Abstract
Astrocytes are a major constituent of the central nervous system. These glia play a major role in regulating blood-brain barrier function, the formation and maintenance of synapses, glutamate uptake, and trophic support for surrounding neurons and glia. Therefore, maintaining the proper functioning of these cells is crucial to survival. Astrocyte defects are associated with a wide variety of neuropathological insults, ranging from neurodegenerative diseases to gliomas. Additionally, injury to the CNS causes drastic changes to astrocytes, often leading to a phenomenon known as reactive astrogliosis. This process is important for protecting the surrounding healthy tissue from the spread of injury, while it also inhibits axonal regeneration and plasticity. Here, we discuss the important roles of astrocytes after injury and in disease, as well as potential therapeutic approaches to restore proper astrocyte functioning.
1. INTRODUCTION
Astrocytes constitute 50% of all the cells in the central nervous system. They are responsible for a wide variety of functions, from regulating synaptic activity to preventing the spread of injury. Because they are present in such large numbers and perform such a diverse array of functions, astrocytes have received a great deal of attention. In particular, much focus has been given to understanding the role of astrocytes after injury, where they are thought to have both beneficial and detrimental effects. Understanding the complexity of their interactions and functions during development, in a healthy adult, and after trauma could have important therapeutic implications. Manipulating the astrocyte response to enhance their beneficial interactions and minimize their negative properties after injury could promote regeneration and functional recovery.
2. ASTROCYTE DEVELOPMENT
During development, astrocytes arise from radial glia, which reside in the neuroepithelium and express both vimentin and nestin. They are capable of generating both neurons (Malatesta et al., 2000) and astrocytes (Morest and Silver, 2003). Radial glia begin to differentiate into astrocytes after neurogenesis is complete and begin to express brain lipid binding protein (BLBP; Barry and McDermott, 2005), a common marker for astrocyte progenitors, as well as the glutamate transporters GLAST (Shibata et al., 1997) and GLT-1 (Furuta et al., 1997). As radial glia differentiate, they also begin to express A2B5 (Hirano and Goldman, 1988) and low levels of GFAP. During this time, they maintain their contact with the pial surface, but retract their basal attachment to the ventricular zone. As they translocate to their destination in the gray or white matter, they upregulate expression of GFAP as they lose their bipolar morphology and form additional processes (Malatesta et al., 2000; Voigt, 1989; Yang et al., 1993a, 1993b). In the spinal cord, after the initial wave of neurogenesis is complete (~E9–11), progenitors in the ventricular zone begin to differentiate into astrocytes (E12.5). The patterning in development establishes astrocyte heterogeneity, which may have important implications in adulthood and after injury (Chaboub and Deneen, 2012).
Unlike neurons and oligodendrocytes, which become post-mitotic and take on a distinct morphology upon terminal differentiation, astrocytes are able to locally proliferate in the postnatal cortex and spinal cord, and can dramatically alter their shape. Postnatally, astrocytes in the forebrain also continue to arise from subventricular stem cells (Lundgaard et al., 2014). In the intact spinal cord there is limited proliferation of astrocytes and ependymal cells postnatally, but in response to injury, inflammatory cytokines cause adult astrocytes as well as ependymal cell progenitors to proliferate and generate reactive astrocytes (Barnabé-Heider et al., 2010; Magnus et al., 2008).
Astrocytes have evolved in both number and complexity (Freeman and Rowitch, 2013). The nervous system in invertebrates is comprised of only 15% glia, in contrast to mammals which are comprised of almost 90% glia. Therefore, astrocytes in mammals differ from those in lower species. For instance, glial cell development in Drosophila is controlled by the transcription factor glial cell missing (gcm). When gcm is mutated in Drosophila, progenitors are shifted from a glial to neuronal fate and ectopic expression of gcm forces all CNS cells to become glia (Hosoya et al., 1995). However, manipulating the homologue of this transcription factor in mouse models has no effect on glial cell development (Kim et al., 1998), suggesting that mammalian astrocytes develop differently from those in more primitive species. There are also important differences between species with regards to their response to injury. Injury to the CNS induces adult mammalian astrocytes to form walls around the lesion and produce a large amount of chondroitin sulfate proteoglycans (CSPGs). Conversely, glial cells in lower vertebrates do not produce large amounts of reactive matrix and, in addition, they migrate into the lesion environment and form bridges to promote axon regeneration, a behavior that better reflects that of immature mammalian astrocytes (Goldshmit et al., 2012; Zukor et al., 2011). Finally, there is currently little evidence of heterogeneity of astrocytes in lower organisms such as Drosophila or C. elegans (Freeman and Rowitch, 2013), whereas astrocytes in vertebrates have regional diversification that leads to a high degree of functional and morphological heterogeneity.
3. ASTROCYTE HETEROGENEITY
Astrocytes have long been characterized as two distinct classes, based primarily on morphology, as first described by Ramon y Cajal (Ramon y Cajal, 1909). However, these two classes of astrocytes also differ in their developmental origin, their location, and their antigenic phenotype (Miller and Raff, 1984). Protoplasmic astrocytes are found in the gray matter, have long processes, and are very bushy. These cells do not express easily detectable levels of GFAP, as assessed by immunohistochemistry, in an uninjured setting. They are commonly identified using the marker S100β (Chaboub and Deneen, 2012). They envelop neuronal cell bodies and synapses. Protoplasmic astrocytes are also largely arranged in non-overlapping domains (Bushong et al., 2002). Conversely, fibrous astrocytes have short branched processes, populate the white matter, and express GFAP. The majority of fibrous astrocytes also express A2B5 (Miller and Raff, 1984). Because they are located in the white matter, they are able to interact with nodes of Ranvier (Ffrench-Constant et al., 1986; Raine, 1984).
Different brain regions possess astrocytes with different functions and morphologies. Recent studies have demonstrated that the genetic and molecular expression profiles of astrocytes isolated from different brain regions vary significantly. Using RNA microarray analyses on astrocytes isolated from postnatal day 1 optic nerve, cerebellum, brainstem, and neocortex, Yeh and colleagues reported that astrocytes from these different brain regions could be individually identified on the basis of their distinct molecular patterns (Yeh et al., 2009). The inherent diversity of astrocytes based on their local environment may have important implications for their functions and response to insult or disease. For instance, region-specific pathology in AD may result, in part, from regional differences in reactive gliosis (Höke et al., 1994). Astrocytes cultured from different CNS regions, including the cerebral cortex, hippocampus, cerebellum, and spinal cord showed different responsiveness to substrate bound beta-amyloid peptide in vitro. Only hippocampal and cortical astrocytes matured in vitro had reactive morphological changes, increased CSPG deposition, and alterations in proteoglycan metabolism when cultured on substrate-bound beta-amyloid peptide, suggesting regional differences in astrocyte populations can trigger different responses to amyloid insult.
There are many studies suggesting that functional differences also exist between immature and mature astrocytes in mammals. One major difference between immature and mature astrocytes is their ability to support or inhibit axon growth. During development, neuronal growth cones, especially near crossing points at the midline, are closely associated with astrocytes, suggesting that immature astrocytes provide a highly favorable substrate for axon outgrowth (Grafe and Schoenfeld, 1982; Silver et al., 1982, 1993). These astrocytes retain their primitive radial architecture and express high levels of GFAP. In certain circumscribed regions (eg. the roof plate of the spinal cord and tectum) CSPGs secreted by radial astrocytes provide guidance boundaries for developing axons as they migrate toward their targets (Powell and Geller, 1999; Powell et al., 1997a, 1997b; Snow et al., 1990; Wu et al., 1998).
Mature astrocytes do not appear to have the same ability to promote robust neurite outgrowth as their embryonic counterparts, but they do have growth promoting capacities (Davies et al., 2011; Filous et al., 2010). In adult mammals, injury to the CNS causes mature astrocytes to become reactive and assume a phenotype similar to boundary astrocytes during development, leading to the formation of a glial scar that surrounds the damaged tissue (Cregg et al., 2014; discussed below). However, such injuries in immature mammals or lower species result in a more reparative type of reactive astrogliosis. After CNS injury in immature mammals, reactive astrocytes support regeneration through or around the lesion (Barrett et al., 1984). Whereas mature astrocytes in the glial scar have a well-established role in inhibiting regeneration, transplantation of fetal rat spinal cord astrocytes is able to improve the regenerative capacities of adult rat neurons after spinal cord injury (SCI) (Reier et al., 1986). Similarly, hippocampal neurons cultured on explanted scar tissue isolated from either immature or mature animals displayed more extensive neurite outgrowth when cultured on scar tissue from younger animals (Rudge and Silver, 1990). Retinal ganglion cells isolated from either rats or fish had the ability to grow on embryonic astrocytes, but this growth was diminished on monolayers of astrocytes from adult animals (Bähr et al., 1995), again suggesting inherent differences between immature and mature glia. These differences may have crucial implications in the context of injury. One study reported that transplanted immature cortical astrocytes have the ability to suppress scar formation in the adult rat brain. Additionally, immature astrocytes were able to migrate into surrounding CNS tissue and become associated with host blood vessels. Transplanting mature cortical astrocytes did not affect scar formation, nor were they able to migrate from their site of implantation. Furthermore, transplanted mature astrocytes were more susceptible to phagocytosis by the host immune system (Smith and Miller, 1991). Another study also reported the ability of transplanted immature, but not mature, astrocytes to migrate into the site of injury, where they form permissive bridges that enable axon regeneration (Filous et al., 2010). Similar effects were observed when transplanting immature astrocytes derived from fetal glial-restricted progenitors (Davies et al., 2006; Haas and Fischer, 2013). It is also important to note that, unlike mature astrocytes, immature astrocytes do not produce massive amounts of chondroitin sulfate proteoglycans (CSPGs), a major inhibitory component of the glial scar, in response to injury (Dow et al., 1994). Together, these data strongly suggest inherent differences that arise in astrocytes as they mature.
4. NORMAL FUNCTIONS OF ASTROCYTES
Astrocytes were once thought to be passive support cells for neurons. It is now clear that these cells play an active role in regulating key functions of the nervous system. Astrocytes are important for maintaining homeostasis, formation of the blood brain barrier (BBB), regulating synaptic formation and function, and neuronal trophic support.
4.1 Role in the BBB
Astrocytes are known to play an active role in the function and maintenance of the BBB (for review see Alvarez et al., 2013). The BBB serves to regulate and limit the exchange between the nervous system and the vasculature. Endothelial cells of the vasculature in the brain form tight junctions, which limit the passive diffusion between the blood and CNS. Astrocytic endfeet contact the endothelial cells of the brain vasculature and are responsible for their formation of tight junctions (Janzer and Raff, 1987). The presence of astrocytes or astrocyte-conditioned media was sufficient to induce tight junctions and BBB behavior in cultured endothelial cells (Alvarez et al., 2011; Neuhaus et al., 1991). Because astrocyte processes both enwrap synapses and contact the endothelial cells of the BBB, they are able to modulate blood flow based on neural activity. Glutamate-mediated calcium signaling within astrocytes increases blood flow in the cortex (Zonta et al., 2003).
CNS injury often leads to disruption of the BBB, which ultimately affects the behavior of astrocytes. After CNS injury, Bardehle et al. found that only astrocytes adjacent to the vasculature proliferate (Bardehle et al., 2013), consistent with the idea that these cells act as injury sensors due to their polarized endfeet contacts with endothelial cells of the vasculature. Disruption of the interaction between astrocytes and the CNS vasculature leads to reactive astrogliosis. Conditionally deleting beta 1-integrin in astrocytes, a protein at the interface between endfeet and the basement membrane of the BBB, led to hypertrophy of astrocytes as characterized by upregulation of GFAP and vimentin, as well as increased secretion of CSPGs (Robel et al., 2009), suggesting a possible role for this receptor in mediating partial activation of astrocytes in response to injury. Disruption of the BBB also allows for the influx of the soluble plasma protein, fibrinogen, which induces TGFβ signaling and leads to activation of astrocytes, as well as scar formation (Schachtrup et al., 2010, see below). It should also be noted that astrogliosis itself may disrupt the interaction between astrocytes and CNS vasculature, which may further exacerbate the injury response.
4.2 Role in synaptogenesis and synaptic function
Astrocyte processes, together with neuronal pre- and post-synaptic regions, form the tripartite synapse. Astrocytes play an active role in modifying synaptic strength and function through their expression of neurotransmitter receptors and release of various neurotransmitters, such as glutamate, GABA, ATP and D-serine. Neuronal activity results in neurotransmitter release, which signals through neurotransmitter receptors expressed on astrocytes to trigger calcium signals within these cells, a phenomenon known as astrocyte excitability. Ultimately, this signaling leads to the release of different gliotransmitters such as prostaglandins, allowing them to alter synaptic function (Shigetomi et al., 2008). The glutamate transporters expressed by astrocytes, including GLAST and GLT-1, are important for removing glutamate from the synaptic cleft and reducing excitotoxicity (Rothstein et al., 1996). Neurons activate nuclear factor kappa B (NFκB) signaling in astrocytes, leading to upregulation of GLT-1 expression (Ghosh et al., 2011). Brain expression of glutamine synthase is found exclusively in astrocytes (Norenberg and Martinez-Hernandez, 1979). This enzyme is responsible for converting glutamate to glutamine, maintaining glutamate homeostasis (Rose et al., 2013). Disruption of this astrocyte function has critical consequences in injury and disease, as discussed below.
Astrocytes also facilitate synaptogenesis. Work from Ben Barres’s lab has demonstrated that astrocytes are necessary for the formation of mature, functional synapses in the CNS and are required for the maintenance of these synapses (Christopherson et al., 2005; Ullian et al., 2001, 2004). These findings translate to a variety of neuronal subtypes, including spinal motor neurons (Ullian et al., 2004). Follow-up work found that synaptogenesis was mediated by the astrocyte-secreted factor, thrombospondin (Christopherson et al., 2005), an important matrix molecule which is induced by purinergic signaling (Tran and Neary, 2006). Astrocytes are also able to mediate the formation of inhibitory synapses (Elmariah et al., 2005). While synaptogenesis ends in development, reactive astrocytes can help to restore synapses after injury (Emirandetti et al., 2006; Tyzack et al., 2014) and may provide a target for promoting plasticity and recovery.
In addition to their constructive role at the synapse, astrocytes also play a role in synaptic plasticity and pruning. Astrocytes have been found to be important in presynaptic muting of hippocampal neurons, again through their expression of thrombospondins (Crawford et al., 2012). Another astrocyte-secreted protein, hevin, has been implicated in regulating cortical connectivity during development by refining dendritic spines (Risher et al., 2014). They also play a role in targeting synapses for elimination (Stevens et al., 2007).
5. ASTROCYTE RESPONSE TO INJURY
Injury to the CNS causes a cascade of cellular and molecular changes that alter the local environment and impede regeneration. The most immediate effects of CNS injury are bleeding, followed by intense local inflammation that leads to progressive cavitation of the lesioned area (Fitch et al., 1999; Horn et al., 2008). The response of astrocytes varies by location in relation to the severity of the injury (described below), but those in the vicinity of the injured tissue become hypertrophic and begin to form a dense scar tissue to wall off the area of damage from the surrounding healthy tissue (Cregg et al., 2014). Although there is an abundance of astrocytes in scar tissue and they have traditionally been thought to be the major scar-forming component, recent work from the Frisen and Jae Lee labs demonstrates that fibroblasts actually outnumber astrocytes in the scar and play a critical role in its formation as well (Göritz et al., 2011; Soderblom et al., 2013). Soon after a crush injury of the spinal cord, astrocytes near the lesion had elongated morphologies with overlapping processes, whereas astrocytes more distant from the lesion maintained their stellate morphology and non-overlapping domains, suggesting a heterogeneity in reactive astrogliosis based on the proximity to the lesion center (Wanner et al., 2013). Later on, processes of the elongated reactive astrocytes near the lesion edge adjacent to the fibroblast-like pericyte population form mesh-like structures that lead to scar formation. The scar itself contains two distinct regions: the lesion core, which is comprised mostly of NG2 glia (Busch et al., 2010; Filous et al., 2014), fibroblasts/pericytes (Zhu et al., 2015), and macrophages (Busch et al., 2009; Horn et al., 2008) and the penumbra, which is formed primarily by reactive astrocytes and activated microglia (Evans et al., 2014). NG2 glia are also found immediately adjacent to the lesion core, with the ability to form a bridge into the center of the lesion (Busch et al., 2010; Cregg et al., 2014; Filous et al., 2014). Within the first week after spinal cord injury, microglia/macrophages and NG2+ cells proliferate in the injured white matter and begin to occupy the lesion core (Zai and Wrathall, 2005). The density of astrocytes and astrocyte proliferation in the penumbra of the lesion nearly doubles compared to that of uninjured tissue (Wanner et al., 2013). Traditionally, the glial scar has been viewed primarily as a major impediment to regeneration, but more recent evidence suggests that this structure is necessary to prevent the spread of injury (Faulkner et al., 2004). The exact role of astrocytes after injury may be better characterized by their distance from the site of injury and the severity of the insult.
The protein expression pattern of astrocytes also changes in response to injury. It is well-established that astrocytes near the site of injury upregulate their expression of GFAP (Vijayan et al., 1990). They also increase their expression of S100β (Rothermundt et al., 2003). Some reports suggest they increase their expression the glutamate transporters, GLT-1 and GLAST (Vera-Portocarrero et al., 2002), while other studies have found that GLT-1 expression is actually reduced after injury (Lepore et al., 2011). Injury also induces astrocytes to re-express their developmental filament decorating proteins vimentin (Miyake et al., 1988) and nestin (Clarke et al., 1994). The altered protein expression in astrocytes after injury may help their motility away from the lesion core into the penumbra and allow the cells to assume a variety of shapes as they undertake specific mechanical as well as biochemical roles during their attempt to wall off the lesion.
