Skip to main content
Cellular and Molecular Neurobiology logoLink to Cellular and Molecular Neurobiology
. 2021 Oct 26;42(8):2629–2642. doi: 10.1007/s10571-021-01159-3

Heterogeneity and Molecular Markers for CNS Glial Cells Revealed by Single-Cell Transcriptomics

Junjie Sun 1, Yixing Song 1, Zhiheng Chen 1, Jiaying Qiu 3, Shunxing Zhu 2, Liucheng Wu 2,, Lingyan Xing 1,
PMCID: PMC11421601  PMID: 34704168

Abstract

Glial cells, including astrocytes, oligodendrocytes, and microglia, are the major components in the central nervous system (CNS). Studies have revealed the heterogeneity of each glial cell type and that they each may play distinct roles in physiological processes and/or neurological diseases. Single-cell sequencing (scRNA-seq) technology developed in recent years has extended our understanding of glial cell heterogeneity from the perspective of transcriptome profiling. This review summarizes the marker genes of major glial cells in the CNS and reveals their heterogeneity in different species, CNS regions, developmental stages, and pathological states (Alzheimer’s disease and spinal cord injury), expanding our knowledge of glial cell heterogeneity on both molecular and functional levels.

Keywords: Glial cell, ScRNA-seq, Spatial and temporal specific, Marker gene, Astrocyte, Microglia, Oligodendrocyte, Alzheimer’s disease, Spinal cord injury

Introduction

The central nervous system (CNS) is mainly composed of neurons and glial cells. Microglial, astrocytic, and oligodendrocyte (OL) lineage cells comprise the major glial cells in the CNS. With new features such as secreting and uptaking discovered, glial cells are now considered critical for neuronal metabolism, regeneration, and homeostasis maintenance of the CNS, instead of solely supporting connective tissues.

Each type of glial cell has diverse states or functions. Microglia are macrophages that reside in the CNS, with properties of both glial and immune cells. In response to environmental changes, microglia can rapidly switch from a “resting state” to a “reactive state,” secreting cytokines and engaging in phagocytosis. Multiple subtypes of microglia and their corresponding marker genes have been identified in different states (Hammond et al. 2019; Masuda et al. 2019). “Reactive” cells are the ones with the defining feature of morphologically protruding branches (Kreutzberg 1996). Notably, the “reactive state” of microglia is dynamic and the complete molecular signatures are currently lacking.

As the most abundant cells in the CNS, astrocytes, identified by stellate projections on their surfaces, have diverse function. They can regulate synaptic transmission by tightly enwrapping neurons and communicate with fast and point-precise neural circuits relying on Ca2+ waves (Allen et al. 2012; Christopherson et al. 2005). They can also protect neurons by metabolic coordination (Ioannou et al. 2019; Rusakov 2015). In addition, astrocytes are important in blood–brain barrier (BBB) formation (Abbott et al. 2010), neuroinflammation (Linnerbauer et al. 2020; Xing et al. 2019), memory encoding (Santello et al. 2019), and generation of circadian rhythms (Hastings et al. 2018). Astrocytes exist as multiple subtypes in different brain regions, which may be the basis of their functional diversity (Batiuk et al. 2020).

OL lineage cells are the only myelin-forming cells in the CNS. The OL lineage includes OL precursor cells (OPCs, also called NG2-glia), committed OL precursors (COPs), mature oligodendrocytes (MOLs), and other intermediate state subtypes. The developmental lineage is OPCs-COPs-MOLs. Notably, a significant pool of OPCs is retained in the adult CNS and can differentiate into myelinating MOLs (Bercury and Macklin 2015). Interestingly, OLs also provide metabolic support to neurons (Philips and Rothstein 2017). Aberrant interaction between OPCs and vessels can disrupt the BBB in multiple sclerosis (Niu et al. 2019). Multiple markers in OL lineage cells have been elucidated (Marques et al. 2016), but the specific role of each subpopulation remains elusive.

The function and maintenance of homeostasis by glial cells are currently hot topics, and several recent reviews have provided incisive summaries of the progress in this field. Greenhalgh et al. summarized the function of CNS glial cells as immune cells (Greenhalgh et al. 2020), Allen et al. reviewed the structure and function of CNS glial cells (Allen and Lyons 2018), Akay et al. described the fates and function of OPCs in the brain (Akay et al. 2021), Liddelow et al. highlighted the role of the communication of microglia and astrocytes in homeostasis and disease (Liddelow et al. 2020), Bohlen et al. discussed how microglia affect brain development, homeostasis, and their role in neurodegeneration (Bohlen et al. 2019a, b), Madore et al. discussed the effects of lifestyle and environmental factors on microglia (Madore et al. 2020), Linnerbauer et al. reviewed the multifaceted role of astrocytes in CNS inflammation (Linnerbauer et al. 2020), and Elbaz et al. discussed the mechanisms of transcriptional control of the developmental lineage of OLs during myelination (Elbaz and Popko 2019). This article will fill a gap in the review literature by focusing on summarizing the progress on marker genes and cellular heterogeneity in glial cells.

Advances in Glial Marker Research

Cell identification and isolation are a prerequisite for cell function studies. Traditional methods rely on antigen–antibody reactions, such as immunohistochemistry (IHC) and fluorescence-activated cell sorting (FACS). Transgenic animals generated for specific cell type labeling have significantly accelerated the process of glial cell research. For example, Zhang et al. purified various types of neurons, glia, and vascular cells by FACS from different transgenic mice and generated a bulk RNA-seq database for each cell type (Zhang et al. 2014). However, these methods are limited in that they only achieve relatively crude labeling of the cell populations and do not accurately reflect the heterogeneity of the cells.

Single-cell RNA sequencing (scRNA-seq) technology is a powerful approach to decipher cellular heterogeneity, which reveals genome-wide gene expression at single-cell resolution (Papalexi and Satija 2018). This technique was first developed in 2009 by Tang et al. and was employed on a large scale by 2014. The applications of this technique have increased exponentially (Svensson et al. 2018). Cell markers are the basis for cell type identification and single-cell analysis. In Table 1, we summarize the markers for major classes of glial cells.

Table 1.

Marker genes used to identify major classes of glial cells in scRNA-seq

Cell type Marker genes Ref.
Microglia Cx3cr1, Trem2, P2ry12, Fcrls, C1qa, CD45, CD11b Blum et al. (2021), Hammond et al. (2019), Keren-Shaul et al. (2017), Masuda et al. (2019), Rosenberg et al. (2018), van der Poel et al. (2019), Zeisel et al. (2018)
Astrocyte Gfap, Aqp4, Aldh1l1, Slc1a3 Batiuk et al. (2020), Blum et al. (2021), Rosenberg et al. (2018), Xu et al. (2018), Zeisel et al. (2018)
OPC Pdgfra, Cspg4, Ptprz1 Blum et al. (2021), Floriddia et al. (2020), Marques et al. (2016), Rosenberg et al. (2018), Xu et al. (2018), Zeisel et al. (2018)
OL Mbp, Mobp, Olig2, Mag, Mog Blum et al. (2021), Floriddia et al. (2020), Marques et al. (2016), Rosenberg et al. (2018), Xu et al. (2018), Zeisel et al. (2018)
OL lineage Aspa, Sox10 Floriddia et al. (2020)

The table shows the marker genes most commonly used in glial cells. These genes are used to define cell types in single-cell sequencing studies. Representative references are listed in the last column

Microglia

Multiple markers of microglia have been identified in past studies. CD45 and CD11b (gene name Itgam) are the most common cell surface markers of microglia, and using these markers for FACS, microglia can easily be sorted from other glial cells (Garcia et al. 2014). In pathology studies, ionized calcium-binding adapter (induction of brown adipocytes 1, Iba-1) is the most widely used activated microglial cell marker. Subsequent studies have shown that microglia specifically express the cell surface marker transmembrane protein 119 (Tmem119) (Bennett et al. 2016). With the development of antibodies targeting exposed epitopes in the N-terminal Tmem119 protein, microglia have been successfully purified (Bennett et al. 2018; Bohlen et al. 2019a, b). Chemokine (C-X3-C motif) receptor 1 (Cx3cr1) is a specific receptor for the neural actin CX3C chemokine fractalkine, which serves as a checkpoint for microglia activation. Genetic deletion of Cx3cr1 has uncovered multiple functions of microglia in the CNS (Jung et al. 2000; Li et al. 2020; Parkhurst et al. 2013; Wu et al. 2020). Except Cx3cr1, recent scRNA-seq papers reveal that many genes, including Trem2, P2ry12, Fcrls, C1qa, CD45, and CD11b, can be used to recognize microglia (Blum et al. 2021; Hammond et al. 2019; Keren-Shaul et al. 2017; Masuda et al. 2019; Rosenberg et al. 2018; van der Poel et al. 2019; Zeisel et al. 2018). These marker genes were located in the plasma membrane or extracellular space, responsible to the rapid activation and secretion of microglia. Caution is needed when applying specific markers of microglia, because microglia and peripheral macrophages share a few markers (e.g., Fcrls, P2ry12, Tmem119, and Trem2) in early development and chronic neuroinflammation. (Grassivaro et al. 2020).

Astrocytes

Unlike microglia, astrocytes are isolated by the immune panning method, in which other glial cells are removed by specific antibodies, leaving behind the pure astrocytes (Foo 2013). Recently, an immunoaffinity-based method was developed for sorting ultrapure astrocytes based on the high affinity of the acetyl–CoA synthetase (Acsa-2) antibody to the astrocytic surface marker Atp1b2 (a Na + /K + transporting ATPase, beta 2 polypeptide) (Batiuk et al. 2017; Kantzer et al. 2017). Glial fibrillary acidic protein (Gfap), an intermediate filament protein, is the best known marker for astrocytes. It plays a role in almost all known functions of astrocytes, including proliferation, migration, myelination, nerve growth, and blood–brain barrier permeability (Middeldorp and Hol 2011). Genetic manipulations of Gfap and another astrocytic marker aldehyde dehydrogenase 1 family member L1 (Aldh1l1) have been widely used in transgenic mice for astrocyte labeling and functional studies (Messing et al. 1998; Srinivasan et al. 2016; Zhuo et al. 2001). Detailed descriptions of the experimental tools used to study astrocytes and microglia have been published (Guttenplan and Liddelow 2019; Yu et al. 2020).

