Abstract
Cerebral ischemia is defined as little or no blood flow in cerebral circulation, characterized by low tissue oxygen and glucose levels, which promotes neuronal mitochondria dysfunction leading to cell death. A strategy to counteract cerebral ischemia-induced neuronal cell death is ischemic preconditioning (IPC). IPC results in neuroprotection, which is conferred by a mild ischemic challenge prior to a normally lethal ischemic insult. Although many IPC-induced mechanisms have been described, many cellular and subcellular mechanisms remain undefined. Some reports have suggested key signal transduction pathways of IPC, such as activation of protein kinase C epsilon, mitogen-activated protein kinase, and hypoxia-inducible factors, that are likely involved in IPC-induced mitochondria mediated-neuroprotection. Moreover, recent findings suggest that signal transducers and activators of transcription (STATs), a family of transcription factors involved in many cellular activities, may be intimately involved in IPC-induced ischemic tolerance. In this review, we explore current signal transduction pathways involved in IPC-induced mitochondria mediated-neuroprotection, STAT activation in the mitochondria as it relates to IPC, and functional significance of STATs in cerebral ischemia. Antioxid. Redox Signal. 14, 1853–1861.
Introduction
Cerebral ischemia is defined as little or no blood flow in cerebral circulation, characterized by low tissue oxygen and glucose levels, and by the accumulation of metabolic products (25). Due to high energy demands, the brain and the heart are most vulnerable during ischemia. Energy consumption is highest in the brain due to its innate physiological activities, and as a consequence, energy failure has severe consequences in the brain, such as loss of electrical activity, depletion of high energy intermediates (43), and loss of ion gradients (21), which results in the release of excitatory neurotransmitters (i.e., glutamate) causing calcium excitotoxicity and irreversible pathologies (18). Global cerebral ischemia affects whole-brain vascular dynamics, promotes neuronal cell death in many brain regions including the hippocampus (12).
It is well accepted that reperfusion after cerebral ischemia causes increased ischemic injury characterized by two phases: rapid hyperemia (increased blood flow) and delayed hypoperfusion (decreased blood flow) (77). The hyperemia phase leads to ischemia-induced cell death to different areas of the brain (87) as well as decreased blood flow (hypoperfusion) (5) resulting in yet another possible ischemic/hypoxic condition. Upon reperfusion, hyperemia and subsequent hypoperfusion of cerebral blood vessels (25) leads to enhanced superoxide generation (87).
Of the complicated cellular processes that occur during and after cerebral ischemia, dysfunction of the mitochondria is well accepted to play a central role in ischemic injury (15). Mitochondria are not only affected during the ischemic insult where they are deprived of substrates and oxygen, but also in the post-ischemic state where changes in redox activity of the respiratory chain components occur as represented by hyperoxidation of electron carriers (59) facilitating enhanced reactive oxygen species (ROS) (50) generation; all have been linked to reperfusion following cerebral ischemia. Consequently, hyperoxidation may result in release of cytochrome c from the mitochondria initiating the apoptotic cascade (6). Additional evidence of mitochondrial dysfunction was described in studies obtained from isolated brain mitochondria exhibiting decreased state 3 respiratory rates of ∼ 70% nicotinamide adenine dinucleotide (NAD)-linked respiratory substrates (66). Moreover, nonsynaptosomal mitochondria were insensitive to ischemia, but became dysfunctional in the late reperfusion phase (4). Mitochondria from synaptic terminals were greatly affected by ischemia, but partially recovered during reperfusion. In addition, in a rat model of forebrain transient ischemia, the rate of oxygen consumption decreased in the CA1, CA3, and CA4 regions of the hippocampus in the late reperfusion phase (67). This study was performed in brain homogenates from different brain subregions (52).
Many therapeutic approaches against cerebral ischemia emerged from understanding these pathways that lead to mitochondrial dysfunction. However, up to this point, not much success has been achieved. A different approach in the field of cerebral ischemia has emerged. It is now believed that by understanding endogenous metabolic adaptations that make sensitive organs like the heart and brain highly resistant to ischemia, we will be able to provide more effective therapies. One such adaptation is termed ‘ischemic tolerance’ or ‘ischemic preconditioning’.
