Feng, J. Modeling Parkinson’s disease using patient-specific induced pluripotent stem cells. J. Parkinsons Dis. 2018, 8, 479–493.
  57. Sundberg, M.; Bogetofte, H.; Lawson, T.; Jansson, J.; Smith, G.; Astradsson, A.; Moore, M.; Osborn, T.; Cooper, O.; Spealman, R.; et al. Improved cell therapy protocols for Parkinson′s disease based on differentiation efficiency and safety of hESC-, hiPSC-, and non-human primate iPSC-derived dopaminergic neurons. Stem Cells 2013, 31, 1548–1562.
  58. Kikuchi, T.; Morizane, A.; Doi, D.; Magotani, H.; Onoe, H.; Hayashi, T.; Mizuma, H.; Takara, S.; Takahashi, R.; Inoue, H.; et al. Human iPS cell-derived dopaminergic neurons function in a primate Parkinson’s disease model. Nature 2017, 548, 592–596.
  59. Takahashi, J. iPS cell-based therapy for Parkinson’s disease: A Kyoto trial. Regen. Ther. 2020, 13, 18–22.
  60. Eisen, A. Amyotrophic lateral sclerosis—Evolutionary and other perspectives. Muscle Nerve 2009, 40, 297–304.
  61. Andersen, P.M.; Al-Chalabi, A. Clinical genetics of amyotrophic lateral sclerosis: What do we really know? Nat. Rev. Neurol. 2011, 7, 603–615.
  62. Laferriere, F.; Polymenidou, M. Advances and challenges in understanding the multifaceted pathogenesis of amyotrophic lateral sclerosis. Swiss Med. Wkly 2015, 30, 145:w14054.
  63. lsultan, A.A.; Waller, R.; Heath, P.R.; Kirby, J. The genetics of amyotrophic lateral sclerosis: Current insights. Degener. Neurol. Neuromuscul. Dis. 2016, 6, 49–64.
  64. Mejzini, R.; Flynn, L.L.; Pitout, I.L.; Fletcher, S.; Wilton, S.D.; Akkari, P.A. ALS genetics, mechanisms, and therapeutics: Where are we now? Front. Neurosci. 2019, 13, 1310.
  65. Lee, J.; Hyeon, S.J.; Im, H.; Ryu, H.; Kim, Y.; Ryu, H. Astrocytes and microglia as non-cell autonomous players in the pathogenesis of ALS. Exp. Neurobiol. 2016, 25, 233–240.
  66. Nagai, M.; Re, D.B.; Nagata, T.; Chalazonitis, A.; Jessell, T.M.; Wichterle, H.; Przedborski, S. Astrocytes expressing ALS-linked mutated SOD1 release factors selectively toxic to motor neurons. Nat. Neurosci. 2007, 10, 615–622.
  67. Yamanaka, K.; Chun, S.J.; Boillee, S.; Fujimori-Tonou, N.; Yamashita, H.; Gutmann, D.H.; Takahashi, R.; Misawa, H.; Cleveland, D.W. Astrocytes as determinants of disease progression in inherited amyotrophic lateral sclerosis. Nat. Neurosci. 2008, 11, 251–253.
  68. Bruijn, L.I.; Becher, M.W.; Lee, M.K.; Anderson, K.L.; Jenkins, N.A.; Copeland, N.G.; Sisodia, S.S.; Rothstein, J.D.; Borchelt, D.R.; Price, D.L.; et al. ALS-linked SOD1 mutant G85R mediates damage to astrocytes and promotes rapidly progressive disease with SOD1-containing inclusions. Neuron 1997, 18, 327–338.
  69. Kia, A.; McAvoy, K.; Krishnamurthy, K.; Trotti, D.; Pasinelli, P. Astrocytes expressing ALS-linked mutant FUS induce motor neuron death through release of tumor necrosis factor-alpha. Glia 2018, 66, 1016–1033.
