Abstract
The importance of glial cells, mainly astrocytes, oligodendrocytes, and microglia, in the central nervous system (CNS) has been increasingly appreciated. Recent advances have demonstrated the diversity of glial cells and their contribution to human CNS development, normal CNS functions, and disease progression. The uniqueness of human glial cells is also supported by multiple lines of evidence. With the discovery of induced pluripotent stem cells (iPSCs) and the progress of generating glial cells from human iPSCs, there are numerous studies to model CNS diseases using human iPSC-derived glial cells. Here we summarize the basic characteristics of glial cells, with the focus on their classical functions, heterogeneity, and uniqueness in human species. We further review the findings from recent studies that use iPSC-derived glial cells for CNS disease modeling. We conclude with promises and future directions of using iPSC-derived glial cells for CNS disease modeling.
Keywords: glial cells, iPSC, neurological diseases, disease modeling, astrocytes, oligodendrocytes, microglia
1. Introduction
Glia in general refers to non-neuronal cell types including astrocytes, oligodendrocytes, and microglia in the central nervous system (CNS). Other non-neuronal cell types also exist in the CNS, for example, ependymal cells that line up ventricles. Glial cells were traditionally viewed as cells that simply glue neurons and CNS tissues together. With increasing knowledge of their functions, each cell type is highly heterogeneous, exhibiting regionality and functional differences. It stands to reason that such abundant and highly specified cell populations play important roles in the physiology and pathology of the CNS. Indeed, the importance of glial cells to the CNS has been supported by numerous studies using animal models as well as human tissues [1–3]. Interestingly, it is shown that glial cells exhibit phenotypical and functional difference across species, and human glial cells possess unique features compared to other mammalian counterparts [4–6]. The divergence of human glial cells from other species may partially lead to the functional difference between the CNS of human and other species. This species difference poses challenges to study human CNS disorders using animal models.
Human induced pluripotent stem cells (iPSCs), which are generated by reprogramming of human somatic cells, provide a superior resource for researchers to derive cell types of interest and study diseases in a dish [7, 8]. The human iPSC platforms are particularly powerful to study neurological diseases due to the uniqueness of the human CNS system and the challenges to access and maintain human CNS tissues and cells in vitro. Because glial cells play an important part in the pathogenesis of neurological diseases, here we review the key properties of glial cells and the contribution of iPSC-derived glial cells to our understanding of CNS diseases (Figure 1). We will also discuss the limitations to overcome when using iPSC-derived glial cells to study CNS diseases based on current knowledge of glial biology.
Figure 1. When glial cells meet iPSCs.
Glial cells mainly include three cell types, astrocytes, oligodendrocytes and microglia. Human glial cells play critical roles in brain functions, are highly heterogeneous, and possess species-specific features. Human iPSCs generated from somatic cell reprogramming can be differentiated into glial cells for disease modeling.
2. Glial cell function, heterogeneity, and their uniqueness in human
Glial cells in the human CNS maintain tissue homeostasis, form myelin sheath, and respond to injuries and pathogens. The dysfunction of glial cells due to genetic mutations, aging or exogenous insult contributes to various CNS diseases. In this section, we will provide an up-to-date overview of the functions of the three major glial cell types (astrocytes, oligodendrocytes and microglia) under physiological conditions, their heterogeneity and species-specific features.
2.1. Astrocytes
Astrocytes are the most abundant cell type and involved in various functions in the CNS. They are highly heterogeneous in terms of morphology, molecular profiles, and functions depending on their location as well as reactive states. Grey matter astrocytes, also named protoplasmic astrocytes, exhibit highly branched morphology and are in close contact with neurons and blood vessels; white matter astrocytes, also named fibrous astrocytes, have less branches and simpler processes and are in contact with oligodendrocytes and axons in the nodes of Ranvier [1]. Astrocytes switch from a non-reactive state to a reactive state in response to stimuli. Reactive astrocytes respond differentially to different insults and can be divided into neurotoxic A1 and neuroprotective A2 subtypes [9, 10]. Astrocyte reactivity is accompanied by a dynamic yet reversible change in gene signature, morphology, and functions [11].
Astrocytes play important roles in synapse formation, clearance and function [12, 13]. The support of synapse formation by astrocytes has been observed across different species [14–16], in different subtypes of neurons [17–19], and through different mediators, for example, cholesterol [20, 21], thrombospondins [22, 23], and Chrdl1 [24]. Astrocytes can phagocytose synapses directly through the phagocytic receptors Mertk and Megf10 to avoid excessive synapse accumulation [25] or signal microglia to clear synapses through molecules such as TGFβ and IL-33 [26]. Astrocytes form tripartite synapses as an addition to pre- and post-synaptic neuron communications to allow bidirectional information flow between astrocytes and synapses [27, 28]. Astrocytes generate intracellular Ca2+ oscillations in response to synaptic neurotransmitters and release neuroactive molecules, such as glutamate, acetylcholine, adenosine, ATP, and γ-aminobutyric acid (GABA), through vesicle and lysosomal exocytosis [28]. These molecules in turn influence neuronal and synaptic activity.
Astrocytes act as sensors for various metabolic signals, such as glutamate, O2, CO2, glucose and endocrine hormones [29–31]. Glutamate is one major excitatory neurotransmitter released from neuronal presynaptic terminals [32]. The accumulation of glutamate at the synapse space leads to neuronal injury through Ca2+ influx and subsequent nitric oxide synthesis, the generation of free radicals, the activation of autophagy/lysosomal pathways, and programmed cell death [33]. Astrocytes express EAATs (excitatory amino acid transporters), which allows glutamate uptake from the extracellular space [32], protecting the brain from glutamate-induced excitotoxicity. The endfeet of astrocytes attach to the microvessel wall in the brain, named the blood-brain barrier (BBB), which selectively permits or excludes molecules in the blood stream to enter the brain. Specialized features in astrocyte endfeet, such as high density of the water channel aquaporin 4 (AQP4) and the Kir4.1 K+ channel, are involved in ion and fluid regulation of the brain [34, 35].
Although mouse models contribute tremendously to our understanding of astrocytes, mouse astrocytes differ from human astrocytes in various ways. Astrocytes in human brains exhibit more complex morphology [4] and possess a set of genes uniquely expressed compared to murine astrocytes [36]. Functionally, human astrocytes can generate calcium waves in response to glutamate, whereas murine astrocytes cannot [36], which may contribute to behavioral level differences between species [37, 38]. Moreover, human astrocytes can grow better than mouse astrocytes after being transplanted into mouse brains. Finally, they gradually replace endogenous astrocytes in mouse brains and improve cognitive functions of the recipient mice [39].
2.2. Oligodendrocytes
Oligodendrocytes are the cell type that perform myelination in the CNS. Oligodendrocytes are developed from PDGFRα+ and NG2+ oligodendrocyte progenitor cells (OPCs) [40]. OPCs can differentiate into O4+ late OPCs (late OPCs) or preoligodendrocytes [41, 42], followed by O1+ premyelinating OLs or immature oligodendrocytes [42]. Immature oligodendrocytes can terminally differentiate into mature myelinating oligodendrocytes, which are marked by the expression of myelin proteins, such as MBP and PLP1 [43, 44]. The process of myelin wrapping around axons is called myelination. At least four important steps occur during the process of myelination: 1) selection of axons by oligodendrocytes for wrapping and initial oligodendrocyte-axon contact; 2) stabilization of oligodendrocyte-axon contact and formation of nodes of Ranvier; 3) myelin wrapping of axons and compaction; and 4) longitudinal extension of myelin segments in response to postnatal axon extension [45].
The impacts of myelination on CNS functions are beyond a passive insulator. The speed of signal conduction along axons is tuned by subtle structural changes of myelin, for example, myelin sheath thickness and the length of myelin segments. This fine adjustment of myelin parameters is crucial for motor skill establishment and cognitive function [46, 47]. In addition, oligodendrocytes provide metabolic products, such as lactate and pyruvate, to support mitochondrial functions and neurotrophic factors to maintain the integrity of axons [48–50]. Emerging evidence supports the heterogeneity of OPCs and oligodendrocytes. At the OPC stage, they differ in terms of origin (dorsal versus ventral), rate of self-renewal (white-matter OPCs proliferate faster than grey-matter OPCs), differentiation potential (white-matter OPCs differentiate more efficiently than grey-matter OPCs), and regenerative capacity (dorsal OPCs make more contribution to remyelination than ventral OPCs, however, under aging, their regeneration properties are reversed) [36]. In addition, Marques et al. analyzed mouse oligodendrocytes from different brain regions using single-cell RNA-sequencing (scRNA-seq) and identified six subgroups of oligodendrocyte subpopulations [51]. Single-cell electrophysiological recordings also demonstrated ion channel changes of OPCs between different brain regions and ages [52].
Studies of human oligodendrocytes and OPCs are relatively rare due to technical challenges of obtaining and processing human postmortem brain tissues. Nevertheless, Jäkel et al. recently performed single-nucleus RNA sequencing (snRNA-seq) of white matter brain tissues from multiple sclerosis patients and unaffected controls [53] and identified 7 subclusters of oligodendrocytes with different transcriptome signatures, supporting the heterogeneity of human oligodendrocytes. The comparison between multiple sclerosis and control snRNA-seq data indicates mature oligodendrocytes also contribute to remyelination in humans, which is different from rodents, in which the recruitment of OPCs followed by the induction into oligodendrocytes is the only driver for remyelination. Retrospective carbon-14 birth-dating analysis of oligodendrocytes in human brains also suggests that mature oligodendrocytes contribute to myelin remodeling rather than newly generated oligodendrocytes [54].