Ischemic Preconditioning
Many molecular and cellular signaling factors modulate neuronal homeostasis in the ischemic brain. Signaling molecules such as protein kinase B (Akt) and C are involved in neuroprotection by modulating apoptotic factors such as B cell lymphoma-2-associated death promoter (BAD), caspases, and p53 among others (3). Other factors such as mitogen-activated protein kinases (MAPKs), hypoxia-inducible factor 1α (HIF-1α), and signal transducers and activator of transcription (STATs) are also considered to be involved in mitochondria dysfunction (14). It is the influence of the mitochondria in the production of peroxide free radicals and enhanced levels of calcium that causes the activation of downstream effectors to induce cellular apoptosis/necrosis.
Resistance to these detrimental effects of neuronal cell death (acute or delayed) after ischemia has emerged from a widely accepted concept, named ischemic preconditioning (IPC), which is an innate neuroprotective mechanism whereby a sublethal ischemic insult protects against a subsequent lethal ischemic attack. First described in the heart by Murry and in brain slices by Schurr nearly 23 years ago (47, 64), IPC has been demonstrated in numerous tissues and species, including humans (78). IPC is characterized by an early or “classical” window of protection, which is sustained for several hours followed by a second window of protection 24–48 hours later, which can last for several days. The early window of protection is mediated by alteration in protein function whereas the delayed window of protection is mediated by changes in gene expression.
The therapeutic potential of IPC has led to numerous investigations into the subcellular mediators and effector pathways activated by the preconditioning stimuli. Some mediators of preconditioning include neuroactive cytokines (40), glutamate (17), γ-aminobutyric acid (13), adenosine (88), adenosine triphosphate (ATP)-sensitive potassium (K+-ATP) channels (51), and hypoxia (41). Many effector signaling pathways have been demonstrated to induce ischemic tolerance. However, it is unlikely that one pathway alone is sufficient to induce a preconditioned response. Instead, ischemic tolerance is likely the result of a culmination of multiple signaling pathways acting in concert to target many of the pathophysiological mechanisms leading to ischemia-induced cell death.
Signaling Pathways Activated in Ischemic Preconditioning
The effector pathways activated by preconditioning typically result in the activation of protein kinase cascades such as protein kinases C (PKC), Akt, and the MAPK signaling cascade, including extracellular signal-regulated kinases (ERK), c-Jun N-terminal protein kinases (JNK), and p38 (Fig. 1). Akt is a serine/threonine kinase implicated in many cellular survival pathways. Akt exerts neuroprotection by phosphorylation and inactivation of many pro-apoptotic proteins including BAD, glycogen synthase kinase 3 beta (GSK3β), caspase 9, and apoptosis signal-regulating kinase 1. Akt is activated during ischemia-reperfusion in both preconditioned and nonconditioned animals. However, the duration of Akt activation is significantly extended by preconditioning. In fact, Akt activation in preconditioned animals reduced apoptosis within the penumbral region reducing infarct expansion (48). In rat cerebellar granule neurons, hypoxic preconditioning increases Akt phosphorylation as well as the expression of vascular endothelial growth factor (VEGF) and its receptor VEGF receptor-2 (Flk-1), which is required for Akt phosphorylation. Inhibition of VEGF, Flk-1, or Akt was sufficient to prevent hypoxia preconditioning (80).
FIG. 1.