Similar to Gfap and Aldh1l1, Aqp4 and Slc1a3 are also common astrocytic markers, which have been validated by both bulk RNA-seq and scRNA-seq (Batiuk et al. 2020; Blum et al. 2021; Rosenberg et al. 2018; Xu et al. 2018; Zeisel et al. 2018; Zhang et al. 2014). The aquaporin Aqp4 can coordinate water balance and osmotic pressure in the CNS (Vandebroek and Yasui 2020). Slc1a3 encodes a high affinity glutamate transporter protein, which can regulate excitatory synapse activity by neuronal glutamate uptake (Nagelhus and Ottersen 2013). However, few reports are available about their specific roles in astrocytes. Notably, astrocytes resemble satellite glial cells (SGC) in the peripheral nervous system, although expression of Kcnj10/Kir4 (potassium inwardly-rectifying channel, subfamily J, member 10) and Fabp7 (fatty acid binding protein 7) differs between these two cells (Avraham et al. 2020).

OLs

The immune panning method has been used to isolate OPCs [platelet-derived growth factor receptor alpha (Pdgfra) positive], myelinating MOLs [myelin oligodendrocyte glycoprotein (Mog) positive], and post-mitotic OLs [galactosylceramidase (GalC) positive] with cell surface markers bound to specific antibodies (Emery and Dugas 2013). By gene manipulation, transgenic animals targeting sex determining region Y box 10 (Sox10), oligodendrocyte transcription factor 2 (Olig2), and Pdgfra have been generated to specifically label all OL lineage cells, MOLs, and OPCs, respectively (Kang et al. 2010; Kessaris et al. 2006; Lu et al. 2016). Generally, myelin basic protein (Mbp), myelin-associated oligodendrocytic basic protein (Mobp), Olig2, myelin-associated glycoprotein (Mag), and Mog are used to identify MOLs, and Pdgfra, chondroitin sulfate proteoglycan 4 (Cspg4), and protein tyrosine phosphatase receptor type Z1 (Ptprz1) are used for OPCs (Blum et al. 2021; Floriddia et al. 2020; Marques et al. 2016; Rosenberg et al. 2018; Xu et al. 2018; Zeisel et al. 2018). COPs are OPC-like cells which express Neu4 and genes maintaining the undifferentiated state (e.g., Sox6, Bmp4, and Gpr17), but unlike OPCs, they lack Pdgfra and Cspg4 (Marques et al. 2016). In line with these, marker genes in MOL are critical for myelination, axon supporting, and signal conduction (Philips and Rothstein 2017); marker genes in OPCs act to maintain the proliferation characteristics of OPCs (Dang et al. 2019; Kucharova and Stallcup 2018; Fujikawa and Noda 2016). Notely, all Sox10-positive cells express aspartoacylase (Aspa) during development and thus Aspa and Sox10 are pan-markers of the OL lineages (Floriddia et al. 2020).

Species Heterogeneity of Glial Cells

Rodents are a common animal model for glial cell research, and despite the highly conserved nature of their genes and those of humans, the genetic consequences of subtle differences cannot be ignored. One study pointed out that heterogeneity among cohorts is widespread even for the same species and the same disease (Neff et al. 2021). Microglia have significant heterogeneity in different genetic backgrounds, including differences in the expression of several genes associated with Alzheimer’s disease (AD) (e.g., Apoe, Trem2, and Sorl1) and in the number of interferon-responding microglia (Yang et al. 2021). The idea of comparing multiple model organisms has been used in regenerative medicine research (Jorstad et al. 2017; Letelier et al. 2018; Wang et al. 2020). However, for mechanistic studies and drug screening, few, if any, have considered the heterogeneity of glial cells in different species or strains.

It is almost certain that differences in the subtypes and transcriptional profiles of human and murine glial cells exist (Han et al. 2013; Hodge et al. 2019; Song et al. 2020). Xu et al. compared the gene expression profiles of several common glial cells in mouse and human brain using scRNA-seq, noting that humans and mice share several common glial cell markers (see Table 1) but they are differentially expressed, and some non-marker genes are also differentially expressed between similar cells of different species (Table 2) (Xu et al. 2018). Another study found limited overlap in gene expression between mouse and human microglia during aging, with overlapping genes containing microglia marker genes, such as Cx3cr1, P2ry12, and CD11B. A number of immune-related receptor genes, including TLR, Fcγ, and SIGLEC, and proliferation and cell cycle regulators TAL1 and IFI16 were up-regulated in human but not mouse microglia (Galatro et al. 2017). In summary, humans and mice have transcriptomic differences, but their marker genes are identical.

Table 2.

Species-specific genes of glial cells identified by scRNA-seq

Cell type Marker genes Ref.
Microglia No publish
Astrocyte RGCC/human Xu et al. (2018)
Mybpc1/mouse
OPC SCN9A/human Xu et al. (2018)
Lpcat2 1/mouse
OL GLDN/human Xu et al. (2018)
Kcnk13/mouse

For the same type of glial cells, the genes in the table are only expressed in one species of human and mouse. The species-specific genes of microglia were not shown because there are no published studies

Temporal Heterogeneity of Glial Cells

The heterogeneity of CNS glial cells throughout development has been a major area of scientific inquiry. Recent scRNA-seq studies have done much to advance knowledge in this field. Here, we summarize the latest progress in understanding the heterogeneity of glial cells from developmental initiation through the embryonic (E), postnatal (P), juvenile, adult, and elderly stages (Table 3).

Table 3.

Time-specific gene expression characteristics of glial cells identified by scRNA-seq

Cell type Marker genes and stages Ref.
Microglia Ms4a7, Ccr1, Ms4a6c/embryonic Hammond et al. (2019), Masuda et al. (2019)
Spp1, Gpnmb, Igf1/postnatal day 4 and 5
Ctsb, Ctsd, Lamp1/embryonic
CST3 −, SPARC − high percentage/embryonic
Tmem119, Selplg, Slc2a5/postnatal
Astrocyte No publish
OPC Ptprz1 high percentage/postnatal day 20 Floriddia et al. (2020)
Ptprz1 high percentage/postnatal day 60
OL Ptgds high percentage/postnatal day 60 Floriddia et al. (2020)

“+” and “−” represent the positive and negative signal of immunofluorescence imaging, respectively; “high/low percentage” indicates that the proportion of cells with this characteristic in this type of glial cells is high or low compared with other times

Microglia

In vivo lineage tracing studies in the mouse model have confirmed that microglia originate from primitive myeloid progenitor cells that appear before embryonic day 8 and later transfer to central maturation (Ginhoux et al. 2010). Unlike other glial cells, microglia have no other precursor cells throughout their lifespan (Gomez Perdiguero et al. 2015; Grassivaro et al. 2020; Kierdorf et al. 2013). Li et al. found high microglial cell heterogeneity in the early postnatal period, compared with little heterogeneity in adult homeostasis-state microglia (Li et al. 2019). Hammond et al. used scRNA-seq to analyze the transcriptional profiles of microglia throughout the lifespan of the mouse brain (E14.5, P4/5, P30, P100, P540) and developed the gene expression profile data for each stage into a visual website (Hammond et al. 2019). In this study the authors found at least nine distinct subpopulations of microglia. In the embryonic period, microglia are characterized by Ms4a7-specific expression; in P4/5, microglia specifically express Spp1; in the early postnatal brain, microglia are characterized as metabolically active and proliferative due to high expression of Birc5, Hist1h2bc, Mcm6, Rrm2, and other genes; and finally, microglial aging is characterized by increased inflammatory and interferon responses and an increased proportion of Ccl4-positive cells (Hammond et al. 2019). Another study reported that high expression of Tmem119, Selplg, and Slc2a5 is a feature of juvenile and adult but not embryonic microglia; Ctsb expression is a feature of microglia in the embryonic period; and lack of Cst3 and Sparc expression is also a feature of microglia in the embryonic period (Masuda et al. 2019). One reason why the development-specific markers of microglia found in the above two studies are not consistent is that the mice used were not exactly the same age, which suggest that the study of microglia developmental heterogeneity may require a more detailed time window.

Astrocytes

Two pathways can renew the mature astrocyte pool; one is the differentiation of radial glial cells acting as precursor cells and the other is the self-renewal of mature astrocytes through their own division and differentiation (Chen et al. 2020; Ge et al. 2012; Morel et al. 2017; Noctor et al. 2004). There has been no clear check point for the developmental initiation and maturation process of astrocytes (Molofsky and Deneen 2015; Molofsky et al. 2012). In the developing spinal cord, as early as E12.5, astrocytes are demarcated by induction of Slc1a3 (GLAST, glutamate/aspartate transporter), Aldh1l1, and Nfia (Cahoy et al. 2008; Deneen et al. 2006). Transcriptome sequencing of astrocytes located in the mouse spinal cord at E13.5–18.5 combined with biochemical experiments revealed that Sox9 promotes astrocyte and OL maturation by regulating Nfe2l1 (Molofsky et al. 2013). Transcriptome sequencing of samples at multiple developmental time points from E12.5 to P7 demonstrated that Asef, Tom1l1, Mfge8, and Gpr37l1 can serve as novel markers for intermediate stages of spinal astrocyte development (Chaboub et al. 2016). Boisvert et al. analyzed gene expression profiles of astrocytes in 4-month-old and 24-month-old mice and found that aging astrocytes exhibited significant molecular differences with increased expression of inflammatory and synapse elimination pathway genes (Boisvert et al. 2018). The above findings were obtained from bulk RNA sequencing in astrocytes isolated by FACS. No single-cell resolution study that accurately reflects the heterogeneity of astrocytes in a temporal time scale is available, which may be an important future direction to explore.

