Abstract
Focal ischemic stroke (FIS) is a leading cause of human debilitation and death. Following the onset of a FIS, the brain experiences a series of spatiotemporal changes which are exemplified in different pathological processes. One prominent feature of FIS is development of reactive astrogliosis and glial scar formation in the peri-infarct region (PIR). During the subacute phase, astrocytes in PIR are activated, referred to as reactive astrocytes (RAs), exhibit changes in morphology (hypotrophy), show increased proliferation capacity, and altered gene expression, a phenomenon known as reactive astrogliosis. Subsequently, the morphology of RAs remains stable, and proliferation starts to decline together with the formation of glial scars. Reactive astrogliosis and glial scar formation eventually cause substantial tissue remodeling and changes in permanent structure around the PIR. Glial cell line-derived neurotrophic factor (GDNF) was originally isolated from a rat glioma cell-line and regarded as a potent survival neurotrophic factor. Under normal conditions, GDNF is expressed in neurons but is upregulated in RAs after FIS. This review briefly describes properties of GDNF, its receptor-mediated signaling pathways, as well as recent studies regarding the role of RAs-derived GDNF in neuronal protection and brain recovery. These results provide evidence suggesting an important role of GDNF in intrinsic brain repair and recovery after FIS, and thus targeting GDNF in RAs may be effective for stroke therapy.
Keywords: GDNF, Focal ischemic stroke, Reactive astrocytes, Neuronal protection, Brain repair
1. Focal Ischemic Stroke and Reactive Astrocytes
Cerebral focal ischemic stroke (FIS) is a leading neurological disorder associated with neuronal death and brain damage, disabilities leading to a major impact on public health. FIS is induced by development of a thrombus in the arteries supplying blood to the brain, which is a complex process leading to changes in microenvironment in the Central Nervous System (CNS). Patients with FIS may suddenly experience paralysis, impairment of speech, and loss of vision. Most survivors develop disabilities which are often permanent. Despite of intense basic and translational research being taken towards FIS, treatment options are still limited. Currently, tissue plasminogen activator (tPA) is the only FDA approved drug which has limited application with a window of 3-5 hours after the onset of FIS. Consequently, improvement of stroke outcomes and recovery of survivors frequently depend on self-brain repair and rehabilitation. Recent advances on mechanisms of post-stroke recovery include improving growth factor release, intrinsic antioxidant capacity, and enhanced neurogenesis and angiogenesis (1-4).
Astrocytes are a major and the most diverse glial cell type in CNS. They are highly heterogeneous in morphology and function and demonstrate remarkable adaptive plasticity that defines the functional maintenance of the CNS during development and aging, and under pathological conditions (5). With their intimate contact with neurons, these cells fulfill many functions including maintenance of ion homeostasis, storage and distribution of energy substrates, and controlling the development of neural cells and synaptogenesis (6-10). Brain injury and other chronical neurological disorders cause the activation of astrocytes, a phenomenon generally known as reactive astrogliosis. The activated astrocytes is called reactive astrocytes (RAs). Reactive astrogliosis is observed in most if not all of the brain disorders. The commons molecular and morphological features of RAs include hypertrophy of astrocyte processes and upregulation of glial fibrillary acidic protein (GFAP), the key constituent of astrocyte intermediate filaments. However, the properties of RAs are disease context dependent and also depend on disease stages and brain regions (e.g., brain injury vs neurodegenerative diseases, mild vs severe injury, and acute vs chronic stage). For comprehensive review of reactive astrogliosis and RAs, readers can consult many nice review articles (5, 11, 12).
The pathological process of FIS has unique features compared with neurodegenerative diseases due to its focal decrease of blood flow in the brain. Following the onset of a FIS, the brain experiences a series of spatiotemporal changes associated with different pathological processes. In the ischemic core, neurons die rapidly after the onset of FIS due mainly to the impairment of energy production and breakdown of ion homeostasis. In the peri-infarct region (PIR) (also commonly called penumbra), brain tissue is hypo-perfused with collateral blood flow as an attempt to preserve partial energy metabolism (Figure 1). Accordingly, the primary goal for stroke therapy is to salvage the PIR tissue after FIS. During the subacute phase (days after a FIS), surviving neurons in the PIR exhibit active structural and functional changes in synaptic wiring and mapping, and these changes may contribute to long-term neural regeneration and functional recovery (13-17). Another hallmark of FIS is glial scar formation in the PIR. During the subacute phase, RAs in the PIR exhibit spatiotemporal changes in morphology (hypotrophy) and proliferation capacity, resulting from altered metabolic functions and gene expression (18-20). Under this condition, RAs become hypertrophic with large processes that can be revealed by large upregulation of GFAP (Figure 1). Upon prolonged time (in chronic phase) following FIS, proliferation of RAs starts to decline and glial scars are stabilized (18-23). Reactive astrogliosis and glial scar formation eventually cause substantial tissue remodeling and structural changes in the PIR. Under normal conditions, astrocytes are quiescent in term of proliferation, but after FIS, RAs have high capacity to proliferate and reaching a peak around 3-4 days (18, 19). The highly active RAs on 3-4 days post-stroke are needed to meet the high energetic and biosynthetic demands for proliferation. RAs have been demonstrated to offer protective effects after FIS and injury (24-27). Proliferating RAs may impact tissue preservation, repair/remodeling, and functional outcome. Specific deletion of proliferating RAs after brain injury has been shown to prevent repair of the blood–brain barrier, increase immune cell infiltration as well as neuronal degeneration(28). The molecular phenotype exhibiting rapid and transient induction of gene expression in RAs after ischemia supports the notion that RAs may offer beneficial or protective effects to neurons (29).
Figure 1. Brain damage and reactive astrogliosis after FIS.
A) Nissl staining showing brain damage in the mouse cortex after PT. B) Double staining of Brdu and GFAP in the cortices of a control (left) and an ischemic mouse 4 days after PT (right). IC-Ischemic core; PIR-Peri-infarct region. Notice that GFAP is highly upregulated in RAs, which is a hallmark of FIS. Adapted from reference Li H. eat al. (16).
Since neuron-centric strategies have not achieved major breakthroughs in stroke therapy, astrocytes have been indicated as a promising target for neural repair in stroke and other brain diseases through either cell or non-cell autonomous effects on neuronal survival and regeneration (9, 20, 21, 24, 30-38). The potential beneficial effects of RAs on brain recovery have been discussed in many reviews (20, 21, 24, 39, 40); however, molecular mechanisms underlying their protective effects remain elusive. Recent studies have suggested that a potential mechanism for beneficial effects is the ability for RAs to release different soluble factors including BDNF, NDF, GDNF, CNTF and CNDF that are important for brain to recover (24, 41-43). This article is intended to focus on GDNF and its effects on brain protection and recovery after FIS.
2. GDNF Ligand Family, Receptor Systems, and Signaling
2.1. GDNF function in the CNS
GDNF is a member of transforming growth factor (TGF)-β super family; it is also a potent neurotrophic factor that promotes neuronal survival, neural progenitor differentiation, and synapse formation (44-49). GDNF was originally isolated from the supernatant of a rat glioma cell-line (44). Initially, it was thought to act as a trophic factor for embryonic midbrain dopaminergic neurons, but was later found to have pronounced effects on other neuronal subpopulations (50). GDNF family proteins were first purified and proved to promote the survival of midbrain dopaminergic neurons. Later, there is more evidence showing that not only dopaminergic neurons, but many other cell types can also be targeted by GDNF. Moreover, the role of GDNF can be changed in different stages. For example, GDNF and its receptors exert a non-survival function during neuronal development (51) and favors different types of neuronal precursors by promoting axonal growth. After differentiation, GDNF functions as a survival factor (52), through regulating the expression of key enzymes to promote survival of neurons and mediate dendritic or electrical maturation, particularly those related to dopaminergic phenotype. GDNF can also promote neurogenesis, neuronal maturation and mediate neuromuscular junction function in motor neuron system (53). Moreover, the ability of GDNF to modulate proliferation of neuroblastoma has been shown to closely associate with conditions such as depression or chronic pain (52).
2.2. The GDNF protein
GDNF and GDNF-family ligands are comprised of neurturin, artemin and persephin, and are thought to belong to the TGF-β superfamily (47). In their structure, all have cysteine knot motif and three disulphide bonds, and they usually contain seven cysteine residues with same relative spacing. This kind of proteins normally function as homodimers (Figure 2).
Figure 2. GDNF family ligands and their receptors.
The four members of GDNF family ligands include GDNF, neurturin, artemin, and persephin. They are homodimers and bind with high affinity to one of the four members of the GDNF receptor α family (GFRα1-4). These receptor-ligand complexes can interact with and activate the canonical GDNF receptor ‘rearranged during transfection’ (RET), which is a receptor tyrosine kinase (TK). GDNF can also activate alternative GDNF receptors, such as the neuronal cell adhesion molecule (NCAM). The intracellular domain of RET can be phosphorylated and ubiquitinylated. Adapted from Edgar R. Kramer and Birgit Liss (155).
Because all GDNF family proteins are glycosylated, intracellular modification is needed for maturation of these proteins. Similar to other secreted proteins, GDNF is produced in the form of a precursor, which is then cleaved and activated upon secretion. The mature form of GDNF has 134 amino acids, with a molecular weight around 30-42 KD. The N-terminal consists of a heparin-binding domain which can bind with heparan-sulphate extracellular proteoglygan (54). During the past decades, synthesis and modification of GDNF have been investigated extensively. However, knowledge on specific enzyme(s) that cleaves and activates GDNF protein is still limited and thus needing future investigation.