Schematic diagram of the effector signaling pathways activated during ischemia preconditioning. Multiple signaling pathways are activated by ischemia preconditioning (IPC) that target different pathological features of the ischemic brain, leading to neuroprotection. Ischemia exposure allows for the accumulation of HIF-1α and the transcriptional activation of hypoxia regulated genes such as erythropoietin, inducible nitric oxide synthase, glycolytic proteins, and vascular endothelial growth factor (VEGF). Activation of the phosphoinositide 3 kinase (PI3K)-Akt pathway acts as a negative regulator of many pro-apoptotic proteins such as GSK3(and BAD; thereby maintaining the mitochondrial permeability transition pore (mPTP) in a closed configuration. Epsilon PKC (ɛPKC) phosphorylation is associated with the opening of the mitochondrial ATP sensitive potassium channels (mtK+-ATP), which provides mitochondrial protection. Epsilon PKC activates the RAS–RAF–MEK–ERK1/2 signaling pathway, leading to STAT3 phosphorylation and nuclear translocation through receptor tyrosine kinase activation (RTK). In the nucleus, STAT3 enhances COX-2 expression, leading to neuroprotection. Recent research suggests that STATs may also play a role in mitochondrial bioenergetics, suggesting a possible role of mitochondrial localized STATs in IPC-mediated neuroprotection.
HIF-1α is a hypoxia-regulated transcription factor that controls the expression of many hypoxia associated genes. HIF-1α is rapidly degraded by the ubiquitin-proteasome pathway so that little to no HIF-1α protein is present under normal physiological oxygen concentrations. However, during hypoxia, HIF-1α protein degradation is halted, allowing for protein accumulation and nuclear translocation. In the nucleus, HIF-1α forms a heterodimer with HIF-1β, which allows for the binding of the activated transcription factor to promoter elements in hypoxia- regulated target genes. Some of the target genes regulated by HIF-1α include VEGF, erythropoietin, inducible nitric oxide synthase (iNOS), glucose transporter-1, a variety of glycolytic proteins, and other proteins that provide neuroprotection. Since HIF-1α plays a pivotal role in the cellular response to oxygen deprivation, it is not surprising that HIF-1α has been implicated in ischemic tolerance. For example, neuronal-specific inactivation of HIF-1α resulted in increased focal cerebral ischemia-induced cell death (73). Furthermore, expression of HIF-1α and its target genes have been demonstrated in numerous models of ischemia and hypoxic preconditioning (28, 55).
STATs are a family of transcription factors involved in many cellular activities. The transcriptional activity of STATs is regulated by phosphorylation-dependent dimerization that allows for DNA binding and recruitment of transcriptional coactivators. STAT-mediated ischemic tolerance in the heart and brain are well characterized. Our laboratory has demonstrated phosphorylation and nuclear translocation of STAT3 in response to preconditioning by oxygen and glucose deprivation (OGD) in mixed cortical/glial co-cultures (32) (Fig. 2). STAT3 was found to exert neuroprotection through the transcriptional upregulation of cyclooxygenase-2 (COX-2). Inhibition of STAT3 activity with the STAT3 inhibitory peptide (PpYLKTK) prevented COX-2 expression and neuroprotection (32). We have recently demonstrated that COX-2 expression during IPC is also regulated by nuclear factor kappa B (NF-κB). Similar to STAT3, NF-κB was found to translocate to the nucleus in response to preconditioning and was found to regulate COX-2 expression leading to neuroprotection (33).
FIG. 2.
Epsilon PKC and ERK1/2 are essential for IPC-mediated neuroprotection. (A) Cell death was determined by propidium iodide (PI) staining in organotypic slices exposed to 40 min of oxygen and glucose deprivation (ischemia) alone or 48 h after IPC (15 min of oxygen and glucose deprivation) (modified from Ref. 56). The neuroprotection provided by IPC is lost when slices were exposed to the ɛPKC antagonist, ɛV1-2. Similarly pharmacological preconditioning with the ɛPKC agonist, ψɛRACK, increases cell survival during oxygen and glucose deprivation. (B) The role of ERK1/2 in the preconditioning response was determined by treating organotypic slices with the MEK1/2 inhibitor PD-98059 during IPC (modified from Ref. 38). Inhibition of ERK1/2 activation significantly increased cell death when compared to IPC vehicle-treated control groups. These results indicate that ERK1/2 is activated by IPC and contributes to ischemic tolerance.