2.3. Microglia
Microglia are the innate immune cells of the CNS. Microglia originate from a subset of primitive myeloid progenitors traveling to neural tube at the yolk sac stage during primitive hematopoiesis from approximately embryonic day 8 (E8) in mouse [55] and gestation week 4.5 in human [56–58]. Microglia progenitors proliferate and mature within brain tissues [59], a process tightly regulated by neural cues in the CNS [60] and brain-gut interactions [59, 61].
As innate immune cells, microglia express pattern recognition receptors, such as the Toll-like receptor 4 (TLR4) [62], the activation of which triggers downstream signaling pathways, e.g. the NF-kB pathway, and the secretion of pro-inflammation cytokines and complement factors. These secreted molecules recruit other immune cells, such as T cells and macrophages, from the blood stream into the CNS and continue immune reactions under pathological conditions [62]. Microglia are regulators for brain functions at both embryonic and postnatal stages. During development, microglia phagocytose redundant cells to ensure the appropriate quantity of cell types at the right locations [63–65]. They can precisely target and phagocytose unwanted synapses tagged by the complement factor C1q or C3 [66]. Refinement of synapses by microglia persists throughout the lifetime, which is important for neuronal plasticity [67]. Microglia also engulf myelin debris after brain injury and promote remyelination [68].
Depending on the immune reaction status, microglia are categorized using the paradigm of macrophages, M1 as neurotoxic and M2 as neuroprotective [69]. However, with increasing knowledge about the response of microglia and macrophage to immune stimuli, the status of microglia is more complicated beyond the scope of the M1/M2 nomenclature [70]. They may respond differently to different molecular constituents, such as LPS, IFNγ, and interleukin cytokines [71, 72]. With the progress of single cell technology, such as scRNA-seq, microglia have been shown to be diverse in terms of their developmental timing, turnover rate, response to insults, and regional differences. Li et al. performed deep scRNA-seq of microglia isolated from different regions of mouse brains and observed a higher level of heterogeneity at the early postnatal stage than adulthood [73]. Very recently, Masuda et al. comprehensively characterized the subtypes of microglia at different regions in the CNS during development and diseases [74] and found both spatial and temporal heterogeneity exist in microglia populations, with 10 clusters showing distinct transcriptional profiles, some of which mainly consisted of the embryonic stage and others of the postnatal stage. The authors also compared the human microglia data with the mouse microglia data, finding overall similarities between human and mouse microglia, but with non-negligible species differences.
3. Contribution of iPSC-derived glial cells to disease modeling
The human CNS diverges from the CNS of the other species in various ways, but current findings of developmental biology and disease pathology have mainly relied on animal models. To overcome species differences, human brain tissues (mainly postmortem tissues) have been used as a supplement to confirm findings from animal models. Most human brain tissues represent end-stage diseases and undergo extensive processing before analysis, which prevents us from understanding the initiation and progression of disease conditions that are critical for disease prevention and intervention at the early stage. The emergence of the iPSC technology has added to the toolbox for disease study due to the unique advantages of iPSCs. iPSCs maintain genetic features of the parental somatic cells and mimic embryonic stem cells (ESCs) in many aspects, such as potent proliferation and relatively easy manipulation of the genome. These advantages make iPSCs a powerful tool for human disease modeling, especially neurological disease modeling, for which human tissues are not easily accessible.
With the advances of differentiation protocols, iPSC-derived astrocytes, oligodendrocytes, and microglia have been applied to study various CNS diseases and answered questions not possible using animal models. Below, we discuss cellular models using human iPSC-derived glial cells and their contribution to our understanding of CNS diseases. However, iPSC-based modeling systems are not perfect, and like any other models, caution is needed when interpreting the results. We will thus discuss how to overcome these obstacles as well.
3.1. Astrocytes
Astrocyte dysfunction plays a role in a wide range of CNS diseases, not only neurodegeneration, but also neurodevelopmental and neuropsychiatric diseases. For example, astrocyte abnormalities, especially reactive gliosis, have been identified in postmortem patient brain tissues and animal models of Alzheimer’s disease (AD), Parkinson’s disease (PD) and Huntington’s disease [75]. Reactive astrocytes could also lead to gliosis formation after spinal cord injury or brain trauma [76, 77]. In addition, mutations of GFAP, a gene predominantly expressed in astrocytes, are sufficient to cause the fatal disorder Alexander disease (AxD) [78, 79]. Because astrocytes interact with all the major cell types within the CNS, their abnormalities could affect various functions of other cell types and contribute to diseases. Importantly, iPSC-derived astrocytes allow researchers to uncover the roles of astrocytes in the pathogenesis of diseases that remain challenging to understand due to the lack of reliable animal models or the absence of disease-relevant phenotypes in animal models. Neurodegenerative diseases, such as AD and PD, are of particular interest, because it has been challenging to dissect the complications of these diseases and develop effective treatments using traditional models. Rare neurodevelopmental diseases are also being more studied using human iPSC models because of the easy access of cellular resources derived from iPSCs and the flexibility of genome editing of iPSCs (summarized in Table 1 by disease type).
Table 1.
Selected Studies Investigating Neurological Diseases Using iPSC-derived Astrocytes
| Disease | Disease Features of Interest | Disease iPSC Characteristics | Cellular Model | Major Findings |
|---|---|---|---|---|
|
| ||||
| Aicardi-Goutières syndrome (AGS) | Three-prime repair exonuclease 1 (TREX1) dysfunction and consequential accumulation extrachromosomal DNA [88] | TREXl-mutant human ESCs generated by CRISPR and TREXl-mutant iPSCs from AGS patients | Astrocyte, neurons and brain organoids | Long Interspersed Element-1 retrotransposons is a major source of extranuclear DNA accumulation; TREX1-deficient astrocytes secrete neurotoxic type I interferons and exacerbate the neurotoxicity |
|
| ||||
| Alzheimer Disease (AD) | Astroglial contribution to AD [84] Role of astrocytes in AD pathogenesis [89] APOE4 effects on astrocytes [104] Role of oligomeric forms of amyloid-β peptide (Aβ) in the pathogenesis of AD [105] Defective fatty acid oxidation and mitochondrial dysfunctions in AD [106] Effects of sAD risk factor APOE4 on multiple neural cell types [107] Analyte production of amyloid β (Aβ) and soluble amyloid precursor proteinalpha (sAPPa) in fAD [108] Physiological function of APP in regulating lipoprotein homeostasis in astrocytes [109] |
iPSCs from patients with FAD carrying M146L mutation in PSEN1 and ApoE4+/+ sAD iPSCs of AD patients with PSEN1 ΔE9 mutation and PSEN1 ΔE9-corrected isogenic iPSCs by CRISPR APOE3 and APOE4 iPSCs from cognitively normal individuals iPSCs from patients with a familial amyloid precursor protein (APP)-E693Δ mutation and sAD iPSCs from AD patients with an amyloidogenic mutation in delta 9 exon of PSEN1 (PSEN1ΔE9) Two-way isogenic APOE3 and APOE4 iPSCs generated by CRISPR iPSCs from fAD patients CRISPR-knockout and mutant APP iPSCs |
Astrocytes Astrocyte-neuron co-culture Astrocyte-neuron co-culture Astrocyte-neuron co-culture Astrocytes Astrocytes Astrocytes, forebrain neurons, GABAergic interneurons Astrocytes |
Less complex morphology, atrophy and abnormal S100B localization in sAD and fAD astrocytes Altered mitochondrial metabolism followed with increases in oxidative stress in AD astrocytes; reduced calcium signaling activity of healthy neurons Fewer lipidated lipoprotein particles with less effectiveness in neurotrophic functions in APOE4 compared to APOE3 astrocytes Endoplasmic reticulum (ER) and oxidative stress in neurons and astrocytes due to Aβ oligomers accumulation; stress alleviated by docosahexaenoic acid (DHA) Impairment in fatty acid oxidation in human astrocytes; alleviated by PPARβ/5- agonism Altered cholesterol metabolism and impaired Aβ clearance in APOE4 iPSC-derived astrocytes Astrocytes secrete high levels of Aβ Full-length APP in astrocytes is required for cholesterol homeostasis and Aβ clearance |
|
| ||||
| Alexander Disease (AxD) | Effects of GFAP mutation on astrocytes and myelination [94] Effects of GFAP mutation on astrocytes [99] Effects of GFAP mutation on astrocytes [100] |
iPSCs from AxD patients and isogenic iPSCs generated by CRISPR iPSCs from AxD patients and isogenic iPSCs generated by CRISPR iPSCs of AxD patients |
Astrocyte-OPC or astrocyte-oligodendrocyte co-culture Astrocytes Astrocytes |
AxD patient iPSC-derived astrocytes inhibit OPC proliferation by secreting CHI3L1 and decrease oligodendrocyte myelination Impaired extracellular ATP release leading to attenuated calcium wave propagation Increased secretion of inflammatory factors |
|
| ||||
| Amyotrophic lateral sclerosis (ALS) | Astrocyte response to ALS motor neuron injury [82] Transactive response DNA-binding protein (TDP-43) proteinopathy [90] Effects of astrocytes from sALS patients on motor neurons [97] |
SOD1 D90A mutant iPSCs from ALS patients and isogenic SOD1 D90D control iPSCs TDP-43 mutation iPSCs from sALS patients |
Astrocytes Astrocyte-neuron co-culture Spinal neural progenitors transplanted into mouse spinal cord |
Failure of the neuroprotective EphB1-ephrin-B1-STAT3 signal transduction pathway in SOD1-mutant astrocytes Astrocyte cell autonomous effects: TDP-43 increase, subcellular mislocalization, and decreased cell survival Reduced number of non-motor neurons followed by reduced number of motor neurons in sALS-transplanted host spinal cord |
|
| ||||
| Autism spectrum disorder (ASD) | Interplay between neurons and astrocytes from nonsyndromic ASD individuals [101] | iPSCs from three individuals with non-syndromic ASD | Astrocyte-neuron co-culture | Neuronal synaptogenesis interference by ASD astrocytes due to augmented IL6 secretion |
|
| ||||
| Parkinson Disease (PD) | Role of astrocytes on the development of hiPSC-derived dopaminergic neurons [86] G2019S mutation in LRRK2 (leucine-rich repeat kinase 2), a risk factor for fPD [87] Effect of G2019S mutation in LRRK2 on fPD astrocytes [110] |
iPSCs from clinically normal individuals iPSCs from LRRK2 G2019S mutant fPD patients iPSCs from LRRK2 G2019S mutant fPD patients |
Astrocyte-neuron co-culture Astrocyte-neuron co-culture Astrocytes |
Dopaminergic neuron neurogenesis defects caused by mitochondrial respiratory chain disruption Impaired autophagy in PD astrocytes; PD astrocytes accumulate and transfer α-synuclein to healthy DA neurons Down-regulation of MMP2 and TGFB1 in LRRK2 G2019S mutant astrocytes |
|
| ||||
| Rett syndrome (RTT) | Non-cell autonomous role of RTT astrocytes in RTT pathogenesis [102] Mecp2-deficient astrocytes in RTT [111] |
iPSC from RTT patients with X-linked methyl-CpG binding protein 2 (MECP2) mutation and isogenic control ipSCs iPSC from RTT patients with MECP2 p.Arg294 mutation |
Astrocyte-neuron co-culture Astrocytes |
Astrocyte non-cell autonomous adverse effects on neuron morphology and electrophysiology Microtubule-dependent vesicle transport defects in MECP2-deficient astrocytes; rescued by Epothilone D treatment |
|
| ||||
| Schizophrenia (SCZ) | Effect of glial dysfunction on the pathogenesis of SCZ [37] | iPSC from childhood-onset SCZ patients | Glial progenitor cells transplanted into Shiverer mice | Delayed astrocytic differentiation and abnormal astrocytic morphologies |
3.1.1. Physiological Models
Progress has been made in studying the role of astrocytes in disease pathogenesis due to the development of astrocyte differentiation protocols from iPSCs [80, 81]. Different cellular models including human iPSC-derived astrocytes have been established to meet different needs, including astrocytes-only cultures [82–85], astrocytes co-cultured with other neural cell types [86–92], astrocyte-containing 3D organoids [93], and mouse models in which human astrocytes are transplanted [39].