C6 glioma cells is considered as astrocyte-like cell (55). GDNF can be highly synthesized and released from these cells. In fact, glioma C6 cells are widely used as a model to investigate the regulatory mechanisms of GDNF synthesis. Many studies on GDNF release measure GDNF level in the culture medium. Hisaoka et al. demonstrated that the antidepressant, amitriptyline, could increase GDNF mRNA expression and GDNF release, and may be regulated by the MEK/MAPK (mitogen-activated protein kinase) pathway (56). However, phosphoinositositide-3-kinase (PI-3K)/AKT pathway was shown to play a positive role in FGF-2-stimulated GDNF release in C6 glioma cells , but this release was independent of the p44/p42 MAPK or stress-activated protein kinase/c-Jun N-terminal kinase (SAPK/JNK) (57). Verity et al. demonstrated that diverse biological factors were capable of modulating the levels of GDNF and that multiple signal transduction systems could regulate GDNF synthesis and/or release from rat C6 glioblastoma (58). Proinflammatory cytokines such as interleukin-1β, interleukin-6, tumor necrosis factor-α, and lipopolysaccharides could largely elevate GDNF release. Activation of protein kinase C (PKC) and Ca2+ ionophore can also enhance GDNF release. Study by Tanabe et al. also demonstrated that IL-1β could stimulate GDNF release through the pathways of IκB-nuclear factor kappa B (NF-κB), p38 MAPK, p44/p42 MAPK and JAK-STAT3 (59).
GDNF can be released by astrocytes or other types of gilal cells to support the survival and function of neurons, especially under injured conditions. Kuno et al. examined the regulatory mechanisms of GDNF production induced by tumor necrosis factor-α (TNF-α) in cultured astrocytes(60). Their study showed that cultured astrocytes expressed both TNF-α receptor 1 (TNFR1) and TNFR2, and that activation of these receptors by TNF-α could promote expression of both NGF and GDNF. In addition, not only exogenous TNF-α but also TNF-α produced by astrocytes could induce NGF and GDNF production in astrocytes. Their studies suggest that TNF-α can contribute to the production of neurotrophic factors in response to inflammation.
While the function of GDNF has been studied extensively, less is known about the mechanism(s) underlying its release in physiological and pathological conditions. Lonka-Nevalaita et al. demonstrated that KCl-induced depolarization to cultured neurons could increase the secretion of β precursor of GDNF and its corresponding mature GDNF in a Ca2+-dependent manner (61). Using neuron-like PC-6.3 cells, these investigators further showed that both β precursor GDNF and mature GDNF were localized primarily in secretogranin II and Rab3A-positive vesicles of the regulated secretory pathway. However, it is not clear whether GDNF is released from astrocytes through similar pathways, since astrocytes are non-excitable cells. Therefore, different secretary pathways may depend on cell type and stimulation.
2.3. GDNF receptors
RET is the most common signaling receptors for GDNF, and the RET gene is a proto-oncogene which can be activated by a DNA rearrangement mechanism (47, 62, 63) (Figure 2). This gene is primarily expressed in the peripheral enteric, sympathetic and sensory neurons, as well as in motor and dopamine neurons (63). RET receptor is a tyrosine kinase (TK) with four cadherin-like repeats and a typical TK domain (47). However, RET can be activated when GDNF firstly binds with a novel class of protein called GDNF family receptors (GFR)-α receptors located in the plasma membrane through the glycosylphosphatidyl inositol (GPI)-anchored protein (64, 65). Currently, four different GFRα receptors have been identified: GFRα1, GFRα2, GFRα3 and GFRα4, and these can bind with GDNF, neurturin (NRTN), artemin (ARTN) and persephin (PSPN), respectively(64). GDNF family proteins first form a complex with one of the four GFRα proteins, which trigger aggregation of two RET molecules, and subsequently result in transphosphorylation of specific tyrosine residues in the TK domain which then initiate intracellular pathways (47, 64, 65). In addition to GFRα, effective RET phosphorylation needs the presence of heparan sulphate, which can facilitate the interaction between GDNF and RET (54, 66). This phenomenon can also be observed upon using a high concentration of GDNF in the absence of heparan sulphate (66).
In recent years, there is evidence that other binding mechanisms or receptors can also mediate GDNF activity. Interestingly, since GDNF mutants deficient in GFRα1 binding activity could still activate RET (67), this result suggests that RET activation is independent of GFRα. Apart from RET, GDNF can trigger intracellular pathways through other receptors. In tissues like forebrain or inner ear, RET is not present, despite of existence of GFRα, thus suggesting that other transmembrane proteins can mediate GDNF activity (68). Neural cell adhesion protein (NCAM), has been proved as a transmembrane protein serving as a second GDNF receptor (69). GDNF binding to NCAM can facilitate Schwann cell migration and axonal growth in hippocampus and cortex (64). Although new activating mechanisms and receptors have been identified, our knowledge is still limited and future studies are highly warranted.
2.4. The GDNF signaling pathways
RET-dependent signaling pathways.
After binding with GDNF-GFRα complex, RET can activate many downstream signaling pathways inside and outside the lipid raft, a membrane domain essential for cell survival, differentiation, proliferation, neurite outgrowth and synaptic plasticity. Lipid rafts present on the cell surface are essential signaling compartments for cell adhesion, axon guidance, and are crucial for signal transduction (70, 71). Lipid rafts consist of dynamic assemblies of cholesterol and sphingolipids on the outer leaflet, and phospholipids in the inner leaflet (47, 70, 71). Lipid rafts also contain interactive proteins that can change in size or composition in response to stimulation of signaling pathways (47, 70, 71). Many proteins such as the GPI-anchored proteins show high affinity for rafts. Signal transduction of RET-dependent pathways appear to rely on the colocalization of RET and GFRα in the lipid rafts (72). GFRα, by virtue of GPI anchors, is normally located in lipid rafts, whereas RET is usually outside of rafts (73, 74). GDNF binding with GFRα could recruit RET to the lipid rafts, which is essential for activating downstream pathways (47, 73, 74).
Once RET binds with GFRα-GDNF complex, the tyrosine residues of RET can be phosphorylated and become activated (75). Activated RET can serve as binding sites for various intracellular proteins within the cells (76, 77). Tyr1062 can bind at least five different proteins: Shc (Src homologous and collagen-like proteins), FRS2 (fibroblast growth factor receptor substrate 2), DOk4/5 (downstream of TK 4/5), IRS1/2 (insulin receptor substrate 1/2) and enigma (47, 76, 77). These proteins have many downstream targets. In addition, activated RET can also activate PI-3K and MAPK and their downstream pathways (78-80). These pathways are known to contribute to neuronal differentiation and neurogenesis. In recent years, there is evidence that activated RET can also activate the phospholipase C-γ (PLC-γ) pathway, and thus enhance neurotransmission together with the PI-3K pathway (47, 81).
Interestingly, phosphorylation of RET can take place in different tyrosine residues after binding with the GDNF-GRFα complex. Four key residues have been identified: Tyr905, Tyr1015, Tyr1062 and Tyr1096 (64). Phosphorylation of these four residues led to stimulate a similar profile of downstream signaling pathways MAPKs ERK1 and ERK2 and the AKT kinase (75). Although different GDNF-GFRα complexes can trigger phosphorylation at different sites, it appears that different isoforms of RET can initiate specific signaling pathway (47, 75, 82). There are two isoforms of RET receptors, namely, RET9 and RET51, which differ in carboxyl terminal (72). These two isoforms do not associate with each other, and their signaling pathways are quite different. As compared with RET9, the longer one, RET51, interacts more strongly with ubiquitin ligase Cb1(83). RET51 can also interact with CrK1 while RET9 cannot. Mice lacking the long isoform appear normal in RET null mutation; however, lacking the shorter form led to abnormal kidney and enteric aganglionosis (84). These abnormalities could be rescued by RET9 isoform, suggesting that the short form is important in RET signaling in embryonic development.
RET independent signaling pathways
GDNF can activate intracellular signaling independent of the RET receptor (47). In a RET deficient cell line and primary neurons, GDNF-GFRα complexes can activate another protein called SFK (72, 85). Activated SFK was shown to phosphorylate a series of proteins such as MAPK, PCL-γ, CREB (cAMP response element binding proteins) and FOS. GDNF can promote biochemical and biological responses in conditionally immortalized neuronal precursors that express high levels of GFRαs but not RET (85). GDNF treatment did not activate the Ras/ERK pathway in these RET deficient cells, but stimulated a GFRα1-associated Src-like kinase activity in the detergent-insoluble membrane compartments, which induced rapid phosphorylation of cAMP response element-binding protein, up-regulation of c-fos mRNA, and cell survival. GDNF can also initiate signal transduction through other receptors such as NACM (69). In epithelial cells, GDNF-GFRα complexes can interact with NCAM receptor leading to activation of Fyn, a Src like protein. Fyn can in turn activate MET and initiate downstream signaling pathways. However, how Fyn is activated or how activated MET can initiate the signaling pathway is not fully understood. The RET independent pathway presents novel signaling mechanisms directly or indirectly mediated by GFRα receptors acting on a cell-autonomous manner, and thus uncovering an unexpected intersection between short- and long-range mechanisms of intercellular communication. The mechanism of RET independent signal transduction needs future investigation.
In sum, there is strong evidence suggesting that GDNF can act through a variety of pathways to mediate development and differentiation, and thus offering protection for impaired neurons after FIS.