5.1 Mild, Moderate, Severe Astrogliosis
The degree of astrogliosis has recently been defined as mild, moderate, or severe, based on the molecular, cellular, and functional changes that occur in the astrocytes, as well as the severity of the insult (Sofroniew and Vinters, 2010). Astrocyte responses are not all-or-none phenomena. The severity of reactive astrogliosis may determine its effects on regeneration and recovery.
Less severe injuries result in mild to moderate reactive astrogliosis. Although astrocytes upregulate expression of GFAP, they do not proliferate in response to minor insults nor do they overlap neighboring astrocytes (Sofroniew, 2009). Astrocytes distant from the lesion environment may be defined as mildly activated. Many of the changes induced by minor insults are reversible. However, severe insults, such as focal lesions, infections, or chronic neurodegeneration, cause astrocytes to upregulate GFAP and other genes, as well as to proliferate and overlap neighboring astrocyte domains (Sofroniew and Vinters, 2010). These changes lead to long-term changes in the tissue structure and over time lead to extremely dense accumulations of cells that appear to be mechanically obstructive to axon regeneration (Silver and Miller, 2004).
5.2 Glial Scar Formation
The molecular triggers of reactive astrogliosis vary by the type of insult and are incompletely understood (for review see Sofroniew, 2009). Injury-induced cytokines, such as ciliary neurotrophic factor (CNTF), interleukin-6, transforming growth factor alpha, and fibroblastic growth factor-2, together with epidermal growth factor have been reported to enhance astrocyte proliferation, potentially contributing to glial scar formation (Levison et al., 2000). Using a brain injury mouse model, Vartak-Sharma and Ghorpade found that GFAP and astrocyte-elevated gene 1 (AEG-1) colocalized at the site of injury and that knocking down AEG-1 reduced astrocyte migration and proliferation in the lesion (Vartak-Sharma and Ghorpade, 2012). Another factor implicated in triggering astrogliosis is endothelin-1 (ET-1). ET-1 is upregulated along the same time course as astrocyte proliferation and GFAP expression in the corpus callosum after lysolecithin-induced focal demyelination and is able to induce astrocyte proliferation in culture (Gadea et al., 2008). Inhibiting matrix metalloproteinase 9 activity prevents the migration of astrocytes through the disruption of actin cytoskeleton dynamics (Hsu et al., 2008). LPS and other Toll-like receptor ligands (Farina et al., 2007), as well as neurotransmitters such as glutamate (Bekar et al., 2008) may also play a role in signaling for reactive astrogliosis. Purinergic signaling through protein kinase cascades has also been implicated in stimulating astrocyte proliferation in response to injury (Neary et al., 2006). Several studies have also shown a correlation between TGFβ signaling and astrocyte activation after SCI (Kohta et al., 2009; O’Brien et al., 1994). More recently, TGFβ has been linked to the blood protein fibrinogen in signaling for glial scar formation. After disruption of the BBB, fibrinogen enters the CNS, where it signals through Smad2 and TGFβ in astrocytes to cause CSPG deposition into the extracellular matrix, leading to inhibition of neurite outgrowth (Schachtrup et al., 2010). Depleting fibrinogen or inhibiting TGFβ signaling blocked glial activation and CSPG expression, providing convincing evidence of the role of these molecules in triggering reactive astrogliosis. TGFβ has also been shown to activate Smad3, leading to glial scar formation after a cortical stab injury. Inhibiting Smad3 signaling reduced the number of immune cells, as well as NG2+ cells, and astrocytes around the lesion area after traumatic brain injury, while also reducing laminin and fibronectin expression (Wang et al., 2007). Conditional deletion of Smad3 reduced expression of CSPGs, collagens, and GFAP one week after contusive spinal cord injury in mice (McKillop et al., 2013). Together, these molecules provide potential targets for altering astrocyte proliferation and migration in response to injury.
Neuroinflammation may play a major role in triggering astrogliosis. Inflammation is one of the earliest responses to injury, occurring within minutes, and is later followed by reactive astrogliosis (Evans et al., 2014). A strong correlation also exists between the appearance of astrogliosis and the accumulation of reactive microglia/macrophages (Balasingam et al., 1996). To demonstrate the importance of inflammatory cytokines in astrogliosis, Balasingam et al. utilized a neonatal mouse model system, in which astrogliosis does not normally occur and the immune system is still developing. A stab wound into the neonatal cortex, immediately followed by microinjection of a variety of cytokines, including interferon-γ, interleukin-1, interleukin-2, interleukin-6, tumor necrosis factor-α, and macrophage colony stimulating factor, resulted in a significant increase in astrocyte reactivity, as assessed by upregulation of GFAP expression (Balasingam et al., 1994). Blocking interleukin-6 signaling immediately following contusive spinal cord injury in rats prevented the differentiation of neural stem/progenitor cells into astrocytes, while also reducing the number of invading inflammatory cells and the formation of the glial scar (Okada et al., 2004). Similar results using double transgenic mice (Brunello et al., 2000) and interleukin-6 deficient mice (Klein et al., 1997) implicate interleukin-6 in the selective activation of astrocytes. Interleukin-1 also has the ability to induce GFAP upregulation and astrocyte hypertrophy when injected directly into the cerebral cortex of adult rats. It has also been reported that activated macrophages stimulate upregulation of inhibitory CSPGs after injury (Fitch and Silver, 1997). Conversely, anti-inflammatory cytokines such as interleukin-10 (Balasingam and Yong, 1996) and type 1 interferon β (Ito et al., 2009) are able to reduce astrocyte reactivity, further suggesting that neuroinflammation plays a major role in modulating astrogliosis.
5.3 Scar Inhibition of Regeneration
Reactive astrocytes have a well-established role in inhibiting neurite outgrowth and regeneration after injury (Cregg et al., 2014; Silver and Miller, 2004). Much of the inhibitory nature associated with the glial scar is attributed to the extracellular matrix CSPGs produced by mature reactive astrocytes. CSPGs, present in high concentrations around the lesioned area only in the adult CNS, are associated with reactive astrocytes. Scar tissue isolated from the lesioned adult CNS was unable to support neurite outgrowth, which correlated with the expression of CSPGs in this tissue (McKeon et al., 1991). When adult rat DRG neurons were microtransplanted directly into white matter tracts of the CNS, they were capable of extensive regeneration, unless they encountered increased concentrations of CSPGs in the extracellular matrix at the site of transplantation (Davies et al., 1997) or at a distal lesion site (Davies et al., 1999), establishing the inhibitory nature of these molecules. High concentrations of CSPGs in a gradient cause growing axons to become dystrophic and stall, preventing regeneration (Lang et al., 2015; Tom et al., 2004a). Targeting of these molecules to promote regeneration into and beyond the glial scar has been a major field of study (Bradbury and Carter, 2011; McKeon et al., 1995). One of the most common methods of degrading CSPGs experimentally has been through the use of an enzyme known as chondroitinase ABC. This enzyme cleaves the inhibitory glycosaminoglycan side chains from the protein core, removing the inhibitory portion of the CSPG molecule that binds to its receptors (Fisher et al., 2011; Shen et al., 2009) and allowing for regeneration/plasticity. As such, this enzyme has been used in a variety of studies to improve regeneration or plasticity after spinal cord injury (Bradbury et al., 2002; Massey et al., 2006; Tom et al., 2009), brain injury (Moon et al., 2001), and in combinatorial strategies (Alilain et al., 2011; Filous et al., 2010; Zhao and Fawcett, 2013; Zhao et al., 2013). Work with this enzyme demonstrates that manipulating the CSPG composition of the glial scar may be an important step in overcoming regeneration failure.
Studies ablating astrocytes or altering expression of intermediate filament decorating proteins have shown mixed results in regards to regeneration and functional recovery. Because GFAP is abundantly expressed throughout the glial scar, studies were performed using GFAP null mice to examine glial scar formation. Surprisingly, there were no differences in scar formation or the upregulation of vimentin in these mice compared to wildtype controls after a cortical needle stab injury (Pekny et al., 1995). Therefore, although GFAP upregulation is a hallmark of astrogliosis, its elimination alone is insufficient to totally disrupt formation of a glial scar. However, GFAP upregulation does play a role in the ability of astrocytes to respond to beta amyloid, since GFAP null astrocytes in culture upon beta amyloid substrates responded more slowly and were unable to form tight bundles (Xu et al., 1999), suggesting that certain aspects of tight wall formation do appear to be dependent on GFAP levels.
A study using a knife wound to the dorsal funiculus of the spinal cord found that mice lacking two of the major proteins of the astrocyte cytoskeleton, GFAP and vimentin, form less dense scars in response to CNS injury (Pekny et al., 1999). Knocking out just one of these intermediate filaments did not affect scar formation, suggesting both are necessary for this process to occur. Because knocking out both proteins has major effects on astrocyte reactivity in response to injury, these mice have been used in a variety of studies to explore the role of astrogliosis. Using aged GFAP(−/−)Vimentin(−/−) mice, lacking the major proteins of the astrocyte cytoskeleton, Larsson et al. demonstrated that cell survival and neurogenesis in the hippocampus were enhanced compared to their age-matched controls, suggesting that astrocyte reactivity may limit hippocampal neurogenesis (Larsson et al., 2004). Other studies using these mice found that astrocyte hypertrophy was reduced after a lesion of the entorhinal cortex, allowing for enhanced synaptic regeneration in the hippocampus (Wilhelmsson et al., 2004). Similar results were found using these same mice to explore the effects of reducing astrogliosis after a hemisection of the spinal cord. GFAP(−/−)Vimentin(−/−) mice had reduced astrocyte reactivity, which was associated with increased sprouting of supraspinal fibers and increased functional recovery (Menet et al., 2003), suggesting that reactive astrocytes may play a role in limiting plasticity after spinal cord injury. However, complete ablation of astrocytes has been reported to worsen the outcome after mild to moderate SCI or after a major stroke (discussed below; Faulkner et al., 2004). Therefore, simply depleting reactive astrocytes to improve recovery after CNS insult may deprive the system of valuable astrocyte functions important for neuroprotection and repair.
5.4 Beneficial Effects of Astrocytes in the Glial Scar
Although extensive research has focused on the glial scar as a physical and molecular impediment to axonal regeneration, research over the last decade has confirmed what evolutionary conservation suggests, that the glial scar is essential for preventing the spread of damage to neighboring tissue. Studies ablating astrocytes or markedly inhibiting the formation of the glial scar have convincingly demonstrated that the scar is necessary to contain the injury and spare the fragile surrounding tissue. By conditionally ablating dividing reactive astrocytes after either stab or crush injuries, Faulkner et al. demonstrated that without these cells, the effects of mild to moderate SCI were exacerbated, leading to persistent blood-brain barrier disruption, more pronounced leukocyte infiltration into the lesioned area, and more pronounced cellular death of neighboring neurons and oligodendrocytes resulting in more severe demyelination and ultimately leading to increased functional deficits (Faulkner et al., 2004). Similar studies were performed to confirm these results in traumatic brain injury. Selectively ablating astrocytes in the vicinity of a forebrain stab injury prevented the repair of the BBB, chronically increased the infiltration of leukocytes, and enhanced neuron degeneration. However, neurite outgrowth was somewhat increased at the site of injury, confirming the dual role of the glial scar in preventing the spread of damage, while also inhibiting regeneration (Bush et al., 1999). Ablating astrocytes after moderate cortical contusion caused a greater loss of cortical tissue, more neuronal degeneration, and increased inflammation, again suggesting that astrocytes are necessary for tissue preservation after CNS injury (Myer et al., 2006). Using mice with a selective deletion of protein signal transducer and activator of transcription 3 (Stat3) under the control of the Nes promoter-enhancer, Stat3 has been implicated as a key regulator of reactive astrocytes and their beneficial role in wound healing after CNS injury (Okada et al., 2006). Ablating Stat3 in astrocytes prevented their migration in response to injury, and also caused demyelination, pronounced immune cell infiltration, and exacerbated functional deficits. Together, these data strongly suggest that astrocytes are necessary to promote wound repair, even though their presence in the scar may also contribute to regeneration failure.
Many studies suggest that reactive astrocytes are also neuroprotective after stroke (Liu et al., 2014). Not only do astrocytes upregulate GFAP and vimentin, but they also begin to re-express nestin in response to stroke or brain injury (Li and Chopp, 1999). Mice lacking both GFAP and vimentin saw a 2.1–3.5 increase in infarct volume compared to WT controls following middle cerebral artery occlusion (Li et al., 2008). Astrocytes cultured from GFAP(−/−)Vimentin(−/−) mice were more susceptible to apoptosis and less supportive of neurons in co-culture compared to WT controls in an oxygen-deprivation/reperfusion model of stroke in vitro, suggesting reactive astrocytes may be necessary to provide neuroprotection (de Pablo et al., 2013). Because of the reports that astrocytes may play a crucial role in neuroprotection, they have become model cells to target to improve functional recovery after ischemia (Li et al., 2014).
The astrocyte response to injury is multifaceted and may promote neuroprotection and repair through a variety of mechanisms. Reactive astrocytes upregulate expression of a variety of molecules that act to support the injured neurons directly. In addition to their production of inhibitory CSPGs, reactive astrocytes produce growth permissive extracellular matrix molecules as well, such as laminin (Canning et al., 1996; Frisén et al., 1995) and fibronectin (Tom et al., 2004b). These molecules provide a supportive substrate for injured neurons as they attempt to regenerate. Reactive astrocytes also upregulate the glutamate transporters GLAST and GLT-1 (Rothstein et al., 1996), which helps protect spared tissue from excitotoxicity. Evidence from co-cultures of astrocytes and neurons suggests that astrocytes produce glutathione, which protects neurons from nitric oxide neurotoxicity (Chen et al., 2001). The neurotrophic factors produced by astrocytes, such as CNTF, (Lee et al., 1998) and brain-derived neurotrophic factor (BDNF, Ikeda et al., 2001) also help support the survival of neurons. A recent study demonstrated that astrocyte conditioned media containing glial derived neurotrophic factor (GDNF) was able to abolish Zymosan-induced activation of microglia, in turn promoting neuron survival by reducing neuroinflammation (Rocha et al., 2012). Therefore, astrocytes may provide crucial support for neurons which helps in the survival of tissue surrounding an injury. It is also important to stress that while reactive astrocytes are largely inhibitory to the passage of axons in vivo due to their mesh-like configuration as well as the production of inhibitory extracellular CSPG containing matrices, the membrane surface of intensely reactive astrocytes (excluding or lacking CSPGs) when presented as a 2 dimensional substrate can be growth supportive (Canning et al., 1996).
5.5 Astrocyte Effects on Other Glia
In addition to affecting neurons, astrocytes play an important role in the regulation of other glial cell types after injury. Understanding their effects on these cells is important for understanding their complex role after injury or disease.
5.5.1 Oligodendrocytes and NG2+ cells/OPCs
Under normal conditions, astrocytes have been shown to promote myelination in vitro (Sorensen et al., 2008). In vivo, astrocytes have been shown to release the cytokine leukemia inhibitory factor (LIF) in response to ATP released from active neurons, and LIF is able to promote myelination by mature oligodendrocytes (Ishibashi et al., 2006). During development, expression of tissue inhibitor of metalloproteinases 1 (TIMP-1) is high and promotes both astrocyte proliferation and oligodendrocyte progenitor cell (OPC) differentiation (Moore et al., 2011). Expression of TIMP-1 increases in response to CNS insult, where it may influence astrocyte behavior once again. The importance of astrocytes in myelination is further demonstrated by altering GFAP expression. GFAP-null mice had disrupted white matter architecture and abnormal myelination, as observed in the optic nerve and spinal cord (Liedtke et al., 1996), providing strong evidence that normal astrocyte function is necessary for proper myelination to occur. GFAP mutations are also associated with Alexander disease, which leads to myelination deficits and loss of oligodendrocytes (Alexander, 1949). In Alexander disease, GFAP mutations cause a disruption in glutamate transport in astrocytes, leading to oligodendrocyte death (Tian et al., 2010). Together, these data demonstrate that astrocytes play a key role in regulating oligodendrocyte behavior.
Reactive astrocytes play an important role in regulating oligodendrocyte behavior and myelination after injury as well. Mild astrogliosis may be beneficial to myelination and oligodendrogenesis (White and Jakeman, 2008). Nash and colleagues used an in vitro model to compare myelination of neuronal fibers grown on a monolayer of quiescent astrocytes to myelination when grown on a monolayer of mildly reactive astrocytes. Astrocytes plated with CNTF to mimic a reactive state were able to promote myelination in culture, whereas quiescent astrocytes were not (Nash et al., 2011), suggesting that mildly reactive astrocytes may play a crucial role in promoting remyelination after injury. A study using ethidium bromide-induced demyelination of the adult spinal cord found that although OPCs were successfully recruited to the lesion area, they were unable to successfully remyelinate axons in areas devoid of astrocytes (Talbott et al., 2005). A study using a contusive spinal cord injury model found that reactive astrocytes secrete bone morphogenetic proteins (BMPs) which actually inhibit OPC differentiation into oligodendrocytes, but rather promotes their differentiation into astrocytes (Wang et al., 2011). Inhibiting the expression of NFκB specifically in astrocytes, using GFAP-IκBα-dn mice, enhanced the proliferation of oligodendrocytes and the expression of myelin-associated proteins after spinal cord injury (Bracchi-Ricard et al., 2013), further supporting the idea that intensely reactive astrocytes in the spinal cord may inhibit remyelination after injury. Astrocytes have also been shown to play a role in models of multiple sclerosis (MS). There is increasing evidence that the extracellular matrix and particularly astroglial or OPC-produced CSPGs play a critical role in regulating OPC re-myelinating capabilities (Lau et al., 2012, 2013). Astrocyte expression of the chemokine CXCL10 in experimental autoimmune encephalomyelitis (EAE), serves as a chemoattractant for immune cells during disease progression (Omari et al., 2005; Ransohoff et al., 1993) leading to demyelination. Reactive astrogliosis has also been reported to cause a loss of gap junctions and a disconnection of oligodendrocytes from astrocytes in a model of MS, leading to myelination deficits (Markoullis et al., 2014). Therefore astrogliosis may have varying effects on oligodendrocyte behavior based on the severity of the insult (Franklin and Ffrench-Constant, 2008).