These models have revealed novel links between disease-causing gene mutations and astrocyte functions, such as inflammatory cytokines secretion (e.g. IL1-b, IL6, TNF-a) [83, 84], decreased neurotrophic factor secretion (e.g. GDNF) [85], and neuroprotective signaling pathways (e.g. STAT3) [82]. In addition, astrocyte-neuron co-culture models have revealed a broad range of astrocytic effects on neuronal properties, for example, neurogenesis, neuronal survival, calcium signals and mitochondrial function in neurons [86–92]. 3D brain organoids are comprehensive models that have enriched the toolbox for studying neural diseases. Astrocytes are an essential component in brain organoids and have been shown to mimic fetal astrocyte features at the molecular and functional levels [93].
Interactions between astrocytes and other glial cells are relatively less studied in animal- and iPSC-based models. Modeling AxD is a good demonstration of the necessity of using iPSCs to improve our understanding on astrocyte interactions with other glial cells. AxD is a type of leukodystrophy disease caused by GFAP mutations. It is defined as a primary astrocyte disease based on its pathological hallmark of AxD, the Rosenthal fiber, which is only identified in astrocytes from patient brain tissues. Our group has recently established a human iPSC-based modeling system consisting of human iPSC-derived astrocytes and OPCs as well as oligodendrocytes to study astrocyte-OPC and astrocyte-oligodendrocyte interactions in the context of AxD [94, 95], which demonstrated a novel regulatory role of astrocytes in regulating OPC proliferation and oligodendrocyte myelination.
Additionally, emerging evidence suggests astrocytes alone could have important functions in memory formation through calcium signaling [96]. Overall, it would be interesting to study their roles in human species and disease context; however, it is challenging to confirm the functional output without an in vivo environment. To resolve this issue, studies by Windrem et al. and Qian et al. combined the advantages of iPSCs and mouse models [37, 39, 97] to establish glial chimeras by transplanting human iPSC-derived glial progenitor cells into mouse brains, demonstrating the effects of human astrocytes on animal behaviors, such as memory and sleep.
3.1.2. Neurodevelopmental and Neuropsychiatric Disorders
Using iPSC-derived astrocyte models, researchers have looked into the roles of astrocytes in neurodevelopmental and neuropsychiatric diseases. For example, Thomas et al. studied Aicardi-Goutières syndrome (AGS), an autoimmune disease that affects neurodevelopment, using iPSC-derived neural cells [88]. Dysfunction of three-prime repair exonuclease 1 (TREX1) in AGS causes an accumulation of extrachromosomal DNA with unknown mechanisms and impairs the intellectual and physical functions of the patients [98]. Using iPSC-derived neurons, astrocytes and 3D brain organoids, Thomas et al. identified endogenous Long Interspersed Element-1 retrotransposons as a major source of extranuclear DNA accumulation in neurons, astrocytes and neural progenitor cells (NPCs). They also found that TREX1-deficient human astrocytes could secrete neurotoxic type I interferons, which further exacerbate the neurotoxicity. Barbar et al. identified CD49f as a novel marker for functional astrocytes derived from iPSCs [92]. The same study showed that CD49f+ astrocytes could exhibit an A1 reactive state in response to inflammatory cytokine stimulation and cause neurotoxicity.
We have used a co-culture system consisting of human iPSC-derived astrocytes and OPCs as well as oligodendrocytes in combination with postmortem patient brain tissues to study AxD. We demonstrated that the AxD iPSC-derived astrocytes could inhibit OPC proliferation by secreting more CHI3L1 (Chitinase-3-like protein 1; also known as YKL-40), a neuroinflammatory molecule, which in turn contributes to decreased myelination. Interestingly, the expression patterns of CHI3L1 differ between mouse and human according to RNA-seq analysis of different brain cell types in these two species. In human, CHI3L1 is predominantly expressed in mature astrocytes. However, it is detected at similar levels in both astrocytes and OPCs in mouse [36]. This may indicate different roles of CHI3L1 in human and mouse brains that need to be considered when studying its function using mouse models. AxD has also been studied using iPSC-derived astrocytes alone by other groups. AxD-iPSC-derived astrocytes exhibit an increased secretion of inflammatory factors and impaired extracellular ATP release [99, 100]. Astrocytes derived from neuropsychiatric patient iPSCs, such as Autism spectrum disorder (ASD) and Rett Syndrome (RTT), showed adverse effects on neuronal functions, such as neuronal development and survival, neurite length, and synapse formation [101, 102]. Finally, the human-mouse glial chimera model mentioned in 3.1.1 was applied to study the contribution of astrocytes to the pathogenesis of schizophrenia (SCZ). The chimera mice exhibited delayed astrocyte maturation and abnormal behaviors, including excessive anxiety, antisocial traits and disturbed sleep [37].
3.1.3. Neurodegenerative Disorders
Neurodegenerative diseases, such as AD and PD, are under the spotlight of iPSC modeling because of the increasing recognition of human and rodent model differences. One PD model included both iPSC-derived astrocytes and dopaminergic neurons with or without the G2019S mutation in LRRK2 (leucine-rich repeat kinase 2), one of the most common disease-causing mutations for familial PD [87]. Through comparison of the two cellular systems in the presence or absence of the G2019S mutation, the authors demonstrated that astrocytes carrying the G2019S mutation could secret α-synuclein, a pathological hallmark of PD, and exert toxic effects on dopaminergic neurons.
AD risk factors such as APOE4 and familial genes such as PSEN1 and APP have been studied using iPSC-derived astrocyte models as well [89, 91, 102–106]. For example, Lin et al. differentiated multiple neural cell types, including astrocytes, from iPSCs carrying the APOE4 allele, which is a major risk factor for AD. A comparison with ApoE3 iPSC-derived astrocytes revealed altered cholesterol metabolism and impaired Aβ clearance in the ApoE4 iPSC-derived astrocytes [107]. Liao et al. provided evidence showing that astrocytes could secrete high levels of Aβ using single cell analytical technology [108]. Fong et al. generated isogenic APP-knockout and mutant iPSCs to study the physiological function of APP in regulating lipoprotein homeostasis in astrocytes and demonstrated that full-length APP in astrocytes is required for cholesterol homeostasis and Aβ clearance [109].
The amyotrophic lateral sclerosis (ALS) mutations SOD1 and TDP-43 have also been modeled using iPSC-derived astrocytes. These models demonstrated failed neuroprotection in SOD1-mutant astrocytes and decreased viability of TDP-43-mutant astrocytes, respectively [82, 90]. Qian et al. found astrocytes reduced the number of motor neurons after sporadic (s)ALS-iPSC-derived spinal NPCs were transplanted into mouse brain [97].