3. GDNF in Astrocytes and Neuronal Protection after FIS
3.1. Expression of GDNF and its receptor in RAs
The ability for GDNF to protect dopaminergic and spinal motor neurons has led to consideration for this compound as a drug candidate for the treatment of Parkison’s Disease (PD) as well as other neurodegenerative diseases (86, 87). GDNF could be released from cultured astrocytes (88-91), possible because cultured astrocytes are more akin to RAs (92). GDNF is secreted to extracellular space as a glycosylated mature protein and can mediate cellular responses through GFRs (86). For in vivo studies based on mRNA analysis, GDNF was expressed in the striatum and skeletal muscle (62), targeted fields for dopaminergic substantia neurons and motor neurons, respectively. In normal brain, GDNF is mostly confined to neurons and is absent from or expressed at low levels in astrocytes (93-95). Consistently, RNA-seq study also showed that mature mouse neurons express higher GDNF mRNA than mature mouse astrocytes (96). However, many studies have demonstrated upregulation of GDNF in RAs after FIS (93, 97-100). After stroke, a 3-fold increased expression of GDNF was mainly limited to RAs (100, 101). In situ hybridization study also indicated elevation of mRNA of GDNF and the increased number of GDNF mRNA-expressing cells in the cortex and striatum after middle cerebral artery occlusion (MCAO), whereas no changes in mRNA expression for neurturin or persephin were detected (100). At 3-7 days after MCAO, both RAs and surviving neurons showed increase in GDNF (93). In the PIR of cerebral cortex and striatum, expression of GDNF mRNA and protein increased as early as 2 h after ischemia-reperfusion, and this was followed by a decline and another increase at 72 h (99). Double staining showed that the earlier peak of GDNF expression was of neuronal origin and the later peak of glial origin. Using Western blot analysis, Zhang et al. showed that GDNF levels increased for a sustained period after photothrombosis (PT)-induced FIS, and immunostaining of GDNF and GFAP indicated significantly increase in RAs (102).
Besides expression of GFRs in cultured cortical astrocytes and neurons (103), the expression of receptors for GDNF family proteins were also increased after stroke (100, 101). After a 2-h MCAO, both c-Ret, GFRα1 and GFRα2 mRNA levels were markedly increased outside the ischemic lesion in the ipsilateral cortex at 6–24 h, whereas GFRα2 expression was decreased in the cortical areas both within and outside the lesion (100). In a rat model of occlusion of the right carotid arteries, both RET and GFRα mRNA level were markedly increased in the cortex, striatum and hippocampus 6-24 hours after ischemia (100). Astrocytes in rats treated with quinolinic acid or kainic acid also showed increased expression of GDNF as well as its receptor GFRα1 (101, 104). Similarly, after mechanical injury of spinal cord in adult rats, GDNF and GFRα1 were also upregulated in RAs (105). Upregulation of GDNF and its receptor signaling pathways in RAs after stroke strongly suggests that RAs may serve as an important source of GDNF to enhance neuroprotective and neuroregenerative responses during the post-ischemic time.
3.2. The role and mechanisms of astrocyte-derived GDNF in stroke outcomes
The brain tissue is known to have a remarkable capacity for spontaneous recovery after FIS, although such mechanism(s) remains largely unclear. Substantial interest has focused on the potential protective effects of GDNF owning to its upregulated production as well as expression of its receptors RET and GFRα-1 (93, 95, 97-100). In fact, based on treatment using recombinant GDNF and molecular genetic methods, multiple mechanisms for GDNF to protect neurons and brain after stroke have been investigated (Figure 3). For example, recombinant GDNF was shown to offer protection after MCAO (106-111). GDNF could reduce apoptosis via the caspase-3 dependent pathway (112), through upregulation of anti-apoptotic Bcl-2 and Bcl-Xl (108, 113), and potentiate and prolong activation of the p-AKT pathway (109). After permanent MCAO, GDNF treatment in cerebral cortex could significantly reduce the immunoreactivity of caspases-1 and caspases-3 in TUNEL positive apoptotic cells, suggesting that GDNF could inhibit DNA fragmentation in apoptotic pathways (108). GDNF could also inhibit excitotoxicity after stroke (114). In an in vivo excitotoxicity model induced by administration of a unilateral dose of N-methyl-d-aspartate (NMDA) to the hippocampus (a mechanism that could induce lesion in the CA1 region), prior transduction of a lentiviral vector expressing GDNF showed protection against neuronal loss and significantly reduced lesion size, thus protecting the hippocampus from excitotoxic damage. Administration of GDNF in the injured cortex could also reduced NMDA-induced calcium influx (103). In hippocampal brain slices, pretreatment with GDNF was shown to significantly attenuate the NMDA-induced cell death (106). The protective response was related to the increased expression of GDNF after administration of NMDA (115). There is also evidence showing that GDNF could attenuate slowing of NMDA-induced excitotoxic neuronal death through activation of the MAPK pathway (103).
Figure 3. Potential mechanisms of RAs-derived GDNP on neuronal protection after stroke.
Astrocytes under normal condition express and release little GDNF. After a FIS, GDNF is upregulated and released from reactive astrocytes (RAs), which can inhibit excitotoxicity, apoptosis, oxidative stress, and modulate synaptic loss of neurons, thereby promoting neuronal survival and brain repair, and improving long-term stroke outcome. Red dots represent GDNF molecules.
Increased oxidative stress is known to contribute to neuronal death after stroke, and thus affecting brain recovery. Along this line, recombinant GDNF was shown to reduce oxidative stress and protect neurons via transcriptional regulation of glutathione synthesis (116). Increased oxidative stress can be attributed to two important oxidative species, namely, the ROS (reactive oxide species) and RNS (reactive nitrogen species) (1, 117). Under ischemic conditions, impaired ability of neurons to maintain normal ionic gradients can lead to excessive influx of calcium, and subsequently activating nitric oxide synthase and NADPH oxidase to produce more RNS and ROS (1, 118). After stroke, GDNF was shown to prevent degeneration of motor neurons through inhibiting the production of RNS and ROS (119). Indeed, treatment of GDNF after stroke could inhibit nitric oxide synthase (NOS) and thus decreased production of RNS (120). GDNF could also downregulate ROS and prevent the over influx of calcium (103).
GSH is known to serve as an antioxidant within cells to defend against oxidative stress-induced injury. Activation of the pentose phosphate pathway (PPP) is considered as the first line of defense response against oxidative stress-induced cell death (121, 122). The production of GSH in this pathway is controlled by NADPH which is mainly generated through the PPP. G6PD is the rate-limiting enzyme in the oxidative stage of PPP and is of central importance to provide NADPH. G6PD converts G6P to 6-p-gluconolactone and generates NADPH, which is required for regeneration of GSH in antioxidant pathways to reduce oxidative stress (123, 124). GDNF treatment can increase the level of reduced glutathione (GSH) (125), suggesting that GDNF not only can directly reduce oxidative species but also increase the antioxidative capability of cells and tissues.
Studies using recombinant GDNF have demonstrated that the neuronal and brain protective effects of this factor can be a promising candidate for stroke therapy. However, there are few weaknesses exist in term of mechanism. First, local delivery of recombinant GDNF protein can only affect a small region of tissue surrounding the injection site. Secondly, most studies have focused on evaluating the effects of GDNF on brain infarction and cell death in the acute phase, but whether these effects prevail upon long-term neural regeneration and motor functional recovery have not been studied in detail. Thirdly, GDNF administration is not cell-type specific, therefore, leaving the respective roles of neuron- or astrocyte-derived GDNF in brain protection and recovery unclear. Lastly, some toxic effects were found upon continuous administration of recombinant GDNF to the primate PD model (86).