5.5.2 Neuroinflammation
As described above, neuroinflammation plays a major role in signaling for astrocyte reactivity in response to injury. However, astrocytes play a role in regulating inflammation as well. Culturing blood-derived monocytes on a monolayer of astrocytes caused the monocytes to deactivate and take on microglial-like properties, such as a ramified morphology and microglia related membrane currents (Schmidtmayer et al., 1994; Sievers et al., 1994a, 1994b), suggesting that astrocytes play a role in modulating immune cell behavior. It is much more difficult to determine the direct effects of astrocytes on microglia and macrophages in vivo, because both cell types act on one another and it is difficult to distinguish the source of various cytokines. Astrocytes have been shown to dramatically alter microglial behavior in response to amyloid plaques (DeWitt et al., 1998a). When astrocytes respond vigorously to aggregated amyloid, they surround the plaque and further seclude it by synthesizing a CSPG rich matrix (Canning et al., 1993; DeWitt and Silver, 1996; DeWitt et al., 1993, 1994, 1998b). Such reactive astrocyte activity, in turn, helps to shield the plaque from an aggressive engulfment by microglia, which increases the presence of plaque material within the brain. This data suggests that astrocytes may play a role in Alzheimer’s disease by altering the immune response. Reactive astrocytes secrete other factors in response to injury that play a role in modifying immune cell behavior. ATP released from astrocytes in response to local injury has been shown to dramatically affect the morphology of infiltrating microglia, acting as a chemoattractant (Davalos et al., 2005). Astrocyte-released ATP was able to stimulate microglia through purinergic receptor P2X(7), causing their rapid migration toward the lesion, followed by an increase in membrane permeability and ultimately microglial apoptosis, suggesting that ATP secreted from astrocytes in response to CNS injury may alter microglial function and number (Verderio and Matteoli, 2001). Stimulating ATP release from astrocytes was also able to induce vesicle shedding and the release of interleukin-1β from microglia in culture (Bianco et al., 2005). Glutamate released from astrocytes in a Ca2+-dependent fashion has also been shown to affect microglial activation. Reactive astrocytes are known to secrete various neurotrophic factors, such as IGF, NGF, BDNF, CNTF, and NT-3, which can support surrounding cells (Escartin and Bonvento, 2008) and alter immune cell behavior. Recently, it has been suggested that zinc released from astrocytes under hypo-osmotic conditions can alter microglial activation as well, as defined by morphology (Segawa et al., 2014). The transcription factor NFκB upregulates a variety of genes in response to trauma and disease, in astrocytes in particular, and has a well-established role in regulating inflammation. Transgenic mice in which NFκB translocation into the nucleus is blocked in astrocytes showed significant functional recovery after a contusive SCI, which correlated with reduced proinflammatory cytokines and reduced expression of CSPGs (Brambilla et al., 2005). There is even some evidence to suggest that CSPGs and other extracellular matrix molecules produced by reactive astrocytes may affect immune cells by binding the chemoattractants and growth factors necessary for recruitment and activation of macrophages (Hayashi et al., 2001; Rolls et al., 2008) and dendritic cells (Kodaira et al., 2000), thereby causing a focal concentration of these factors to enhance immune cell infiltration at the site of injury.
6. ASTROCYTES IN NEUROPATHOLOGIES
6.1 Alzheimer’s Disease
Alzheimer’s disease (AD) is a progressive neurodegenerative disease characterized by the presence of extracellular amyloid beta (Aβ) plaques, intracellular neurofibrillary tau tangles, and the decline of cognitive function. As a major homeostatic cell type in the CNS, astrocytes have been implicated in AD pathology. Although they are far less efficient at clearing amyloid than are microglia and they mostly tend to allow for amyloid deposition rather than its removal (see discussion above, DeWitt et al., 1998a), astrocytes are able to phagocytose and accumulate small amounts of Aβ-42, and the level of Aβ-42 accumulation correlates with the severity of AD pathology (Nagele et al., 2003). Similar results were found when astrocytes were cultured on Aβ-plaque burdened brain slices from an AD mouse model (Wyss-Coray et al., 2003), suggesting that restoring astrocyte deficits in Aβ clearance may help provide a possible treatment for AD; however, improving microglial clearance capacity will be more effective (Cramer et al., 2012). Low-density lipoprotein receptor (LDLR), a cell surface receptor for apolipoprotein E (apoE), mediates Aβ uptake and degradation by astrocytes (Basak et al., 2012; Koistinaho et al., 2004). Therefore, reactive astrocytes serve to protect neurons from plaques through this phagocytosis (Mathur et al., 2015). Fluorescently labeled astrocytes from adult, but not neonatal, mice transplanted into an AD mouse brain were able to migrate and aggregate near Aβ plaques in the hippocampus, where they were able to internalize Aβ (Pihlaja et al., 2008). However, astrocytes often lyse as a result of Aβ accumulation, resulting in the formation of astrocyte-derived Aβ-plaques in the cortical molecular layer, contributing smaller plaques to the overall plaque load in the AD brain (Nagele et al., 2003). In addition to deficits in Aβ clearance, glutamate excitotoxicity may also lead to neurodegeneration. Using cultured rat brain astroctyes, Matos et al. found that glutamate uptake, particularly through GLT-1 is reduced in astrocytes cultured in the presence of Aβ1–40 peptide (Matos et al., 2008). A recent study looked to see if glutamate uptake or glutamate metabolism were altered in an AD mouse model. Researchers found that expression of the glutamate transporter GLT-1 was unaltered, suggesting that glutamate uptake from the synaptic cleft was unabated. Conversely, expression of glutamine synthase was reduced over time, leading to a gradual decline in astrocyte-dependent glutamate homeostasis. This disruption in glutamate levels results in failed synaptic connectivity and ultimately cognitive and memory deficits (Kulijewicz-Nawrot et al., 2013). Calcium homeostasis in reactive astrocytes may also be altered in AD. Using a mouse model of AD, Kuchibhotla et al. used multiphoton fluorescence microscopy to image calcium homeotstasis in astrocytes and found that these astrocytes exhibited elevated resting calcium, as well as intracellular calcium waves in astrocytes near plaques, suggesting that the astrocyte network could contribute to AD pathology (Kuchibhotla et al., 2009). Additionally, gap junctions between astrocytes are altered in AD, as evidenced by increased expression of the gap junctional protein connexin 43 (Nagy et al., 1996). Altered gap junction expression has also been linked to increased glutamate and ATP release, leading to neuronal death (Orellana et al., 2011), suggesting that blocking hemichannels on neurons could be neuroprotective in AD. These studies provide multiple approaches for restoring astrocyte function in AD to protect neurons.
6.2 ALS
Amyotrophic lateral sclerosis (ALS) is another chronic progressive neurodegenerative disorder, primarily causing the death of motor neurons in the cerebral cortex and spinal cord. Astrocytes play a major role in ALS pathology (for review see Vargas and Johnson, 2010). Patients with ALS have been reported to have impaired glutamate metabolism, caused by reduced glutamate uptake by astrocyte-associated glutamate transporters (Rothstein et al., 1992). A recent study found that the inflammatory cytokine TNF-α is able to increase the expression of GLT-1 in astrocytes cultured from wild-type rats, but not those cultured from a rat model of ALS (Dumont et al., 2014). Another study has linked astrocytes, inflammation, and ALS pathology, reporting that in both mouse and human ALS, astrocytes have upregulated expression of the anti-inflammatory cytokine transforming growth factor-β1 (TGF-β1), which prevents microglial and T cell production of IGF-1, leading to accelerated disease progression due to the loss of inflammatory-mediated neuroprotection (Endo et al., 2015). Astrocytes from both familial and sporadic ALS patients postmortem have been shown to be toxic to motor neurons (Haidet-Phillips et al., 2011). This may in part be due to upregulation of iNOS and other markers of oxidative stress by reactive astrocytes in ALS and ALS models (Almer et al., 1999; Sasaki et al., 2001). Additionally, a recent study found that astrocytes from ALS mice had a reduced ability to support neurons, similar to what is seen in aged astrocytes (Das and Svendsen, 2015), suggesting that their functions could be targeted for therapeutic intervention to reduce motor neuron death. Finally, over 95% of ALS cases present with aggregations of transactive response DNA binding protein (TDP-43), which leads to cellular toxicity and, importantly, the TDP-43 dysfunction has recently been linked to astrocytes (Yang et al., 2014).
6.3 Parkinson’s Disease
Loss of dopaminergic neurons in the substantia nigra leads to progressive neurodegeneration known as Parkinson’s disease (PD). PD is characterized by the presence of Lewy bodies (intranuclear aggregates of α-synuclein). Like the other neurodegenerative diseases described so far, astrocytes are able to take up α-synuclein, which ultimately disrupts astrocyte function. It has been suggested that affected cortico-striatal and cortico-thalamic neurons release α-synuclein from their axon terminals, where the protein is then taken up by surrounding astrocytes (Braak et al., 2007). Astrocyte endocytosis of neuron-released α-synuclein activates expression of inflammatory genes, leading to upregulation of pro-inflammatory cytokines and chemokines (Lee et al., 2010). Immunohistochemistry of different regions of PD brains revealed that protoplasmic astrocytes, but not fibrous astrocytes, accumulate α-synuclein and that this protein accumulation altered astrocyte reactivity (Song et al., 2009). Unlike in AD, where astrogliosis coincides with declining cognition, PD severity does not correlate with cortical astrogliosis (van den Berge et al., 2012), consistent with the finding that the accumulation of α-synuclein may hinder astrocyte reactivity and function. However, other studies selectively-expressing A53T α-synuclein in astrocytes showed increased paralysis in mice, which correlated with increased reactive astrogliosis, and impaired astrocyte function, such as reduced glutamate transport and cerebral hemorrhaging (Gu et al., 2010). Accumulation of α-synuclein in astrocytes led to microglial activation and neuron death (Gu et al., 2010), suggesting possible therapeutic opportunities for reducing this inflammation to preserve vulnerable neurons. Another study found that astrocyte activation, through ATF6α, is crucial for neuronal survival (Hashida et al., 2012). These results suggest that a careful manipulation of astrocyte activation and function may provide therapeutic potential for PD patients.
A variety of different genes have been implicated in PD. Although a deficiency of DJ-1 has been linked to familial PD, patients with sporadic PD have been found to have abundant expression of DJ-1 in reactive astrocytes, which has been found to be a compensatory neuroprotective mechanism (Mullett and Hinkle, 2009; Mullett et al., 2013). Co-culturing neurons with astrocytes overexpressing DJ-1 protected neurons from oxidative stress caused by rotenone, a chemical known to increase the risk of PD (Mullett et al., 2013). Targeting DJ-1 may have therapeutic benefits in protecting neurons in PD patients. Expression profiling was used to identify genes affected in reactive astrocytes of the striatum after dopamine depletion (Nakagawa and Schwartz, 2004). This study identified 29 genes in astrocytes with enhanced expression and 2 genes with decreased expression, providing a variety of astrocyte-specific targets to alter PD pathology.
6.4 Huntington’s Disease
Huntington’s disease (HD) is caused by the mutant protein, huntington. Its accumulation in both astrocytes and neurons leads to neuronal death. A wide variety of astrocyte functions have been implicated in facilitating neuronal death in HD, including glutamate toxicity, impaired GABA release, impaired secretion of trophic factors, increased inflammatory signaling, and reduced anti-inflammatory signaling. The R6 mouse model of HD shows reduced mRNA levels of GLT-1 in the striatum and reduced glutamate uptake prior to the development of neurodegeneration, suggesting astrocyte-mediated glutamate excitotoxicity leads to neuronal death (Liévens et al., 2001). Reduced expression of glutamate transporters correlates with the accumulation of mutant huntington protein in astrocytes (Shin et al., 2005). The level of glutamate transporter expression decreased corresponding with disease severity (Faideau et al., 2010). Interestingly, the excitotoxic stress in HD has been reported to stimulate striatal astrocytes to take on a pluripotent form to become neuroblasts (Nato et al., 2015), providing a possible mechanism for neuronal replacement. In addition to reduced glutamate uptake, astrocytes in HD models also have reduced GABA release, resulting in impaired tonic inhibition (Wójtowicz et al., 2013). Both patients and mouse models of HD show increased activation of the NFκB signaling in astrocytes, leading to enhanced inflammation (Hsiao et al., 2013). Inhibition of astrocyte-mediated inflammatory signaling through TNFα enhanced motor function and reduced aggregates of mutant huntington in a mouse model of HD, suggesting anti-inflammatory treatments may help slow the progression of HD (Hsiao et al., 2014). Recent findings report that cholesterol biosynthesis by astrocytes is reduced in HD, leading to neuronal deficits (Valenza et al., 2015). Additionally, accumulation of huntington aggregates in astrocytes reduced astrocytes secretion of brain derived neurotrophic factor (BDNF) (Wang et al., 2012). Alternatively, reactive astrocytes secrete pro-NGF, which leads to apoptosis of motor neurons (Domeniconi et al., 2007).
6.5 Epilepsy
Astrocyte dysfunction also plays a crucial role in epilepsy, as demonstrated in studies of patients with temporal lobe epilepsy and epilepsy models (Coulter and Steinhäuser, 2015). Disruption of K+ homeostasis, alterations in channel expression, dysfunctional gap junctions, and deficits in glutamate uptake together lead to seizures. Breakdown of the BBB leads to albumin accumulation, which stimulates upregulation of GFAP (David et al., 2009). A recent study suggests that this reactive astrogliosis is sufficient to induce seizures in mice (Robel et al., 2015), due to impaired glutamate uptake. Conditional deletion of the glutamate receptor GLT-1 in reactive astrocytes resulted in seizures and lower body weight (Petr et al., 2015). Elevated glutamate-mediated calcium signaling in astrocytes makes them hyperexcitable, leading to enhanced excitatory neurotransmission in epileptic hippocampal slices (Álvarez-Ferradas et al., 2015). Astrocytes in hippocampal slices of human patients suffering from temporal lobe epilepsy displayed prolonged depolarization and reduced inward rectifier currents (Hinterkeuser et al., 2000), implicating K+ homeostasis as an important contributor to epilepsy. Optogenetics may provide promising therapeutic potential to correct astrocytic glutamate and K+ uptake in epilepsy (Ji and Wang, 2015). Furthermore, proper functioning of gap junctions and inter-astrocytic communication are believed to play a role in the development of epilepsy, but their exact role is still unknown. Some reports suggest that astrocyte coupling is increased in murine epileptic models (Samoilova et al., 2003; Takahashi et al., 2010), whereas others suggest that gap junctions are decreased (Bedner et al., 2015; Xu et al., 2009). Expression studies have been conducted to determine the levels of connexin 43, the major gap junction protein, in epilepsy, but the results have also been mixed. Increased, decreased, and unaltered connexin expression have been reported in human epilepsy and animal models, making the results difficult to interpret without accompanying functional coupling analysis (Coulter and Steinhäuser, 2015).
6.6 Gliomas
Astrocyte interactions with glioma cells are important for glioma invasion, leading to poor survival. An interesting study looking at miRNA transfer found that gap junction mediated miRNA transfer between a glioma cell and another glioma cell does not allow for glioma invasion. In contrast, when miRNA transfer occurs between a glioma cell and an astrocyte, it promotes invasion in an in vitro transwell invasion assay (Hong et al., 2015). This finding suggests that gap junctions in astrocytes could be a crucial target for manipulation in preventing glioma invasion and promote patient survival. Using co-cultures of glioma cells and astrocytes, a separate study found that normal functioning astrocytes were able to reduce the rate of proliferation of glioma cells, but if astrocytes become outnumbered by glioma cells, they are no longer able to protect neurons from glioma cell glutamate secretion and excitotoxicity (Yao et al., 2014). Unlike lower grade gliomas, where non-neural metastases are self-contained, high grade tumor cells are able to infiltrate throughout the brain. The difference in the invasiveness of these tumors can be partly attributed to differences in the CSPG environment and surrounding astrocytes. In lower gliomas, a rich CSPG environment accompanied by the presence of LAR family receptors, along with an astrocytic capsule prevents the spread of the tumor. In highly invasive glioblastoma, the CSPG matrix as well as its receptors and astrocyte encapsulation are absent, favoring tumor cell invasion (Silver et al., 2013). A better understanding of astrocyte contribution to glioma growth and invasion could provide valuable therapeutic options for patients.
7. TARGETING ASTOCYTES FOR CNS REPAIR
7.1 Astrocytes in transplantation and bridge formation
Because of their reported benefits after injury and their important role in normal physiology, astrocytes have been used in transplantation studies to try to improve functional recovery. In particular, immature astrocytes have been used because of their growth-permissive properties without the negative effects of scar formation. In fact, transplanted immature, but not mature, astrocytes have the ability to suppress glial scar formation and thereby enhance neurite outgrowth in the mouse brain (Smith and Silver, 1988). Transplanted immature astrocytes are also more motile and associate better with blood vessels than mature astrocytes (Smith and Miller, 1991), which may account for their ability to suppress scar formation. Immature astrocytes also have a better ability to grow on high concentrations of CSPGs in an MMP-2 dependent manner (Filous et al., 2010).