3.2. Oligodendrocytes
As the only cell type that performs the function of myelination, oligodendrocyte dysfunction is associated with a variety of CNS diseases. The causes of oligodendrocyte dysfunction could be due to gene mutations interrupting oligodendrocyte lineage differentiation, proliferation or myelination and are mainly seen in leukodystrophy diseases such as Canavan disease (CD) caused by ASPA gene mutations [112, 113], Krabe Disease (KD) caused by GalC mutations [114], Pelizaeus-Merzbacher disease (PMD) caused by PLP1 gene mutations [115, 116], and Vanishing White Matter disease (VWM) caused by EIF2B gene mutations [117]. Multiple sclerosis is the most common demyelinating disease caused by autoimmunity targeting myelin components, e.g. MBP and PLP [118]. White matter degeneration has been observed in many neurodegenerative and neuropsychiatric diseases, most often secondary to other insults, as OLs are very vulnerable to environmental disturbances such as oxidative stress due to a high metabolic rate, high iron level and low antioxidative glutathione level during maturation and myelination [119, 120].
3.2.1. Physiological Models
Due to the broad effects of oligodendrocyte dysfunctions on brain functions and many unknowns of human oligodendrocyte lineage development and functions, there are increasing studies using iPSCs to model oligodendrocyte defects (Table 2). Protocols for the differentiation of oligodendrocytes from iPSCs have been established by multiple groups following in vivo developmental principles [44, 121–123]. A co-culture model consisting of oligodendrocytes and neurons has been applied to examine the effects of gene mutations in oligodendrocytes on motor neuron survival [124]. Our group replaced neurons with nanofibers to study the interplay between astrocytes and OPCs/oligodendrocytes [94]. Recently, studies have successfully generated oligodendrocyte-containing 3D organoid/spheroid models, which also include neurons to provide scaffolds for myelination and astrocytes to interact with the oligodendrocyte lineage cells [125, 126].
Table 2.
Selected Studies Investigating Neurological Diseases Using iPSC-derived Oligodendrocytes
| Disease | Disease Features of Interest | Disease iPSC Characteristics | Cellular Model | Major Findings |
|---|---|---|---|---|
|
| ||||
| Alexander disease (AxD) | Effects of GFAP mutations on astrocytes and myelination [94] | iPSCs from AxD patients and isogenic iPSCs generated by CRISPR | Astrocyte-OPC and astrocyte-oligodendrocyte co-culture | AxD iPSC-derived astrocytes inhibit OPC proliferation by secreting CHI3L1 and decrease myelination |
|
| ||||
| Amyotrophic lateral sclerosis (ALS) | Role of oligodendrocytes in AL S [124] | iPSC from s/f ALS patients | Oligodendrocyte-motor neuron co-culture | Mutant superoxide dismutase 1 (SOD1) oligodendrocytes induce wild-type motor neuron death |
|
| ||||
| Pelizaeus-Merzbacher disease (PMD) | Oligodendrocyte dysfunction in PMD [126] Characterize phenotypes caused by different types of PLP1 mutations [128] Pathogenetic mechanism of PMD [129] |
iPSCs from a PMD patient carrying PLP1 c.254T>G point mutation and isogenic correction by CRISPR iPSCs from 12 PMD patients carrying PLP1 point mutation, duplication, or deletion iPSCs from PMD patients carrying PLP1 missense mutations 757 T > A and, 643 C > T |
Oligocortical spheriods Oligodendrocytes and OPCs Oligodendrocytes |
PLP1 point mutation leads to a reduced generation of oligodendrocytes and protein mislocalization associated with ER stress. Variable oligodendrocyte phenotypes across PLP1 mutation types; ER stress targeting improves a subset of mutation- induced defects Increased ER stress involved in the apoptosis of PMD oligodendrocytes |
|
| ||||
| Schizophrenia (SCZ) | Glial dysfunction contributes to the pathogenesis of SCZ [37] Abnormal oligodendrocyte development and function and lower myelination in SCZ brains [130] Genetic and cellular defects in SCZ brains [131] |
iPSC from childhood-onset SCZ patients iPSCs from SCZ patients iPSCs with Chondroitin Sulfate Proteoglycan 4 (CSPG4) mutations (c.391G > A and c.2702T > G) mutations (c.391G > A and c.2702T > G) |
Glial progenitor cells transplanted into Shiverer mice Oligodendrocytes OPCs |
Hypomyelination and behavioral abnormalities in SCZ chimeras Fewer oligodendrocytes in SCZ concordant with abnormalities seen by in vivo brain MR studies Mutant OPCs with abnormal morphology, impaired viability, and defective maturation to oligodendrocytes |
3.2.2. Neurodevelopmental and Neuropsychiatric Disorders
Marton et al. generated oligodendrocyte spheroids from iPSCs that mimic human primary oligodendrocytes [125]. These oligodendrocytes are electrophysiologically mature and can myelinate neuron axons. OL loss induced by lysolecithin treatment provides a proof of principle for studying leukodystrophy diseases using this spheroid model [125]. In addition to this study, Madhavan et al. developed an OL spheroid protocol that allowed the generation and maturation of OLs [126]. They also modeled a monogenic leukodystrophy PMD using oligodendrocyte spheroids. In addition to disease modeling, oligodendrocyte spheroids have been applied to study brain development. Kim et al. patterned region-specific organoids by activating or inhibiting the sonic hedgehog (SHH) signaling pathway, demonstrating the ventral and dorsal origins of human OPCs and the competitive advantage of dorsal OPCs over ventral OPCs [127]. Besides using the organoid approach to study oligodendrocyte lineage and model diseases, Windrem et al. combined the advantages of human iPSC-based disease models and animal models in their study of SCZ [37]. They transplanted human iPSC-derived glial progenitors into Shiverer mice that lack endogenous myelin and generated glia chimeras of human and mouse. When comparing chimeras that had SCZ or control iPSC-derived glia progenitors transplanted into Shiverer mice, the authors observed cell differentiation defects, hypomyelination and behavioral abnormalities in the SCZ chimeras, indicating the contribution of glial cells to the SCZ pathogenesis [37].
3.2.3. Neurodegenerative Disorders
Ferraiuolo et al. generated oligodendrocytes from sporadic and familial ALS patients and controls. They observed toxicity on motor neurons caused by ALS oligodendrocytes through lactate and potential soluble factors [124].
3.3. Microglia
The role of microglia in CNS diseases has been increasingly appreciated. As the resident immune cells in the brain, they are the mediators of inflammatory responses in the brain to various insults. Inflammation is almost a ubiquitous phenotype observed in neurodegenerative diseases, such as AD and PD, as well as CNS injuries, for example, cerebral ischemia and traumatic brain injury [132]. Overactivation of microglia has been observed in these diseases. Therefore, it is critical to study how human microglia respond to environmental changes and contribute to disease initiation and progression. Besides immune functions, microglia also sculpt neural circuits by engulfing synaptosomes and cleaning protein debris through phagocytosis. Because over-excitation is a key phenotype of many psychiatric diseases [133], such as SCZ and ASD, the dysfunction of microglia phagocytosis could be important for the pathogenesis of psychiatric disorders. We will review the application of iPSC-derived microglia for disease modeling below (summarized in Table 3).
Table 3.
Selected Studies Investigating Neurological Diseases Using iPSC-derived Microglia
| Disease | Disease Features of Interest | Disease iPSC Characteristics | Cellular Model | Major Findings |
|---|---|---|---|---|
|
| ||||
| Alzheimer disease (AD) | Effects of sAD risk factor APOE4 on multiple neural cell types [107] Effects of AD-predisposing genetic backgrounds including APOE4, PSEN1ΔE9, and APPswe on the functionality of iMGs[138] Effects of a surrogate brain environment on human microglia [143] |
Two-way isogenic APOE3 and APOE4 iPSCs generated by CRISPR APOE4, PSEN1ΔE9, and APPswe iPSCs; PSEN1ΔE9, and APPswe were corrected Isogenic iPSCs by CRISPR control iPSCs and isogenic TREM2 R47H mutant generated by CRISPR |
Microglia in vitro Microglia in vitro Microglia ex vivo (xMG; transplantation of iPSC-derived hematopoietic progenitors into the postnatal brain of humanized, immune-deficient mice) |
Morphological changes of APOE4 microglia with reduced Aβ phagocytosis Impaired phagocytosis, migration and metabolic activity and increased cytokine secretion in APOE4 microglia; PSEN1ΔE9 and APPswe had minor effects xMGs in contact with Aβ plaques adopted an activated, ameboid morphology |
|
| ||||
| Nasu-Hakola disease (NHD) | Role of TREM2 mutation in NHD [144] | iPSCs from NHD patients with TREM2 T66M and W50C mutations | Microglia in vitro | Impairment in the phagocytosis of apoptotic bodies, but not in Escherichia coli or zymosan substrate in mutant microglia |
|
| ||||
| ZIKA virus (ZIKV) infection | Effect of ZIKV on different neural cell types [140] Effects of microglia antiviral immune response on NPCs [139] |
iPSCs from one individual iPSCs from healthy individuals |
Microglia infected with MR766 strain of ZIKV and integrated into brain organoids Microglia infected with Brazil strain of ZIKV co-cultured with NPCs |
Microglia acted as the reservoir for ZIKV that invaded the neural tissue and initiated the infection Microglia infected by ZIKV transmitted the virus to NPCs and increased NPC apoptosis |
|
| ||||
| Schizophrenia (SCZ) | Synapse density reduction in SCZ brains [145] | iPSCs from SCZ patients | Mic roglia in vitro | Increased synapse elimination by SCZ microglia |
3.3.1. Physiological Models
To study human microglial functions and their roles in diseases in vitro, multiple protocols have been developed to derive microglia-like cells (iMGs) from human ESCs and iPSCs [134–137]. iMG-based models have been applied to study the phagocytotic capability of the cells for synapses in vitro [107, 138]. There are also studies focusing on the roles of microglia in early neural development by co-culturing iMGs with NPCs or integrating iMGs into 3D brain organoids [139, 140]. It is noteworthy that iMGs still differ from primary microglia in certain aspects. Microglia as an immune cell type are sensitive to in vitro culture conditions, therefore, microglia (both iMGs and primary microglia isolated from tissues) are prone to act in the active state rather than the resting state. Transcriptomic analysis of microglia cultured in vitro demonstrated that the cells exhibit significant changes of gene expression levels when cultured in vitro for 6 hours, including the down-regulation of microglia cell type-specific genes, such as TMEM119 and P2RY12, and up-regulation of microglia activation genes [5, 141]. These limitations have motivated studies aimed at maintaining ground-state microglia by transplanting iPSCs-derived intermediate progenitors into mouse brains. These iMG progenitors were transplanted into NOD-scid gamma (NSG) mice expressing human cytokines IL3, SCF, GM-CSF and CSF1 [142]. Under the in vivo environment, the microglia precursors not only matured into microglia that possess a similar morphology and gene expression signatures as primary microglia, but they also showed cellular heterogeneity, as evidenced by scRNA-seq analysis.