  70. Tripathi, P.; Rodriguez-Muela, N.; Klim, J.R.; de Boer, A.S.; Agrawal, S.; Sandoe, J.; Lopes, C.S.; Ogliari, K.S.; Williams, L.A.; Shear, M.; et al. Reactive astrocytes promote ALS-like degeneration and intracellular protein aggregation in human motor neurons by disrupting autophagy through TGF-beta1. Stem Cell Rep. 2017, 9, 667–680.
  71. Rojas, F.; Gonzalez, D.; Cortes, N.; Ampuero, E.; Hernández, D.E.; Fritz, E.; Abarzua, S.; Martinez, A.; Elorza, A.A.; Alvarez, A.; et al. Reactive oxygen species trigger motoneuron death in non-cell-autonomous models of ALS through activation of c-Abl signaling. Front. Cell Neurosci. 2015, 9, 203.
  72. Cho, I.K.; Yang, B.; Forest, C.; Qian, L.; Chan, A.W. Amelioration of Huntington’s disease phenotype in astrocytes derived from iPSC-derived neural progenitor cells of Huntington’s disease monkeys. PLoS ONE. 2019, 14, e0214156.
  73. Garcia, V.J.; Rushton, D.J.; Tom, C.M.; Allen, N.D.; Kemp, P.J.; Svendsen, C.N.; Mattis, V.B. Huntington’s disease patient-derived astrocytes display electrophysiological impairments and reduced neuronal support. Front. Neurosci. 2019, 13, 669.
  74. Tabrizi, S.J.; Flower, M.D.; Ross, C.A.; Wild, E.J. Huntington disease: New insights into molecular pathogenesis and therapeutic opportunities. Nat. Rev. Neurol. 2020, 16, 529–546.
  75. McColgan, P.; Tabrizi, S.J. Huntington′s disease: A clinical review. Eur. J. Neurol. 2018, 5, 24–34.
  76. Evans, S.J.; Douglas, I.; Rawlins, M.D.; Wexler, N.S.; Tabrizi, S.J.; Smeeth, L. Prevalence of adult Huntington′s disease in the UK based on diagnoses recorded in general practice records. J. Neurol. Neurosurg. Psychiatr. 2013, 84, 1156–1160.
  77. Ross, C.A.; Aylward, E.H.; Wild, E.J.; Langbehn, D.R.; Long, J.D.; Warner, J.H.; Scahill, R.I.; Leavitt, B.R.; Stout, J.C.; Paulsen, J.S.; et al. Huntington disease: Natural history, biomarkers and prospects for therapeutics. Nat. Rev. Neurol. 2014, 10, 204.
  78. Biagioli, M.; Ferrari, F.; Mendenhall, E.M.; Zhang, Y.; Erdin, S.; Vijayvargia, R.; Vallabh, S.M.; Solomos, N.; Manavalan, P.; Ragavendran, A.; et al. Htt CAG repeat expansion confers pleiotropic gains of mutant huntingtin function in chromatin regulation. Hum. Mol. Genet. 2015, 24, 2442–2457.
  79. Shannon, K.M. Recent advances in the treatment of Huntington’s disease: Targeting DNA and RNA. CNS Drugs 2020, 34, 219–228.
  80. Wilton, D.K.; Stevens, B. The contribution of glial cells to Huntington′s disease pathogenesis. Neurobiol. Dis. 2020, 143, 104963.
  81. Phatnani, H.; Maniatis, T. Astrocytes in neurodegenerative disease. Cold Spring Harb Perspect Biol. 2015, 7, a020628.
  82. Ponath, G.; Park, C.; Pitt, D. The role of astrocytes in multiple sclerosis. Front. Immunol. 2018, 9, 217.
  83. Cree, B.A.; Mares, J.; Hartung, H.P. Current therapeutic landscape in multiple sclerosis: An evolving treatment paradigm. Curr. Opin. Neurol. 2019, 32, 365–377.