In addition to promoting regeneration directly, astrocytes have been transplanted as a way to alter the behavior of other glia as well. Transplanting type-1 astrocytes into ethidium bromide lesions in the white matter of the spinal cord was able to enhance remyelination (Franklin et al., 1991).
Enhancing the beneficial neuroprotective effects of astrocytes after injury, while minimizing the negative effects on regeneration is a major interest in the field. One study used an intraparenchymal adeno-associated virus injected at the site of injury to induce overexpression TGFα (White et al., 2011). TGFα was able to transform neighboring astrocytes to a growth permissive phenotype that enhanced cell proliferation, altered their distribution, and led to increased regeneration to the rostral end of the lesion, suggesting that manipulation of astrocytes, rather than their ablation, may provide a promising avenue for therapy in the future.
Many studies have focused on the possibility of astrocytes to form bridges across the lesion core after injury. A study using a microlesion of the cingulate gyrus found that microtransplanting immature astrocytes along with the enzyme chondroitinase ABC to aid in proteoglycan degradation provided a bridge across the lesion environment that allowed for injured axons to regenerate just past the lesion (Filous et al., 2010). Suppressing the expression of phosphate and tenascin homolog (PTEN) with short hairpin RNA in mouse corticospinal neurons enabled injured fibers to cross the lesion along bridge-forming astrocytes, believed to be derived from mature astrocytes rather than ependymal cells, after spinal cord injury (Zukor et al., 2013). An earlier study suggested that adult cortical astrocytes retain the ability to revert to a more immature, even radial-glial like, state that was capable of directing the migration of transplanted immature neurons (Leavitt et al., 1999).
7.2 Modifying endogenous astrocytes to promote repair
Although in the normal CNS astrocytes are not generally thought to be neural stem cells, after injury, astrocytes have been found to de-differentiate and upregulate nestin expression (Lang et al., 2004; Shibuya et al., 2002). When cultured, these astrocytes had the ability to generate neurons, astrocytes, and oligodendrocytes (Lang et al., 2004), suggesting these cells take on neural stem cell properties in response to spinal cord injury (Götz et al., 2015). Other studies suggest that while astrocytes are able to proliferate in response to injury in vivo, their ability to generate neurospheres may simply be an in vitro phenomenon (Buffo et al., 2008). However, more recent work suggests that more invasive injuries, such as a stab wound or cerebral ischemia, are necessary to elicit the multipotency of reactive astrocytes in vivo, and that sonic hedgehog is necessary and sufficient to induce astrocytes to take on these stem-cell like properties (Sirko et al., 2013). The presence of endogenous stem cells after injury holds the promise of providing therapeutic options for repair.
7.3 Targeting specific molecules to enhance repair
7.3.1 Glutamate excitotoxicity
Astrocytes are important for regulating glutamate excitotoxicity within the synaptic cleft. Deficiencies in astrocyte uptake of glutamate has been linked to various neurodegenerative disorders, such as ALS (Rothstein et al., 1992). Impairments in glutamate transport and metabolism in astrocytes has also been implicated in epilepsy (Eid et al., 2013), oligodendrocyte death (Murugan et al., 2013), tauopathies (Dabir et al., 2006), and schizophrenia (Hu et al., 2015; Toro et al., 2006), whereas stroke actually leads to an increased expression of glutamate transporters (Yatomi et al., 2013). Therefore, modifying the action or expression of these glutamate transporters may provide a therapeutic target for these conditions. Various molecules have entered clinical trials to test the feasibility of manipulating astrocyte-mediated glutamate uptake in reducing neurodegeneration in stroke and ALS. Rothstein et al. discovered that beta-lactam antibiotics stimulate GLT-1 expression by increasing its transcription, making these FDA-approved drugs strong candidates to target glutamate excitotoxicity (Rothstein et al., 2005). One of these antibiotics, ceftriaxone, promoted EAAT2 activation through NFκB signaling (Lee et al., 2008). Furthermore, ceftriaxone is able to reduce glutamate mediated oxidative stress through the activation of antioxidant pathways (Lewerenz et al., 2009), further suggesting the possible therapeutic benefits of this drug in reducing glutamate toxicity. Alternatively, because astrocyte excitability is mediated through calcium signaling, targeting this pathway can be used to alter glutamatergic synapses.
7.3.2 Altering gap junctions
Gap junctions connect astrocytes and allow for intracellular communication. Disruptions of this signaling has been implicated in a variety of insults, making it an attractive target for therapeutic intervention. For example, ischemia leads to improper opening of connexin43 hemichannels, which make up gap junctions, leading to apoptotic cell death. However, inhibiting this signaling by pre-treating with either Gap26 or Gap27 reduced cerebral infarct volume and enhanced functional recovery (Li et al., 2015). Loss of connexin43 has also been linked to the progression of MS (Masaki, 2015). Working to restore proper glial communication could provide valuable therapeutic options for patients with many neuropathologies, including those described above.
8. CONCLUSION
It is clear that astrocytes are not mere bystanders in the complicated network of the nervous system. Their active participation in a variety of functions and pathways makes their proper functioning crucial to survival. Enhancing their beneficial roles while minimizing their deleterious effects holds enormous therapeutic potential in many diseases and insults.
Highlights.
Astrocytes function in blood-brain barrier formation and synaptogenesis under normal physiological conditions.
Astrocytes undergo varying degrees of astrogliosis in response to the severity of injury that has occurred.
Astrocytes in the glial scar have both beneficial and harmful effects on regeneration.
Astrocytes play a significant role in a variety of diseases, including Alzheimer’s disease, Amytrophic lateral sclerosis, Huntington’s disease, Parkinson’s disease, and gliomas.
Astrocytes can be targeted to repair the CNS after injury.
Acknowledgments
This work was supported by NINDS NS025713.
Glossary
- AD
Alzheimer’s disease
- AEG-1
Astrocyte-elevated gene 1
- ALS
Amyotrophic lateral sclerosis
- ApoE
Apolipoprotein E
- BBB
Blood-brain barrier
- BDNF
Brain-derived neurotrophic factor
- BLBP
Brain lipid binding protein
- BMP
bone morphogenetic protein
- CNTF
ciliary neurotrophic factor
- CSPG
Chondroitin sulfate proteoglycan
- ET-1
Endothelin-1
- GABA
γ-Aminobutyric acid
- Gcm
Glial cell missing
- GDNF
Glial derived neurotrophic factor
- GFAP
Glial fibrillary acidic protein
- GLAST
Glutamate aspartate transporter
- GLT-1
Glutamate transporter 1
- HD
Huntington’s disease
- IGF
Insulin-like growth factor
- LDLR
Low-density lipoprotein receptor
- LIF
leukemia inhibitory factor
- NFκB
nuclear factor kappa B
- NGF
Nerve growth factor
- NT-3
Neurotrophin 3
- OPC
Oligodendrocyte progenitor cell
- PD
Parkinson’s disease
- PTEN
Phosphate and tenascin homolog
- SCI
Spinal cord injury
- TDP-43
Transactive response DNA binding protein
- TGFβ
transforming growth factor
- TIMP-1
Tissue inhibitor of metalloproteinases 1
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 citable 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.
Contributor Information
Angela R. Filous, Department of Neurosciences, Case Western Reserve University, Cleveland, OH, Arn29@case.edu, 216-368-4615
Jerry Silver, Department of Neurosciences, Case Western Reserve University, Cleveland, OH, Jxs10@case.edu, 216-368-2150
References
- Alexander WS. Progressive fibrinoid degeneration of fibrillary astrocytes associated with mental retardation in a hydrocephalic infant. Brain J. Neurol. 1949;72:373–381. doi: 10.1093/brain/72.3.373. 3 pl. [DOI] [PubMed] [Google Scholar]
- Alilain WJ, Horn KP, Hu H, Dick TE, Silver J. Functional regeneration of respiratory pathways after spinal cord injury. Nature. 2011;475:196–200. doi: 10.1038/nature10199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Almer G, Vukosavic S, Romero N, Przedborski S. Inducible nitric oxide synthase up-regulation in a transgenic mouse model of familial amyotrophic lateral sclerosis. J. Neurochem. 1999;72:2415–2425. doi: 10.1046/j.1471-4159.1999.0722415.x. [DOI] [PubMed] [Google Scholar]
- Alvarez JI, Dodelet-Devillers A, Kebir H, Ifergan I, Fabre PJ, Terouz S, Sabbagh M, Wosik K, Bourbonnière L, Bernard M, et al. The Hedgehog pathway promotes blood-brain barrier integrity and CNS immune quiescence. Science. 2011;334:1727–1731. doi: 10.1126/science.1206936. [DOI] [PubMed] [Google Scholar]
- Alvarez JI, Katayama T, Prat A. Glial influence on the Blood Brain Barrier. Glia. 2013;61:1939–1958. doi: 10.1002/glia.22575. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Álvarez-Ferradas C, Morales JC, Wellmann M, Nualart F, Roncagliolo M, Fuenzalida M, Bonansco C. Enhanced astroglial Ca(2+) signaling increases excitatory synaptic strength in the epileptic brain. Glia. 2015 doi: 10.1002/glia.22817. [DOI] [PubMed] [Google Scholar]
- Bähr M, Przyrembel C, Bastmeyer M. Astrocytes from adult rat optic nerves are nonpermissive for regenerating retinal ganglion cell axons. Exp. Neurol. 1995;131:211–220. doi: 10.1016/0014-4886(95)90043-8. [DOI] [PubMed] [Google Scholar]
- Balasingam V, Yong VW. Attenuation of astroglial reactivity by interleukin-10. J. Neurosci. Off. J. Soc. Neurosci. 1996;16:2945–2955. doi: 10.1523/JNEUROSCI.16-09-02945.1996. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Balasingam V, Tejada-Berges T, Wright E, Bouckova R, Yong VW. Reactive astrogliosis in the neonatal mouse brain and its modulation by cytokines. J. Neurosci. Off. J. Soc. Neurosci. 1994;14:846–856. doi: 10.1523/JNEUROSCI.14-02-00846.1994. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Balasingam V, Dickson K, Brade A, Yong VW. Astrocyte reactivity in neonatal mice: apparent dependence on the presence of reactive microglia/macrophages. Glia. 1996;18:11–26. doi: 10.1002/(SICI)1098-1136(199609)18:1<11::AID-GLIA2>3.0.CO;2-6. [DOI] [PubMed] [Google Scholar]
- Bardehle S, Krüger M, Buggenthin F, Schwausch J, Ninkovic J, Clevers H, Snippert HJ, Theis FJ, Meyer-Luehmann M, Bechmann I, et al. Live imaging of astrocyte responses to acute injury reveals selective juxtavascular proliferation. Nat. Neurosci. 2013;16:580–586. doi: 10.1038/nn.3371. [DOI] [PubMed] [Google Scholar]
- Barnabé-Heider F, Göritz C, Sabelström H, Takebayashi H, Pfrieger FW, Meletis K, Frisén J. Origin of new glial cells in intact and injured adult spinal cord. Cell Stem Cell. 2010;7:470–482. doi: 10.1016/j.stem.2010.07.014. [DOI] [PubMed] [Google Scholar]
- Barrett CP, Donati EJ, Guth L. Differences between adult and neonatal rats in their astroglial response to spinal injury. Exp. Neurol. 1984;84:374–385. doi: 10.1016/0014-4886(84)90234-6. [DOI] [PubMed] [Google Scholar]
- Barry D, McDermott K. Differentiation of radial glia from radial precursor cells and transformation into astrocytes in the developing rat spinal cord. Glia. 2005;50:187–197. doi: 10.1002/glia.20166. [DOI] [PubMed] [Google Scholar]
- Basak JM, Verghese PB, Yoon H, Kim J, Holtzman DM. Low-density lipoprotein receptor represents an apolipoprotein E-independent pathway of Aβ uptake and degradation by astrocytes. J. Biol. Chem. 2012;287:13959–13971. doi: 10.1074/jbc.M111.288746. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bedner P, Dupper A, Hüttmann K, Müller J, Herde MK, Dublin P, Deshpande T, Schramm J, Häussler U, Haas CA, et al. Astrocyte uncoupling as a cause of human temporal lobe epilepsy. Brain J. Neurol. 2015;138:1208–1222. doi: 10.1093/brain/awv067. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bekar LK, He W, Nedergaard M. Locus Coeruleus α-Adrenergic–Mediated Activation of Cortical Astrocytes In Vivo. Cereb. Cortex N. Y. NY. 2008;18:2789–2795. doi: 10.1093/cercor/bhn040. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Van den Berge SA, Kevenaar JT, Sluijs JA, Hol EM. Dementia in Parkinson’s Disease Correlates with α-Synuclein Pathology but Not with Cortical Astrogliosis. Park. Dis. 2012;2012:420957. doi: 10.1155/2012/420957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bianco F, Pravettoni E, Colombo A, Schenk U, Möller T, Matteoli M, Verderio C. Astrocyte-derived ATP induces vesicle shedding and IL-1 beta release from microglia. J. Immunol. Baltim. Md 1950. 2005;174:7268–7277. doi: 10.4049/jimmunol.174.11.7268. [DOI] [PubMed] [Google Scholar]
- Braak H, Sastre M, Del Tredici K. Development of alpha-synuclein immunoreactive astrocytes in the forebrain parallels stages of intraneuronal pathology in sporadic Parkinson’s disease. Acta Neuropathol. (Berl.) 2007;114:231–241. doi: 10.1007/s00401-007-0244-3. [DOI] [PubMed] [Google Scholar]
- Bracchi-Ricard V, Lambertsen KL, Ricard J, Nathanson L, Karmally S, Johnstone J, Ellman DG, Frydel B, McTigue DM, Bethea JR. Inhibition of astroglial NF-kappaB enhances oligodendrogenesis following spinal cord injury. J. Neuroinflammation. 2013;10:92. doi: 10.1186/1742-2094-10-92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bradbury EJ, Carter LM. Manipulating the glial scar: chondroitinase ABC as a therapy for spinal cord injury. Brain Res. Bull. 2011;84:306–316. doi: 10.1016/j.brainresbull.2010.06.015. [DOI] [PubMed] [Google Scholar]
- Bradbury EJ, Moon LDF, Popat RJ, King VR, Bennett GS, Patel PN, Fawcett JW, McMahon SB. Chondroitinase ABC promotes functional recovery after spinal cord injury. Nature. 2002;416:636–640. doi: 10.1038/416636a. [DOI] [PubMed] [Google Scholar]
- Brambilla R, Bracchi-Ricard V, Hu W-H, Frydel B, Bramwell A, Karmally S, Green EJ, Bethea JR. Inhibition of astroglial nuclear factor kappaB reduces inflammation and improves functional recovery after spinal cord injury. J. Exp. Med. 2005;202:145–156. doi: 10.1084/jem.20041918. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Brunello AG, Weissenberger J, Kappeler A, Vallan C, Peters M, Rose-John S, Weis J. Astrocytic alterations in interleukin-6/Soluble interleukin-6 receptor alpha double-transgenic mice. Am. J. Pathol. 2000;157:1485–1493. doi: 10.1016/s0002-9440(10)64787-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Buffo A, Rite I, Tripathi P, Lepier A, Colak D, Horn A-P, Mori T, Götz M. Origin and progeny of reactive gliosis: A source of multipotent cells in the injured brain. Proc. Natl. Acad. Sci. U. S. A. 2008;105:3581–3586. doi: 10.1073/pnas.0709002105. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Busch SA, Horn KP, Silver DJ, Silver J. Overcoming Macrophage-Mediated Axonal Dieback Following CNS Injury. J. Neurosci. Off. J. Soc. Neurosci. 2009;29:9967–9976. doi: 10.1523/JNEUROSCI.1151-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Busch SA, Horn KP, Cuascut FX, Hawthorne AL, Bai L, Miller RH, Silver J. Adult NG2+ Cells are Permissive to Neurite Outgrowth and Stabilize Sensory Axons During Macrophage-Induced Axonal Dieback After Spinal Cord Injury. J. Neurosci. Off. J. Soc. Neurosci. 2010;30:255. doi: 10.1523/JNEUROSCI.3705-09.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Bush TG, Puvanachandra N, Horner CH, Polito A, Ostenfeld T, Svendsen CN, Mucke L, Johnson MH, Sofroniew MV. Leukocyte infiltration, neuronal degeneration, and neurite outgrowth after ablation of scar-forming, reactive astrocytes in adult transgenic mice. Neuron. 1999;23:297–308. doi: 10.1016/s0896-6273(00)80781-3. [DOI] [PubMed] [Google Scholar]