3.3.2. Neurodegenerative and Neuropsychiatric Disorders
Microglia have been shown to play significant roles in neurodegenerative and neuropsychiatric diseases [62, 133]. A study by Lin et al. examined multiple cell types derived from AD patient iPSCs carrying the APOE4 allele including iMGs and observed morphological changes of the ApoE4 microglia that is associated with reduced Aβ phagocytosis [107]. Konttinen et al. observed additional functional changes of ApoE4 iMGs, including impaired phagocytosis, migration and metabolic activity and increased cytokine secretion [138]. Hasselmann et al. transplanted iPSC-derived hematopoietic progenitors into mouse brains and observed microglia generation in vivo, which exhibited different gene signatures compared to mouse microglia in response to Aβ plaques [143]. Garcia-Reiboeck et al. modeled an early-onset dementia, named Nasu-Hakola disease (NHD), caused by homozygous TREM2 mutation using iMGs [144]. They found that the TREM2 mutation could reduce iMG survival and the phagocytosis of apoptotic bodies. Sellgren et al. investigated the role of microglia in SCZ using iMGs and found an increased elimination of synapses in patient iMGs that could be reduced using the antibiotic minocycline [145].
3.3.3. Environmental Factors
iMGs have also been used to investigate the effects of environmental factors, such as Zika virus (ZIKV), on neural function and development. Muffat et al. investigated the infection of ZIKV on different neural cell types using iPSC-derived cells, including NPCs, astrocytes, and microglia [140]. They identified microglia as the reservoir for ZIKV that invaded the neural tissues and initiated the infection, providing a mechanism for how ZIKV occurs at the early stage of pregnancy. Mesci et al. also investigated how the antiviral immune response of microglia could affect NPCs using a novel microglia-NPC co-culture system. They found microglia infected by ZIKV could transmit the virus to NPCs and increase NPC apoptosis. Importantly, they also demonstrated that Sofosbuvir (SOF), an FDA-approved drug against Hepatitis C infection, was able to reduce ZIKV-induced NPC death [139].
4. Future Directions
Glial cells derived from human iPSCs have expanded our knowledge about their importance in physiology and contributions to CNS diseases, especially in diseases that are challenging to study using animal models. Continuous efforts have been made to optimize in vitro culture conditions in order to better mimic the in vivo environment. An emerging new tool to achieve this goal is brain organoids, which are pluripotent stem cell-derived self-organized architectures composed of progenitors, neurons, and glial cells [146–151]. Brain organoids mimic many aspects of brain development, such as the neuroepithelial structure and molecular and cellular developmental trajectories [152], therefore, they are a promising tool for researchers to study multiple neural cell type interactions under physiological and disease conditions. A number of studies have described protocols to generate astrocytes, oligodendrocytes and microglia in brain organoids and studied their features during development or under disease conditions [93, 125, 126, 153]. Sloan et al. took advantage of the 3D organoid structure to study the transcriptional and functional maturation of human astrocytes, which has been challenging due to the difficulty of obtaining human brain tissues at early and late gestation stages [93]. They observed transcriptional maturation from fetal to mature astrocytes over time, accompanied with functional alterations such as a decrease in the capacity to phagocytose synapses. These findings will facilitate the application of iPSC-derived astrocytes to development and disease research. Because microglia originate from the hematopoietic system, which is from the mesodermal layer of the embryo, whereas other CNS neural cell types originate from the neuroectoderm layer, the majority of brain organoid protocols do not generate microglia. However, the flexibility of in vitro cultures allows integrating microglia into brain organoids [107, 134] to study neuro-immune interactions. A recent study has described a protocol that generates cells with typical microglial molecular phenotype, morphology, and function in brain organoids [153]. As another important component of the brain, the vasculature can be integrated into brain organoids in vivo [154] and in vitro [155], which improves the nutrient supply and growth potential of brain organoids and enables modeling interactions between the neural and blood systems. Glial cells are highly heterogenous. One of the biggest challenges of using iPSC-derived glial cells to study CNS diseases is to accurately generate spatially and temporally defined glial subtypes. It is widely acknowledged that specific neuron subtypes are needed to study diseases that affect corresponding brain regions. Accordingly, glial subtypes are also critical to accurately study the role of the glial type in disease. Due to the heterogeneity, different subtypes of glial cells possess different transcript profiles and perform distinct functions in their territories at specific developmental timing. White-matter disease models need white-matter glial cells to study glial cell interactions with each other and neurons. Similar to the establishment of cell-type differentiation protocols, which benefits from developmental biology, the progress of generating subtypes of glial cells from iPSCs will also greatly rely on the progress of glial biology through in vivo systems such as animal models and primary tissues.
Already, there is an increasing number of studies aimed at generating specific subtypes of glial cells, mainly astrocytes, to model diseases affected by the corresponding regions of the CNS. Krencik et al. has patterned human pluripotent stem cell-derived astrocytes to rostral-caudal and dorsal-ventral identities by supplementing the morphogens retinoic acid and SHH [156]. A recent study by the same group further refined the region-specific astrocyte differentiation from human pluripotent stem cells and demonstrated gene signature specificities and functional diversities of astrocyte subtypes. Xiang et al. developed a fast protocol for generating subtypes of astrocytes by introducing the astrocyte transcriptional factors NFIA and SOX9 into regionally patterned NPCs [157]. These studies have pioneered the work of generating defined astrocyte subtypes. A recent study generated white-matter and grey-matter astrocytes from both human and mouse iPSCs to model the leukodystrophy disease VWM by co-culturing the astrocytes with mouse oligodendrocytes. That study demonstrated the inhibitory effects on oligodendrocyte maturation specifically by white-matter astrocytes [158]. We expect more studies in the near future will generate defined glial cell subtypes and delineate the subtype-specific role of astrocytes in CNS diseases.
With the refinement of glial cell regionality, identity, and functionality, modeling CNS diseases using iPSC-derived glial cells will become increasingly accurate and informative, which will also benefit drug discovery. In addition, iPSC-derived glial cells will benefit glial biology by providing us more information about the species-specific features of human glial cell functions and helping us understand how the evolution of glial cells contributed to the divergence of human brains from that of the other species.
Highlights.