OLs

OL development involves several well-defined steps, starting with the differentiation of neural progenitor cells into proliferating OPCs, moving on to differentiation into COPs, and finally developing into MOLs with myelinogenic functions (Richardson et al. 2006). In the brain, the origin of OPCs is restricted to 3 waves in mice, occurring at E11.5–12.5, E16.5, and after birth. Spatially, these OPCs develop in the ventricular zone of the medial ganglionic eminence and anterior entopeduncular area, the Gsh2-expressing ventricular zone of the lateral and central ganglionic eminences, and the cerebral cortex, respectively (Kessaris et al. 2006). In the spinal cord, OPC formation initiates in the ventrally localized motor neuron domain (pMN domain) at E12.5 and in the dorsal precursor region of the spinal cord at E15.5, but by birth the ventral region rather than the dorsal is the primary source of OPCs (Fogarty et al. 2005; Ravanelli and Appel 2015; Richardson et al. 2000). OPCs first appear in humans at approximately 10–15 weeks of gestation (Jakovcevski et al. 2009). OL maturation and myelin formation in mice begin at E18.5 and continue into adulthood (Akay et al. 2021; Michalski and Kothary 2015). The mechanisms of developmental initiation and myelin formation in OPCs have been summarized in detail (Akay et al. 2021; van Tilborg et al. 2018).

Temporal heterogeneity of OPCs and OLs were investigated by multiple studies. Branco’s group sequenced single cells from Pdgfra-positive populations in adolescent and adult mice and found that OPCs that developed at different times are not transcriptionally heterogeneous, but they can differentiate into MOL populations with temporal and spatial specificity (Marques et al. 2016). Similarly, results from study did not report significant transcriptional program differences in OPCs isolated from embryonic and postnatal periods (Marques et al. 2018). However, Spitzer et al. studied the function of mouse OPCs at several time points from E13 to 9 months and found that although OPCs originated from homogeneous populations, they were spatially and temporally different in electrophysiological properties (Spitzer et al. 2019).

Spatial Heterogeneity of Glial Cells

Although glial cells exist throughout the entire CNS, differential developmental origins can lead to regionalization of glial cell populations. Anatomically, numerous neurological diseases have foci that occur only in specific brain regions, implying that the spatial heterogeneity of glial cells is closely related to disease progression. This section provides a summary of recent advances in glial cell spatial specificity. We summarized the progress of spatial heterogeneity of glial cells identified by scRNA-seq (Table 4).

Table 4.

Spatial-specific gene expression characteristics of glial cells identified by scRNA-seq

Cell type Marker genes Ref.
Microglia CST3 + SPARC + high percentage/cortex Masuda et al. (2019)
CST3 + SPARC + low percentage/cerebellum
CST3 + SPARC − low percentage/cortex
CST3 + SPARC − high percentage/cerebellum
Astrocyte Frzb, Ascl1/cortex Batiuk et al. (2020)
Unc13c/hippocampus
OPC No publish
OL Klk6 high percentage/dorsal spinal cord Floriddia et al. (2020)
Klk6 low percentage/cortex and corpus callosum

“+” and “−” represent the positive and negative signal of immunofluorescence imaging, respectively; “high/low percentage” indicates that the proportion of cells with this characteristic in this type of glial cells is high or low compared with other times

Microglia

Grabert et al. proposed that microglia in different brain regions (striatum, hippocampus, cerebral cortex, and cerebellum) of adult mice have distinct transcriptomic profiles in terms of immune response and energy metabolism as determined by microarray analysis (Grabert et al. 2016). Ayata et al. demonstrated that microglia in the cerebellum have stronger scavenging activity compared to the cortex and striatum, due to polycomb repressive complex 2 (PRC2), which limits the expression of genes with scavenging activity in the striatum through an epigenetic mechanism (Ayata et al. 2018). ScRNA-seq transcriptome of microglia in eight anatomical regions at three developmental time points revealed strong variation between different regions of the CNS in microglia (Masuda et al. 2019). However, it has also been argued that the spatial heterogeneity of microglia is not absolute. One study used scRNA-seq to analyze the transcriptome of microglia from different brain regions at multiple developmental time points. The authors found that the transcriptomes of adults, but not embryonic and postnatal microglia, are very similar regardless of the brain region (Li et al. 2019). Another study performed scRNA-seq on CD45-positive cells isolated from several CNS compartments (including the leptomeninges, the perivascular space and parenchyma, and the choroid plexus) and found that the disease-associated subpopulation had stronger heterogeneity in microglia than the homeostatic subpopulation. Although the major marker genes were expressed population-wide, the disease-associated microglia clusters showed lower expression of P2ry12, Maf, and Slc2a5 and higher expression of Ccl2, Cxcl10, Ly86, and Mki67; in inflammatory diseases and spinal cord injury, expression of P2ry12, Tmem119, and Selplg was strongly down-regulated and Ly86 expression was up-regulated (Jordao et al. 2019).

Astrocytes

Previous findings provide evidence that astrocytes exhibit functional and molecular heterogeneity in a region-dependent manner (Bayraktar et al. 2014; Zhang and Barres 2010). Subsequent studies have also analyzed in detail the region-specific nature of astrocytes at the transcriptional, translational, and epigenetic levels as well as by novel FACS methods (Chai et al. 2017; Herrero-Navarro et al. 2021; John Lin et al. 2017; Morel et al. 2017; Sharma et al. 2015). The rehabilitative effect of certain activities, such as exercise, on astrocytes also occurs in a region-dependent manner (Lundquist et al. 2019). Not only that, several studies imply that regional specificity leads to functional limitations of astrocytes, e.g., astrocytes from different regions cannot compensate for each other’s functions (Tsai et al. 2012), and astrocytes from different regions have varying abilities to differentiate into neurons (Mattugini et al. 2019). Advances have been made in identifying spatially specific subpopulations of astrocytes using scRNA-seq. Batiuk et al. analyzed the astrocyte single-cell transcriptome in the hippocampus and cortex of adult mice and found significant heterogeneity in astrocytes in both regions, with the hippocampus but not the cortex containing subpopulations associated with neural regeneration; more than 70% of the enriched genes were subtype specific (Batiuk et al. 2020). Moreover, Bayraktar et al. performed a novel high-resolution spatial transcriptome analysis and found differential laminar distribution and varying transcriptome characteristics of astrocytes in the mouse cortex (Bayraktar et al. 2020).

OLs

Results from recent studies have shown significant molecular heterogeneity and spatial preference in MOLs (Floriddia et al. 2020; Marques et al. 2016). However, it is controversial whether or not OPCs originating from different regions are heterogeneous. Using scRNA-seq, Floriddia et al. found that OPCs from different regions show similar transcriptomic features during embryonic development (Floriddia et al. 2020). However, inconsistent results have been observed: cells of the OL lineages generated by separate OPC pools during development have varying responsiveness to demyelination in vivo and show heterogeneous migration and differentiation abilities in vitro (Crawford et al. 2016). The most recent view is that the functional heterogeneity of MOLs is independent of OPCs and may depend on the spatial environment or neuron type in which they are located and this may partially explain the controversy (Zonouzi et al. 2019). The results of Floriddia et al. also supported the idea that the intrinsic controls of OPCs do not determine the heterogeneity of MOLs (Floriddia et al. 2020). Sun et al. proposed a molecular mechanism that could explain the region- and time-specific myelin formation in the CNS, i.e., cell-autonomous programmed death of MOL subpopulations mediated by the transcription factor EB (TFEB) (Sun et al. 2018). However, the results of Floriddia et al. indicated that certain MOL subpopulations in the spinal cord are not produced by TFEB regulation (Floriddia et al. 2020). Therefore, more evidence is needed to resolve these controversies.

Heterogeneity of Glial Cells in the Pathogenesis of AD and Spinal Cord Injury (SCI)

CNS lesions are accompanied by changes in the number and gene expression of glial cells (Greenhalgh et al. 2020). Recently, advances have been made in understanding the molecular and cellular complexity of glial cells in diseases. Using AD and SCI as a representative neurodegenerative diseases and CNS mechanical injury, respectively, we summarize the progress of research in glial cell heterogeneity in disease.

AD

AD is the most common neurodegenerative disease. Extensive work has been done on the pathogenesis, clinical phenotypes, research methods, biomarkers, etc. of the disease (Lashley et al. 2018; Mantzavinos and Alexiou 2017; Penney et al. 2020; Scheltens et al. 2016). However, it is unclear on the molecular level how individual glial cell types are associated with AD. Four representative studies that use single-cell nuclear sequencing (snRNA-seq) to analyze the transcriptomes in different regions of the cerebral cortex of postmortem AD patients have not only provided new insights into cellular heterogeneity in the AD process but more importantly have generated a valuable data resource (Del-Aguila et al. 2019; Grubman et al. 2019; Mathys et al. 2019; Zhou et al. 2020). Pathology-associated glial cell types and genes were identified by scRNA-seq in AD patients: the AD-pathology-associated OL lineage cells characterized by high expression of CRYAB or QDPR, GLUL, and CLU are preferentially expressed in AD-pathology-associated astrocyte subtype (Mathys et al. 2019). These genes have the potential to become markers of AD pathology-associated glial cell subtypes. It is noteworthy that the heterogeneity and function of disease-related reactive astrocytes have also been implicated in a variety of neurodegenerative diseases (Escartin et al. 2021). In addition to the discussion of the abundance and differentially expressed genes of glial cells in AD patients, there are several interesting findings from these studies. Zhou et al. noted that changes in the expression of disease-related genes of glial cells in AD mouse models do not match these in AD patients (Zhou et al. 2020), Grubman et al. reported that the AD risk gene Apoe was specifically repressed in OPCs and astrocytes and up-regulated in disease-specific microglia in AD patients (Grubman et al. 2019), and Mathys et al. found significant gender differences in AD-associated transcriptomic alterations, with a higher proportion of AD-associated glial cell subtypes in female patients (Mathys et al. 2019). It has been shown that p2y12 is a marker of homeostatic microglia (Mildner et al. 2017); however, Walker et al. observed high expression levels of p2y12 in brain sections from patients with severe AD (activated microglia), emphasizing the heterogeneity of microglia in AD patients (Walker et al. 2020). Two studies focused on microglial heterogeneity in two separate AD mouse models using scRNA-seq and identified new disease-associated microglia subtypes as well as marker genes and reprogramming trajectories associated with these subtypes (Keren-Shaul et al. 2017; Mathys et al. 2017). Although both astrocytes and OL lineages have been reported to play a role in the AD process (Carter et al., 2019; Zhang et al. 2020), there are no studies on these two cell types in mouse models of AD with single-cell resolution, which would likely be a fruitful avenue of future research. We summarized the Alzheimer’s disease-specific differentially expressed genes of glial cells identified by scRNA-seq (Table 5).