- Bushong EA, Martone ME, Jones YZ, Ellisman MH. Protoplasmic astrocytes in CA1 stratum radiatum occupy separate anatomical domains. J. Neurosci. Off. J. Soc. Neurosci. 2002;22:183–192. doi: 10.1523/JNEUROSCI.22-01-00183.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Canning DR, McKeon RJ, DeWitt DA, Perry G, Wujek JR, Frederickson RC, Silver J. beta-Amyloid of Alzheimer’s disease induces reactive gliosis that inhibits axonal outgrowth. Exp. Neurol. 1993;124:289–298. doi: 10.1006/exnr.1993.1199. [DOI] [PubMed] [Google Scholar]
- Canning DR, Höke A, Malemud CJ, Silver J. A potent inhibitor of neurite outgrowth that predominates in the extracellular matrix of reactive astrocytes. Int. J. Dev. Neurosci. Off. J. Int. Soc. Dev. Neurosci. 1996;14:153–175. doi: 10.1016/0736-5748(96)00004-4. [DOI] [PubMed] [Google Scholar]
- Chaboub LS, Deneen B. Developmental Origins of Astrocyte Heterogeneity: The final frontier of CNS development. Dev. Neurosci. 2012;34:379–388. doi: 10.1159/000343723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Chen Y, Vartiainen NE, Ying W, Chan PH, Koistinaho J, Swanson RA. Astrocytes protect neurons from nitric oxide toxicity by a glutathione-dependent mechanism. J. Neurochem. 2001;77:1601–1610. doi: 10.1046/j.1471-4159.2001.00374.x. [DOI] [PubMed] [Google Scholar]
- Christopherson KS, Ullian EM, Stokes CCA, Mullowney CE, Hell JW, Agah A, Lawler J, Mosher DF, Bornstein P, Barres BA. Thrombospondins are astrocyte-secreted proteins that promote CNS synaptogenesis. Cell. 2005;120:421–433. doi: 10.1016/j.cell.2004.12.020. [DOI] [PubMed] [Google Scholar]
- Clarke SR, Shetty AK, Bradley JL, Turner DA. Reactive astrocytes express the embryonic intermediate neurofilament nestin. Neuroreport. 1994;5:1885–1888. doi: 10.1097/00001756-199410000-00011. [DOI] [PubMed] [Google Scholar]
- Coulter DA, Steinhäuser C. Role of astrocytes in epilepsy. Cold Spring Harb. Perspect. Med. 2015;5:a022434. doi: 10.1101/cshperspect.a022434. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cramer PE, Cirrito JR, Wesson DW, Lee CYD, Karlo JC, Zinn AE, Casali BT, Restivo JL, Goebel WD, James MJ, et al. ApoE-directed therapeutics rapidly clear β-amyloid and reverse deficits in AD mouse models. Science. 2012;335:1503–1506. doi: 10.1126/science.1217697. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crawford DC, Jiang X, Taylor A, Mennerick S. Astrocyte-derived thrombospondins mediate the development of hippocampal presynaptic plasticity in vitro. J. Neurosci. Off. J. Soc. Neurosci. 2012;32:13100–13110. doi: 10.1523/JNEUROSCI.2604-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Cregg JM, DePaul MA, Filous AR, Lang BT, Tran A, Silver J. Functional regeneration beyond the glial scar. Exp. Neurol. 2014;253:197–207. doi: 10.1016/j.expneurol.2013.12.024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dabir DV, Robinson MB, Swanson E, Zhang B, Trojanowski JQ, Lee VM-Y, Forman MS. Impaired glutamate transport in a mouse model of tau pathology in astrocytes. J. Neurosci. Off. J. Soc. Neurosci. 2006;26:644–654. doi: 10.1523/JNEUROSCI.3861-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Das MM, Svendsen CN. Astrocytes show reduced support of motor neurons with aging that is accelerated in a rodent model of ALS. Neurobiol. Aging. 2015;36:1130–1139. doi: 10.1016/j.neurobiolaging.2014.09.020. [DOI] [PubMed] [Google Scholar]
- Davalos D, Grutzendler J, Yang G, Kim JV, Zuo Y, Jung S, Littman DR, Dustin ML, Gan W-B. ATP mediates rapid microglial response to local brain injury in vivo. Nat. Neurosci. 2005;8:752–758. doi: 10.1038/nn1472. [DOI] [PubMed] [Google Scholar]
- David Y, Cacheaux LP, Ivens S, Lapilover E, Heinemann U, Kaufer D, Friedman A. Astrocytic Dysfunction in Epileptogenesis: Consequence of Altered Potassium and Glutamate Homeostasis? J. Neurosci. 2009;29:10588–10599. doi: 10.1523/JNEUROSCI.2323-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davies JE, Huang C, Proschel C, Noble M, Mayer-Proschel M, Davies SJ. Astrocytes derived from glial-restricted precursors promote spinal cord repair. J. Biol. 2006;5:7. doi: 10.1186/jbiol35. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davies SJ, Fitch MT, Memberg SP, Hall AK, Raisman G, Silver J. Regeneration of adult axons in white matter tracts of the central nervous system. Nature. 1997;390:680–683. doi: 10.1038/37776. [DOI] [PubMed] [Google Scholar]
- Davies SJ, Goucher DR, Doller C, Silver J. Robust regeneration of adult sensory axons in degenerating white matter of the adult rat spinal cord. J. Neurosci. Off. J. Soc. Neurosci. 1999;19:5810–5822. doi: 10.1523/JNEUROSCI.19-14-05810.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Davies SJA, Shih C-H, Noble M, Mayer-Proschel M, Davies JE, Proschel C. Transplantation of specific human astrocytes promotes functional recovery after spinal cord injury. PloS One. 2011;6:e17328. doi: 10.1371/journal.pone.0017328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- DeWitt DA, Silver J. Regenerative failure: a potential mechanism for neuritic dystrophy in Alzheimer’s disease. Exp. Neurol. 1996;142:103–110. doi: 10.1006/exnr.1996.0182. [DOI] [PubMed] [Google Scholar]
- DeWitt DA, Silver J, Canning DR, Perry G. Chondroitin sulfate proteoglycans are associated with the lesions of Alzheimer’s disease. Exp. Neurol. 1993;121:149–152. doi: 10.1006/exnr.1993.1081. [DOI] [PubMed] [Google Scholar]
- DeWitt DA, Richey PL, Praprotnik D, Silver J, Perry G. Chondroitin sulfate proteoglycans are a common component of neuronal inclusions and astrocytic reaction in neurodegenerative diseases. Brain Res. 1994;656:205–209. doi: 10.1016/0006-8993(94)91386-2. [DOI] [PubMed] [Google Scholar]
- DeWitt DA, Perry G, Cohen M, Doller C, Silver J. Astrocytes regulate microglial phagocytosis of senile plaque cores of Alzheimer’s disease. Exp. Neurol. 1998a;149:329–340. doi: 10.1006/exnr.1997.6738. [DOI] [PubMed] [Google Scholar]
- DeWitt DA, Perry G, Cohen M, Doller C, Silver J. Astrocytes regulate microglial phagocytosis of senile plaque cores of Alzheimer’s disease. Exp. Neurol. 1998b;149:329–340. doi: 10.1006/exnr.1997.6738. [DOI] [PubMed] [Google Scholar]
- Domeniconi M, Hempstead BL, Chao MV. Pro-NGF secreted by astrocytes promotes motor neuron cell death. Mol. Cell. Neurosci. 2007;34:271–279. doi: 10.1016/j.mcn.2006.11.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dow KE, Ethell DW, Steeves JD, Riopelle RJ. Molecular correlates of spinal cord repair in the embryonic chick: heparan sulfate and chondroitin sulfate proteoglycans. Exp. Neurol. 1994;128:233–238. doi: 10.1006/exnr.1994.1132. [DOI] [PubMed] [Google Scholar]
- Dumont AO, Goursaud S, Desmet N, Hermans E. Differential regulation of glutamate transporter subtypes by pro-inflammatory cytokine TNF-α in cortical astrocytes from a rat model of amyotrophic lateral sclerosis. PloS One. 2014;9:e97649. doi: 10.1371/journal.pone.0097649. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Eid T, Tu N, Lee T-SW, Lai JCK. Regulation of astrocyte glutamine synthetase in epilepsy. Neurochem. Int. 2013;63:670–681. doi: 10.1016/j.neuint.2013.06.008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Elmariah SB, Oh EJ, Hughes EG, Balice-Gordon RJ. Astrocytes regulate inhibitory synapse formation via Trk-mediated modulation of postsynaptic GABAA receptors. J. Neurosci. Off. J. Soc. Neurosci. 2005;25:3638–3650. doi: 10.1523/JNEUROSCI.3980-04.2005. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Emirandetti A, Graciele Zanon R, Sabha M, de Oliveira ALR. Astrocyte reactivity influences the number of presynaptic terminals apposed to spinal motoneurons after axotomy. Brain Res. 2006;1095:35–42. doi: 10.1016/j.brainres.2006.04.021. [DOI] [PubMed] [Google Scholar]
- Endo F, Komine O, Fujimori-Tonou N, Katsuno M, Jin S, Watanabe S, Sobue G, Dezawa M, Wyss-Coray T, Yamanaka K. Astrocyte-Derived TGF-β1 Accelerates Disease Progression in ALS Mice by Interfering with the Neuroprotective Functions of Microglia and T Cells. Cell Rep. 2015;11:592–604. doi: 10.1016/j.celrep.2015.03.053. [DOI] [PubMed] [Google Scholar]
- Escartin C, Bonvento G. Targeted activation of astrocytes: a potential neuroprotective strategy. Mol. Neurobiol. 2008;38:231–241. doi: 10.1007/s12035-008-8043-y. [DOI] [PubMed] [Google Scholar]
- Evans TA, Barkauskas DS, Myers JT, Hare EG, You JQ, Ransohoff RM, Huang AY, Silver J. High-resolution intravital imaging reveals that blood-derived macrophages but not resident microglia facilitate secondary axonal dieback in traumatic spinal cord injury. Exp. Neurol. 2014;254:109–120. doi: 10.1016/j.expneurol.2014.01.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Faideau M, Kim J, Cormier K, Gilmore R, Welch M, Auregan G, Dufour N, Guillermier M, Brouillet E, Hantraye P, et al. In vivo expression of polyglutamine-expanded huntingtin by mouse striatal astrocytes impairs glutamate transport: a correlation with Huntington’s disease subjects. Hum. Mol. Genet. 2010;19:3053–3067. doi: 10.1093/hmg/ddq212. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Farina C, Aloisi F, Meinl E. Astrocytes are active players in cerebral innate immunity. Trends Immunol. 2007;28:138–145. doi: 10.1016/j.it.2007.01.005. [DOI] [PubMed] [Google Scholar]
- Faulkner JR, Herrmann JE, Woo MJ, Tansey KE, Doan NB, Sofroniew MV. Reactive Astrocytes Protect Tissue and Preserve Function after Spinal Cord Injury. J. Neurosci. 2004;24:2143–2155. doi: 10.1523/JNEUROSCI.3547-03.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ffrench-Constant C, Miller RH, Kruse J, Schachner M, Raff MC. Molecular specialization of astrocyte processes at nodes of Ranvier in rat optic nerve. J. Cell Biol. 1986;102:844–852. doi: 10.1083/jcb.102.3.844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Filous AR, Miller JH, Coulson-Thomas YM, Horn KP, Alilain WJ, Silver J. Immature astrocytes promote CNS axonal regeneration when combined with chondroitinase ABC. Dev. Neurobiol. 2010;70:826–841. doi: 10.1002/dneu.20820. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Filous AR, Tran A, Howell CJ, Busch SA, Evans TA, Stallcup WB, Kang SH, Bergles DE, Lee S, Levine JM, et al. Entrapment via synaptic-like connections between NG2 proteoglycan+ cells and dystrophic axons in the lesion plays a role in regeneration failure after spinal cord injury. J. Neurosci. Off. J. Soc. Neurosci. 2014;34:16369–16384. doi: 10.1523/JNEUROSCI.1309-14.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fisher D, Xing B, Dill J, Li H, Hoang HH, Zhao Z, Yang X-L, Bachoo R, Cannon S, Longo FM, et al. Leukocyte common antigen-related phosphatase is a functional receptor for chondroitin sulfate proteoglycan axon growth inhibitors. J. Neurosci. Off. J. Soc. Neurosci. 2011;31:14051–14066. doi: 10.1523/JNEUROSCI.1737-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Fitch MT, Silver J. Activated macrophages and the blood-brain barrier: inflammation after CNS injury leads to increases in putative inhibitory molecules. Exp. Neurol. 1997;148:587–603. doi: 10.1006/exnr.1997.6701. [DOI] [PubMed] [Google Scholar]
- Fitch MT, Doller C, Combs CK, Landreth GE, Silver J. Cellular and molecular mechanisms of glial scarring and progressive cavitation: in vivo and in vitro analysis of inflammation-induced secondary injury after CNS trauma. J. Neurosci. Off. J. Soc. Neurosci. 1999;19:8182–8198. doi: 10.1523/JNEUROSCI.19-19-08182.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Franklin RJM, Ffrench-Constant C. Remyelination in the CNS: from biology to therapy. Nat. Rev. Neurosci. 2008;9:839–855. doi: 10.1038/nrn2480. [DOI] [PubMed] [Google Scholar]
- Franklin RJ, Crang AJ, Blakemore WF. Transplanted type-1 astrocytes facilitate repair of demyelinating lesions by host oligodendrocytes in adult rat spinal cord. J. Neurocytol. 1991;20:420–430. doi: 10.1007/BF01355538. [DOI] [PubMed] [Google Scholar]
- Freeman MR, Rowitch DH. Evolving Concepts of Gliogenesis: A Look Way Back and Ahead to the Next 25 Years. Neuron. 2013;80:613–623. doi: 10.1016/j.neuron.2013.10.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Frisén J, Haegerstrand A, Risling M, Fried K, Johansson CB, Hammarberg H, Elde R, Hökfelt T, Cullheim S. Spinal axons in central nervous system scar tissue are closely related to lamininimmunoreactive astrocytes. Neuroscience. 1995;65:293–304. doi: 10.1016/0306-4522(94)00467-j. [DOI] [PubMed] [Google Scholar]
- Furuta A, Rothstein JD, Martin LJ. Glutamate Transporter Protein Subtypes Are Expressed Differentially during Rat CNS Development. J. Neurosci. 1997;17:8363–8375. doi: 10.1523/JNEUROSCI.17-21-08363.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gadea A, Schinelli S, Gallo V. Endothelin-1 regulates astrocyte proliferation and reactive gliosis via a JNK/c-Jun signaling pathway. J. Neurosci. Off. J. Soc. Neurosci. 2008;28:2394–2408. doi: 10.1523/JNEUROSCI.5652-07.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ghosh M, Yang Y, Rothstein JD, Robinson MB. Nuclear factor-κB contributes to neuron-dependent induction of glutamate transporter-1 expression in astrocytes. J. Neurosci. Off. J. Soc. Neurosci. 2011;31:9159–9169. doi: 10.1523/JNEUROSCI.0302-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Goldshmit Y, Sztal TE, Jusuf PR, Hall TE, Nguyen-Chi M, Currie PD. Fgf-dependent glial cell bridges facilitate spinal cord regeneration in zebrafish. J. Neurosci. Off. J. Soc. Neurosci. 2012;32:7477–7492. doi: 10.1523/JNEUROSCI.0758-12.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Göritz C, Dias DO, Tomilin N, Barbacid M, Shupliakov O, Frisén J. A pericyte origin of spinal cord scar tissue. Science. 2011;333:238–242. doi: 10.1126/science.1203165. [DOI] [PubMed] [Google Scholar]
- Götz M, Sirko S, Beckers J, Irmler M. Reactive astrocytes as neural stem or progenitor cells: In vivo lineage, In vitro potential, and Genome-wide expression analysis. Glia. 2015 doi: 10.1002/glia.22850. n/a – n/a. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Grafe MR, Schoenfeld TA. Radial glial cells in the postnatal olfactory tubercle of hamsters. Brain Res. 1982;256:115–118. doi: 10.1016/0165-3806(82)90103-1. [DOI] [PubMed] [Google Scholar]