The MAPK cascade is comprised of three protein kinases consisting of MAPK, MAPK kinase (MEK), and MAPK kinase kinase (MEKK) and results in the activation of ERK, p38, or JNK. There is increasing evidence that IPC regulates the activity of the MAPK family. For example, our laboratory has shown that preconditioning by OGD induces ERK1/2-dependent neuroprotection in cortical–glial co-cultures (32) (Fig. 2). Similarly, Choi et al. demonstrated ERK1/2 activation by IPC in the CA1 region of the rat hippocampus (10). A neuroprotective role for ERK1/2 in preconditioning has been demonstrated by numerous pharmacological studies using ERK1/2-specific inhibitors (20). In contrast to ERK1/2, JNK activation is associated with cellular apoptosis during cerebral ischemia (26). Interestingly, IPC can prevent JNK activation via ERK1/2 by mechanisms not fully understood (86).
The PKC family of serine/threonine kinases is comprised of 11 isoforms which are classified as either conventional, novel, or atypical, based upon their requirement for calcium and/or lipids for activation. Recent research in our laboratory and others has demonstrated a vital role for epsilon PKC (ɛPKC) in neuroprotection during IPC (46, 56). The neuroprotection initiated by IPC is lost in the presence of ɛPKC inhibitors and can be simulated with activators of ɛPKC (56) (Fig. 2). A vital role of ɛPKC in preconditioning is especially demonstrated in ɛPKC knockout mice, which failed to exhibit an IPC response (61). It is thought that ePKC serves as a central signaling protein in many preconditioning models.
Many of the effector pathways discussed above confers protection in the mitochondria. Zhang et al. demonstrated that IPC prevented mitochondrial swelling and preserved membrane integrity and metabolism during middle cerebral artery occlusion (MCAO) in rats (85). Our laboratory has demonstrated that preconditioning only preserves mitochondrial function in the delayed, but not the early window of protection (52). The mechanism by which these effector pathways protect the mitochondria are not fully understood, but is thought to involve inhibiting the opening of the mitochondrial permeability transition pore (mPTP) via opening of ATP-sensitive potassium channels and by upregulating mitochondrial uncoupling proteins. Recently, it was shown that STAT3 serine phosphorylation induces mitochondrial translocation and enhanced mitochondrial oxidative phosphorylation (57). Therefore, STAT3 may provide ischemic tolerance by altering mitochondrial function. In the next sections, we will examine the role of STATs on mitochondria and cell death.
Overview of STATs
STATs are a family of transcription factors that modulates diverse biological activities throughout the body. STATs consist of seven family members (STAT1–4, STAT5a, STAT5b, and STAT6) (62) which can be activated by cytokines/growth factors, free radicals, neurotransmitters, and inflammatory mediators in the event of injury (1). STAT1, STAT3, and STAT5 have been shown to be activated in the event of cellular injury; therefore, these factors will be the focus of discussion (53, 74, 84). The mechanisms of STAT activation have been well characterized. Growth factor or cytokine receptors are present on cell surface membranes to serve as signaling molecules to the cell producing a variety of functions (i.e., immune modulation, inflammation, cellular repair from injury). Janus kinase (JAK) (a tyrosine kinase), located in the cytosol, is closely linked to these growth factor/cytokine receptors. Upon ligand binding to its respective receptor, activated JAK kinases phosphorylate tyrosine residues on the cytosolic portion of the receptor complex. STATs, which are inactive in the cytosol, dock with the receptor-phosphorylated tyrosine residues, are then phosphorylated by JAKs, resulting in STAT activation. STAT activation can also be derived from other receptor tyrosine kinase receptors and Src family of kinases (63). Upon activation, STATs translocate to the nucleus to induce gene transcription resulting in cell survival or cell degeneration pathways (71) (summarized in Fig. 3). In this review, we will discuss the beneficial role of STAT3, STAT5, and STAT6 as anti-apoptotic transcription factors as it relates to cerebral ischemia, while STAT1 promotes pro-apoptotic events thought to be detrimental to the brain post-ischemia.
FIG. 3.
Schematic diagram of general STAT activation pathway leading to neuroprotection.