There is increasing evidence indicating a non-cell autonomous effect of astrocytes or RAs on neurodegeneration and death under different pathological conditions (35, 126). Genetic strategies such as viral transduction to increase local production of GDNF have been tested for stroke therapy (127-131). However, few studies have used molecular genetic approach to test whether and how astrocyte specific GDNF could provide neuronal and brain protection after FIS. With the availability of genetic tools such as astrocyte-specific driver transgenic mouse lines and astrocyte-specific adeno-associated virus (AAV) for gene deletion and overexpression (132, 133), dissection of exclusive roles of astrocytes in neuronal circuit and in diseases associated with neurodegeneration is becoming feasible. For example, GLAST-CreERT2 mediated recombination has been used to express fluorescent reporters to study the morphology and Ca2+ signaling in astrocytes, and to label adult-born granule cells (134-136). To study the effects of RA-derived GDNF on brain protection and recovery, Zhang et al. generated astrocyte-specific, inducible and conditional GDNF knockout (cKO) mice, i.e., GLAST-GDNF−/− cKO mice, by crossing floxed GDNF (GDNFf/f) mice with GLAST-CreERT2 driver line (102). This study showed the effects of endogenous astrocytic GDNF on neuronal degeneration, brain infarction, reactive astrogliosis, oxidative stress and behavioral deficits after PT. This study provided several novel findings on the GLAST-GDNF−/− cKO mice and PT-induced FIS mouse model: (1) GDNF is highly upregulated in the brain and RAs after PT, and the increased level was maintained within a sustained time during the post-FIS period, suggesting that GDNF is a encoded by a stress-responsive gene. In addition, the secreted GDNF could act on neurons to mediate protective effect after FIS. (2) Deletion of GDNF in astrocytes was shown to exacerbate brain and hippocampal damage and neuronal death. Brain infarction and neuronal death at PIR were significantly increased in GLAST-GDNF−/− cKO mice. These results are consistent with previous study on a MCAO rat model showing that application of recombinant GDNF could significantly reduce both infarct size and brain edema (108). (3) RAs-derived GDNF could also affect reactive astrogliosis after PT. Using confocal imaging on immunostained Brdu and Ki67, the GLAST-GDNF−/− cKO mice exhibited a significant reduction in Brdu+ and Ki67+ proliferating cells in the PIR after PT as compared with the WT mice. Here, the GLAST-GDNF−/− cKO mice also showed less GFAP+ RAs and lower GFAP immunofluorescent signals than the WT mice, especially at 2 and 4 days after PT. Moreover, the GLAST-GDNF−/− cKO mice showed less Brdu+ cells and GFAP+Brdu+ double positive cells in proliferating RAs, lower ratio of GFAP+ proliferating RAs among total proliferating cells (i.e., GFAP+Brdu+/Brdu+ ratio), and lower ratios of GFAP+ proliferating RAs (i.e., GFAP+Brdu+/GFAP+ ratio) as compared with the control mice after PT. However, cell proliferation became largely stabilized at 14 days after PT, and after this time, no major differences were observed between the control and cKO mice. These results indicate that the RAs-derived GDNF can affect the dynamics of reactive astrogliosis through a cell autonomous manner (102). These results also corroborated with the data showing reduced adult neurogenesis in the GLAST-GDNF−/− cKO mice under normal conditions, suggesting that reduced reactive astrogliosis can lead to worsening motor function recovery in the cKO mice. (4) GLAST-GDNF−/− cKO mice showed reduced adult neurogenesis in the hippocampus in normal brain. In addition to having proliferative capacity, RAs also exhibit many other stem cell-like properties (137-144). For example, they express neural stem cell markers such as nestin, Sox2 and DCX (141, 143, 145, 146). Given that GLAST is also expressed in neural stem cells, Zhang et al. examined the effects of GDNF deletion in astrocytes on adult neurogenesis in the dentate gyrus (DG) (102). Normally, neurogenesis in the adult brain exists in two neurogenic regions in the brain, namely, the subgranular zone (SGZ) of DG in the hippocampus, and the subventricular zone (SVZ) along the walls of the lateral ventricle (147, 148). Unique microenvironments (i.e., ‘niches’) in these two regions include soluble factors, membrane-bound molecules and extracellular matrix, permitting stem cells to self-renewal and progenitor differentiation into neurons in adult stage. Neurogenesis and survival of newly generated neurons can be regulated by various factors. GLAST is an astrocyte-specific glutamate transporter and is expressed in type I neural stem cell. GLAST-lineage radial glia-like cells (RGCs) can contribute to adult hippocampal neurogenesis (136). In the GLAST-GDNF−/− cKO mice, GDNF was reduced in hippocampus under normal conditions albeit without statistical significance(102). Study by Zhang et al. also found that the cKO mice exhibit reduced adult neurogenesis in the DG under normal conditions, suggesting that GDNF in GLAST-lineage RGCs is an important growth factor to facilitate adult neurogenesis. The results of neurogenesis and GDNF levels are not contradictive, since in normal adult brain, GDNF is mostly expressed in neurons and only very low in astrocytes (93-95). On the other hand, growing evidence suggests that RAs play an important role in neurogenesis in the SVZ and SGZ in DG (149, 150). Ischemic stroke can cause substantial reactive astrogliosis in SVZ, and there is an association between endogenous neurogenesis and improved stroke outcomes (151). Therefore, RAs can play a role in stroke recovery through modulating neurogenesis via GDNF. Consistently, intracerebral infusion of GDNF was shown to promote striatal neurogenesis after stroke (107). (5) Deletion of GDNF in astrocyte could decrease G6PD and increase ROS production. Using double immunostaining of G6PD and GFAP, study by Zhang et al. showed that G6PD was largely upregulated in RAs after PT, but deletion of GDNF in astrocytes suppressed the upregulation of G6PD in RAs (102). In vivo DHE labeling confirmed that the GDNF deficient mice have much higher levels of ROS in the PIR than WT mice, further suggesting that through activating an antioxidant mechanism via PPP, RAs-derived GDNF can exert a neuronal and brain protective effect after FIS (102). These results are consistent with the in vitro and in vivo studies on Parkinson’s disease models (125, 152), showing that through analysis of oxidative stress markers such as protein carbonyls and 4-hydroxynonenal, astrocytes overexpressing GDNF or recombinant GDNF can reduce oxidative stress in 6-hydroxydopamine (6-OHDA)-induced mouse Parkinson’s disease model (152). Both markers were significantly reduced in GDNF treated animals. In addition, GDNF was shown to protect against loss of striatal and nigral tissue levels of dopamine. Taken together, these results agree with the notion that the protective effects of GDNF against 6-OHDA involves a reduction in oxidative stress. (6) GLAST-GDNF−/− cKO mice exhibited higher motor function deficits than the control mice after PT, suggesting that RA-derived GDNF may play an important role in intrinsic brain repair and recovery in the chronic phase after FIS. Taken together, these results further demonstrate that astrocytes- and/or RAs-derived GDNF is beneficial to protect against neuronal death and brain damage after FIS, stimulate reactive astrogliosis, promote anti-oxidative stress mechanism, and improve long-term stroke outcomes. These findings firmly indicate the role of endogenous GDNF in astrocytes/RAs as an essential factor in brain repair processes.
RAs-derived GDNF may promote neuronal survival through inhibition of microglia activation. Microglial activation in stroke has been implicated as a major source of inflammation that contribute to neuronal dysfunction and degeneration (19). Factors secreted by astrocytes have shown to reduce the phagocytic activity and the production of ROS by activated microglia. Using Zymosan A-stimulated midbrain microglia cultures, Sandra et al. showed that astrocytes conditioned media was capable of modulating microglial activation, including increase in phagocytic activity and production of ROS (89). Both parameters were diminished in microglia culture incubated with astrocytes conditioned media, but not in conditioned media silenced with GDNF. These results indicate that astrocyte derived GDNF can play a major role in the control of microglial activation in stroke, and that GDNF can protect against neurodegeneration through inhibition of neuroinflammation.
Since GDNF is a potent neurotropic factor capable of promoting neuronal synaptic formation (45), it is also expected that RAs-derived GDNF may directly stimulate synaptogenesis and enhance function of surviving neurons in the PIR after FIS. However, information in this area is limited and future studies involving electrophysiology and in vivo two-photon imaging on dendritic spines may shed new lights.
4. Concluding Remarks