- Gu X-L, Long C-X, Sun L, Xie C, Lin X, Cai H. Astrocytic expression of Parkinson’s disease-related A53T alpha-synuclein causes neurodegeneration in mice. Mol. Brain. 2010;3:12. doi: 10.1186/1756-6606-3-12. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Haas C, Fischer I. Human Astrocytes Derived from Glial Restricted Progenitors Support Regeneration of the Injured Spinal Cord. J. Neurotrauma. 2013;30:1035–1052. doi: 10.1089/neu.2013.2915. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Haidet-Phillips AM, Hester ME, Miranda CJ, Meyer K, Braun L, Frakes A, Song S, Likhite S, Murtha MJ, Foust KD, et al. Astrocytes from familial and sporadic ALS patients are toxic to motor neurons. Nat. Biotechnol. 2011;29:824–828. doi: 10.1038/nbt.1957. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hashida K, Kitao Y, Sudo H, Awa Y, Maeda S, Mori K, Takahashi R, Iinuma M, Hori O. ATF6alpha promotes astroglial activation and neuronal survival in a chronic mouse model of Parkinson’s disease. PloS One. 2012;7:e47950. doi: 10.1371/journal.pone.0047950. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hayashi K, Kadomatsu K, Muramatsu T. Requirement of chondroitin sulfate/dermatan sulfate recognition in midkine-dependent migration of macrophages. Glycoconj. J. 2001;18:401–406. doi: 10.1023/a:1014864131288. [DOI] [PubMed] [Google Scholar]
- Hinterkeuser S, Schröder W, Hager G, Seifert G, Blümcke I, Elger CE, Schramm J, Steinhäuser C. Astrocytes in the hippocampus of patients with temporal lobe epilepsy display changes in potassium conductances. Eur. J. Neurosci. 2000;12:2087–2096. doi: 10.1046/j.1460-9568.2000.00104.x. [DOI] [PubMed] [Google Scholar]
- Hirano M, Goldman JE. Gliogenesis in rat spinal cord: evidence for origin of astrocytes and oligodendrocytes from radial precursors. J. Neurosci. Res. 1988;21:155–167. doi: 10.1002/jnr.490210208. [DOI] [PubMed] [Google Scholar]
- Höke A, Canning DR, Malemud CJ, Silver J. Regional differences in reactive gliosis induced by substrate-bound beta-amyloid. Exp. Neurol. 1994;130:56–66. doi: 10.1006/exnr.1994.1185. [DOI] [PubMed] [Google Scholar]
- Hong X, Sin WC, Harris AL, Naus CC. Gap junctions modulate glioma invasion by direct transfer of microRNA. Oncotarget. 2015 doi: 10.18632/oncotarget.3904. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Horn KP, Busch SA, Hawthorne AL, van Rooijen N, Silver J. Another barrier to regeneration in the CNS: Activated macrophages induce extensive retraction of dystrophic axons through direct physical interactions. J. Neurosci. Off. J. Soc. Neurosci. 2008;28:9330–9341. doi: 10.1523/JNEUROSCI.2488-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hosoya T, Takizawa K, Nitta K, Hotta Y. glial cells missing: a binary switch between neuronal and glial determination in Drosophila. Cell. 1995;82:1025–1036. doi: 10.1016/0092-8674(95)90281-3. [DOI] [PubMed] [Google Scholar]
- Hsiao H-Y, Chen Y-C, Chen H-M, Tu P-H, Chern Y. A critical role of astrocyte-mediated nuclear factor-κB-dependent inflammation in Huntington’s disease. Hum. Mol. Genet. 2013;22:1826–1842. doi: 10.1093/hmg/ddt036. [DOI] [PubMed] [Google Scholar]
- Hsiao H-Y, Chiu F-L, Chen C-M, Wu Y-R, Chen H-M, Chen Y-C, Kuo H-C, Chern Y. Inhibition of soluble tumor necrosis factor is therapeutic in Huntington’s disease. Hum. Mol. Genet. 2014;23:4328–4344. doi: 10.1093/hmg/ddu151. [DOI] [PubMed] [Google Scholar]
- Hsu J-YC, Bourguignon LYW, Adams CM, Peyrollier K, Zhang H, Fandel T, Cun CL, Werb Z, Noble-Haeusslein LJ. Matrix metalloproteinase-9 facilitates glial scar formation in the injured spinal cord. J. Neurosci. Off. J. Soc. Neurosci. 2008;28:13467–13477. doi: 10.1523/JNEUROSCI.2287-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hu W, MacDonald ML, Elswick DE, Sweet RA. The glutamate hypothesis of schizophrenia: evidence from human brain tissue studies. Ann. N. Y. Acad. Sci. 2015;1338:38–57. doi: 10.1111/nyas.12547. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ikeda O, Murakami M, Ino H, Yamazaki M, Nemoto T, Koda M, Nakayama C, Moriya H. Acute up-regulation of brain-derived neurotrophic factor expression resulting from experimentally induced injury in the rat spinal cord. Acta Neuropathol. (Berl.) 2001;102:239–245. doi: 10.1007/s004010000357. [DOI] [PubMed] [Google Scholar]
- Ishibashi T, Dakin KA, Stevens B, Lee PR, Fields RD, Kozlov SV, Stewart CL. Astrocytes Promote Myelination in Response to Electrical Impulses. Neuron. 2006;49:823–832. doi: 10.1016/j.neuron.2006.02.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ito M, Natsume A, Takeuchi H, Shimato S, Ohno M, Wakabayashi T, Yoshida J. Type I interferon inhibits astrocytic gliosis and promotes functional recovery after spinal cord injury by deactivation of the MEK/ERK pathway. J. Neurotrauma. 2009;26:41–53. doi: 10.1089/neu.2008.0646. [DOI] [PubMed] [Google Scholar]
- Janzer RC, Raff MC. Astrocytes induce blood-brain barrier properties in endothelial cells. Nature. 1987;325:253–257. doi: 10.1038/325253a0. [DOI] [PubMed] [Google Scholar]
- Ji Z-G, Wang H. Optogenetic control of astrocytes: Is it possible to treat astrocyte-related epilepsy? Brain Res. Bull. 2015;110:20–25. doi: 10.1016/j.brainresbull.2014.10.013. [DOI] [PubMed] [Google Scholar]
- Kim J, Jones BW, Zock C, Chen Z, Wang H, Goodman CS, Anderson DJ. Isolation and characterization of mammalian homologs of the Drosophila gene glial cells missing. Proc. Natl. Acad. Sci. U. S. A. 1998;95:12364–12369. doi: 10.1073/pnas.95.21.12364. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Klein MA, Möller JC, Jones LL, Bluethmann H, Kreutzberg GW, Raivich G. Impaired neuroglial activation in interleukin-6 deficient mice. Glia. 1997;19:227–233. doi: 10.1002/(sici)1098-1136(199703)19:3<227::aid-glia5>3.0.co;2-w. [DOI] [PubMed] [Google Scholar]
- Kodaira Y, Nair SK, Wrenshall LE, Gilboa E, Platt JL. Phenotypic and functional maturation of dendritic cells mediated by heparan sulfate. J. Immunol. Baltim. Md 1950. 2000;165:1599–1604. doi: 10.4049/jimmunol.165.3.1599. [DOI] [PubMed] [Google Scholar]
- Kohta M, Kohmura E, Yamashita T. Inhibition of TGF-beta1 promotes functional recovery after spinal cord injury. Neurosci. Res. 2009;65:393–401. doi: 10.1016/j.neures.2009.08.017. [DOI] [PubMed] [Google Scholar]
- Koistinaho M, Lin S, Wu X, Esterman M, Koger D, Hanson J, Higgs R, Liu F, Malkani S, Bales KR, et al. Apolipoprotein E promotes astrocyte colocalization and degradation of deposited amyloid-beta peptides. Nat. Med. 2004;10:719–726. doi: 10.1038/nm1058. [DOI] [PubMed] [Google Scholar]
- Kuchibhotla KV, Lattarulo CR, Hyman BT, Bacskai BJ. Synchronous Hyperactivity and Intercellular Calcium Waves in Astrocytes in Alzheimer Mice. Science. 2009;323:1211–1215. doi: 10.1126/science.1169096. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kulijewicz-Nawrot M, Syková E, Chvátal A, Verkhratsky A, Rodríguez JJ. Astrocytes and glutamate homoeostasis in Alzheimer’s disease: a decrease in glutamine synthetase, but not in glutamate transporter-1, in the prefrontal cortex. ASN Neuro. 2013;5:273–282. doi: 10.1042/AN20130017. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lang B, Liu HL, Liu R, Feng GD, Jiao XY, Ju G. Astrocytes in injured adult rat spinal cord may acquire the potential of neural stem cells. Neuroscience. 2004;128:775–783. doi: 10.1016/j.neuroscience.2004.06.033. [DOI] [PubMed] [Google Scholar]
- Lang BT, Cregg JM, DePaul MA, Tran AP, Xu K, Dyck SM, Madalena KM, Brown BP, Weng Y-L, Li S, et al. Modulation of the proteoglycan receptor PTPσ promotes recovery after spinal cord injury. Nature. 2015;518:404–408. doi: 10.1038/nature13974. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Larsson A, Wilhelmsson U, Pekna M, Pekny M. Increased cell proliferation and neurogenesis in the hippocampal dentate gyrus of old GFAP(−/−)Vim(−/−) mice. Neurochem. Res. 2004;29:2069–2073. doi: 10.1007/s11064-004-6880-2. [DOI] [PubMed] [Google Scholar]
- Lau LW, Keough MB, Haylock-Jacobs S, Cua R, Döring A, Sloka S, Stirling DP, Rivest S, Yong VW. Chondroitin sulfate proteoglycans in demyelinated lesions impair remyelination. Ann. Neurol. 2012;72:419–432. doi: 10.1002/ana.23599. [DOI] [PubMed] [Google Scholar]
- Lau LW, Cua R, Keough MB, Haylock-Jacobs S, Yong VW. Pathophysiology of the brain extracellular matrix: a new target for remyelination. Nat. Rev. Neurosci. 2013;14:722–729. doi: 10.1038/nrn3550. [DOI] [PubMed] [Google Scholar]
- Leavitt BR, Hernit-Grant CS, Macklis JD. Mature astrocytes transform into transitional radial glia within adult mouse neocortex that supports directed migration of transplanted immature neurons. Exp. Neurol. 1999;157:43–57. doi: 10.1006/exnr.1999.6982. [DOI] [PubMed] [Google Scholar]
- Lee H-J, Suk J-E, Patrick C, Bae E-J, Cho J-H, Rho S, Hwang D, Masliah E, Lee S-J. Direct transfer of alpha-synuclein from neuron to astroglia causes inflammatory responses in synucleinopathies. J. Biol. Chem. 2010;285:9262–9272. doi: 10.1074/jbc.M109.081125. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lee MY, Kim CJ, Shin SL, Moon SH, Chun MH. Increased ciliary neurotrophic factor expression in reactive astrocytes following spinal cord injury in the rat. Neurosci. Lett. 1998;255:79–82. doi: 10.1016/s0304-3940(98)00710-1. [DOI] [PubMed] [Google Scholar]
- Lee S-G, Su Z-Z, Emdad L, Gupta P, Sarkar D, Borjabad A, Volsky DJ, Fisher PB. Mechanism of Ceftriaxone Induction of Excitatory Amino Acid Transporter-2 Expression and Glutamate Uptake in Primary Human Astrocytes. J. Biol. Chem. 2008;283:13116–13123. doi: 10.1074/jbc.M707697200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lepore AC, O’Donnell J, Bonner JF, Paul C, Miller ME, Rauck B, Kushner RA, Rothstein JD, Fischer I, Maragakis NJ. Spatial and temporal changes in promoter activity of the astrocyte glutamate transporter GLT1 following traumatic spinal cord injury. J. Neurosci. Res. 2011;89:1001–1017. doi: 10.1002/jnr.22624. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Levison SW, Jiang FJ, Stoltzfus OK, Ducceschi MH. IL-6-type cytokines enhance epidermal growth factor-stimulated astrocyte proliferation. Glia. 2000;32:328–337. doi: 10.1002/1098-1136(200012)32:3<328::aid-glia110>3.0.co;2-7. [DOI] [PubMed] [Google Scholar]
- Lewerenz J, Albrecht P, Tien M-LT, Henke N, Karumbayaram S, Kornblum HI, Wiedau-Pazos M, Schubert D, Maher P, Methner A. Induction of Nrf2 and xCT are involved in the action of the neuroprotective antibiotic ceftriaxone in vitro. J. Neurochem. 2009;111:332–343. doi: 10.1111/j.1471-4159.2009.06347.x. [DOI] [PubMed] [Google Scholar]
- Li Y, Chopp M. Temporal profile of nestin expression after focal cerebral ischemia in adult rat. Brain Res. 1999;838:1–10. doi: 10.1016/s0006-8993(99)01502-4. [DOI] [PubMed] [Google Scholar]
- Li L, Lundkvist A, Andersson D, Wilhelmsson U, Nagai N, Pardo AC, Nodin C, Ståhlberg A, Aprico K, Larsson K, et al. Protective role of reactive astrocytes in brain ischemia. J. Cereb. Blood Flow Metab. Off. J. Int. Soc. Cereb. Blood Flow Metab. 2008;28:468–481. doi: 10.1038/sj.jcbfm.9600546. [DOI] [PubMed] [Google Scholar]
- Li X, Zhao H, Tan X, Kostrzewa RM, Du G, Chen Y, Zhu J, Miao Z, Yu H, Kong J, et al. Inhibition of connexin43 improves functional recovery after ischemic brain injury in neonatal rats. Glia. 2015 doi: 10.1002/glia.22826. [DOI] [PubMed] [Google Scholar]
- Li Y, Liu Z, Xin H, Chopp M. The Role of Astrocytes in Mediating Exogenous Cell-Based Restorative Therapy for Stroke. Glia. 2014;62:1–16. doi: 10.1002/glia.22585. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Liedtke W, Edelmann W, Bieri PL, Chiu FC, Cowan NJ, Kucherlapati R, Raine CS. GFAP is necessary for the integrity of CNS white matter architecture and long-term maintenance of myelination. Neuron. 1996;17:607–615. doi: 10.1016/s0896-6273(00)80194-4. [DOI] [PubMed] [Google Scholar]
- Liévens JC, Woodman B, Mahal A, Spasic-Boscovic O, Samuel D, Kerkerian-Le Goff L, Bates GP. Impaired glutamate uptake in the R6 Huntington’s disease transgenic mice. Neurobiol. Dis. 2001;8:807–821. doi: 10.1006/nbdi.2001.0430. [DOI] [PubMed] [Google Scholar]
- Liu Z, Xin H, Chopp M. Reactive astrocytes promote axonal remodeling and neurological recovery after stroke. Neural Regen. Res. 2014;9:1874–1875. doi: 10.4103/1673-5374.145343. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lundgaard I, Osório MJ, Kress BT, Sanggaard S, Nedergaard M. White matter astrocytes in health and disease. Neuroscience. 2014;276:161–173. doi: 10.1016/j.neuroscience.2013.10.050. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Magnus T, Carmen J, Deleon J, Xue H, Pardo AC, Lepore AC, Mattson MP, Rao MS, Maragakis NJ. Adult glial precursor proliferation in mutant SOD1G93A mice. Glia. 2008;56:200–208. doi: 10.1002/glia.20604. [DOI] [PubMed] [Google Scholar]
- Malatesta P, Hartfuss E, Götz M. Isolation of radial glial cells by fluorescent-activated cell sorting reveals a neuronal lineage. Dev. Camb. Engl. 2000;127:5253–5263. doi: 10.1242/dev.127.24.5253. [DOI] [PubMed] [Google Scholar]
- Markoullis K, Sargiannidou I, Schiza N, Roncaroli F, Reynolds R, Kleopa KA. Oligodendrocyte gap junction loss and disconnection from reactive astrocytes in multiple sclerosis gray matter. J. Neuropathol. Exp. Neurol. 2014;73:865–879. doi: 10.1097/NEN.0000000000000106. [DOI] [PubMed] [Google Scholar]
- Masaki K. Early disruption of glial communication via connexin gap junction in multiple sclerosis, Baló’s disease and neuromyelitis optica. Neuropathol. Off. J. Jpn. Soc. Neuropathol. 2015 doi: 10.1111/neup.12211. [DOI] [PubMed] [Google Scholar]
- Massey JM, Hubscher CH, Wagoner MR, Decker JA, Amps J, Silver J, Onifer SM. Chondroitinase ABC digestion of the perineuronal net promotes functional collateral sprouting in the cuneate nucleus after cervical spinal cord injury. J. Neurosci. Off. J. Soc. Neurosci. 2006;26:4406–4414. doi: 10.1523/JNEUROSCI.5467-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mathur R, Ince PG, Minett T, Garwood CJ, Shaw PJ, Matthews FE, Brayne C, Simpson JE, Wharton SB MRC Cognitive Function and Ageing Neuropathology Study Group. A reduced astrocyte response to β-amyloid plaques in the ageing brain associates with cognitive impairment. PloS One. 2015;10:e0118463. doi: 10.1371/journal.pone.0118463. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Matos M, Augusto E, Oliveira CR, Agostinho P. Amyloid-beta peptide decreases glutamate uptake in cultured astrocytes: involvement of oxidative stress and mitogen-activated protein kinase cascades. Neuroscience. 2008;156:898–910. doi: 10.1016/j.neuroscience.2008.08.022. [DOI] [PubMed] [Google Scholar]
- McKeon RJ, Schreiber RC, Rudge JS, Silver J. Reduction of neurite outgrowth in a model of glial scarring following CNS injury is correlated with the expression of inhibitory molecules on reactive astrocytes. J. Neurosci. Off. J. Soc. Neurosci. 1991;11:3398–3411. doi: 10.1523/JNEUROSCI.11-11-03398.1991. [DOI] [PMC free article] [PubMed] [Google Scholar]
- McKeon RJ, Höke A, Silver J. Injury-induced proteoglycans inhibit the potential for laminin-mediated axon growth on astrocytic scars. Exp. Neurol. 1995;136:32–43. doi: 10.1006/exnr.1995.1081. [DOI] [PubMed] [Google Scholar]
- McKillop WM, Dragan M, Schedl A, Brown A. Conditional Sox9 ablation reduces chondroitin sulfate proteoglycan levels and improves motor function following spinal cord injury. Glia. 2013;61:164–177. doi: 10.1002/glia.22424. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Menet V, Prieto M, Privat A, Ribotta MGy. Axonal plasticity and functional recovery after spinal cord injury in mice deficient in both glial fibrillary acidic protein and vimentin genes. Proc. Natl. Acad. Sci. U. S. A. 2003;100:8999–9004. doi: 10.1073/pnas.1533187100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miller RH, Raff MC. Fibrous and protoplasmic astrocytes are biochemically and developmentally distinct. J. Neurosci. Off. J. Soc. Neurosci. 1984;4:585–592. doi: 10.1523/JNEUROSCI.04-02-00585.1984. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Miyake T, Hattori T, Fukuda M, Kitamura T, Fujita S. Quantitative studies on proliferative changes of reactive astrocytes in mouse cerebral cortex. Brain Res. 1988;451:133–138. doi: 10.1016/0006-8993(88)90757-3. [DOI] [PubMed] [Google Scholar]