Glial cell heterogeneity and species specificity
iPSC-derived astrocytes in neurological diseases
iPSC-derived oligodendrocytes in neurological diseases
iPSC-derived microglia in neurological diseases
Acknowledgements
We thank Dr. P. Karagiannis for editing the manuscript. This work was supported by the Herbert Horvitz Family, Sidell Kagan Foundation, California Institute for Regenerative Medicine TRAN1-08525, and the National Institute of Aging of the National Institutes of Health R01 AG056305, RF1 AG061794, and R56 AG061171. Research reported in this publication was also supported by the National Cancer Institute of the National Institutes of Health under award number P30CA33572. The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References:
- 1.Sofroniew MV and Vinters HV, Astrocytes: biology and pathology. Acta Neuropathol, 2010. 119(1): p. 7–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Bradl M. and Lassmann H, Oligodendrocytes: biology and pathology. Acta Neuropathol, 2010. 119(1): p. 37–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Sominsky L, De Luca S, and Spencer SJ, Microglia: Key players in neurodevelopment and neuronal plasticity. Int J Biochem Cell Biol, 2018. 94: p. 56–60. [DOI] [PubMed] [Google Scholar]
- 4.Oberheim NA, et al. , Astrocytic complexity distinguishes the human brain. Trends Neurosci, 2006. 29(10): p. 547–53. [DOI] [PubMed] [Google Scholar]
- 5.Gosselin D, et al. , An environment-dependent transcriptional network specifies human microglia identity. Science, 2017. 356(6344). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Galatro TF, et al. , Transcriptomic analysis of purified human cortical microglia reveals age-associated changes. Nat Neurosci, 2017. 20(8): p. 1162–1171. [DOI] [PubMed] [Google Scholar]
- 7.Takahashi K, et al. , Induction of pluripotent stem cells from adult human fibroblasts by defined factors. Cell, 2007. 131(5): p. 861–72. [DOI] [PubMed] [Google Scholar]
- 8.Shi Y, et al. , Induced pluripotent stem cell technology: a decade of progress. Nat Rev Drug Discov, 2017. 16(2): p. 115–130. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Liddelow SA, et al. , Neurotoxic reactive astrocytes are induced by activated microglia. Nature, 2017. 541(7638): p. 481–487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Clarke LE, et al. , Normal aging induces A1-like astrocyte reactivity. Proc Natl Acad Sci U S A, 2018. 115(8): p. E1896–E1905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Anderson MA, Ao Y, and Sofroniew MV, Heterogeneity of reactive astrocytes. Neurosci Lett, 2014. 565: p. 23–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Allen NJ and Eroglu C, Cell Biology of Astrocyte-Synapse Interactions. Neuron, 2017. 96(3): p. 697–708. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Pfrieger FW and Barres BA, Synaptic efficacy enhanced by glial cells in vitro. Science, 1997. 277(5332): p. 1684–7. [DOI] [PubMed] [Google Scholar]
- 14.Colon-Ramos DA, Margeta MA, and Shen K, Glia promote local synaptogenesis through UNC-6 (netrin) signaling in C. elegans. Science, 2007. 318(5847): p. 103–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Muthukumar AK, Stork T, and Freeman MR, Activity-dependent regulation of astrocyte GAT levels during synaptogenesis. Nat Neurosci, 2014. 17(10): p. 1340–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Ullian EM, et al. , Control of synapse number by glia. Science, 2001. 291(5504): p. 657–61. [DOI] [PubMed] [Google Scholar]
- 17.Elmariah SB, et al. , Astrocytes regulate inhibitory synapse formation via Trk-mediated modulation of postsynaptic GABAA receptors. J Neurosci, 2005. 25(14): p. 3638–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Cuevas ME, et al. , The presence of glia stimulates the appearance of glycinergic synaptic transmission in spinal cord neurons. Mol Cell Neurosci, 2005. 28(4): p. 770–8. [DOI] [PubMed] [Google Scholar]
- 19.Cao G. and Ko CP, Schwann cell-derived factors modulate synaptic activities at developing neuromuscular synapses. J Neurosci, 2007. 27(25): p. 6712–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Goritz C, Mauch DH, and Pfrieger FW, Multiple mechanisms mediate cholesterol-induced synaptogenesis in a CNS neuron. Mol Cell Neurosci, 2005. 29(2): p. 190–201. [DOI] [PubMed] [Google Scholar]
- 21.Mauch DH, et al. , CNS synaptogenesis promoted by glia-derived cholesterol. Science, 2001. 294(5545): p. 1354–7. [DOI] [PubMed] [Google Scholar]
- 22.Christopherson KS, et al. , Thrombospondins are astrocyte-secreted proteins that promote CNS synaptogenesis. Cell, 2005. 120(3): p. 421–33. [DOI] [PubMed] [Google Scholar]
- 23.Kucukdereli H, et al. , Control of excitatory CNS synaptogenesis by astrocyte-secreted proteins Hevin and SPARC. Proc Natl Acad Sci U S A, 2011. 108(32): p. E440–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Blanco-Suarez E, et al. , Astrocyte-Secreted Chordin-like 1 Drives Synapse Maturation and Limits Plasticity by Increasing Synaptic GluA2 AMPA Receptors. Neuron, 2018. 100(5): p. 1116–1132 e13. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Chung WS, et al. , Astrocytes mediate synapse elimination through MEGF10 and MERTK pathways. Nature, 2013. 504(7480): p. 394–400. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Vainchtein ID, et al. , Astrocyte-derived interleukin-33 promotes microglial synapse engulfment and neural circuit development. Science, 2018. 359(6381): p. 1269–1273. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Araque A, et al. , Tripartite synapses: glia, the unacknowledged partner. Trends Neurosci, 1999. 22(5): p. 208–15. [DOI] [PubMed] [Google Scholar]
- 28.Perea G, Navarrete M, and Araque A, Tripartite synapses: astrocytes process and control synaptic information. Trends Neurosci, 2009. 32(8): p. 421–31. [DOI] [PubMed] [Google Scholar]
- 29.Angelova PR, et al. , Functional Oxygen Sensitivity of Astrocytes. J Neurosci, 2015. 35(29): p. 10460–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Garcia-Caceres C, et al. , Astrocytic Insulin Signaling Couples Brain Glucose Uptake with Nutrient Availability. Cell, 2016. 166(4): p. 867–880. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Marina N, et al. , Brain metabolic sensing and metabolic signaling at the level of an astrocyte. Glia, 2018. 66(6): p. 1185–1199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Rose CR, et al. , Astroglial Glutamate Signaling and Uptake in the Hippocampus. Front Mol Neurosci, 2017. 10: p. 451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Dong XX, Wang Y, and Qin ZH, Molecular mechanisms of excitotoxicity and their relevance to pathogenesis of neurodegenerative diseases. Acta Pharmacol Sin, 2009. 30(4): p. 379–87. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Daneman R. and Prat A, The blood-brain barrier. Cold Spring Harb Perspect Biol, 2015. 7(1): p. a020412. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Verkman AS, Aquaporin water channels and endothelial cell function. J Anat, 2002. 200(6): p. 617–27. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Zhang Y, et al. , Purification and Characterization of Progenitor and Mature Human Astrocytes Reveals Transcriptional and Functional Differences with Mouse. Neuron, 2016. 89(1): p. 37–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Windrem MS, et al. , Human iPSC Glial Mouse Chimeras Reveal Glial Contributions to Schizophrenia. Cell Stem Cell, 2017. 21(2): p. 195–208 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Chen H, et al. , Human-derived neural progenitors functionally replace astrocytes in adult mice. J Clin Invest, 2015. 125(3): p. 1033–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Windrem MS, et al. , A competitive advantage by neonatally engrafted human glial progenitors yields mice whose brains are chimeric for human glia. J Neurosci, 2014. 34(48): p. 16153–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Rivers LE, et al. , PDGFRA/NG2 glia generate myelinating oligodendrocytes and piriform projection neurons in adult mice. Nat Neurosci, 2008. 11(12): p. 1392–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Bansal R, et al. , Multiple and novel specificities of monoclonal antibodies O1, O4, and R-mAb used in the analysis of oligodendrocyte development. J Neurosci Res, 1989. 24(4): p. 548–57. [DOI] [PubMed] [Google Scholar]
- 42.Sommer I. and Schachner M, Monoclonal antibodies (O1 to O4) to oligodendrocyte cell surfaces: an immunocytological study in the central nervous system. Dev Biol, 1981. 83(2): p. 311–27. [DOI] [PubMed] [Google Scholar]
- 43.Emery B, et al. , Myelin gene regulatory factor is a critical transcriptional regulator required for CNS myelination. Cell, 2009. 138(1): p. 172–85. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Goldman SA and Kuypers NJ, How to make an oligodendrocyte. Development, 2015. 142(23): p. 3983–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Nave KA and Werner HB, Myelination of the nervous system: mechanisms and functions. Annu Rev Cell Dev Biol, 2014. 30: p. 503–33. [DOI] [PubMed] [Google Scholar]
- 46.Liu J, et al. , Impaired adult myelination in the prefrontal cortex of socially isolated mice. Nat Neurosci, 2012. 15(12): p. 1621–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Makinodan M, et al. , A critical period for social experience-dependent oligodendrocyte maturation and myelination. Science, 2012. 337(6100): p. 1357–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Philips T. and Rothstein JD, Oligodendroglia: metabolic supporters of neurons. J Clin Invest, 2017. 127(9): p. 3271–3280. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Lee Y, et al. , Oligodendroglia metabolically support axons and contribute to neurodegeneration. Nature, 2012. 487(7408): p. 443–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Funfschilling U, et al. , Glycolytic oligodendrocytes maintain myelin and long-term axonal integrity. Nature, 2012. 485(7399): p. 517–21. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Marques S, et al. , Oligodendrocyte heterogeneity in the mouse juvenile and adult central nervous system. Science, 2016. 352(6291): p. 1326–1329. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Spitzer SO, et al. , Oligodendrocyte Progenitor Cells Become Regionally Diverse and Heterogeneous with Age. Neuron, 2019. 101(3): p. 459–471 e5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Jakel S, et al. , Altered human oligodendrocyte heterogeneity in multiple sclerosis. Nature, 2019. 566(7745): p. 543–547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Yeung MS, et al. , Dynamics of oligodendrocyte generation and myelination in the human brain. Cell, 2014. 159(4): p. 766–74. [DOI] [PubMed] [Google Scholar]