Evidence from clinical studies suggest that the brain is ‘primed’ to recovery during the subacute phase of ischemic stroke (153, 154). Therefore, understanding its mechanism will help to identify potential strategies to facilitate brain recovery and improve stroke outcomes. In this respect, GDNF released from RAs may offer multiple functions to mediate recovery of damaged neurons after FIS. Studies on glia-neuron interactions in health and disease reach an inflection point in which cell-specific in vivo approaches are beginning to validate evidence obtained from pharmacological approaches. With recent advance in genetic tools, such as availability of astrocyte specific transgenic mouse lines and astrocyte specific viral transduction, it is possible to look at exclusive roles of astrocytes/RAs in neuronal circuitry during neuronal degeneration/death. Further studies on the role and mechanism of GDNF in RAs may help to better understand the intrinsic brain repair mechanisms through the non-cell and cell autonomous effects, and in the context of neuron-glia interactions. These observations further demonstrate that targeting RAs can be a potentially important strategy in stroke restorative therapies.
Acknowledgements
This work was supported by the National Institute of Health [National Institute of Neurological Disorders and Stroke (NINDS) grants R01NS069726 and R01NS094539 to SD] and the America Heart Association [Midwest Affiliate Grant-in-Aid (16GRNT31280014), and NCRG-IRG 16IRG27780023 to SD].
Footnotes
Conflict of interest
The authors declare no conflict of interest.
References
- 1.Dirnagl U, Iadecola C, and Moskowitz MA 1999. Pathobiology of ischaemic stroke: an integrated view. Trends Neurosci 22:391–397. [DOI] [PubMed] [Google Scholar]
- 2.Dirnagl U 2012. Pathobiology of injury after stroke: the neurovascular unit and beyond. Annals of the New York Academy of Sciences 1268:21–25. [DOI] [PubMed] [Google Scholar]
- 3.Dirnagl U, Simon RP, and Hallenbeck JM 2003. Ischemic tolerance and endogenous neuroprotection. Trends Neurosci 26:248–254. [DOI] [PubMed] [Google Scholar]
- 4.Campbell BCV, De Silva DA, Macleod MR, Coutts SB, Schwamm LH, Davis SM, and Donnan GA 2019. Ischaemic stroke. Nature Reviews Disease Primers 5:70. [DOI] [PubMed] [Google Scholar]
- 5.Verkhratsky A, and Nedergaard M 2017. Physiology of Astroglia. Physiol. Rev 98:239–389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Halassa MM, Fellin T, and Haydon PG The tripartite synapse: roles for gliotransmission in health and disease. Trends in Molecular Medicine In Press, Corrected Proof. [DOI] [PubMed] [Google Scholar]
- 7.Haydon PG 2001. GLIA: listening and talking to the synapse. [Review] [90 refs]. Nature Reviews Neuroscience 2:185–193. [DOI] [PubMed] [Google Scholar]
- 8.Haydon PG, and Carmignoto G 2006. Astrocyte Control of Synaptic Transmission and Neurovascular Coupling. Physiol. Rev 86:1009–1031. [DOI] [PubMed] [Google Scholar]
- 9.Kimelberg H, and Nedergaard M 2010. Functions of astrocytes and their potential as therapeutic targets. Neurotherapeutics 7:338–353. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Zuchero JB, and Barres BA 2015. Glia in mammalian development and disease. Development 142:3805–3809. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Pekny M, Pekna M, Messing A, Steinh+ñuser C, Lee JM, Parpura V, Hol EM, Sofroniew MV, and Verkhratsky A 2016. Astrocytes: a central element in neurological diseases. Acta Neuropathologica 131:323–345. [DOI] [PubMed] [Google Scholar]
- 12.Verkhratsky A, Zorec R, and Parpura V 2017. Stratification of astrocytes in healthy and diseased brain. Brain Pathology 27:629–644. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Brown CE, Aminoltejari K, Erb H, Winship IR, and Murphy TH 2009. In Vivo Voltage-Sensitive Dye Imaging in Adult Mice Reveals That Somatosensory Maps Lost to Stroke Are Replaced over Weeks by New Structural and Functional Circuits with Prolonged Modes of Activation within Both the Peri-Infarct Zone and Distant Sites. The Journal of Neuroscience 29:1719–1734. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Clarkson AN, Lopez-Valdes HE, Overman JJ, Charles AC, Brennan KC, and Thomas Carmichael S 2013. Multimodal examination of structural and functional remapping in the mouse photothrombotic stroke model. J Cereb Blood Flow Metab 33:716–723. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Murphy TH, and Corbett D 2009. Plasticity during stroke recovery: from synapse to behaviour. Nat Rev Neurosci 10:861–872. [DOI] [PubMed] [Google Scholar]
- 16.Mostany R, Chowdhury TG, Johnston DG, Portonovo SA, Carmichael ST, and Portera-Cailliau C 2010. Local Hemodynamics Dictate Long-Term Dendritic Plasticity in Peri-Infarct Cortex. J. Neurosci 30:14116–14126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Winship IR, and Murphy TH 2008. In Vivo Calcium Imaging Reveals Functional Rewiring of Single Somatosensory Neurons after Stroke. J. Neurosci 28:6592–6606. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Barreto GE, Sun X, Xu L, and Giffard RG 2011. Astrocyte Proliferation Following Stroke in the Mouse Depends on Distance from the Infarct. PLoS ONE 6:e27881. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Li H et al. 2014. Histological, cellular and behavioral assessments of stroke outcomes after photothrombosis-induced ischemia in adult mice. BMC Neuroscience 15:58. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Choudhury GR, and Ding S Reactive astrocytes and therapeutic potential in focal ischemic stroke. Neurobiol. Dis 85: 234–244. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Ding S 2014. Dynamic reactive astrocytes after focal ischemia. Neural Regenerative Research 9:2048–2052. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Burda J, and Sofroniew M 2014. Reactive Gliosis and the Multicellular Response to CNS Damage and Disease. Neuron 81:229–248. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Voskuhl RR, Peterson RS, Song B, Ao Y, Morales LB, Tiwari-Woodruff S, and Sofroniew MV 2009. Reactive Astrocytes Form Scar-Like Perivascular Barriers to Leukocytes during Adaptive Immune Inflammation of the CNS. J. Neurosci 29:11511–11522. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Linnerbauer M, and Rothhammer V 2020. Protective Functions of Reactive Astrocytes Following Central Nervous System Insult. Frontiers in Immunology 11:2571. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Myer DJ, Gurkoff GG, Lee SM, Hovda DA, and Sofroniew MV 2006. Essential protective roles of reactive astrocytes in traumatic brain injury. Brain 129:2761–2772. [DOI] [PubMed] [Google Scholar]
- 26.Faulkner JR, Herrmann JE, Woo MJ, Tansey KE, Doan NB, and Sofroniew MV 2004. Reactive Astrocytes Protect Tissue and Preserve Function after Spinal Cord Injury. The Journal of Neuroscience 24:2143–2155. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Anderson MA, Burda JE, Ren Y, Ao Y Ö'Shea TM, Kawaguchi R, Coppola G, Khakh BS, Deming TJ, and Sofroniew MV 2016. Astrocyte scar formation aids central nervous system axon regeneration. Nature 532:195–200. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Bush TG, Puvanachandra N, Horner CH, Polito A, Ostenfeld T, Svendsen CN, Mucke L, Johnson MH, and Sofroniew MV 1999. Leukocyte Infiltration, Neuronal Degeneration, and Neurite Outgrowth after Ablation of Scar-Forming, Reactive Astrocytes in Adult Transgenic Mice. Neuron 23:297–308. [DOI] [PubMed] [Google Scholar]
- 29.Zamanian JL, Xu L, Foo LC, Nouri N, Zhou L, Giffard RG, and Barres BA 2012. Genomic Analysis of Reactive Astrogliosis. The Journal of Neuroscience 32:6391–6410. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Zhao Y, and Rempe D 2010. Targeting astrocytes for stroke therapy. Neurotherapeutics 7:439–451. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Gleichman AJ, and Carmichael ST 2014. Astrocytic therapies for neuronal repair in stroke. Neuroscience Letters 565:47–52. [DOI] [PubMed] [Google Scholar]
- 32.Chouchane M, and Costa MR 2012. Cell therapy for stroke: use of local astrocytes. Frontiers in Cellular Neuroscience 6:49. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Escartin C, and Bonvento G 2008. Targeted Activation of Astrocytes: A Potential Neuroprotective Strategy. Mol Neurobiol 38:231–241. [DOI] [PubMed] [Google Scholar]
- 34.Lobsiger CS, and Cleveland DW 2007. Glial cells as intrinsic components of non-cell-autonomous neurodegenerative disease. Nat Neurosci 10:1355–1360. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Jolly S, Journiac N, Naudet F.d.r., Gautheron V, Mariani J, and Vernet-der Garabedian B 2011. Cell-Autonomous and Non-Cell-Autonomous Neuroprotective Functions of RORα in Neurons and Astrocytes during Hypoxia. J. of Neurosci 31:14314–14323. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Li H, Xie Y, Zhang N, Yu Y, Zhang Q, and Ding S 2015. Disruption of IP3R2-mediated Ca2+ signaling pathway in astrocytes ameliorates neuronal death and brain damage while reducing behavioral deficits after focal ischemic stroke. Cell Calcium 58:565–576. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Maragakis NJ, and Rothstein JD 2006. Mechanisms of Disease: astrocytes in neurodegenerative disease. Nat Clin Pract Neuro 2:679–689. [DOI] [PubMed] [Google Scholar]
- 38.Pecho-Vrieseling E, Rieker C, Fuchs S, Bleckmann D, Esposito MS, Botta P, Goldstein C, Bernhard M, Galimberti I, Muller M, Luthi A, Arber S, Bouwmeester T, van der Putten H, and Di Giorgio FP 2014. Transneuronal propagation of mutant huntingtin contributes to non-cell autonomous pathology in neurons. Nat Neurosci 17:1064–1072. [DOI] [PubMed] [Google Scholar]
- 39.Barreto GE, Gonzalez J, Torres Y, and Morales L 2011. Astrocytic-neuronal crosstalk: Implications for neuroprotection from brain injury. Neuroscience Research 71:107–113. [DOI] [PubMed] [Google Scholar]