- Moon LD, Asher RA, Rhodes KE, Fawcett JW. Regeneration of CNS axons back to their target following treatment of adult rat brain with chondroitinase ABC. Nat. Neurosci. 2001;4:465–466. doi: 10.1038/87415. [DOI] [PubMed] [Google Scholar]
- Moore CS, Milner R, Nishiyama A, Frausto RF, Serwanski DR, Pagarigan RR, Whitton JL, Miller RH, Crocker SJ. Astrocytic TIMP-1 Promotes Oligodendrocyte Differentiation and Enhances CNS Myelination. J. Neurosci. Off. J. Soc. Neurosci. 2011;31:6247–6254. doi: 10.1523/JNEUROSCI.5474-10.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Morest DK, Silver J. Precursors of neurons, neuroglia, and ependymal cells in the CNS: what are they? Where are they from? How do they get where they are going? Glia. 2003;43:6–18. doi: 10.1002/glia.10238. [DOI] [PubMed] [Google Scholar]
- Mullett SJ, Hinkle DA. DJ-1 knock-down in astrocytes impairs astrocyte-mediated neuroprotection against rotenone. Neurobiol. Dis. 2009;33:28–36. doi: 10.1016/j.nbd.2008.09.013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Mullett SJ, Di Maio R, Greenamyre JT, Hinkle DA. DJ-1 expression modulates astrocyte-mediated protection against neuronal oxidative stress. J. Mol. Neurosci. MN. 2013;49:507–511. doi: 10.1007/s12031-012-9904-4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Murugan M, Ling E-A, Kaur C. Dysregulated glutamate uptake by astrocytes causes oligodendroglia death in hypoxic perventricular white matter damage. Mol. Cell. Neurosci. 2013;56:342–354. doi: 10.1016/j.mcn.2013.07.005. [DOI] [PubMed] [Google Scholar]
- Myer DJ, Gurkoff GG, Lee SM, Hovda DA, Sofroniew MV. Essential protective roles of reactive astrocytes in traumatic brain injury. Brain J. Neurol. 2006;129:2761–2772. doi: 10.1093/brain/awl165. [DOI] [PubMed] [Google Scholar]
- Nagele RG, D’Andrea MR, Lee H, Venkataraman V, Wang H-Y. Astrocytes accumulate A beta 42 and give rise to astrocytic amyloid plaques in Alzheimer disease brains. Brain Res. 2003;971:197–209. doi: 10.1016/s0006-8993(03)02361-8. [DOI] [PubMed] [Google Scholar]
- Nagy JI, Li W, Hertzberg EL, Marotta CA. Elevated connexin43 immunoreactivity at sites of amyloid plaques in Alzheimer’s disease. Brain Res. 1996;717:173–178. doi: 10.1016/0006-8993(95)01526-4. [DOI] [PubMed] [Google Scholar]
- Nakagawa T, Schwartz JP. Gene expression profiles of reactive astrocytes in dopaminedepleted striatum. Brain Pathol. Zurich Switz. 2004;14:275–280. doi: 10.1111/j.1750-3639.2004.tb00064.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nash B, Thomson CE, Linington C, Arthur AT, McClure JD, McBride MW, Barnett SC. Functional duality of astrocytes in myelination. J. Neurosci. Off. J. Soc. Neurosci. 2011;31:13028–13038. doi: 10.1523/JNEUROSCI.1449-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nato G, Caramello A, Trova S, Avataneo V, Rolando C, Taylor V, Buffo A, Peretto P, Luzzati F. Striatal astrocytes produce neuroblasts in an excitotoxic model of Huntington’s disease. Dev. Camb. Engl. 2015;142:840–845. doi: 10.1242/dev.116657. [DOI] [PubMed] [Google Scholar]
- Neary JT, Kang Y, Shi Y, Tran MD, Wanner IB. P2 receptor signalling, proliferation of astrocytes, and expression of molecules involved in cell-cell interactions. Novartis Found. Symp. 2006;276:131–143. discussion 143–147, 233–237, 275–281. [PubMed] [Google Scholar]
- Neuhaus J, Risau W, Wolburg H. Induction of blood-brain barrier characteristics in bovine brain endothelial cells by rat astroglial cells in transfilter coculture. Ann. N. Y. Acad. Sci. 1991;633:578–580. doi: 10.1111/j.1749-6632.1991.tb15667.x. [DOI] [PubMed] [Google Scholar]
- Norenberg MD, Martinez-Hernandez A. Fine structural localization of glutamine synthetase in astrocytes of rat brain. Brain Res. 1979;161:303–310. doi: 10.1016/0006-8993(79)90071-4. [DOI] [PubMed] [Google Scholar]
- O’Brien MF, Lenke LG, Lou J, Bridwell KH, Joyce ME. Astrocyte response and transforming growth factor-beta localization in acute spinal cord injury. Spine. 1994;19:2321–2329. doi: 10.1097/00007632-199410150-00012. discussion 2330. [DOI] [PubMed] [Google Scholar]
- Okada S, Nakamura M, Mikami Y, Shimazaki T, Mihara M, Ohsugi Y, Iwamoto Y, Yoshizaki K, Kishimoto T, Toyama Y, et al. Blockade of interleukin-6 receptor suppresses reactive astrogliosis and ameliorates functional recovery in experimental spinal cord injury. J. Neurosci. Res. 2004;76:265–276. doi: 10.1002/jnr.20044. [DOI] [PubMed] [Google Scholar]
- Okada S, Nakamura M, Katoh H, Miyao T, Shimazaki T, Ishii K, Yamane J, Yoshimura A, Iwamoto Y, Toyama Y, et al. Conditional ablation of Stat3 or Socs3 discloses a dual role for reactive astrocytes after spinal cord injury. Nat. Med. 2006;12:829–834. doi: 10.1038/nm1425. [DOI] [PubMed] [Google Scholar]
- Omari KM, John GR, Sealfon SC, Raine CS. CXC chemokine receptors on human oligodendrocytes: implications for multiple sclerosis. Brain J. Neurol. 2005;128:1003–1015. doi: 10.1093/brain/awh479. [DOI] [PubMed] [Google Scholar]
- Orellana JA, Froger N, Ezan P, Jiang JX, Bennett MVL, Naus CC, Giaume C, Sáez JC. ATP and glutamate released via astroglial connexin 43 hemichannels mediate neuronal death through activation of pannexin 1 hemichannels. J. Neurochem. 2011;118:826–840. doi: 10.1111/j.1471-4159.2011.07210.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- De Pablo Y, Nilsson M, Pekna M, Pekny M. Intermediate filaments are important for astrocyte response to oxidative stress induced by oxygen-glucose deprivation and reperfusion. Histochem. Cell Biol. 2013;140:81–91. doi: 10.1007/s00418-013-1110-0. [DOI] [PubMed] [Google Scholar]
- Pekny M, Levéen P, Pekna M, Eliasson C, Berthold CH, Westermark B, Betsholtz C. Mice lacking glial fibrillary acidic protein display astrocytes devoid of intermediate filaments but develop and reproduce normally. EMBO J. 1995;14:1590–1598. doi: 10.1002/j.1460-2075.1995.tb07147.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pekny M, Johansson CB, Eliasson C, Stakeberg J, Wallén Å, Perlmann T, Lendahl U, Betsholtz C, Berthold C-H, Frisén J. Abnormal Reaction to Central Nervous System Injury in Mice Lacking Glial Fibrillary Acidic Protein and Vimentin. J. Cell Biol. 1999;145:503–514. doi: 10.1083/jcb.145.3.503. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Petr GT, Sun Y, Frederick NM, Zhou Y, Dhamne SC, Hameed MQ, Miranda C, Bedoya EA, Fischer KD, Armsen W, et al. Conditional deletion of the glutamate transporter GLT-1 reveals that astrocytic GLT-1 protects against fatal epilepsy while neuronal GLT-1 contributes significantly to glutamate uptake into synaptosomes. J. Neurosci. Off. J. Soc. Neurosci. 2015;35:5187–5201. doi: 10.1523/JNEUROSCI.4255-14.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pihlaja R, Koistinaho J, Malm T, Sikkilä H, Vainio S, Koistinaho M. Transplanted astrocytes internalize deposited beta-amyloid peptides in a transgenic mouse model of Alzheimer’s disease. Glia. 2008;56:154–163. doi: 10.1002/glia.20599. [DOI] [PubMed] [Google Scholar]
- Powell EM, Geller HM. Dissection of astrocyte-mediated cues in neuronal guidance and process extension. Glia. 1999;26:73–83. doi: 10.1002/(sici)1098-1136(199903)26:1<73::aid-glia8>3.0.co;2-s. [DOI] [PubMed] [Google Scholar]
- Powell EM, Meiners S, DiProspero NA, Geller HM. Mechanisms of astrocyte-directed neurite guidance. Cell Tissue Res. 1997a;290:385–393. doi: 10.1007/s004410050945. [DOI] [PubMed] [Google Scholar]
- Powell EM, Fawcett JW, Geller HM. Proteoglycans provide neurite guidance at an astrocyte boundary. Mol. Cell. Neurosci. 1997b;10:27–42. doi: 10.1006/mcne.1997.0629. [DOI] [PubMed] [Google Scholar]
- Raine CS. On the association between perinodal astrocytic processes and the node of Ranvier in the C.N.S. J. Neurocytol. 1984;13:21–27. doi: 10.1007/BF01148316. [DOI] [PubMed] [Google Scholar]
- Ramon y Cajal S. Hitologie du systeme nerveux de l’homme et des vertebres. Paris. 1909 [Google Scholar]
- Ransohoff RM, Hamilton TA, Tani M, Stoler MH, Shick HE, Major JA, Estes ML, Thomas DM, Tuohy VK. Astrocyte expression of mRNA encoding cytokines IP-10 and JE/MCP-1 in experimental autoimmune encephalomyelitis. FASEB J. Off. Publ. Fed. Am. Soc. Exp. Biol. 1993;7:592–600. doi: 10.1096/fasebj.7.6.8472896. [DOI] [PubMed] [Google Scholar]
- Reier PJ, Bregman BS, Wujek JR. Intraspinal transplantation of embryonic spinal cord tissue in neonatal and adult rats. J. Comp. Neurol. 1986;247:275–296. doi: 10.1002/cne.902470302. [DOI] [PubMed] [Google Scholar]
- Risher WC, Patel S, Kim IH, Uezu A, Bhagat S, Wilton DK, Pilaz L-J, Singh Alvarado J, Calhan OY, Silver DL, et al. Astrocytes refine cortical connectivity at dendritic spines. eLife. 2014;3 doi: 10.7554/eLife.04047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Robel S, Mori T, Zoubaa S, Schlegel J, Sirko S, Faissner A, Goebbels S, Dimou L, Götz M. Conditional deletion of beta1-integrin in astroglia causes partial reactive gliosis. Glia. 2009;57:1630–1647. doi: 10.1002/glia.20876. [DOI] [PubMed] [Google Scholar]
- Robel S, Buckingham SC, Boni JL, Campbell SL, Danbolt NC, Riedemann T, Sutor B, Sontheimer H. Reactive Astrogliosis Causes the Development of Spontaneous Seizures. J. Neurosci. 2015;35:3330–3345. doi: 10.1523/JNEUROSCI.1574-14.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rocha SM, Cristovão AC, Campos FL, Fonseca CP, Baltazar G. Astrocyte-derived GDNF is a potent inhibitor of microglial activation. Neurobiol. Dis. 2012;47:407–415. doi: 10.1016/j.nbd.2012.04.014. [DOI] [PubMed] [Google Scholar]
- Rolls A, Shechter R, London A, Segev Y, Jacob-Hirsch J, Amariglio N, Rechavi G, Schwartz M. Two faces of chondroitin sulfate proteoglycan in spinal cord repair: a role in microglia/macrophage activation. PLoS Med. 2008;5:e171. doi: 10.1371/journal.pmed.0050171. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rose CF, Verkhratsky A, Parpura V. Astrocyte glutamine synthetase: pivotal in health and disease. Biochem. Soc. Trans. 2013;41:1518–1524. doi: 10.1042/BST20130237. [DOI] [PubMed] [Google Scholar]
- Rothermundt M, Peters M, Prehn JHM, Arolt V. S100B in brain damage and neurodegeneration. Microsc. Res. Tech. 2003;60:614–632. doi: 10.1002/jemt.10303. [DOI] [PubMed] [Google Scholar]
- Rothstein JD, Martin LJ, Kuncl RW. Decreased glutamate transport by the brain and spinal cord in amyotrophic lateral sclerosis. N. Engl. J. Med. 1992;326:1464–1468. doi: 10.1056/NEJM199205283262204. [DOI] [PubMed] [Google Scholar]
- Rothstein JD, Dykes-Hoberg M, Pardo CA, Bristol LA, Jin L, Kuncl RW, Kanai Y, Hediger MA, Wang Y, Schielke JP, et al. Knockout of glutamate transporters reveals a major role for astroglial transport in excitotoxicity and clearance of glutamate. Neuron. 1996;16:675–686. doi: 10.1016/s0896-6273(00)80086-0. [DOI] [PubMed] [Google Scholar]
- Rothstein JD, Patel S, Regan MR, Haenggeli C, Huang YH, Bergles DE, Jin L, Dykes Hoberg M, Vidensky S, Chung DS, et al. Beta-lactam antibiotics offer neuroprotection by increasing glutamate transporter expression. Nature. 2005;433:73–77. doi: 10.1038/nature03180. [DOI] [PubMed] [Google Scholar]
- Rudge JS, Silver J. Inhibition of neurite outgrowth on astroglial scars in vitro. J. Neurosci. 1990;10:3594–3603. doi: 10.1523/JNEUROSCI.10-11-03594.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Samoilova M, Li J, Pelletier MR, Wentlandt K, Adamchik Y, Naus CC, Carlen PL. Epileptiform activity in hippocampal slice cultures exposed chronically to bicuculline: increased gap junctional function and expression. J. Neurochem. 2003;86:687–699. doi: 10.1046/j.1471-4159.2003.01893.x. [DOI] [PubMed] [Google Scholar]
- Sasaki S, Warita H, Abe K, Iwata M. Inducible nitric oxide synthase (iNOS) and nitrotyrosine immunoreactivity in the spinal cords of transgenic mice with a G93A mutant SOD1 gene. J. Neuropathol. Exp. Neurol. 2001;60:839–846. doi: 10.1093/jnen/60.9.839. [DOI] [PubMed] [Google Scholar]
- Schachtrup C, Ryu JK, Helmrick MJ, Vagena E, Galanakis DK, Degen JL, Margolis RU, Akassoglou K. Fibrinogen triggers astrocyte scar formation by promoting the availability of active TGF-beta after vascular damage. J. Neurosci. Off. J. Soc. Neurosci. 2010;30:5843–5854. doi: 10.1523/JNEUROSCI.0137-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schmidtmayer J, Jacobsen C, Miksch G, Sievers J. Blood monocytes and spleen macrophages differentiate into microglia-like cells on monolayers of astrocytes: membrane currents. Glia. 1994;12:259–267. doi: 10.1002/glia.440120403. [DOI] [PubMed] [Google Scholar]
- Segawa S, Nishiura T, Furuta T, Ohsato Y, Tani M, Nishida K, Nagasawa K. Zinc is released by cultured astrocytes as a gliotransmitter under hypoosmotic stress-loaded conditions and regulates microglial activity. Life Sci. 2014;94:137–144. doi: 10.1016/j.lfs.2013.11.007. [DOI] [PubMed] [Google Scholar]
- Shen Y, Tenney AP, Busch SA, Horn KP, Cuascut FX, Liu K, He Z, Silver J, Flanagan JG. PTPsigma is a receptor for chondroitin sulfate proteoglycan, an inhibitor of neural regeneration. Science. 2009;326:592–596. doi: 10.1126/science.1178310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shibata T, Yamada K, Watanabe M, Ikenaka K, Wada K, Tanaka K, Inoue Y. Glutamate Transporter GLAST Is Expressed in the Radial Glia–Astrocyte Lineage of Developing Mouse Spinal Cord. J. Neurosci. 1997;17:9212–9219. doi: 10.1523/JNEUROSCI.17-23-09212.1997. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shibuya S, Miyamoto O, Auer RN, Itano T, Mori S, Norimatsu H. Embryonic intermediate filament, nestin, expression following traumatic spinal cord injury in adult rats. Neuroscience. 2002;114:905–916. doi: 10.1016/s0306-4522(02)00323-8. [DOI] [PubMed] [Google Scholar]
- Shigetomi E, Bowser DN, Sofroniew MV, Khakh BS. Two forms of astrocyte calcium excitability have distinct effects on NMDA receptor-mediated slow inward currents in pyramidal neurons. J. Neurosci. Off. J. Soc. Neurosci. 2008;28:6659–6663. doi: 10.1523/JNEUROSCI.1717-08.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Shin J-Y, Fang Z-H, Yu Z-X, Wang C-E, Li S-H, Li X-J. Expression of mutant huntingtin in glial cells contributes to neuronal excitotoxicity. J. Cell Biol. 2005;171:1001–1012. doi: 10.1083/jcb.200508072. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sievers J, Schmidtmayer J, Parwaresch R. Blood monocytes and spleen macrophages differentiate into microglia-like cells when cultured on astrocytes. Ann. Anat. Anat. Anz. Off. Organ Anat. Ges. 1994a;176:45–51. doi: 10.1016/s0940-9602(11)80414-0. [DOI] [PubMed] [Google Scholar]
- Sievers J, Parwaresch R, Wottge HU. Blood monocytes and spleen macrophages differentiate into microglia-like cells on monolayers of astrocytes: morphology. Glia. 1994b;12:245–258. doi: 10.1002/glia.440120402. [DOI] [PubMed] [Google Scholar]
- Silver J, Miller JH. Regeneration beyond the glial scar. Nat. Rev. Neurosci. 2004;5:146–156. doi: 10.1038/nrn1326. [DOI] [PubMed] [Google Scholar]