- 55.Ginhoux F, et al. , Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science, 2010. 330(6005): p. 841–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Monier A, et al. , Entry and distribution of microglial cells in human embryonic and fetal cerebral cortex. J Neuropathol Exp Neurol, 2007. 66(5): p. 372–82. [DOI] [PubMed] [Google Scholar]
- 57.Monier A, et al. , Distribution and differentiation of microglia in the human encephalon during the first two trimesters of gestation. J Comp Neurol, 2006. 499(4): p. 565–82. [DOI] [PubMed] [Google Scholar]
- 58.Verney C, et al. , Early microglial colonization of the human forebrain and possible involvement in periventricular white-matter injury of preterm infants. J Anat, 2010. 217(4): p. 436–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59.Erny D, et al. , Host microbiota constantly control maturation and function of microglia in the CNS. Nat Neurosci, 2015. 18(7): p. 965–77. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Bennett FC, et al. , A Combination of Ontogeny and CNS Environment Establishes Microglial Identity. Neuron, 2018. 98(6): p. 1170–1183 e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Matcovitch-Natan O, et al. , Microglia development follows a stepwise program to regulate brain homeostasis. Science, 2016. 353(6301): p. aad8670. [DOI] [PubMed] [Google Scholar]
- 62.Colonna M. and Butovsky O, Microglia Function in the Central Nervous System During Health and Neurodegeneration. Annu Rev Immunol, 2017. 35: p. 441–468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 63.Cunningham CL, Martinez-Cerdeno V, and Noctor SC, Microglia regulate the number of neural precursor cells in the developing cerebral cortex. J Neurosci, 2013. 33(10): p. 4216–33. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Wakselman S, et al. , Developmental neuronal death in hippocampus requires the microglial CD11b integrin and DAP12 immunoreceptor. J Neurosci, 2008. 28(32): p. 8138–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Hoshiko M, et al. , Deficiency of the microglial receptor CX3CR1 impairs postnatal functional development of thalamocortical synapses in the barrel cortex. J Neurosci, 2012. 32(43): p. 15106–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Bialas AR and Stevens B, TGF-beta signaling regulates neuronal C1q expression and developmental synaptic refinement. Nat Neurosci, 2013. 16(12): p. 1773–82. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 67.Stevens B, et al. , The classical complement cascade mediates CNS synapse elimination. Cell, 2007. 131(6): p. 1164–78. [DOI] [PubMed] [Google Scholar]
- 68.Lampron A, et al. , Inefficient clearance of myelin debris by microglia impairs remyelinating processes. J Exp Med, 2015. 212(4): p. 481–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Cherry JD, Olschowka JA, and O’Banion MK, Neuroinflammation and M2 microglia: the good, the bad, and the inflamed. J Neuroinflammation, 2014. 11: p. 98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Michell-Robinson MA, et al. , Roles of microglia in brain development, tissue maintenance and repair. Brain, 2015. 138(Pt 5): p. 1138–59. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Mantovani A, et al. , The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol, 2004. 25(12): p. 677–86. [DOI] [PubMed] [Google Scholar]
- 72.Edwards JP, et al. , Biochemical and functional characterization of three activated macrophage populations. J Leukoc Biol, 2006. 80(6): p. 1298–307. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Li Q, et al. , Developmental Heterogeneity of Microglia and Brain Myeloid Cells Revealed by Deep Single-Cell RNA Sequencing. Neuron, 2019. 101(2): p. 207–223 e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Masuda T, et al. , Spatial and temporal heterogeneity of mouse and human microglia at single-cell resolution. Nature, 2019. 566(7744): p. 388–392. [DOI] [PubMed] [Google Scholar]
- 75.Phatnani H. and Maniatis T, Astrocytes in neurodegenerative disease. Cold Spring Harb Perspect Biol, 2015. 7(6). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Burda JE and Sofroniew MV, Reactive gliosis and the multicellular response to CNS damage and disease. Neuron, 2014. 81(2): p. 229–48. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Sofroniew MV, Astrogliosis. Cold Spring Harb Perspect Biol, 2014. 7(2): p. a020420. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Messing A, et al. , Alexander Disease. J Neurosci, 2012. 32(15): p. 5017–5023. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Olabarria M. and Goldman JE, Disorders of Astrocytes: Alexander Disease as a Model. Annu Rev Pathol, 2017. 12: p. 131–152. [DOI] [PubMed] [Google Scholar]
- 80.Li L, Chao J, and Shi Y, Modeling neurological diseases using iPSC-derived neural cells : iPSC modeling of neurological diseases. Cell Tissue Res, 2018. 371(1): p. 143–151. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.de Majo M, et al. , An update on human astrocytes and their role in development and disease. Glia, 2020. [DOI] [PubMed] [Google Scholar]
- 82.Tyzack GE, et al. , A neuroprotective astrocyte state is induced by neuronal signal EphB1 but fails in ALS models. Nat Commun, 2017. 8(1): p. 1164. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Perriot S, et al. , Human Induced Pluripotent Stem Cell-Derived Astrocytes Are Differentially Activated by Multiple Sclerosis-Associated Cytokines. Stem Cell Reports, 2018. 11(5): p. 1199–1210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Jones VC, et al. , Aberrant iPSC-derived human astrocytes in Alzheimer’s disease. Cell Death Dis, 2017. 8(3): p. e2696. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.McGivern JV, et al. , Spinal muscular atrophy astrocytes exhibit abnormal calcium regulation and reduced growth factor production. Glia, 2013. 61(9): p. 1418–1428. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Du F, et al. , Astrocytes Attenuate Mitochondrial Dysfunctions in Human Dopaminergic Neurons Derived from iPSC. Stem Cell Reports, 2018. 10(2): p. 366–374. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.di Domenico A, et al. , Patient-Specific iPSC-Derived Astrocytes Contribute to Non-Cell-Autonomous Neurodegeneration in Parkinson’s Disease. Stem Cell Reports, 2019. 12(2): p. 213–229. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Thomas CA, et al. , Modeling of TREX1-Dependent Autoimmune Disease using Human Stem Cells Highlights L1 Accumulation as a Source of Neuroinflammation. Cell Stem Cell, 2017. 21(3): p. 319–331 e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Oksanen M, et al. , PSEN1 Mutant iPSC-Derived Model Reveals Severe Astrocyte Pathology in Alzheimer’s Disease. Stem Cell Reports, 2017. 9(6): p. 1885–1897. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Serio A, et al. , Astrocyte pathology and the absence of non-cell autonomy in an induced pluripotent stem cell model of TDP-43 proteinopathy. Proc Natl Acad Sci U S A, 2013. 110(12): p. 4697–702. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Russo FB, et al. , Modeling the Interplay Between Neurons and Astrocytes in Autism Using Human Induced Pluripotent Stem Cells. Biol Psychiatry, 2018. 83(7): p. 569–578. [DOI] [PubMed] [Google Scholar]
- 92.Barbar L, et al. , CD49f Is a Novel Marker of Functional and Reactive Human iPSC-Derived Astrocytes. Neuron, 2020. 107(3): p. 436–453 e12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Sloan SA, et al. , Human Astrocyte Maturation Captured in 3D Cerebral Cortical Spheroids Derived from Pluripotent Stem Cells. Neuron, 2017. 95(4): p. 779–790 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Li L, et al. , GFAP Mutations in Astrocytes Impair Oligodendrocyte Progenitor Proliferation and Myelination in an hiPSC Model of Alexander Disease. Cell Stem Cell, 2018. 23(2): p. 239–251 e6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Sofroniew MV, Stem-Cell-Derived Astrocytes Divulge Secrets of Mutant GFAP. Cell Stem Cell, 2018. 23(5): p. 630–631. [DOI] [PubMed] [Google Scholar]
- 96.Adamsky A, et al. , Astrocytic Activation Generates De Novo Neuronal Potentiation and Memory Enhancement. Cell, 2018. 174(1): p. 59–71 e14. [DOI] [PubMed] [Google Scholar]
- 97.Qian K, et al. , Sporadic ALS Astrocytes Induce Neuronal Degeneration In Vivo. Stem Cell Reports, 2017. 8(4): p. 843–855. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 98.Stetson DB, et al. , Trex1 prevents cell-intrinsic initiation of autoimmunity. Cell, 2008. 134(4): p. 587–98. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Jones JR, et al. , Mutations in GFAP Disrupt the Distribution and Function of Organelles in Human Astrocytes. Cell Rep, 2018. 25(4): p. 947–958 e4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Kondo T, et al. , Modeling Alexander disease with patient iPSCs reveals cellular and molecular pathology of astrocytes. Acta Neuropathol Commun, 2016. 4(1): p. 69. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Krasovska V. and Doering LC, Regulation of IL-6 Secretion by Astrocytes via TLR4 in the Fragile X Mouse Model. Front Mol Neurosci, 2018. 11: p. 272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Williams EC, et al. , Mutant astrocytes differentiated from Rett syndrome patients-specific iPSCs have adverse effects on wild-type neurons. Hum Mol Genet, 2014. 23(11): p. 2968–80. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Wada T, et al. , Amyotrophic lateral sclerosis model derived from human embryonic stem cells overexpressing mutant superoxide dismutase 1. Stem Cells Transl Med, 2012. 1(5): p. 396–402. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Zhao J, et al. , APOE epsilon4/epsilon4 diminishes neurotrophic function of human iPSC-derived astrocytes. Hum Mol Genet, 2017. 26(14): p. 2690–2700. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Kondo T, et al. , Modeling Alzheimer’s Disease with iPSCs Reveals Stress Phenotypes Associated with Intracellular Abeta and Differential Drug Responsiveness. Cell Stem Cell, 2013. 12(4): p. 487–96. [DOI] [PubMed] [Google Scholar]