Table 5.

Alzheimer’s disease-specific differentially expressed genes (DEGs) of glial cells identified by scRNA-seq

Cell type Marker genes Ref.
Microglia RPS19, CD163/up-regulated Grubman et al. (2019)
CX3CR1, CD86, P2RY12/down-regulated
Astrocyte MAP1B, S100A6/up-regulated Grubman et al. (2019)
APOE, NRXN1, GABRB1, GRIA2/down-regulated
OPC APOE/down-regulated Grubman et al. (2019)
OL IL1RAPL1/up-regulated Grubman et al. (2019)
All types MT-ND1, CRYAB, HSPA1A/up-regulated Grubman et al. (2019)

The DEGs in this table come from a single-cell sequencing study using AD patients as samples. “All types” means that these genes have consistent expression characteristics in all types of glial cells

In significant recent research, Jiang et al. integrated AD-related scRNA-seq databases and developed the scREAD dataset. scREAD provides a comprehensive analysis of 73 databases from 10 brain regions, including the construction of control maps, cell type prediction, identification of differentially expressed genes, and identification of cell type-specific regulators (Jiang et al. 2020). In addition, several new algorithms were developed to construct complex molecular networks of AD-related glial cells, which could help reveal new therapeutic targets for AD (Wang et al. 2016; Xu et al. 2021).

SCI

The intrinsic growth capacity of neurons and the injury microenvironment regulated by glial cells are two major determinants of the process of neural circuit reconstruction after SCI (He and Jin 2016). Spinal cord regeneration is extremely difficult in mammals, and glial scars formed by reactive astrocytes, fibroblasts, and microglia/macrophages are thought to be important in limiting spinal cord regeneration (Tran et al. 2018; Yang et al. 2020). However, many new findings have challenged this popular opinion. In the early stages of SCI, rapidly proliferating and activated microglia promote the formation of glial scars that are beneficial for repair, which helps to stop the spread of the lesion. This was confirmed by the results of subsequent experiments using transgenic mice or specific drugs depleting microglia, resulting in inhibition of spinal cord regeneration (Bellver-Landete et al. 2019; Fu et al. 2020). Li et al. used scRNA-seq and bioinformatic analysis to identify a subtype of microglia suitable for promoting scarless wound healing and suggested that fibronectin derived from this population of microglia may act as a bridge at the injury to promote wound healing (Li et al. 2020). Astrocytes are the main component of glial scars, and it is controversial whether they form a “wall” or a “bridge” for axonal regeneration after SCI. Astrocytes can produce chondroitin sulfate proteoglycans (CSPGs) that inhibit axonal growth (Silver and Miller 2004). With a rigorous experimental design, Anderson et al. demonstrated that scar-forming astrocytes allow, but do not prevent, CNS axon regeneration with appropriate stimulation (Anderson et al. 2016). There is widespread heterogeneity in astrocytes, but no study has yet indicated which subtypes have regeneration-inhibiting properties.

SCI leads to demyelination of nerve axons, and subsequent remodeling of spinal cord circuits requires remyelination. Information on how OL lineage cells change after SCI, as well as their function and ultimate fate has been summarized previously (Duncan et al. 2020). Spectral tracking experiments after SCI showed that OPCs are the main source of myelinogenic OLs (Assinck et al. 2017; Bartus et al. 2019) and that OPCs can also differentiate into astrocytes to facilitate glial scar formation (Hackett et al. 2018; Sellers et al. 2009). However, these results are insufficient to confirm whether there is heterogeneity in (1) the OPCs after injury and (2) between the original resident OLs and the newly formed OLs. Floriddia et al. used scRNA-seq to identify the effects of SCI on the transcriptome of MOLs and found that different subtypes of MOLs from healthy mice responded differently to SCI, but there was no evidence to indicate which MOL subtype promoted spinal cord regeneration (Floriddia et al. 2020).

In summary, glial cells dynamically change after SCI and contain complex lineages (Barnabe-Heider et al. 2010). Little is known about the heterogeneity of glial cells after SCI, and strategies to activate beneficial subtypes and inhibit deleterious ones might be utilized in the future to meet the challenges of mammalian spinal cord regeneration.

Conclusions and Perspectives

Recently, especially since the broad application of scRNA-seq technology, our knowledge of the molecular characteristics and spatiotemporal heterogeneity of glial cells has been greatly expanded. This will accelerate the breakthroughs in drug development targeting glial cells and BBB permeability and will lay a solid foundation for precision therapies (Yao et al. 2020). The enormous amount of data that can be generated by single-cell sequencing is a treasure trove to be mined and only a tiny fraction of it has been utilized so far. Here, we present a summary of representative scRNA-seq online data retrieval sites related to glial cells (Table 6), which is useful for understanding of the molecular basis and the biology of many glial cell-related diseases.

Table 6.

Online retrieval sites of visualized single-cell transcriptional atlas

Website Cell type Species Ref. Keywords
www.microgliasinglecell.com Microglia Mouse Hammond et al. (2019) Time specific; brain injury
www.brainrnaseq.org Microglia Mouse Li et al. (2019) Time and region specific
https://holt-sc.glialab.org/ Astrocyte Mouse Batiuk et al. (2020) Region specific
https://ki.se/en/mbb/oligointernode MOL Mouse Floriddia et al. (2020) Region specific; spinal cord injury
http://linnarssonlab.org/oligodendrocytes/ OL Mouse Marques et al. (2016) Time and region specific
http://spinalcordatlas.org Neuron Mouse Blum et al. (2021) Spinal cord
http://mousebrain.org All CNS cell Mouse Zeisel et al. (2018) Region specific
http://ngi.pub/snuclRNA-seq All CNS cell Human Del-Aguila et al. (2019) Alzheimer’s disease
http://adsn.ddnetbio.com All CNS cell Human Grubman et al. (2019) Alzheimer’s disease
https://bmbls.bmi.osumc.edu/scread/ All CNS cell Human and mouse Jiang et al. (2020) An integrated database for Alzheimer’s disease

This table summarizes the online database of single-cell sequencing related to CNS glial cells. The information in the table includes website address, cell type, species, references, and database topics

There are complex interactions between glial cells and between glial cells and neurons that together maintain CNS homeostasis (Konishi et al. 2020; Patel et al. 2019). The massive amount of single-cell sequencing data has led to increased research efforts in bioinformatics, and a variety of cell–cell communication analysis methods based on ligand and receptor interaction databases have been proposed (Armingol et al. 2021; Jin et al. 2021; Noel et al. 2021). Notably, Clark et al. developed a viral vector-based scRNA-seq technology, RABID-seq, which enables effective molecular target identification of cell-to-cell interactions and will enrich the database of ligand–receptor interactions (Clark et al. 2021).

At present, the mechanisms regulating the generation, maintenance, and transformation of glial cells in temporal, spatial, and species specificity are not well understood. Some studies have been undertaken to examine these (Huang et al. 2020; Lee et al. 2008; Sun et al. 2018), but none of them are at single-cell levels. Single-cell ATAC-seq (assay for transposase-accessible chromatin with high-throughput sequencing) can obtain the chromatin accessibility profiles of individual cells and can resolve the transcriptional data of key genes in cells (Buenrostro et al. 2015; Cusanovich et al. 2018). This technology has the power to yield new breakthroughs in understanding glial cell heterogeneity in the near future.

Acknowledgements

We gratefully acknowledge the National Science Foundation of China (Grant No. 81701127 to XL, Grant No. 32000841 to SJ), the Municipal Health Commission of Nantong (Grant No. MA2020019 to QJ), and the Science and Technology Bureau of Nantong (Grant No. JC2020101 to QJ).

Author Contributions

SJ, XL, and WL provided ideas and wrote the paper. SJ, SY, CZ, QJ, and ZS were responsible for the retrieval of papers. All authors read the manuscript.

Declarations

Conflict of interest

The authors have read this review and state that there was no conflict of interest in the publication of it.

Consent for Publication

Written informed consent for publication was obtained from all participants.

Footnotes

Publisher's Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Contributor Information

Liucheng Wu, Email: hnwulc@ntu.edu.cn.

Lingyan Xing, Email: xlyan011@163.com.