- 40.Becerra-Calixto A, and Cardona-Gomez GP 2017. The Role of Astrocytes in Neuroprotection after Brain Stroke: Potential in Cell Therapy. Frontiers in Molecular Neuroscience 10:88. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Poyhonen S, Er S, Domanskyi A, and Airavaara M 2019. Effects of Neurotrophic Factors in Glial Cells in the Central Nervous System: Expression and Properties in Neurodegeneration and Injury. Frontiers in Physiology 10:486. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Duarte E.l.P., Curcio M, Canzoniero LM, and Duarte CB 2012. Neuroprotection by GDNF in the ischemic brain. Growth Factors 30:242–257. [DOI] [PubMed] [Google Scholar]
- 43.Harvey BK, Hoffer BJ, and Wang Y 2005. Stroke and TGF-α proteins: glial cell line-derived neurotrophic factor and bone morphogenetic protein. Pharmacology & Therapeutics 105:113–125. [DOI] [PubMed] [Google Scholar]
- 44.Nakajima K, Hida H, Shimano Y, Fujimoto I, Hashitani T, Kumazaki M, Sakurai T, and Nishino H 2001. GDNF is a major component of trophic activity in DA-depleted striatum for survival and neurite extension of DAergic neurons. Brain Research 916:76–84. [DOI] [PubMed] [Google Scholar]
- 45.Ledda F, Paratcha G, Sandoval-Guzman T, and Ibanez CF 2007. GDNF and GFRα1 promote formation of neuronal synapses by ligand-induced cell adhesion. Nat Neurosci 10:293–300. [DOI] [PubMed] [Google Scholar]
- 46.Chen Y, Ai Y, Slevin JR, Maley BE, and Gash DM 2005. Progenitor proliferation in the adult hippocampus and substantia nigra induced by glial cell line-derived neurotrophic factor. Experimental Neurology 196:87–95. [DOI] [PubMed] [Google Scholar]
- 47.Airaksinen MS, and Saarma M 2002. The GDNF family: Signalling, biological functions and therapeutic value. Nat Rev Neurosci 3:383–394. [DOI] [PubMed] [Google Scholar]
- 48.Pascual A, Hidalgo-Figueroa M, Piruat JI, Pintado CO, Gomez-Diaz R, and Lopez-Barneo J 2008. Absolute requirement of GDNF for adult catecholaminergic neuron survival. Nat Neurosci 11:755–761. [DOI] [PubMed] [Google Scholar]
- 49.Uesaka T, Nagashimada M, and Enomoto H 2013. GDNF Signaling Levels Control Migration and Neuronal Differentiation of Enteric Ganglion Precursors. The Journal of Neuroscience 33:16372–16382. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50.Lin LF, Doherty DH, Lile JD, Bektesh S, and Collins F 1993. GDNF: a glial cell line-derived neurotrophic factor for midbrain dopaminergic neurons. Science 260:1130–1132. [DOI] [PubMed] [Google Scholar]
- 51.Hellmich HL, Kos L, Cho ES, Mahon KA, and Zimmer A 1996. Embryonic expression of glial cell-line derived neurotrophic factor (GDNF) suggests multiple developmental roles in neural differentiation and epithelial-mesenchymal interactions. Mechanisms of Development 54:95–105. [DOI] [PubMed] [Google Scholar]
- 52.Cortes D, Carballo-Molina OA, Castellanos-Montiel M. a.J., and Velasco I 2017. The Non-Survival Effects of Glial Cell Line-Derived Neurotrophic Factor on Neural Cells. Frontiers in Molecular Neuroscience 10:258. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Cortes D, Robledo-Arratia Y, Hernandez-Martinez R, Escobedo-Avila I, Bargas J, and Velasco I 2016. Transgenic GDNF Positively Influences Proliferation, Differentiation, Maturation and Survival of Motor Neurons Produced from Mouse Embryonic Stem Cells. Frontiers in Cellular Neuroscience 10:217. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Bespalov MM, Sidorova YA, Tumova S, Ahonen-Bishopp A, Magalhaes AC, Kulesskiy E, Paveliev M, Rivera C, Rauvala H, and Saarma M 2011. Heparan sulfate proteoglycan syndecan-3 is a novel receptor for GDNF, neurturin, and artemin. J Cell Biol 192:153–169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Saavedra A, Baltazar G, and Duarte E.l.P. 2008. Driving GDNF expression: The green and the red traffic lights. Progress in Neurobiology 86:186–215. [DOI] [PubMed] [Google Scholar]
- 56.Hisaoka K, Nishida A, Koda T, Miyata M, Zensho H, Morinobu S, Ohta M, and Yamawaki S 2001. Antidepressant drug treatments induce glial cell line-derived neurotrophic factor (GDNF) synthesis and release in rat C6 glioblastoma cells. Journal of Neurochemistry 79:25–34. [DOI] [PubMed] [Google Scholar]
- 57.Tanabe K, Matsushima-Nishiwaki R, Iida M, Kozawa O, and Iida H 2012. Involvement of phosphatidylinositol 3-kinase/Akt on basic fibroblast growth factor-induced glial cell line-derived neurotrophic factor release from rat glioma cells. Brain Research 1463:21–29. [DOI] [PubMed] [Google Scholar]
- 58.Verity AN, Wyatt TL, Hajos B, Eglen RM, Baecker PA, and Johnson RM 1998. Regulation of Glial Cell Line-Derived Neurotrophic Factor Release from Rat C6 Glioblastoma Cells. Journal of Neurochemistry 70:531–539. [DOI] [PubMed] [Google Scholar]
- 59.Tanabe K, Nishimura K, Dohi S, and Kozawa O 2009. Mechanisms of interleukin-1β-induced GDNF release from rat glioma cells. Brain Research 1274:11–20. [DOI] [PubMed] [Google Scholar]
- 60.Kuno R, Yoshida Y, Nitta A, Nabeshima T, Wang J, Sonobe Y, Kawanokuchi J, Takeuchi H, Mizuno T, and Suzumura A 2006. The role of TNF-alpha and its receptors in the production of NGF and GDNF by astrocytes. Brain Research 1116:12–18. [DOI] [PubMed] [Google Scholar]
- 61.Lonka-Nevalaita L, Lume M, Leppanen S, Jokitalo E, Peranen J, and Saarma M 2010. Characterization of the Intracellular Localization, Processing, and Secretion of Two Glial Cell Line-Derived Neurotrophic Factor Splice Isoforms. The Journal of Neuroscience 30:11403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62.Allen SJ, Watson JJ, Shoemark DK, Barua NU, and Patel NK 2013. GDNF, NGF and BDNF as therapeutic options for neurodegeneration. Pharmacology & Therapeutics 138:155–175. [DOI] [PubMed] [Google Scholar]
- 63.Wang X 2013. Structural studies of GDNF family ligands with their receptors-Insights into ligand recognition and activation of receptor tyrosine kinase RET. Biochimica et Biophysica Acta (BBA) - Proteins and Proteomics 1834:2205–2212. [DOI] [PubMed] [Google Scholar]
- 64.Sariola H, and Saarma M 2003. Novel functions and signalling pathways for GDNF. Journal of Cell Science 116:3855. [DOI] [PubMed] [Google Scholar]
- 65.Ibanez CF, and Andressoo JO 2017. Biology of GDNF and its receptors-Relevance for disorders of the central nervous system. Neurobiology of Disease 97:80–89. [DOI] [PubMed] [Google Scholar]
- 66.Barnett MW, Fisher CE, Perona-Wright G, and Davies JA 2002. Signalling by glial cell line-derived neurotrophic factor (GDNF) requires heparan sulphate glycosaminoglycan. Journal of Cell Science 115:4495. [DOI] [PubMed] [Google Scholar]
- 67.Eketjall S, Fainzilber M, Murray-Rust J, and Ibanez CF 1999. Distinct structural elements in GDNF mediate binding to GFRalpha1 and activation of the GFR-c-Ret receptor complex. The EMBO Journal 18:5901–5910. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Trupp M, Belluardo N, Funakoshi H, and Ibanez CF 1997. Complementary and Overlapping Expression of Glial Cell Line-Derived Neurotrophic Factor (GDNF), c-ret Proto-Oncogene, and GDNF Receptor-α Indicates Multiple Mechanisms of Trophic Actions in the Adult Rat CNS. The Journal of Neuroscience 17:3554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.Paratcha G, Ledda F, and Ibanez CF 2003. The Neural Cell Adhesion Molecule NCAM Is an Alternative Signaling Receptor for GDNF Family Ligands. Cell 113:867–879. [DOI] [PubMed] [Google Scholar]
- 70.Simons K, and Toomre D 2000. Lipid rafts and signal transduction. Nature Reviews Molecular Cell Biology 1:31–39. [DOI] [PubMed] [Google Scholar]
- 71.Tsui-Pierchala BA, Encinas M, Milbrandt J, and Johnson EM 2002. Lipid rafts in neuronal signaling and function. Trends Neurosci 25:412–417. [DOI] [PubMed] [Google Scholar]
- 72.Poteryaev D, Titievsky A, Sun YF, Thomas-Crusells J, Lindahl M, Billaud M, Arum+ñe U, and Saarma M 1999. GDNF triggers a novel Ret-independent Src kinase family-coupled signaling via a GPI-linked GDNF receptor α1. FEBS Letters 463:63–66. [DOI] [PubMed] [Google Scholar]
- 73.Baloh RH, Enomoto H, Johnson EM, and Milbrandt J 2000. The GDNF family ligands and receptors-implications for neural development. Current Opinion in Neurobiology 10:103–110. [DOI] [PubMed] [Google Scholar]
- 74.Takahashi M 2001. The GDNF/RET signaling pathway and human diseases. Cytokine & Growth Factor Reviews 12:361–373. [DOI] [PubMed] [Google Scholar]
- 75.Coulpier M, Anders J, and Ibanez CF 2002. Coordinated Activation of Autophosphorylation Sites in the RET Receptor Tyrosine Kinase: IMPORTANCE OF TYROSINE 1062 FOR GDNF MEDIATED NEURONAL DIFFERENTIATION AND SURVIVAL. J. Biol. Chem 277:1991–1999. [DOI] [PubMed] [Google Scholar]
- 76.Lemmon MA, and Schlessinger J 2010. Cell Signaling by Receptor Tyrosine Kinases. Cell 141:1117–1134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Paratcha G, Ledda F, Baars L, Coulpier M, Besset V, Anders J, Scott R, and Ibanez CF 2001. Released GFRα1 Potentiates Downstream Signaling, Neuronal Survival, and Differentiation via a Novel Mechanism of Recruitment of c-Ret to Lipid Rafts. Neuron 29:171–184. [DOI] [PubMed] [Google Scholar]
- 78.Ibanez CF 2013. Structure and Physiology of the RET Receptor Tyrosine Kinase. Cold Spring Harbor Perspectives in Biology 5:a009134. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Chen ZY, Chai YF, Cao L, Huang AJ, Cui RY, Lu CL, and He C 2001. Glial cell line-derived neurotrophic factor promotes survival and induces differentiation through the phosphatidylinositol 3-kinase and mitogen-activated protein kinase pathway respectively in PC12 cells. Neuroscience 104:593–598. [DOI] [PubMed] [Google Scholar]