- 106.Konttinen H, et al. , PPARbeta/delta-agonist GW0742 ameliorates dysfunction in fatty acid oxidation in PSEN1DeltΔE9 astrocytes. Glia, 2019. 67(1): p. 146–159. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Lin YT, et al. , APOE4 Causes Widespread Molecular and Cellular Alterations Associated with Alzheimer’s Disease Phenotypes in Human iPSC-Derived Brain Cell Types. Neuron, 2018. 98(6): p. 1141–1154 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Liao MC, et al. , Single-Cell Detection of Secreted Abeta and sAPPalpha from Human IPSC-Derived Neurons and Astrocytes. J Neurosci, 2016. 36(5): p. 1730–46. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Fong LK, et al. , Full-length amyloid precursor protein regulates lipoprotein metabolism and amyloid-beta clearance in human astrocytes. J Biol Chem, 2018. 293(29): p. 11341–11357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Booth HDE, et al. , RNA sequencing reveals MMP2 and TGFB1 downregulation in LRRK2 G2019S Parkinson’s iPSC-derived astrocytes. Neurobiol Dis, 2019. 129: p. 56–66. [DOI] [PubMed] [Google Scholar]
- 111.Delepine C, et al. , Altered microtubule dynamics and vesicular transport in mouse and human MeCP2-deficient astrocytes. Hum Mol Genet, 2016. 25(1): p. 146–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Matalon R, et al. , Aspartoacylase deficiency and N-acetylaspartic aciduria in patients with Canavan disease. Am J Med Genet, 1988. 29(2): p. 463–71. [DOI] [PubMed] [Google Scholar]
- 113.Surendran S, et al. , Canavan disease: a monogenic trait with complex genomic interaction. Mol Genet Metab, 2003. 80(1–2): p. 74–80. [DOI] [PubMed] [Google Scholar]
- 114.Wenger DA, et al. , Krabbe disease: genetic aspects and progress toward therapy. Mol Genet Metab, 2000. 70(1): p. 1–9. [DOI] [PubMed] [Google Scholar]
- 115.Inoue K, PLP1-related inherited dysmyelinating disorders: Pelizaeus-Merzbacher disease and spastic paraplegia type 2. Neurogenetics, 2005. 6(1): p. 1–16. [DOI] [PubMed] [Google Scholar]
- 116.Hudson LD, et al. , Mutation of the proteolipid protein gene PLP in a human X chromosome-linked myelin disorder. Proc Natl Acad Sci U S A, 1989. 86(20): p. 8128–31. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Leegwater PA, et al. , The gene for leukoencephalopathy with vanishing white matter is located on chromosome 3q27. Am J Hum Genet, 1999. 65(3): p. 728–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Steinman L, Multiple sclerosis: a coordinated immunological attack against myelin in the central nervous system. Cell, 1996. 85(3): p. 299–302. [DOI] [PubMed] [Google Scholar]
- 119.Giacci M. and Fitzgerald M, Oligodendroglia Are Particularly Vulnerable to Oxidative Damage After Neurotrauma In Vivo. J Exp Neurosci, 2018. 12: p. 1179069518810004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Butts BD, Houde C, and Mehmet H, Maturation-dependent sensitivity of oligodendrocyte lineage cells to apoptosis: implications for normal development and disease. Cell Death Differ, 2008. 15(7): p. 1178–86. [DOI] [PubMed] [Google Scholar]
- 121.Czepiel M, et al. , Differentiation of induced pluripotent stem cells into functional oligodendrocytes. Glia, 2011. 59(6): p. 882–92. [DOI] [PubMed] [Google Scholar]
- 122.Wang S, et al. , Human iPSC-derived oligodendrocyte progenitor cells can myelinate and rescue a mouse model of congenital hypomyelination. Cell Stem Cell, 2013. 12(2): p. 252–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Douvaras P, et al. , Efficient generation of myelinating oligodendrocytes from primary progressive multiple sclerosis patients by induced pluripotent stem cells. Stem Cell Reports, 2014. 3(2): p. 250–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Ferraiuolo L, et al. , Oligodendrocytes contribute to motor neuron death in ALS via SOD1-dependent mechanism. Proc Natl Acad Sci U S A, 2016. 113(42): p. E6496–E6505. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Marton RM, et al. , Differentiation and maturation of oligodendrocytes in human three-dimensional neural cultures. Nat Neurosci, 2019. 22(3): p. 484–491. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Madhavan M, et al. , Induction of myelinating oligodendrocytes in human cortical spheroids. Nat Methods, 2018. 15(9): p. 700–706. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Kim H, et al. , Pluripotent Stem Cell-Derived Cerebral Organoids Reveal Human Oligodendrogenesis with Dorsal and Ventral Origins. Stem Cell Reports, 2019. 12(5): p. 890–905. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Nevin ZS, et al. , Modeling the Mutational and Phenotypic Landscapes of Pelizaeus-Merzbacher Disease with Human iPSC-Derived Oligodendrocytes. Am J Hum Genet, 2017. 100(4): p. 617–634. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 129.Numasawa-Kuroiwa Y, et al. , Involvement of ER stress in dysmyelination of Pelizaeus-Merzbacher Disease with PLP1 missense mutations shown by iPSC-derived oligodendrocytes. Stem Cell Reports, 2014. 2(5): p. 648–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.McPhie DL, et al. , Oligodendrocyte differentiation of induced pluripotent stem cells derived from subjects with schizophrenias implicate abnormalities in development. Transl Psychiatry, 2018. 8(1): p. 230. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.de Vrij FM, et al. , Candidate CSPG4 mutations and induced pluripotent stem cell modeling implicate oligodendrocyte progenitor cell dysfunction in familial schizophrenia. Mol Psychiatry, 2019. 24(5): p. 757–771. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Skaper SD, et al. , An Inflammation-Centric View of Neurological Disease: Beyond the Neuron. Front Cell Neurosci, 2018. 12: p. 72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Mondelli V, et al. , Brain microglia in psychiatric disorders. Lancet Psychiatry, 2017. 4(7): p. 563–572. [DOI] [PubMed] [Google Scholar]
- 134.Abud EM, et al. , iPSC-Derived Human Microglia-like Cells to Study Neurological Diseases. Neuron, 2017. 94(2): p. 278–293 e9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Douvaras P, et al. , Directed Differentiation of Human Pluripotent Stem Cells to Microglia. Stem Cell Reports, 2017. 8(6): p. 1516–1524. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Muffat J, et al. , Efficient derivation of microglia-like cells from human pluripotent stem cells. Nat Med, 2016. 22(11): p. 1358–1367. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Pandya H, et al. , Differentiation of human and murine induced pluripotent stem cells to microglia-like cells. Nat Neurosci, 2017. 20(5): p. 753–759. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Konttinen H, et al. , PSEN1DeltΔE9, APPswe, and APOE4 Confer Disparate Phenotypes in Human iPSC-Derived Microglia. Stem Cell Reports, 2019. 13(4): p. 669–683. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Mesci P, et al. , Modeling neuro-immune interactions during Zika virus infection. Hum Mol Genet, 2018. 27(1): p. 41–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Muffat J, et al. , Human induced pluripotent stem cell-derived glial cells and neural progenitors display divergent responses to Zika and dengue infections. Proc Natl Acad Sci U S A, 2018. 115(27): p. 7117–7122. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Bohlen CJ, et al. , Diverse Requirements for Microglial Survival, Specification, and Function Revealed by Defined-Medium Cultures. Neuron, 2017. 94(4): p. 759–773 e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Svoboda DS, et al. , Human iPSC-derived microglia assume a primary microglia-like state after transplantation into the neonatal mouse brain. Proc Natl Acad Sci U S A, 2019. 116(50): p. 25293–25303. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Hasselmann J, et al. , Development of a Chimeric Model to Study and Manipulate Human Microglia In Vivo. Neuron, 2019. 103(6): p. 1016–1033 e10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Garcia-Reitboeck P, et al. , Human Induced Pluripotent Stem Cell-Derived Microglia-Like Cells Harboring TREM2 Missense Mutations Show Specific Deficits in Phagocytosis. Cell Rep, 2018. 24(9): p. 2300–2311. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Sellgren CM, et al. , Increased synapse elimination by microglia in schizophrenia patient-derived models of synaptic pruning. Nat Neurosci, 2019. 22(3): p. 374–385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Lancaster MA, et al. , Cerebral organoids model human brain development and microcephaly. Nature, 2013. 501(7467): p. 373–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Pasca AM, et al. , Functional cortical neurons and astrocytes from human pluripotent stem cells in 3D culture. Nat Methods, 2015. 12(7): p. 671–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Qian X, et al. , Brain-Region-Specific Organoids Using Mini-bioreactors for Modeling ZIKV Exposure. Cell, 2016. 165(5): p. 1238–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 149.Quadrato G, et al. , Cell diversity and network dynamics in photosensitive human brain organoids. Nature, 2017. 545(7652): p. 48–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Camp JG, et al. , Human cerebral organoids recapitulate gene expression programs of fetal neocortex development. Proc Natl Acad Sci U S A, 2015. 112(51): p. 15672–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Cugola FR, et al. , The Brazilian Zika virus strain causes birth defects in experimental models. Nature, 2016. 534(7606): p. 267–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 152.Trujillo CA, et al. , Complex Oscillatory Waves Emerging from Cortical Organoids Model Early Human Brain Network Development. Cell Stem Cell, 2019. 25(4): p. 558–569 e7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Ormel PR, et al. , Microglia innately develop within cerebral organoids. Nat Commun, 2018. 9(1): p. 4167. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Mansour AA, et al. , An in vivo model of functional and vascularized human brain organoids. Nat Biotechnol, 2018. 36(5): p. 432–441. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Pham MT, et al. , Generation of human vascularized brain organoids. Neuroreport, 2018. 29(7): p. 588–593. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156.Krencik R, et al. , Specification of transplantable astroglial subtypes from human pluripotent stem cells. Nat Biotechnol, 2011. 29(6): p. 528–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Li X, et al. , Fast Generation of Functional Subtype Astrocytes from Human Pluripotent Stem Cells. Stem Cell Reports, 2018. 11(4): p. 998–1008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Leferink PS, et al. , Astrocyte Subtype Vulnerability in Stem Cell Models of Vanishing White Matter. Ann Neurol, 2019. 86(5): p. 780–792. [DOI] [PMC free article] [PubMed] [Google Scholar]