References

  1. Abbott NJ, Patabendige AA, Dolman DE, Yusof SR, Begley DJ (2010) Structure and function of the blood-brain barrier. Neurobiol Dis 37(1):13–25. 10.1016/j.nbd.2009.07.030 [DOI] [PubMed] [Google Scholar]
  2. Akay LA, Effenberger AH, Tsai LH (2021) Cell of all trades: oligodendrocyte precursor cells in synaptic, vascular, and immune function. Genes Dev 35(3–4):180–198. 10.1101/gad.344218.120 [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Allen NJ, Lyons DA (2018) Glia as architects of central nervous system formation and function. Science 362(6411):181–185. 10.1126/science.aat0473 [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Allen NJ, Bennett ML, Foo LC, Wang GX, Chakraborty C, Smith SJ, Barres BA (2012) Astrocyte glypicans 4 and 6 promote formation of excitatory synapses via GluA1 AMPA receptors. Nature 486(7403):410–414. 10.1038/nature11059 [DOI] [PMC free article] [PubMed] [Google Scholar]
  5. Anderson MA, Burda JE, Ren Y, Ao Y, O’Shea TM, Kawaguchi R et al (2016) Astrocyte scar formation aids central nervous system axon regeneration. Nature 532(7598):195–200. 10.1038/nature17623 [DOI] [PMC free article] [PubMed] [Google Scholar]
  6. Armingol E, Officer A, Harismendy O, Lewis NE (2021) Deciphering cell-cell interactions and communication from gene expression. Nat Rev Genet 22(2):71–88. 10.1038/s41576-020-00292-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  7. Assinck P, Duncan GJ, Plemel JR, Lee MJ, Stratton JA, Manesh SB et al (2017) Myelinogenic plasticity of oligodendrocyte precursor cells following spinal cord contusion injury. J Neurosci 37(36):8635–8654. 10.1523/JNEUROSCI.2409-16.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  8. Avraham O, Deng PY, Jones S, Kuruvilla R, Semenkovich CF, Klyachko VA, Cavalli V (2020) Satellite glial cells promote regenerative growth in sensory neurons. Nat Commun 11(1):4891. 10.1038/s41467-020-18642-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  9. Ayata P, Badimon A, Strasburger HJ, Duff MK, Montgomery SE, Loh YE et al (2018) Epigenetic regulation of brain region-specific microglia clearance activity. Nat Neurosci 21(8):1049–1060. 10.1038/s41593-018-0192-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Barnabe-Heider F, Goritz C, Sabelstrom H, Takebayashi H, Pfrieger FW, Meletis K, Frisen J (2010) Origin of new glial cells in intact and injured adult spinal cord. Cell Stem Cell 7(4):470–482. 10.1016/j.stem.2010.07.014 [DOI] [PubMed] [Google Scholar]
  11. Bartus K, Burnside ER, Galino J, James ND, Bennett DLH, Bradbury EJ (2019) ErbB receptor signaling directly controls oligodendrocyte progenitor cell transformation and spontaneous remyelination after spinal cord injury. Glia 67(6):1036–1046. 10.1002/glia.23586 [DOI] [PMC free article] [PubMed] [Google Scholar]
  12. Batiuk MY, de Vin F, Duque SI, Li C, Saito T, Saido T et al (2017) An immunoaffinity-based method for isolating ultrapure adult astrocytes based on ATP1B2 targeting by the ACSA-2 antibody. J Biol Chem 292(21):8874–8891. 10.1074/jbc.M116.765313 [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Batiuk MY, Martirosyan A, Wahis J, de Vin F, Marneffe C, Kusserow C et al (2020) Identification of region-specific astrocyte subtypes at single cell resolution. Nat Commun 11(1):1220. 10.1038/s41467-019-14198-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  14. Bayraktar OA, Fuentealba LC, Alvarez-Buylla A, Rowitch DH (2014) Astrocyte development and heterogeneity. Cold Spring Harb Perspect Biol 7(1):a020362. 10.1101/cshperspect.a020362 [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Bayraktar OA, Bartels T, Holmqvist S, Kleshchevnikov V, Martirosyan A, Polioudakis D et al (2020) Astrocyte layers in the mammalian cerebral cortex revealed by a single-cell in situ transcriptomic map. Nat Neurosci 23(4):500–509. 10.1038/s41593-020-0602-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  16. Bellver-Landete V, Bretheau F, Mailhot B, Vallieres N, Lessard M, Janelle ME et al (2019) Microglia are an essential component of the neuroprotective scar that forms after spinal cord injury. Nat Commun 10(1):518. 10.1038/s41467-019-08446-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  17. Bennett ML, Bennett FC, Liddelow SA, Ajami B, Zamanian JL, Fernhoff NB et al (2016) New tools for studying microglia in the mouse and human CNS. Proc Natl Acad Sci USA 113(12):E1738-1746. 10.1073/pnas.1525528113 [DOI] [PMC free article] [PubMed] [Google Scholar]
  18. Bennett FC, Bennett ML, Yaqoob F, Mulinyawe SB, Grant GA, Hayden Gephart M et al (2018) A combination of ontogeny and CNS environment establishes microglial identity. Neuron 98(6):1170-1183.e1178. 10.1016/j.neuron.2018.05.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Bercury KK, Macklin WB (2015) Dynamics and mechanisms of CNS myelination. Dev Cell 32(4):447–458. 10.1016/j.devcel.2015.01.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  20. Blum JA, Klemm S, Shadrach JL, Guttenplan KA, Nakayama L, Kathiria A et al (2021) Single-cell transcriptomic analysis of the adult mouse spinal cord reveals molecular diversity of autonomic and skeletal motor neurons. Nat Neurosci 24(4):572–583. 10.1038/s41593-020-00795-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Bohlen CJ, Bennett FC, Bennett ML (2019a) Isolation and culture of microglia. Curr Protoc Immunol 125(1):e70. 10.1002/cpim.70 [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Bohlen CJ, Friedman BA, Dejanovic B, Sheng M (2019b) Microglia in brain development, homeostasis, and neurodegeneration. Annu Rev Genet 53:263–288. 10.1146/annurev-genet-112618-043515 [DOI] [PubMed] [Google Scholar]
  23. Boisvert MM, Erikson GA, Shokhirev MN, Allen NJ (2018) The aging astrocyte transcriptome from multiple regions of the mouse brain. Cell Rep 22(1):269–285. 10.1016/j.celrep.2017.12.039 [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Buenrostro JD, Wu B, Litzenburger UM, Ruff D, Gonzales ML, Snyder MP et al (2015) Single-cell chromatin accessibility reveals principles of regulatory variation. Nature 523(7561):486–490. 10.1038/nature14590 [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Cahoy JD, Emery B, Kaushal A, Foo LC, Zamanian JL, Christopherson KS et al (2008) A transcriptome database for astrocytes, neurons, and oligodendrocytes: a new resource for understanding brain development and function. J Neurosci 28(1):264–278. 10.1523/JNEUROSCI.4178-07.2008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  26. Carter SF, Herholz K, Rosa-Neto P, Pellerin L, Nordberg A, Zimmer ER (2019) Astrocyte biomarkers in Alzheimer’s disease. Trends Mol Med 25(2):77–95. 10.1016/j.molmed.2018.11.006 [DOI] [PubMed] [Google Scholar]
  27. Chaboub LS, Manalo JM, Lee HK, Glasgow SM, Chen F, Kawasaki Y et al (2016) Temporal profiling of astrocyte precursors reveals parallel roles for asef during development and after injury. J Neurosci 36(47):11904–11917. 10.1523/JNEUROSCI.1658-16.2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  28. Chai H, Diaz-Castro B, Shigetomi E, Monte E, Octeau JC, Yu X et al (2017) Neural circuit-specialized astrocytes: transcriptomic, proteomic, morphological, and functional evidence. Neuron 95(3):531-549.e539. 10.1016/j.neuron.2017.06.029 [DOI] [PMC free article] [PubMed] [Google Scholar]
  29. Chen J, Poskanzer KE, Freeman MR, Monk KR (2020) Live-imaging of astrocyte morphogenesis and function in zebrafish neural circuits. Nat Neurosci 23(10):1297–1306. 10.1038/s41593-020-0703-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  30. Christopherson KS, Ullian EM, Stokes CC, Mullowney CE, Hell JW, Agah A et al (2005) Thrombospondins are astrocyte-secreted proteins that promote CNS synaptogenesis. Cell 120(3):421–433. 10.1016/j.cell.2004.12.020 [DOI] [PubMed] [Google Scholar]
  31. Clark IC, Gutierrez-Vazquez C, Wheeler MA, Li Z, Rothhammer V, Linnerbauer M et al (2021) Barcoded viral tracing of single-cell interactions in central nervous system inflammation. Science. 10.1126/science.abf1230 [DOI] [PMC free article] [PubMed] [Google Scholar]
  32. Crawford AH, Tripathi RB, Richardson WD, Franklin RJM (2016) Developmental origin of oligodendrocyte lineage cells determines response to demyelination and susceptibility to age-associated functional decline. Cell Rep 15(4):761–773. 10.1016/j.celrep.2016.03.069 [DOI] [PMC free article] [PubMed] [Google Scholar]
  33. Cusanovich DA, Hill AJ, Aghamirzaie D, Daza RM, Pliner HA, Berletch JB et al (2018) A single-cell atlas of in vivo mammalian chromatin accessibility. Cell 174(5):1309-1324.e1318. 10.1016/j.cell.2018.06.052 [DOI] [PMC free article] [PubMed] [Google Scholar]
  34. Dang TC, Ishii Y, Nguyen V, Yamamoto S, Hamashima T, Okuno N et al (2019) Powerful homeostatic control of oligodendroglial lineage by PDGFRalpha in adult brain. Cell Rep 27(4):1073-1089.e1075. 10.1016/j.celrep.2019.03.084 [DOI] [PubMed] [Google Scholar]
  35. Del-Aguila JL, Li Z, Dube U, Mihindukulasuriya KA, Budde JP, Fernandez MV et al (2019) A single-nuclei RNA sequencing study of Mendelian and sporadic AD in the human brain. Alzheimers Res Ther 11(1):71. 10.1186/s13195-019-0524-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Deneen B, Ho R, Lukaszewicz A, Hochstim CJ, Gronostajski RM, Anderson DJ (2006) The transcription factor NFIA controls the onset of gliogenesis in the developing spinal cord. Neuron 52(6):953–968. 10.1016/j.neuron.2006.11.019 [DOI] [PubMed] [Google Scholar]