- 80.Encinas M, Tansey MG, Tsui-Pierchala BA, Comella JX, Milbrandt J, and Johnson EM 2001. c-Src Is Required for Glial Cell Line-Derived Neurotrophic Factor (GDNF) Family Ligand-Mediated Neuronal Survival via a Phosphatidylinositol-3 Kinase (PI-3K)-Dependent Pathway. J of Neurosci 21:1464. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Yang F, He X.p., Feng L, Mizuno K, Liu XW, Russell J, Xiong WC, and Lu B 2001. PI-3 kinase and IP3 are both necessary and sufficient to mediate NT3-induced synaptic potentiation. Nature Neuroscience 4:19–28. [DOI] [PubMed] [Google Scholar]
- 82.Fukuda T, Kiuchi K, and Takahashi M 2002. Novel Mechanism of Regulation of Rac Activity and Lamellipodia Formation by RET Tyrosine Kinase. J. Biol. Chem 277:19114–19121. [DOI] [PubMed] [Google Scholar]
- 83.Richardson DS, Rodrigues DM, Hyndman BD, Crupi MJF, Nicolescu AC, and Mulligan LM 2012. Alternative splicing results in RET isoforms with distinct trafficking properties. MBoC 23:3838–3850. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.de Graaff E, Srinivas S, Kilkenny C, D'Agati V, Mankoo BS, Costantini F, and Pachnis V 2001. Differential activities of the RET tyrosine kinase receptor isoforms during mammalian embryogenesis. Genes & Development 15:2433–2444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Trupp M, Scott R, Whittemore SR, and Ibanez CF 1999. Ret-dependent and -independent Mechanisms of Glial Cell Line-derived Neurotrophic Factor Signaling in Neuronal Cells. J. Biol. Chem 274:20885–20894. [DOI] [PubMed] [Google Scholar]
- 86.Luz M, Mohr E, and Fibiger HC 2016. GDNF-induced cerebellar toxicity: A brief review. NeuroToxicology 52:46–56. [DOI] [PubMed] [Google Scholar]
- 87.Kirik D, Georgievska B, and Bjorklund A 2004. Localized striatal delivery of GDNF as a treatment for Parkinson disease. Nat Neurosci 7:105–110. [DOI] [PubMed] [Google Scholar]
- 88.Yamagata K, Hakata K, Maeda A, Mochizuki C, Matsufuji H, Chino M, and Yamori Y 2007. Adenosine induces expression of glial cell line-derived neurotrophic factor (GDNF) in primary rat astrocytes. Neuroscience Research 59:467–474. [DOI] [PubMed] [Google Scholar]
- 89.Rocha SM, Cristovao AC, Campos FL, Fonseca CP, and Baltazar G 2012. Astrocyte-derived GDNF is a potent inhibitor of microglial activation. Neurobiology of Disease 47:407–415. [DOI] [PubMed] [Google Scholar]
- 90.Yan M, Dai H, Ding T, Dai A, Zhang F, Yu L, Chen G, and Chen Z 2011. Effects of dexmedetomidine on the release of glial cell line-derived neurotrophic factor from rat astrocyte cells. Neurochemistry International 58:549–557. [DOI] [PubMed] [Google Scholar]
- 91.Chen SH, Oyarzabal EA, Sung YF, Chu CH, Wang Q, Chen SL, Lu RB, and Hong JS 2015. Microglial regulation of immunological and neuroprotective functions of astroglia. GLIA 63:118–131. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92.Foo L, Allen N, Bushong E, Ventura P, Chung WS, Zhou L, Cahoy J, Daneman R, Zong H, Ellisman M, and Barres B 2011. Development of a Method for the Purification and Culture of Rodent Astrocytes. Neuron 71:799–811. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93.Miyazaki H, Nagashima K, Okuma Y, and Nomura Y 2001. Expression of glial cell line-derived neurotrophic factor induced by transient forebrain ischemia in rats. Brain Research 922:165–172. [DOI] [PubMed] [Google Scholar]
- 94.Hidalgo-Figueroa M.a., Bonilla S, Gutierrez F, Pascual A, and Lopez-Barneo J 2012. GDNF Is Predominantly Expressed in the PV+ Neostriatal Interneuronal Ensemble in Normal Mouse and after Injury of the Nigrostriatal Pathway. The Journal of Neuroscience 32:864–872. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Kuric E, Wieloch T, and Ruscher K 2013. Dopamine receptor activation increases glial cell line-derived neurotrophic factor in experimental stroke. Experimental Neurology 247:202–208. [DOI] [PubMed] [Google Scholar]
- 96.Zhang Y, Chen K, Sloan SA, Bennett ML, Scholze AR, O'Keeffe S, Phatnani HP, Guarnieri P, Caneda C, Ruderisch N, Deng S, Liddelow SA, Zhang C, Daneman R, Maniatis T, Barres BA, and Wu JQ 2014. An RNA-Sequencing Transcriptome and Splicing Database of Glia, Neurons, and Vascular Cells of the Cerebral Cortex. The Journal of Neuroscience 34:11929–11947. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Abe K, and Hayashi T 1997. Expression of the glial cell line-derived neurotrophic factor gene in rat brain after transient MCA occlusion. Brain Research 776:230–234. [DOI] [PubMed] [Google Scholar]
- 98.Kitagawa H, Sasaki C, Zhang WR, Sakai K, Shiro Y, Warita H, Mitsumoto Y, Mori T, and Abe K 1999. Induction of glial cell line-derived neurotrophic factor receptor proteins in cerebral cortex and striatum after permanent middle cerebral artery occlusion in rats. Brain Research 834:190–195. [DOI] [PubMed] [Google Scholar]
- 99.Wei G.w., Wu G.c., and Cao X.d. 2000. Dynamic expression of glial cell lineGÇÉderived neurotrophic factor after cerebral ischemia. Neuroreport 11. [DOI] [PubMed] [Google Scholar]
- 100.Arvidsson A, Kokaia Z, Airaksinen MS, Saarma M, and Lindvall O 2001. Stroke induces widespread changes of gene expression for glial cell line-derived neurotrophic factor family receptors in the adult rat brain. Neuroscience 106:27–41. [DOI] [PubMed] [Google Scholar]
- 101.Bresjanac M, and Antauer G 2000. Reactive Astrocytes of the Quinolinic Acid-Lesioned Rat Striatum Express GFRα1 as Well as GDNF in Vivo. Experimental Neurology 164:53–59. [DOI] [PubMed] [Google Scholar]
- 102.Zhang N, Zhang Z, He R, Li H, and Ding S 2020. GLAST-CreERT2 mediated deletion of GDNF increases brain damage and exacerbates long-term stroke outcomes after focal ischemic stroke in mouse model. GLIA 68:2395–2414. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Nicole O, Ali C, Docagne F, Plawinski L, MacKenzie ET, Vivien D, and Buisson A 2001. Neuroprotection Mediated by Glial Cell Line-Derived Neurotrophic Factor: Involvement of a Reduction of NMDA-Induced Calcium Influx by the Mitogen-Activated Protein Kinase Pathway. J. of Neurosci 21:3024. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Marco S, Canudas AM, Canals JM, Gavaldá N, Perez-Navarro E, and Alberch J 2002. Excitatory Amino Acids Differentially Regulate the Expression of GDNF, Neurturin, and Their Receptors in the Adult Rat Striatum. Experimental Neurology 174:243–252. [DOI] [PubMed] [Google Scholar]
- 105.Widenfalk J, Lundstr+¦mer K, Jubran M, Brene S, and Olson L 2001. Neurotrophic Factors and Receptors in the Immature and Adult Spinal Cord after Mechanical Injury or Kainic Acid. The Journal of Neuroscience 21:3457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Wang Y, Lin SZ, Chiou AL, Williams LR, and Hoffer BJ 1997. Glial Cell Line-Derived Neurotrophic Factor Protects against Ischemia-Induced Injury in the Cerebral Cortex. The Journal of Neuroscience 17:4341–4348. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Kobayashi T, Ahlenius H, Thored P, Kobayashi R, Kokaia Z, and Lindvall O 2006. Intracerebral Infusion of Glial Cell Line-Derived Neurotrophic Factor Promotes Striatal Neurogenesis After Stroke in Adult Rats. Stroke 37:2361–2367. [DOI] [PubMed] [Google Scholar]
- 108.Kitagawa H, Hayashi T, Mitsumoto Y, Koga N, Itoyama Y, and Abe K 1998. Reduction of Ischemic Brain Injury by Topical Application of Glial Cell Line-Derived Neurotrophic Factor After Permanent Middle Cerebral Artery Occlusion in Rats. Stroke 29:1417–1422. [DOI] [PubMed] [Google Scholar]
- 109.Jin G, Omori N, Li F, Nagano I, Manabe Y, Shoji M, and Abe K 2003. Protection against ischemic brain damage by GDNF affecting cell survival and death signals. International Congress Series 1252:221–231. [DOI] [PubMed] [Google Scholar]
- 110.Horita Y, Honmou O, Harada K, Houkin K, Hamada H, and Kocsis JD 2006. Intravenous administration of glial cell line-derived neurotrophic factor gene-modified human mesenchymal stem cells protects against injury in a cerebral ischemia model in the adult rat. J. Neurosci. Res 84:1495–1504. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 111.Shang J, Deguchi K, Yamashita T, Ohta Y, Zhang H, Morimoto N, Liu N, Zhang X, Tian F, Matsuura T, Funakoshi H, Nakamura T, and Abe K 2010. Antiapoptotic and antiautophagic effects of glial cell line-derived neurotrophic factor and hepatocyte growth factor after transient middle cerebral artery occlusion in rats. J. Neurosci. Res 88:2197–2206. [DOI] [PubMed] [Google Scholar]
- 112.Kilic U, Kilic E, Dietz Gunnar PH, and Bahr M 2003. Intravenous TAT-GDNF Is Protective After Focal Cerebral Ischemia in Mice. Stroke 34:1304–1310. [DOI] [PubMed] [Google Scholar]
- 113.Sawada H, Ibi M, Kihara T, Urushitani M, Nakanishi M, Akaike A, and Shimohama S 2000. Neuroprotective Mechanism of Glial Cell Line-Derived Neurotrophic Factor in Mesencephalic Neurons. Journal of Neurochemistry 74:1175–1184. [DOI] [PubMed] [Google Scholar]
- 114.Wong LF, Scott Ralph G, Walmsley LE, Bienemann AS, Parham S, Kingsman SM, Uney JB, and Mazarakis ND 2005. Lentiviral-Mediated Delivery of Bcl-2 or GDNF Protects against Excitotoxicity in the Rat Hippocampus. Molecular Therapy 11:89–95. [DOI] [PubMed] [Google Scholar]
- 115.Ho A, Gore AC, Weickert CS, and Blum M 1995. Glutamate regulation of GDNF gene expression in the striatum and primary striatal astrocytes. Neuroreport 6. [DOI] [PubMed] [Google Scholar]
- 116.Iwata-Ichikawa E, Kondo Y, Miyazaki I, Asanuma M, and Ogawa N 1999. Glial Cells Protect Neurons Against Oxidative Stress via Transcriptional Up-Regulation of the Glutathione Synthesis. Journal of Neurochemistry 72:2334–2344. [DOI] [PubMed] [Google Scholar]