  84. Kobelt, G.; Thompson, A.; Berg, J.; Gannedahl, M.; Eriksson, J.; MSCOI Study Group; European Multiple Sclerosis Platform. New insights into the burden and costs of multiple sclerosis in Europe. Mult. Scler. J. 2017, 23, 1123–1136.
  85. Martínez-Larrosa, J.; Matute-Blanch, C.; Montalbano, X.; Comabella, M. Modelling multiple sclerosis using induced pluripotent stem cells. J. Neuroimmunol. 2020, 349, 577425.
  86. Michal, I.; Guy, S.S.; Michel, R. Astrocytes in pathogenesis of multiple sclerosis and potential translation into clinic. In Glia Health and Disease; IntechOpen: London, UK, 2019; pp. 1–19.
  87. Brambilla, R. The contribution of astrocytes to the neuroinflammatory response in multiple sclerosis and experimental autoimmune encephalomyelitis. Acta Neuropathol. 2019, 137, 757–783.
  88. Ding, Z.B.; Song, L.J.; Wang, Q.; Kumar, G.; Yan, Y.Q.; Ma, C.G. Astrocytes: A double-edged sword in neurodegenerative diseases. Neural Regen. Res. 2021, 16, 1702.
  89. Ponath, G.; Lincoln, M.R.; Levine-Ritterman, M.; Park, C.; Dahlawi, S.; Mubarak, M.; Sumida, T.; Airas, L.; Zhang, S.; Isitan, C.; et al. Enhanced astrocyte responses are driven by a genetic risk allele associated with multiple sclerosis. Nat. Commun. 2018, 9, 1–9.
  90. Patitucci, T.N.; Ebert, A.D. SMN deficiency does not induce oxidative stress in SMA iPSC-derived astrocytes or motor neurons. Hum. Mol. Genet. 2016, 25, 514–523.
  91. Adami, R.; Bottai, D. Spinal muscular atrophy modeling and treatment advances by induced pluripotent stem cells studies. Stem Cell Rev. Rep. 2019, 15, 795–813.
  92. Rindt, H.; Feng, Z.; Mazzasette, C.; Glascock, J.J.; Valdivia, D.; Pyles, N.; Crawford, T.O.; Swoboda, K.J.; Patitucci, T.N.; Ebert, A.D.; et al. Astrocytes influence the severity of spinal muscular atrophy. Hum. Mol. Genet. 2015, 24, 4094–4102.
  93. McGivern, J.V.; Patitucci, T.N.; Nord, J.A.; Barabas, M.E.; Stucky, C.L.; Ebert, A.D. Spinal muscular atrophy astrocytes exhibit abnormal calcium regulation and reduced growth factor production. Glia 2013, 61, 1418–1428.
  94. Ratni, H.; Ebeling, M.; Baird, J.; Bendels, S.; Bylund, J.; Chen, K.S.; Denk, N.; Feng, Z.; Green, L.; Guerard, M.; et al. Discovery of risdiplam, a selective survival of motor neuron-2 (SMN2) gene splicing modifier for the treatment of spinal muscular atrophy (SMA). J. Med. Chem. 2018, 61, 6501–6517.
  95. Nicolau, S.; Waldrop, M.A.; Connolly, A.M.; Mendell, J.R. Spinal muscular atrophy. Sem. in Ped. Neurol. 2021, 37, 100878.
  96. Sison, S.L.; Patitucci, T.N.; Seminary, E.R.; Villalon, E.; Lorson, C.L.; Ebert, A.D. Astrocyte-produced miR-146a as a mediator of motor neuron loss. Hum. Mol. Genet. 2017, 26, 3409–3420.
More
Information
Subjects: Cell Biology
Contributor MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register :
View Times: 368
Entry Collection: Neurodegeneration
Revisions: 2 times (View History)
Update Date: 15 Mar 2022
1000/1000
Video Production Service