  37. Duncan GJ, Manesh SB, Hilton BJ, Assinck P, Plemel JR, Tetzlaff W (2020) The fate and function of oligodendrocyte progenitor cells after traumatic spinal cord injury. Glia 68(2):227–245. 10.1002/glia.23706 [DOI] [PubMed] [Google Scholar]
  38. Elbaz B, Popko B (2019) Molecular control of oligodendrocyte development. Trends Neurosci 42(4):263–277. 10.1016/j.tins.2019.01.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  39. Emery B, Dugas JC (2013) Purification of oligodendrocyte lineage cells from mouse cortices by immunopanning. Cold Spring Harb Protoc 2013(9):854–868. 10.1101/pdb.prot073973 [DOI] [PubMed] [Google Scholar]
  40. Escartin C, Galea E, Lakatos A, O’Callaghan JP, Petzold GC, Serrano-Pozo A et al (2021) Reactive astrocyte nomenclature, definitions, and future directions. Nat Neurosci 24(3):312–325. 10.1038/s41593-020-00783-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  41. Floriddia EM, Lourenco T, Zhang S, van Bruggen D, Hilscher MM, Kukanja P et al (2020) Distinct oligodendrocyte populations have spatial preference and different responses to spinal cord injury. Nat Commun 11(1):5860. 10.1038/s41467-020-19453-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Fogarty M, Richardson WD, Kessaris N (2005) A subset of oligodendrocytes generated from radial glia in the dorsal spinal cord. Development 132(8):1951–1959. 10.1242/dev.01777 [DOI] [PubMed] [Google Scholar]
  43. Foo LC (2013) Purification of rat and mouse astrocytes by immunopanning. Cold Spring Harb Protoc 2013(5):421–432. 10.1101/pdb.prot074211 [DOI] [PubMed] [Google Scholar]
  44. Fu H, Zhao Y, Hu D, Wang S, Yu T, Zhang L (2020) Depletion of microglia exacerbates injury and impairs function recovery after spinal cord injury in mice. Cell Death Dis 11(7):528. 10.1038/s41419-020-2733-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  45. Fujikawa A, Noda M (2016) Role of pleiotrophin-protein tyrosine phosphatase receptor type Z signaling in myelination. Neural Regen Res 11(4):549–551. 10.4103/1673-5374.180761 [DOI] [PMC free article] [PubMed] [Google Scholar]
  46. Galatro TF, Holtman IR, Lerario AM, Vainchtein ID, Brouwer N, Sola PR et al (2017) Transcriptomic analysis of purified human cortical microglia reveals age-associated changes. Nat Neurosci 20(8):1162–1171. 10.1038/nn.4597 [DOI] [PubMed] [Google Scholar]
  47. Garcia JA, Cardona SM, Cardona AE (2014) Isolation and analysis of mouse microglial cells. Curr Protoc Immunol 104:143511–143515. 10.1002/0471142735.im1435s104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  48. Ge WP, Miyawaki A, Gage FH, Jan YN, Jan LY (2012) Local generation of glia is a major astrocyte source in postnatal cortex. Nature 484(7394):376–380. 10.1038/nature10959 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Ginhoux F, Greter M, Leboeuf M, Nandi S, See P, Gokhan S et al (2010) Fate mapping analysis reveals that adult microglia derive from primitive macrophages. Science 330(6005):841–845. 10.1126/science.1194637 [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Gomez Perdiguero E, Klapproth K, Schulz C, Busch K, Azzoni E, Crozet L et al (2015) Tissue-resident macrophages originate from yolk-sac-derived erythro-myeloid progenitors. Nature 518(7540):547–551. 10.1038/nature13989 [DOI] [PMC free article] [PubMed] [Google Scholar]
  51. Grabert K, Michoel T, Karavolos MH, Clohisey S, Baillie JK, Stevens MP et al (2016) Microglial brain region-dependent diversity and selective regional sensitivities to aging. Nat Neurosci 19(3):504–516. 10.1038/nn.4222 [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Grassivaro F, Menon R, Acquaviva M, Ottoboni L, Ruffini F, Bergamaschi A et al (2020) Convergence between microglia and peripheral macrophages phenotype during development and neuroinflammation. J Neurosci 40(4):784–795. 10.1523/JNEUROSCI.1523-19.2019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  53. Greenhalgh AD, David S, Bennett FC (2020) Immune cell regulation of glia during CNS injury and disease. Nat Rev Neurosci 21(3):139–152. 10.1038/s41583-020-0263-9 [DOI] [PubMed] [Google Scholar]
  54. Grubman A, Chew G, Ouyang JF, Sun G, Choo XY, McLean C et al (2019) A single-cell atlas of entorhinal cortex from individuals with Alzheimer’s disease reveals cell-type-specific gene expression regulation. Nat Neurosci 22(12):2087–2097. 10.1038/s41593-019-0539-4 [DOI] [PubMed] [Google Scholar]
  55. Guttenplan KA, Liddelow SA (2019) Astrocytes and microglia: mod{guttenplan, 2019 #9}els and tools. J Exp Med 216(1):71–83. 10.1084/jem.20180200 [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Hackett AR, Yahn SL, Lyapichev K, Dajnoki A, Lee DH, Rodriguez M et al (2018) Injury type-dependent differentiation of NG2 glia into heterogeneous astrocytes. Exp Neurol 308:72–79. 10.1016/j.expneurol.2018.07.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
  57. Hammond TR, Dufort C, Dissing-Olesen L, Giera S, Young A, Wysoker A et al (2019) Single-cell RNA sequencing of microglia throughout the mouse lifespan and in the injured brain reveals complex cell-state changes. Immunity 50(1):253-271.e256. 10.1016/j.immuni.2018.11.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  58. Han X, Chen M, Wang F, Windrem M, Wang S, Shanz S et al (2013) Forebrain engraftment by human glial progenitor cells enhances synaptic plasticity and learning in adult mice. Cell Stem Cell 12(3):342–353. 10.1016/j.stem.2012.12.015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Hastings MH, Maywood ES, Brancaccio M (2018) Generation of circadian rhythms in the suprachiasmatic nucleus. Nat Rev Neurosci 19(8):453–469. 10.1038/s41583-018-0026-z [DOI] [PubMed] [Google Scholar]
  60. He Z, Jin Y (2016) Intrinsic control of axon regeneration. Neuron 90(3):437–451. 10.1016/j.neuron.2016.04.022 [DOI] [PubMed] [Google Scholar]
  61. Herrero-Navarro A, Puche-Aroca L, Moreno-Juan V, Sempere-Ferrandez A, Espinosa A, Susin R et al (2021) Astrocytes and neurons share region-specific transcriptional signatures that confer regional identity to neuronal reprogramming. Sci Adv. 10.1126/sciadv.abe8978 [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Hodge RD, Bakken TE, Miller JA, Smith KA, Barkan ER, Graybuck LT et al (2019) Conserved cell types with divergent features in human versus mouse cortex. Nature 573(7772):61–68. 10.1038/s41586-019-1506-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  63. Huang AY, Woo J, Sardar D, Lozzi B, Bosquez Huerta NA, Lin CJ et al (2020) Region-specific transcriptional control of astrocyte function oversees local circuit activities. Neuron 106(6):992-1008.e1009. 10.1016/j.neuron.2020.03.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Ioannou MS, Jackson J, Sheu SH, Chang CL, Weigel AV, Liu H et al (2019) Neuron-astrocyte metabolic coupling protects against activity-induced fatty acid toxicity. Cell 177(6):1522-1535.e1514. 10.1016/j.cell.2019.04.001 [DOI] [PubMed] [Google Scholar]
  65. Jakovcevski I, Filipovic R, Mo Z, Rakic S, Zecevic N (2009) Oligodendrocyte development and the onset of myelination in the human fetal brain. Front Neuroanat 3:5. 10.3389/neuro.05.005.2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  66. Jiang J, Wang C, Qi R, Fu H, Ma Q (2020) scREAD: a single-cell RNA-seq database for Alzheimer’s disease. iScience 23(11):101769. 10.1016/j.isci.2020.101769 [DOI] [PMC free article] [PubMed] [Google Scholar]
  67. Jin S, Guerrero-Juarez CF, Zhang L, Chang I, Ramos R, Kuan CH et al (2021) Inference and analysis of cell-cell communication using cell chat. Nat Commun 12(1):1088. 10.1038/s41467-021-21246-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. John Lin CC, Yu K, Hatcher A, Huang TW, Lee HK, Carlson J et al (2017) Identification of diverse astrocyte populations and their malignant analogs. Nat Neurosci 20(3):396–405. 10.1038/nn.4493 [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Jordao MJC, Sankowski R, Brendecke SM, Sagar, Locatelli G, Tai YH et al (2019) Single-cell profiling identifies myeloid cell subsets with distinct fates during neuroinflammation. Science. 10.1126/science.aat7554 [DOI] [PubMed] [Google Scholar]
  70. Jorstad NL, Wilken MS, Grimes WN, Wohl SG, VandenBosch LS, Yoshimatsu T et al (2017) Stimulation of functional neuronal regeneration from Muller glia in adult mice. Nature 548(7665):103–107. 10.1038/nature23283 [DOI] [PMC free article] [PubMed] [Google Scholar]
  71. Jung S, Aliberti J, Graemmel P, Sunshine MJ, Kreutzberg GW, Sher A, Littman DR (2000) Analysis of fractalkine receptor CX(3)CR1 function by targeted deletion and green fluorescent protein reporter gene insertion. Mol Cell Biol 20(11):4106–4114. 10.1128/mcb.20.11.4106-4114.2000 [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Kang SH, Fukaya M, Yang JK, Rothstein JD, Bergles DE (2010) NG2+ CNS glial progenitors remain committed to the oligodendrocyte lineage in postnatal life and following neurodegeneration. Neuron 68(4):668–681. 10.1016/j.neuron.2010.09.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  73. Kantzer CG, Boutin C, Herzig ID, Wittwer C, Reiss S, Tiveron MC et al (2017) Anti-ACSA-2 defines a novel monoclonal antibody for prospective isolation of living neonatal and adult astrocytes. Glia 65(6):990–1004. 10.1002/glia.23140 [DOI] [PubMed] [Google Scholar]
  74. Keren-Shaul H, Spinrad A, Weiner A, Matcovitch-Natan O, Dvir-Szternfeld R, Ulland TK et al (2017) A unique microglia type associated with restricting development of Alzheimer’s disease. Cell 169(7):1276-1290.e1217. 10.1016/j.cell.2017.05.018 [DOI] [PubMed] [Google Scholar]