- 117.Endres M, Laufs U, Liao JK, and Moskowitz MA 2004. Targeting eNOS for stroke protection. Trends Neurosci 27:283–289. [DOI] [PubMed] [Google Scholar]
- 118.Lo EH, Dalkara T, and Moskowitz MA 2003. Mechanisms, challenges and opportunities in stroke. Nat Rev Neurosci 4:399–415. [DOI] [PubMed] [Google Scholar]
- 119.Ikeda T, Xia XY, Xia YX, Ikenoue T, Han B, and Choi BH 2000. Glial cell line-derived neurotrophic factor protects against ischemia/hypoxia-induced brain injury in neonatal rat. Acta Neuropathologica 100:161–167. [DOI] [PubMed] [Google Scholar]
- 120.Wang Y, Chang CF, Morales M, Chiang YH, and HOFFER J 2002. Protective Effects of Glial Cell Line-Derived Neurotrophic Factor in Ischemic Brain Injury. Annals of the New York Academy of Sciences 962:423–437. [DOI] [PubMed] [Google Scholar]
- 121.Hwang S, Mruk K, Rahighi S, Raub AG, Chen CH, Dorn LE, Horikoshi N, Wakatsuki S, Chen JK, and Mochly-Rosen D 2018. Correcting glucose-6-phosphate dehydrogenase deficiency with a small-molecule activator. Nature Communications 9:4045. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Mejias R, Villadiego J, Pintado CO, Vime PJ, Gao L, Toledo-Aral JJ, Echevarrea M, and Lopez-Barneo J 2006. Neuroprotection by Transgenic Expression of Glucose-6-Phosphate Dehydrogenase in Dopaminergic Nigrostriatal Neurons of Mice. The Journal of Neuroscience 26:4500. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Kuehne A, Emmert H, Soehle J, Winnefeld M, Fischer F, Wenck H, Gallinat S, Terstegen L, Lucius R, Hildebrand J, and Zamboni N 2015. Acute Activation of Oxidative Pentose Phosphate Pathway as First-Line Response to Oxidative Stress in Human Skin Cells. Molecular Cell 59:359–371. [DOI] [PubMed] [Google Scholar]
- 124.Jeng W, Loniewska MM, and Wells PG 2013. Brain Glucose-6-phosphate Dehydrogenase Protects against Endogenous Oxidative DNA Damage and Neurodegeneration in Aged Mice. ACS Chem. Neurosci 4:1123–1132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125.Sandhu JK, Gardaneh M, Iwasiow R, Lanthier P, Gangaraju S, Ribecco-Lutkiewicz M, Tremblay R, Kiuchi K, and Sikorska M 2009. Astrocyte-secreted GDNF and glutathione antioxidant system protect neurons against 6OHDA cytotoxicity. Neurobiology of Disease 33:405–414. [DOI] [PubMed] [Google Scholar]
- 126.Marchetto MCN, Muotri AR, Mu Y, Smith AM, Cezar GG, and Gage FH 2008. Non-Cell-Autonomous Effect of Human SOD1G37R Astrocytes on Motor Neurons Derived from Human Embryonic Stem Cells. Cell Stem Cell 3:649–657. [DOI] [PubMed] [Google Scholar]
- 127.Yagi T, Jikihara I, Fukumura M, Watabe K, Ohashi T, Eto Y, Hara M, and Maeda M 2000. Rescue of ischemic brain injury by adenoviral gene transfer of glial cell line-derived neurotrophic factor after transient global ischemia in gerbils. Brain Research 885:273–282. [DOI] [PubMed] [Google Scholar]
- 128.Harvey BK, Chang CF, Chiang YH, Bowers WJ, Morales M, Hoffer BJ, Wang Y, and Federoff HJ 2003. HSV amplicon delivery of glial cell line-derived neurotrophic factor is neuroprotective against ischemic injury. Experimental Neurology 183:47–55. [DOI] [PubMed] [Google Scholar]
- 129.Tsai TH, Chen SL, Chiang YH, Lin SZ, Ma HI, Kuo SW, and Tsao YP 2000. Recombinant Adeno-Associated Virus Vector Expressing Glial Cell Line-Derived Neurotrophic Factor Reduces Ischemia-Induced Damage. Experimental Neurology 166:266–275. [DOI] [PubMed] [Google Scholar]
- 130.Arvidsson A, Kirik D, Lundberg C, Mandel RJ, Andsberg G, Kokaia Z, and Lindvall O 2003. Elevated GDNF levels following viral vector-mediated gene transfer can increase neuronal death after stroke in rats. Neurobiology of Disease 14:542–556. [DOI] [PubMed] [Google Scholar]
- 131.Hermann DM, Kilic E, Kugler S, Isenmann S, and Bahr M 2001. Adenovirus-Mediated GDNF and CNTF Pretreatment Protects against Striatal Injury Following Transient Middle Cerebral Artery Occlusion in Mice. Neurobiology of Disease 8:655–666. [DOI] [PubMed] [Google Scholar]
- 132.Xie Y, Wang T, Sun GY, and Ding S 2010. Specific disruption of astrocytic Ca2+ signaling pathway in vivo by adeno-associated viral transduction. Neuroscience 170:992–1003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Jahn HM, Scheller A, and Kirchhoff F 2015. Genetic control of astrocyte function in neural circuits. Frontiers in Cellular Neuroscience 9:310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134.Bardehle S, Kruger M, Buggenthin F, Schwausch J, Ninkovic J, Clevers H, Snippert HJ, Theis FJ, Meyer-Luehmann M, Bechmann I, Dimou L, and Gotz M 2013. Live imaging of astrocyte responses to acute injury reveals selective juxtavascular proliferation. Nature Neuroscience 16:580. [DOI] [PubMed] [Google Scholar]
- 135.Paukert M, Agarwal A, Cha J, Doze V, Kang J, and Bergles D 2014. Norepinephrine Controls Astroglial Responsiveness to Local Circuit Activity. Neuron 82:1263–1270. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.DeCarolis NA, Mechanic M, Petrik D, Carlton A, Ables JL, Malhotra S, Bachoo R, Gotz M, Lagace DC, and Eisch AJ 2013. In vivo contribution of nestin- and GLAST-lineage cells to adult hippocampal neurogenesis. Hippocampus 23:708–719. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Buffo A, Rite I, Tripathi P, Lepier A, Colak D, Horn AP, Mori T, and Gotz M 2008. Origin and progeny of reactive gliosis: A source of multipotent cells in the injured brain. PNAS 105:3581–3586. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Sirko S, Behrendt G, Johansson P, Tripathi P, Costa MR, Bek S, Heinrich C, Tiedt S, Colak D, Dichgans M, Fischer I, Plesnila N, Staufenbiel M, Haass C, Snapyan M, Saghatelyan A, Tsai LH, Fischer A, Grobe K, Dimou L, and Gotz M 2013. Reactive Glia in the Injured Brain Acquire Stem Cell Properties in Response to Sonic Hedgehog. Cell Stem Cell 12:426–439. [DOI] [PubMed] [Google Scholar]
- 139.Robel S, Berninger B, and Gotz M 2011. The stem cell potential of glia: lessons from reactive gliosis. Nat Rev Neurosci 12:88–104. [DOI] [PubMed] [Google Scholar]
- 140.Dimou L.G.t. 2014. Glial Cells as Progenitors and Stem Cells: New Roles in the Healthy and Diseased Brain. Physiol. Rev 94:709–737. [DOI] [PubMed] [Google Scholar]
- 141.Shimada IS, LeComte MD, Granger JC, Quinlan NJ, and Spees JL 2012. Self-Renewal and Differentiation of Reactive Astrocyte-Derived Neural Stem/Progenitor Cells Isolated from the Cortical Peri-Infarct Area after Stroke. The Journal of Neuroscience 32:7926–7940. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.LeComte MD, Shimada IS, Sherwin C, and Spees JL 2015. Notch1-STAT3-ETBR signaling axis controls reactive astrocyte proliferation after brain injury. PNAS 112:8726–8731. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Shimada IS, Borders A, Aronshtam A, and Spees JL 2011. Proliferating Reactive Astrocytes Are Regulated by Notch-1 in the Peri-Infarct Area After Stroke. Stroke 42:3231–3237. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Gotz M, Sirko S, Beckers J, and Irmler M 2015. Reactive astrocytes as neural stem or progenitor cells: In vivo lineage, In vitro potential, and Genome-wide expression analysis. GLIA 63:1452–1468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Ohab JJ, Fleming S, Blesch A, and Carmichael ST 2006. A Neurovascular Niche for Neurogenesis after Stroke. The Journal of Neuroscience 26:13007–13016. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Magnusson JP, Goritz C, Tatarishvili J, Dias DO, Smith EMK, Lindvall O, Kokaia Z, and Frisen J 2014. A latent neurogenic program in astrocytes regulated by Notch signaling in the mouse. Science 346:237–241. [DOI] [PubMed] [Google Scholar]
- 147.Lois C, and Alvarez-Buylla A 1993. Proliferating subventricular zone cells in the adult mammalian forebrain can differentiate into neurons and glia. PNAS 90:2074. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148.Gage FH, Kempermann G, Palmer TD, Peterson DA, and Ray J 1998. Multipotent progenitor cells in the adult dentate gyrus. J. Neurobiol 36:249–266. [DOI] [PubMed] [Google Scholar]
- 149.Tobin MK, Bonds JA, Minshall RD, Pelligrino DA, Testai FD, and Lazarov O 2014. Neurogenesis and inflammation after ischemic stroke: what is known and where we go from here. J Cereb Blood Flow Metab 34:1573–1584. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Young CC, van der Harg JM, Lewis NJ, Brooks KJ, Buchan AM, and Szele FG 2013. Ependymal Ciliary Dysfunction and Reactive Astrocytosis in a Reorganized Subventricular Zone after Stroke. cercor 23:647–659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.Lagace DC 2012. Does the endogenous neurogenic response alter behavioral recovery following stroke? Behavioural Brain Research 227:426–432. [DOI] [PubMed] [Google Scholar]
- 152.Smith MP, and Cass WA 2007. GDNF reduces oxidative stress in a 6-hydroxydopamine model of Parkinson's disease. Neuroscience Letters 412:259–263. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Ronning OM, and Guldvog B 1998. Outcome of Subacute Stroke Rehabilitation: A Randomized Controlled Trial. Stroke 29:779–784. [DOI] [PubMed] [Google Scholar]
- 154.Poulin V.r., Korner-Bitensky N, Bherer L, Lussier M, and Dawson DR 2016. Comparison of two cognitive interventions for adults experiencing executive dysfunction post-stroke: a pilot study. Disability and Rehabilitation 39:1–13. [DOI] [PubMed] [Google Scholar]
- 155.Kramer ER, and Liss B 2015. GDNF-Ret signaling in midbrain dopaminergic neurons and its implication for Parkinson disease. FEBS Letters 589:3760–3772. [DOI] [PubMed] [Google Scholar]