- Silver DJ, Siebzehnrubl FA, Schildts MJ, Yachnis AT, Smith GM, Smith AA, Scheffler B, Reynolds BA, Silver J, Steindler DA. Chondroitin sulfate proteoglycans potently inhibit invasion and serve as a central organizer of the brain tumor microenvironment. J. Neurosci. Off. J. Soc. Neurosci. 2013;33:15603–15617. doi: 10.1523/JNEUROSCI.3004-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Silver J, Lorenz SE, Wahlsten D, Coughlin J. Axonal guidance during development of the great cerebral commissures: descriptive and experimental studies, in vivo, on the role of preformed glial pathways. J. Comp. Neurol. 1982;210:10–29. doi: 10.1002/cne.902100103. [DOI] [PubMed] [Google Scholar]
- Silver J, Edwards MA, Levitt P. Immunocytochemical demonstration of early appearing astroglial structures that form boundaries and pathways along axon tracts in the fetal brain. J. Comp. Neurol. 1993;328:415–436. doi: 10.1002/cne.903280308. [DOI] [PubMed] [Google Scholar]
- Sirko S, Behrendt G, Johansson PA, Tripathi P, Costa M, Bek S, Heinrich C, Tiedt S, Colak D, Dichgans M, et al. Reactive glia in the injured brain acquire stem cell properties in response to sonic hedgehog. Cell Stem Cell. 2013;12:426–439. doi: 10.1016/j.stem.2013.01.019. [corrected] [DOI] [PubMed] [Google Scholar]
- Smith GM, Miller RH. Immature type-1 astrocytes suppress glial scar formation, are motile and interact with blood vessels. Brain Res. 1991;543:111–122. doi: 10.1016/0006-8993(91)91054-5. [DOI] [PubMed] [Google Scholar]
- Smith GM, Silver J. Transplantation of immature and mature astrocytes and their effect on scar formation in the lesioned central nervous system. Prog. Brain Res. 1988;78:353–361. doi: 10.1016/s0079-6123(08)60304-0. [DOI] [PubMed] [Google Scholar]
- Snow DM, Steindler DA, Silver J. Molecular and cellular characterization of the glial roof plate of the spinal cord and optic tectum: a possible role for a proteoglycan in the development of an axon barrier. Dev. Biol. 1990;138:359–376. doi: 10.1016/0012-1606(90)90203-u. [DOI] [PubMed] [Google Scholar]
- Soderblom C, Luo X, Blumenthal E, Bray E, Lyapichev K, Ramos J, Krishnan V, Lai-Hsu C, Park KK, Tsoulfas P, et al. Perivascular fibroblasts form the fibrotic scar after contusive spinal cord injury. J. Neurosci. Off. J. Soc. Neurosci. 2013;33:13882–13887. doi: 10.1523/JNEUROSCI.2524-13.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sofroniew MV. Molecular dissection of reactive astrogliosis and glial scar formation. Trends Neurosci. 2009;32:638–647. doi: 10.1016/j.tins.2009.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Sofroniew MV, Vinters HV. Astrocytes: biology and pathology. Acta Neuropathol. (Berl.) 2010;119:7–35. doi: 10.1007/s00401-009-0619-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Song YJC, Halliday GM, Holton JL, Lashley T, O’Sullivan SS, McCann H, Lees AJ, Ozawa T, Williams DR, Lockhart PJ, et al. Degeneration in different parkinsonian syndromes relates to astrocyte type and astrocyte protein expression. J. Neuropathol. Exp. Neurol. 2009;68:1073–1083. doi: 10.1097/NEN.0b013e3181b66f1b. [DOI] [PubMed] [Google Scholar]
- Sorensen A, Moffat K, Thomson C, Barnett SC. Astrocytes, but not olfactory ensheathing cells or Schwann cells, promote myelination of CNS axons in vitro. Glia. 2008;56:750–763. doi: 10.1002/glia.20650. [DOI] [PubMed] [Google Scholar]
- Stevens B, Allen NJ, Vazquez LE, Howell GR, Christopherson KS, Nouri N, Micheva KD, Mehalow AK, Huberman AD, Stafford B, et al. The classical complement cascade mediates CNS synapse elimination. Cell. 2007;131:1164–1178. doi: 10.1016/j.cell.2007.10.036. [DOI] [PubMed] [Google Scholar]
- Takahashi DK, Vargas JR, Wilcox KS. Increased coupling and altered glutamate transport currents in astrocytes following kainic-acid-induced status epilepticus. Neurobiol. Dis. 2010;40:573–585. doi: 10.1016/j.nbd.2010.07.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Talbott JF, Loy DN, Liu Y, Qiu MS, Bunge MB, Rao MS, Whittemore SR. Endogenous Nkx2.2+/Olig2+ oligodendrocyte precursor cells fail to remyelinate the demyelinated adult rat spinal cord in the absence of astrocytes. Exp. Neurol. 2005;192:11–24. doi: 10.1016/j.expneurol.2004.05.038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tian R, Wu X, Hagemann TL, Sosunov AA, Messing A, McKhann GM, Goldman JE. Alexander disease mutant glial fibrillary acidic protein compromises glutamate transport in astrocytes. J. Neuropathol. Exp. Neurol. 2010;69:335–345. doi: 10.1097/NEN.0b013e3181d3cb52. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tom VJ, Steinmetz MP, Miller JH, Doller CM, Silver J. Studies on the development and behavior of the dystrophic growth cone, the hallmark of regeneration failure, in an in vitro model of the glial scar and after spinal cord injury. J. Neurosci. Off. J. Soc. Neurosci. 2004a;24:6531–6539. doi: 10.1523/JNEUROSCI.0994-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tom VJ, Doller CM, Malouf AT, Silver J. Astrocyte-associated fibronectin is critical for axonal regeneration in adult white matter. J. Neurosci. Off. J. Soc. Neurosci. 2004b;24:9282–9290. doi: 10.1523/JNEUROSCI.2120-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tom VJ, Kadakia R, Santi L, Houlé JD. Administration of chondroitinase ABC rostral or caudal to a spinal cord injury site promotes anatomical but not functional plasticity. J. Neurotrauma. 2009;26:2323–2333. doi: 10.1089/neu.2009.1047. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Toro CT, Hallak JEC, Dunham JS, Deakin JFW. Glial fibrillary acidic protein and glutamine synthetase in subregions of prefrontal cortex in schizophrenia and mood disorder. Neurosci. Lett. 2006;404:276–281. doi: 10.1016/j.neulet.2006.05.067. [DOI] [PubMed] [Google Scholar]
- Tran MD, Neary JT. Purinergic signaling induces thrombospondin-1 expression in astrocytes. Proc. Natl. Acad. Sci. U. S. A. 2006;103:9321–9326. doi: 10.1073/pnas.0603146103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tyzack GE, Sitnikov S, Barson D, Adams-Carr KL, Lau NK, Kwok JC, Zhao C, Franklin RJM, Karadottir RT, Fawcett JW, et al. Astrocyte response to motor neuron injury promotes structural synaptic plasticity via STAT3-regulated TSP-1 expression. Nat. Commun. 2014;5:4294. doi: 10.1038/ncomms5294. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ullian EM, Sapperstein SK, Christopherson KS, Barres BA. Control of synapse number by glia. Science. 2001;291:657–661. doi: 10.1126/science.291.5504.657. [DOI] [PubMed] [Google Scholar]
- Ullian EM, Harris BT, Wu A, Chan JR, Barres BA. Schwann cells and astrocytes induce synapse formation by spinal motor neurons in culture. Mol. Cell. Neurosci. 2004;25:241–251. doi: 10.1016/j.mcn.2003.10.011. [DOI] [PubMed] [Google Scholar]
- Valenza M, Marullo M, Di Paolo E, Cesana E, Zuccato C, Biella G, Cattaneo E. Disruption of astrocyte-neuron cholesterol cross talk affects neuronal function in Huntington’s disease. Cell Death Differ. 2015;22:690–702. doi: 10.1038/cdd.2014.162. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vargas MR, Johnson JA. ASTROGLIOSIS IN AMYOTROPHIC LATERAL SCLEROSIS: ROLE AND THERAPEUTIC POTENTIAL OF ASTROCYTES. Neurother. J. Am. Soc. Exp. Neurother. 2010;7:471–481. doi: 10.1016/j.nurt.2010.05.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vartak-Sharma N, Ghorpade A. Astrocyte elevated gene-1 regulates astrocyte responses to neural injury: implications for reactive astrogliosis and neurodegeneration. J. Neuroinflammation. 2012;9:195. doi: 10.1186/1742-2094-9-195. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Vera-Portocarrero LP, Mills CD, Ye Z, Fullwood SD, McAdoo DJ, Hulsebosch CE, Westlund KN. Rapid changes in expression of glutamate transporters after spinal cord injury. Brain Res. 2002;927:104–110. doi: 10.1016/s0006-8993(01)03329-7. [DOI] [PubMed] [Google Scholar]
- Verderio C, Matteoli M. ATP mediates calcium signaling between astrocytes and microglial cells: modulation by IFN-gamma. J. Immunol. Baltim. Md. 2001;166:6383–6391. doi: 10.4049/jimmunol.166.10.6383. 1950. [DOI] [PubMed] [Google Scholar]
- Vijayan VK, Lee YL, Eng LF. Increase in glial fibrillary acidic protein following neural trauma. Mol. Chem. Neuropathol. Spons. Int. Soc. Neurochem. World Fed. Neurol. Res. Groups Neurochem. Cerebrospinal Fluid. 1990;13:107–118. doi: 10.1007/BF03159912. [DOI] [PubMed] [Google Scholar]
- Voigt T. Development of glial cells in the cerebral wall of ferrets: direct tracing of their transformation from radial glia into astrocytes. J. Comp. Neurol. 1989;289:74–88. doi: 10.1002/cne.902890106. [DOI] [PubMed] [Google Scholar]
- Wang L, Lin F, Wang J, Wu J, Han R, Zhu L, Difiglia M, Qin Z. Expression of mutant N-terminal huntingtin fragment (htt552-100Q) in astrocytes suppresses the secretion of BDNF. Brain Res. 2012;1449:69–82. doi: 10.1016/j.brainres.2012.01.077. [DOI] [PubMed] [Google Scholar]
- Wang Y, Moges H, Bharucha Y, Symes A. Smad3 null mice display more rapid wound closure and reduced scar formation after a stab wound to the cerebral cortex. Exp. Neurol. 2007;203:168–184. doi: 10.1016/j.expneurol.2006.08.006. [DOI] [PubMed] [Google Scholar]
- Wang Y, Cheng X, He Q, Zheng Y, Kim DH, Whittemore SR, Cao QL. Astrocytes from the Contused Spinal Cord Inhibit Oligodendrocyte Differentiation of Adult Oligodendrocyte Precursor Cells by Increasing the Expression of Bone Morphogenetic Proteins. J. Neurosci. 2011;31:6053–6058. doi: 10.1523/JNEUROSCI.5524-09.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wanner IB, Anderson MA, Song B, Levine J, Fernandez A, Gray-Thompson Z, Ao Y, Sofroniew MV. Glial scar borders are formed by newly proliferated, elongated astrocytes that interact to corral inflammatory and fibrotic cells via STAT3-dependent mechanisms after spinal cord injury. J. Neurosci. Off. J. Soc. Neurosci. 2013;33:12870–12886. doi: 10.1523/JNEUROSCI.2121-13.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- White RE, Jakeman LB. Don’t fence me in: harnessing the beneficial roles of astrocytes for spinal cord repair. Restor. Neurol. Neurosci. 2008;26:197–214. [PMC free article] [PubMed] [Google Scholar]
- White RE, Rao M, Gensel JC, McTigue DM, Kaspar BK, Jakeman LB. Transforming growth factor α transforms astrocytes to a growth-supportive phenotype after spinal cord injury. J. Neurosci. Off. J. Soc. Neurosci. 2011;31:15173–15187. doi: 10.1523/JNEUROSCI.3441-11.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wilhelmsson U, Li L, Pekna M, Berthold C-H, Blom S, Eliasson C, Renner O, Bushong E, Ellisman M, Morgan TE, et al. Absence of Glial Fibrillary Acidic Protein and Vimentin Prevents Hypertrophy of Astrocytic Processes and Improves Post-Traumatic Regeneration. J. Neurosci. 2004;24:5016–5021. doi: 10.1523/JNEUROSCI.0820-04.2004. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wójtowicz AM, Dvorzhak A, Semtner M, Grantyn R. Reduced tonic inhibition in striatal output neurons from Huntington mice due to loss of astrocytic GABA release through GAT-3. Front. Neural Circuits. 2013;7:188. doi: 10.3389/fncir.2013.00188. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wu DY, Schneider GE, Silver J, Poston M, Jhaveri S. A role for tectal midline glia in the unilateral containment of retinocollicular axons. J. Neurosci. Off. J. Soc. Neurosci. 1998;18:8344–8355. doi: 10.1523/JNEUROSCI.18-20-08344.1998. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wyss-Coray T, Loike JD, Brionne TC, Lu E, Anankov R, Yan F, Silverstein SC, Husemann J. Adult mouse astrocytes degrade amyloid-beta in vitro and in situ. Nat. Med. 2003;9:453–457. doi: 10.1038/nm838. [DOI] [PubMed] [Google Scholar]
- Xu K, Malouf AT, Messing A, Silver J. Glial fibrillary acidic protein is necessary for mature astrocytes to react to beta-amyloid. Glia. 1999;25:390–403. doi: 10.1002/(sici)1098-1136(19990215)25:4<390::aid-glia8>3.0.co;2-7. [DOI] [PubMed] [Google Scholar]
- Xu L, Zeng L-H, Wong M. Impaired astrocytic gap junction coupling and potassium buffering in a mouse model of tuberous sclerosis complex. Neurobiol. Dis. 2009;34:291–299. doi: 10.1016/j.nbd.2009.01.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang C, Wang H, Qiao T, Yang B, Aliaga L, Qiu L, Tan W, Salameh J, McKenna-Yasek DM, Smith T, et al. Partial loss of TDP-43 function causes phenotypes of amyotrophic lateral sclerosis. Proc. Natl. Acad. Sci. U. S. A. 2014;111:E1121–E1129. doi: 10.1073/pnas.1322641111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang HY, Lieska N, Glick R, Shao D, Pappas GD. Expression of 300-kilodalton intermediate filament-associated protein distinguishes human glioma cells from normal astrocytes. Proc. Natl. Acad. Sci. U. S. A. 1993a;90:8534–8537. doi: 10.1073/pnas.90.18.8534. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yang HY, Lieska N, Shao D, Kriho V, Pappas GD. Immunotyping of radial glia and their glial derivatives during development of the rat spinal cord. J. Neurocytol. 1993b;22:558–571. doi: 10.1007/BF01189043. [DOI] [PubMed] [Google Scholar]
- Yao P-S, Kang D-Z, Lin R-Y, Ye B, Wang W, Ye Z-C. Glutamate/glutamine metabolism coupling between astrocytes and glioma cells: Neuroprotection and inhibition of glioma growth. Biochem. Biophys. Res. Commun. 2014;450:295–299. doi: 10.1016/j.bbrc.2014.05.120. [DOI] [PubMed] [Google Scholar]
- Yatomi Y, Tanaka R, Shimura H, Miyamoto N, Yamashiro K, Takanashi M, Urabe T, Hattori N. Chronic brain ischemia induces the expression of glial glutamate transporter EAAT2 in subcortical white matter. Neuroscience. 2013;244:113–121. doi: 10.1016/j.neuroscience.2013.04.018. [DOI] [PubMed] [Google Scholar]
- Yeh T-H, Lee DY, Gianino SM, Gutmann DH. Microarray analyses reveal regional astrocyte heterogeneity with implications for neurofibromatosis type 1 (NF1)-regulated glial proliferation. Glia. 2009;57:1239–1249. doi: 10.1002/glia.20845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao R-R, Fawcett JW. Combination treatment with chondroitinase ABC in spinal cord injury--breaking the barrier. Neurosci. Bull. 2013;29:477–483. doi: 10.1007/s12264-013-1359-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zhao R-R, Andrews MR, Wang D, Warren P, Gullo M, Schnell L, Schwab ME, Fawcett JW. Combination treatment with anti-Nogo-A and chondroitinase ABC is more effective than single treatments at enhancing functional recovery after spinal cord injury. Eur. J. Neurosci. 2013;38:2946–2961. doi: 10.1111/ejn.12276. [DOI] [PubMed] [Google Scholar]
- Zhu Y, Soderblom C, Trojanowsky M, Lee D-H, Lee JK. Fibronectin Matrix Assembly after Spinal Cord Injury. J. Neurotrauma. 2015 doi: 10.1089/neu.2014.3703. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zonta M, Angulo MC, Gobbo S, Rosengarten B, Hossmann K-A, Pozzan T, Carmignoto G. Neuron-to-astrocyte signaling is central to the dynamic control of brain microcirculation. Nat. Neurosci. 2003;6:43–50. doi: 10.1038/nn980. [DOI] [PubMed] [Google Scholar]
- Zukor K, Belin S, Wang C, Keelan N, Wang X, He Z. Short Hairpin RNA against PTEN Enhances Regenerative Growth of Corticospinal Tract Axons after Spinal Cord Injury. J. Neurosci. 2013;33:15350–15361. doi: 10.1523/JNEUROSCI.2510-13.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Zukor KA, Kent DT, Odelberg SJ. Meningeal cells and glia establish a permissive environment for axon regeneration after spinal cord injury in newts. Neural Develop. 2011;6:1. doi: 10.1186/1749-8104-6-1. [DOI] [PMC free article] [PubMed] [Google Scholar]