  75. Kessaris N, Fogarty M, Iannarelli P, Grist M, Wegner M, Richardson WD (2006) Competing waves of oligodendrocytes in the forebrain and postnatal elimination of an embryonic lineage. Nat Neurosci 9(2):173–179. 10.1038/nn1620 [DOI] [PMC free article] [PubMed] [Google Scholar]
  76. Kierdorf K, Erny D, Goldmann T, Sander V, Schulz C, Perdiguero EG et al (2013) Microglia emerge from erythromyeloid precursors via Pu.1- and Irf8-dependent pathways. Nat Neurosci 16(3):273–280. 10.1038/nn.3318 [DOI] [PubMed] [Google Scholar]
  77. Konishi H, Okamoto T, Hara Y, Komine O, Tamada H, Maeda M et al (2020) Astrocytic phagocytosis is a compensatory mechanism for microglial dysfunction. EMBO J 39(22):e104464. 10.15252/embj.2020104464 [DOI] [PMC free article] [PubMed] [Google Scholar]
  78. Kreutzberg GW (1996) Microglia: a sensor for pathological events in the CNS. Trends Neurosci 19(8):312–318. 10.1016/0166-2236(96)10049-7 [DOI] [PubMed] [Google Scholar]
  79. Kucharova K, Stallcup WB (2018) Dissecting the multifactorial nature of demyelinating disease. Neural Regen Res 13(4):628–632. 10.4103/1673-5374.230281 [DOI] [PMC free article] [PubMed] [Google Scholar]
  80. Lashley T, Schott JM, Weston P, Murray CE, Wellington H, Keshavan A et al (2018) Molecular biomarkers of Alzheimer’s disease: progress and prospects. Dis Model Mech. 10.1242/dmm.031781 [DOI] [PMC free article] [PubMed] [Google Scholar]
  81. Lee Y, Messing A, Su M, Brenner M (2008) GFAP promoter elements required for region-specific and astrocyte-specific expression. Glia 56(5):481–493. 10.1002/glia.20622 [DOI] [PubMed] [Google Scholar]
  82. Letelier J, de la Calle-Mustienes E, Pieretti J, Naranjo S, Maeso I, Nakamura T et al (2018) A conserved Shh cis-regulatory module highlights a common developmental origin of unpaired and paired fins. Nat Genet 50(4):504–509. 10.1038/s41588-018-0080-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Li Q, Cheng Z, Zhou L, Darmanis S, Neff NF, Okamoto J et al (2019) Developmental heterogeneity of microglia and brain myeloid cells revealed by deep single-Cell RNA sequencing. Neuron 101(2):207-223.e210. 10.1016/j.neuron.2018.12.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Li Y, He X, Kawaguchi R, Zhang Y, Wang Q, Monavarfeshani A et al (2020) Microglia-organized scar-free spinal cord repair in neonatal mice. Nature 587(7835):613–618. 10.1038/s41586-020-2795-6 [DOI] [PMC free article] [PubMed] [Google Scholar]
  85. Liddelow SA, Marsh SE, Stevens B (2020) Microglia and astrocytes in disease: dynamic duo or partners in crime? Trends Immunol 41(9):820–835. 10.1016/j.it.2020.07.006 [DOI] [PubMed] [Google Scholar]
  86. Linnerbauer M, Wheeler MA, Quintana FJ (2020) Astrocyte crosstalk in CNS inflammation. Neuron 108(4):608–622. 10.1016/j.neuron.2020.08.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  87. Lu F, Chen Y, Zhao C, Wang H, He D, Xu L et al (2016) Olig2-dependent reciprocal shift in PDGF and EGF receptor signaling regulates tumor phenotype and mitotic growth in malignant glioma. Cancer Cell 29(5):669–683. 10.1016/j.ccell.2016.03.027 [DOI] [PMC free article] [PubMed] [Google Scholar]
  88. Lundquist AJ, Parizher J, Petzinger GM, Jakowec MW (2019) Exercise induces region-specific remodeling of astrocyte morphology and reactive astrocyte gene expression patterns in male mice. J Neurosci Res 97(9):1081–1094. 10.1002/jnr.24430 [DOI] [PubMed] [Google Scholar]
  89. Madore C, Yin Z, Leibowitz J, Butovsky O (2020) Microglia, lifestyle stress, and neurodegeneration. Immunity 52(2):222–240. 10.1016/j.immuni.2019.12.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Mantzavinos V, Alexiou A (2017) Biomarkers for Alzheimer’s disease diagnosis. Curr Alzheimer Res 14(11):1149–1154. 10.2174/1567205014666170203125942 [DOI] [PMC free article] [PubMed] [Google Scholar]
  91. Marques S, Zeisel A, Codeluppi S, van Bruggen D, Mendanha Falcao A, Xiao L et al (2016) Oligodendrocyte heterogeneity in the mouse juvenile and adult central nervous system. Science 352(6291):1326–1329. 10.1126/science.aaf6463 [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Marques S, van Bruggen D, Vanichkina DP, Floriddia EM, Munguba H, Varemo L et al (2018) Transcriptional convergence of oligodendrocyte lineage progenitors during development. Dev Cell 46(4):504-517.e507. 10.1016/j.devcel.2018.07.005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Masuda T, Sankowski R, Staszewski O, Bottcher C, Amann L et al (2019) Spatial and temporal heterogeneity of mouse and human microglia at single-cell resolution. Nature 566(7744):388–392. 10.1038/s41586-019-0924-x [DOI] [PubMed] [Google Scholar]
  94. Mathys H, Adaikkan C, Gao F, Young JZ, Manet E, Hemberg M et al (2017) Temporal tracking of microglia activation in neurodegeneration at single-cell resolution. Cell Rep 21(2):366–380. 10.1016/j.celrep.2017.09.039 [DOI] [PMC free article] [PubMed] [Google Scholar]
  95. Mathys H, Davila-Velderrain J, Peng Z, Gao F, Mohammadi S, Young JZ et al (2019) Single-cell transcriptomic analysis of Alzheimer’s disease. Nature 570(7761):332–337. 10.1038/s41586-019-1195-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Mattugini N, Bocchi R, Scheuss V, Russo GL, Torper O, Lao CL, Gotz M (2019) Inducing different neuronal subtypes from astrocytes in the injured mouse cerebral cortex. Neuron 103(6):1086-1095.e1085. 10.1016/j.neuron.2019.08.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Messing A, Head MW, Galles K, Galbreath EJ, Goldman JE, Brenner M (1998) Fatal encephalopathy with astrocyte inclusions in GFAP transgenic mice. Am J Pathol 152(2):391–398 [PMC free article] [PubMed] [Google Scholar]
  98. Michalski JP, Kothary R (2015) Oligodendrocytes in a nutshell. Front Cell Neurosci 9:340. 10.3389/fncel.2015.00340 [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Middeldorp J, Hol EM (2011) GFAP in health and disease. Prog Neurobiol 93(3):421–443. 10.1016/j.pneurobio.2011.01.005 [DOI] [PubMed] [Google Scholar]
  100. Mildner A, Huang H, Radke J, Stenzel W, Priller J (2017) P2Y12 receptor is expressed on human microglia under physiological conditions throughout development and is sensitive to neuroinflammatory diseases. Glia 65(2):375–387. 10.1002/glia.23097 [DOI] [PubMed] [Google Scholar]
  101. Molofsky AV, Deneen B (2015) Astrocyte development: a guide for the perplexed. Glia 63(8):1320–1329. 10.1002/glia.22836 [DOI] [PubMed] [Google Scholar]
  102. Molofsky AV, Krencik R, Ullian EM, Tsai HH, Deneen B, Richardson WD et al (2012) Astrocytes and disease: a neurodevelopmental perspective. Genes Dev 26(9):891–907. 10.1101/gad.188326.112 [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Molofsky AV, Glasgow SM, Chaboub LS, Tsai HH, Murnen AT, Kelley KW et al (2013) Expression profiling of Aldh1l1-precursors in the developing spinal cord reveals glial lineage-specific genes and direct Sox9-Nfe2l1 interactions. Glia 61(9):1518–1532. 10.1002/glia.22538 [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Morel L, Chiang MSR, Higashimori H, Shoneye T, Iyer LK, Yelick J et al (2017) Molecular and functional properties of regional astrocytes in the adult brain. J Neurosci 37(36):8706–8717. 10.1523/JNEUROSCI.3956-16.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  105. Nagelhus EA, Ottersen OP (2013) Physiological roles of aquaporin-4 in brain. Physiol Rev 93(4):1543–1562. 10.1152/physrev.00011.2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  106. Neff RA, Wang M, Vatansever S, Guo L, Ming C, Wang Q et al (2021) Molecular subtyping of Alzheimer’s disease using RNA sequencing data reveals novel mechanisms and targets. Sci Adv. 10.1126/sciadv.abb5398 [DOI] [PMC free article] [PubMed] [Google Scholar]
  107. Niu J, Tsai HH, Hoi KK, Huang N, Yu G, Kim K et al (2019) Aberrant oligodendroglial-vascular interactions disrupt the blood-brain barrier, triggering CNS inflammation. Nat Neurosci 22(5):709–718. 10.1038/s41593-019-0369-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  108. Noctor SC, Martinez-Cerdeno V, Ivic L, Kriegstein AR (2004) Cortical neurons arise in symmetric and asymmetric division zones and migrate through specific phases. Nat Neurosci 7(2):136–144. 10.1038/nn1172 [DOI] [PubMed] [Google Scholar]
  109. Noel F, Massenet-Regad L, Carmi-Levy I, Cappuccio A, Grandclaudon M, Trichot C et al (2021) Dissection of intercellular communication using the transcriptome-based framework ICELLNET. Nat Commun 12(1):1089. 10.1038/s41467-021-21244-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Papalexi E, Satija R (2018) Single-cell RNA sequencing to explore immune cell heterogeneity. Nat Rev Immunol 18(1):35–45. 10.1038/nri.2017.76 [DOI] [PubMed] [Google Scholar]
  111. Parkhurst CN, Yang G, Ninan I, Savas JN, Yates JR 3rd, Lafaille JJ et al (2013) Microglia promote learning-dependent synapse formation through brain-derived neurotrophic factor. Cell 155(7):1596–1609. 10.1016/j.cell.2013.11.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Patel DC, Tewari BP, Chaunsali L, Sontheimer H (2019) Neuron-glia interactions in the pathophysiology of epilepsy. Nat Rev Neurosci 20(5):282–297. 10.1038/s41583-019-0126-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
  113. Penney J, Ralvenius WT, Tsai LH (2020) Modeling Alzheimer’s disease with iPSC-derived brain cells. Mol Psychiatry 25(1):148–167. 10.1038/s41380-019-0468-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  114. Philips T, Rothstein JD (2017) Oligodendroglia: metabolic supporters of neurons. J Clin Invest 127(9):3271–3280. 10.1172/JCI90610 [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Ravanelli AM, Appel B (2015) Motor neurons and oligodendrocytes arise from distinct cell lineages by progenitor recruitment. Genes Dev 29(23):2504–2515. 10.1101/gad.271312.115 [DOI] [PMC free article] [PubMed] [Google Scholar]
  116. Richardson WD, Smith HK, Sun T, Pringle NP, Hall A, Woodruff R (2000) Oligodendrocyte lineage and the motor neuron connection. Glia 29(2):136–142. 10.1002/(sici)1098-1136(20000115)29:2%3c136::aid-glia6%3e3.0.co;2-g [DOI] [PubMed] [Google Scholar]
  117. Richardson WD, Kessaris N, Pringle N (2006) Oligodendrocyte wars. Nat Rev Neurosci 7(1):11–18. 10.1038/nrn1826 [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Rosenberg AB, Roco CM, Muscat RA, Kuchina A, Sample P, Yao Z et al (2018) Single-cell profiling of the developing mouse brain and spinal cord with split-pool barcoding. Science 360(6385):176–182. 10.1126/science.aam8999 [DOI] [PMC free article] [PubMed] [Google Scholar]
  119. Rusakov DA (2015) Disentangling calcium-driven astrocyte physiology. Nat Rev Neurosci 16(4):226–233. 10.1038/nrn3878 [DOI] [PubMed] [Google Scholar]
  120. Santello M, Toni N, Volterra A (2019) Astrocyte function from information processing to cognition and cognitive impairment. Nat Neurosci 22(2):154–166. 10.1038/s41593-018-0325-8 [DOI] [PubMed] [Google Scholar]
  121. Scheltens P, Blennow K, Breteler MM, de Strooper B, Frisoni GB, Salloway S, Van der Flier WM (2016) Alzheimer’s disease. Lancet 388(10043):505–517. 10.1016/S0140-6736(15)01124-1 [DOI] [PubMed] [Google Scholar]
  122. Sellers DL, Maris DO, Horner PJ (2009) Postinjury niches induce temporal shifts in progenitor fates to direct lesion repair after spinal cord injury. J Neurosci 29(20):6722–6733. 10.1523/JNEUROSCI.4538-08.2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Sharma K, Schmitt S, Bergner CG, Tyanova S, Kannaiyan N, Manrique-Hoyos N et al (2015) Cell type- and brain region-resolved mouse brain proteome. Nat Neurosci 18(12):1819–1831. 10.1038/nn.4160 [DOI] [PMC free article] [PubMed] [Google Scholar]
  124. Silver J, Miller JH (2004) Regeneration beyond the glial scar. Nat Rev Neurosci 5(2):146–156. 10.1038/nrn1326 [DOI] [PubMed] [Google Scholar]
  125. Song HW, Foreman KL, Gastfriend BD, Kuo JS, Palecek SP, Shusta EV (2020) Transcriptomic comparison of human and mouse brain microvessels. Sci Rep 10(1):12358. 10.1038/s41598-020-69096-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  126. Spitzer SO, Sitnikov S, Kamen Y, Evans KA, Kronenberg-Versteeg D, Dietmann S et al (2019) Oligodendrocyte progenitor cells become regionally diverse and heterogeneous with age. Neuron 101(3):459-471.e455. 10.1016/j.neuron.2018.12.020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  127. Srinivasan R, Lu TY, Chai H, Xu J, Huang BS, Golshani P et al (2016) New transgenic mouse lines for selectively targeting astrocytes and studying calcium signals in astrocyte processes in situ and in vivo. Neuron 92(6):1181–1195. 10.1016/j.neuron.2016.11.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  128. Sun LO, Mulinyawe SB, Collins HY, Ibrahim A, Li Q, Simon DJ et al (2018) Spatiotemporal control of CNS myelination by oligodendrocyte programmed cell death through the TFEB-PUMA axis. Cell 175(7):1811-1826.e1821. 10.1016/j.cell.2018.10.044 [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Svensson V, Vento-Tormo R, Teichmann SA (2018) Exponential scaling of single-cell RNA-seq in the past decade. Nat Protoc 13(4):599–604. 10.1038/nprot.2017.149 [DOI] [PubMed] [Google Scholar]
  130. Tran AP, Warren PM, Silver J (2018) The biology of regeneration failure and success after spinal cord injury. Physiol Rev 98(2):881–917. 10.1152/physrev.00017.2017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Tsai HH, Li H, Fuentealba LC, Molofsky AV, Taveira-Marques R, Zhuang H et al (2012) Regional astrocyte allocation regulates CNS synaptogenesis and repair. Science 337(6092):358–362. 10.1126/science.1222381 [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. van der Poel M, Ulas T, Mizee MR, Hsiao CC, Miedema SSM et al (2019) Transcriptional profiling of human microglia reveals grey-white matter heterogeneity and multiple sclerosis-associated changes. Nat Commun 10(1):1139. 10.1038/s41467-019-08976-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. van Tilborg E, de Theije CGM, van Hal M, Wagenaar N, de Vries LS, Benders MJ et al (2018) Origin and dynamics of oligodendrocytes in the developing brain: implications for perinatal white matter injury. Glia 66(2):221–238. 10.1002/glia.23256 [DOI] [PMC free article] [PubMed] [Google Scholar]
  134. Vandebroek A, Yasui M (2020) Regulation of AQP4 in the central nervous system. Int J Mol Sci 21(5):1603. 10.3390/ijms21051603 [DOI] [PMC free article] [PubMed] [Google Scholar]
  135. Walker DG, Tang TM, Mendsaikhan A, Tooyama I, Serrano GE, Sue LI et al (2020) Patterns of expression of purinergic receptor P2RY12, a putative marker for non-activated microglia, in aged and Alzheimer’s disease brains. Int J Mol Sci 21(2):678. 10.3390/ijms21020678 [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Wang M, Roussos P, McKenzie A, Zhou X, Kajiwara Y, Brennand KJ et al (2016) Integrative network analysis of nineteen brain regions identifies molecular signatures and networks underlying selective regional vulnerability to Alzheimer’s disease. Genome Med 8(1):104. 10.1186/s13073-016-0355-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
  137. Wang W, Hu CK, Zeng A, Alegre D, Hu D, Gotting K et al (2020) Changes in regeneration-responsive enhancers shape regenerative capacities in vertebrates. Science. 10.1126/science.aaz3090 [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Wu W, Li Y, Wei Y, Bosco DB, Xie M, Zhao MG et al (2020) Microglial depletion aggravates the severity of acute and chronic seizures in mice. Brain Behav Immun 89:245–255. 10.1016/j.bbi.2020.06.028 [DOI] [PMC free article] [PubMed] [Google Scholar]
  139. Xing L, Yang T, Cui S, Chen G (2019) Connexin hemichannels in astrocytes: role in CNS disorders. Front Mol Neurosci 12:23. 10.3389/fnmol.2019.00023 [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Xu X, Stoyanova EI, Lemiesz AE, Xing J, Mash DC, Heintz N (2018) Species and cell-type properties of classically defined human and rodent neurons and glia. Elife. 10.7554/eLife.37551 [DOI] [PMC free article] [PubMed] [Google Scholar]
  141. Xu J, Zhang P, Huang Y, Zhou Y, Hou Y, Bekris L et al (2021) Multimodal single-cell/nucleus RNA sequencing data analysis uncovers molecular networks between disease-associated microglia and astrocytes with implications for drug repurposing in Alzheimer’s disease. Genome Res. 10.1101/gr.272484.120 [DOI] [PMC free article] [PubMed] [Google Scholar]
  142. Yang T, Xing L, Yu W, Cai Y, Cui S, Chen G (2020) Astrocytic reprogramming combined with rehabilitation strategy improves recovery from spinal cord injury. FASEB J 34(11):15504–15515. 10.1096/fj.202001657RR [DOI] [PubMed] [Google Scholar]
  143. Yang HS, Onos KD, Choi K, Keezer KJ, Skelly DA, Carter GW, Howell GR (2021) Natural genetic variation determines microglia heterogeneity in wild-derived mouse models of Alzheimer’s disease. Cell Rep 34(6):108739. 10.1016/j.celrep.2021.108739 [DOI] [PMC free article] [PubMed] [Google Scholar]
  144. Yao K, Gong GM, Fu ZX, Wang YQ, Zhang LZ, Li GC, Yang YM (2020) Synthesis and evaluation of cytocompatible alkyne-containing poly(beta-amino ester)-based hydrogels functionalized via click reaction. ACS Macro Lett 9(9):1391–1397. 10.1021/acsmacrolett.0c00545 [DOI] [PubMed] [Google Scholar]
  145. Yu X, Nagai J, Khakh BS (2020) Improved tools to study astrocytes. Nat Rev Neurosci 21(3):121–138. 10.1038/s41583-020-0264-8 [DOI] [PubMed] [Google Scholar]
  146. Zeisel A, Hochgerner H, Lonnerberg P, Johnsson A, Memic F, van der Zwan J et al (2018) Molecular architecture of the mouse nervous system. Cell 174(4):999-1014.e1022. 10.1016/j.cell.2018.06.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Zhang Y, Barres BA (2010) Astrocyte heterogeneity: an underappreciated topic in neurobiology. Curr Opin Neurobiol 20(5):588–594. 10.1016/j.conb.2010.06.005 [DOI] [PubMed] [Google Scholar]
  148. Zhang Y, Chen K, Sloan SA, Bennett ML, Scholze AR, O’Keeffe S et al (2014) An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. J Neurosci 34(36):11929–11947. 10.1523/JNEUROSCI.1860-14.2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  149. Zhang X, Wang R, Hu D, Sun X, Fujioka H, Lundberg K et al (2020) Oligodendroglial glycolytic stress triggers inflammasome activation and neuropathology in Alzheimer’s disease. Sci Adv. 10.1126/sciadv.abb8680 [DOI] [PMC free article] [PubMed] [Google Scholar]
  150. Zhou Y, Song WM, Andhey PS, Swain A, Levy T, Miller KR et al (2020) Human and mouse single-nucleus transcriptomics reveal TREM2-dependent and TREM2-independent cellular responses in Alzheimer’s disease. Nat Med 26(1):131–142. 10.1038/s41591-019-0695-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
  151. Zhuo L, Theis M, Alvarez-Maya I, Brenner M, Willecke K, Messing A (2001) hGFAP-cre transgenic mice for manipulation of glial and neuronal function in vivo. Genesis 31(2):85–94. 10.1002/gene.10008 [DOI] [PubMed] [Google Scholar]
  152. Zonouzi M, Berger D, Jokhi V, Kedaigle A, Lichtman J, Arlotta P (2019) Individual oligodendrocytes show bias for inhibitory axons in the neocortex. Cell Rep 27(10):2799-2808.e2793. 10.1016/j.celrep.2019.05.018 [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Cellular and Molecular Neurobiology are provided here courtesy of Springer

RESOURCES