The Role of STATs on the Mitochondria
A major mechanism by which IPC confers neuroprotection is through the modification of mitochondrial properties. Previous studies have shown that while the JAK/STAT pathways are involved in mediating ischemia/reperfusion injury and apoptosis (45), this pathway is also involved in IPC-mediated mitochondrial protection. In studies of mice hearts, IPC induced nuclear translocation and activation of STAT1 and STAT3 (81), which have been shown to have opposing roles in cell survival. STAT1 activation leads to pro-apoptotic signaling production of ROS (8), and loss of the mitochondrial membrane potential (39). STAT1, along with STAT6 signaling, are involved in the upregulation of adenine nucleotide translocator 3 (27), a mitochondrial protein that exchanges mitochondrial ATP for cytosolic adenosine diphosphate and is linked to apoptosis (44). In contrast, STAT3 inhibits the activity and transcription of STAT1 (24) and opposes STAT1-mediated apoptosis (71). Furthermore, STAT3 regulates mitochondrial function in a neuroprotective manner (60) and is required for the protective mechanisms of IPC (68).
STAT3 modulation of mitochondrial function and metabolism may explain its requirement for IPC-mediated protection (68). When STAT3 is knocked-out or downregulated, impairments in complexes of the electron transport chain are observed (summarized in Fig. 4). For example, lower rates of oxygen consumption in complexes I and II of heart mitochondria were detected in STAT3-/- mice (79). Ras-transformed STAT3-deficient mouse embryonic fibroblasts exhibited decreased complex II and V activity and lowered cellular ATP concentrations (16). Deletion of STAT3 in astrocytes led to similar results, but also caused increased ROS production, enhanced depolarization of the mitochondrial membrane potential, and led to reductions in mitochondrial mass. STAT3 appears to transcriptionally upregulate the activity of manganese superoxide dismutase (MnSOD) (29), an antioxidant that removes free radicals and deletion of STAT3 leads to increased production of ROS (60). Reconstitution of STAT3 in deficient cells reverses some of the mitochondrial impairments on complex activity and respiration that can be mediated by its deletion or downregulation (54, 79). The pervasive mitochondrial dysfunction that results from loss of STAT3 may partially explain the significance of STAT3-mediated neuroprotection during IPC (Fig. 4).
FIG. 4.
STAT3 inhibition leads to mitochondrial dysfunction in the electron transport chain. Inhibition of STAT3 downregulates expression of free radical scavenger MnSOD1 (29), increases levels of ROS2 (60), decreases complex I & II oxygen consumption rates3 (79), decreases activity of complex V4 (16), and reduces cellular ATP levels5 (60).
Recent studies have shown the presence of STAT3 in the mitochondria in a number of tissues including the heart, liver, brain, and kidney (16, 79). Immunoprecipitates of complex I from liver mitochondrial extracts were found to contain STAT3, which suggests a direct interaction of STAT3 with specific proteins on complex I (79). Furthermore, Wegrzyn et al. also found that mitochondria isolated from STAT3 deficient pro-B cells had reduced maximal state 3 oxidation rates, which suggests mitochondria-localized STAT3 plays a major role in modulating mitochondrial respiration. Therefore, STAT3 may have the ability to regulate mitochondrial function independent of its already well-defined role as a transcription factor. However, the functional role of STAT3 as a mitochondrial-mediator and transcription factor as it relates to IPC remains unclear.
STATs and Apoptosis
STAT1 is activated by cytokines, growth factors, and hormones (74). STAT1 has been shown to play a dedicated role in interferon-γ (35) and physiological responses by inducing cyclin-dependent kinase inhibitor (p21WAF1) to promote cell growth arrest (7). STAT1 activation is prevalent in neurons in the cortex and striatum following ischemic injury (53, 74). It is expressed normally in the adult rat brain and is induced after focal ischemia in the ipsilateral cortex up to 15 days post-ischemia (53). Several reports have suggested that STAT1 regulates apoptosis in cardiomyocytes and fibroblasts up-regulating pro-apoptotic factors such as caspases (8). In the heart, STAT1 decreases anti-apoptotic gene expression of B-cell lymphoma-2 (Bcl-2) and B-cell lymphoma-2-associated X protein (Bcl-x) in ischemia/reperfusion injury (71). Only a few reports have suggested the involvement of STAT1 in cerebral ischemia-induced cell death. In MCAO mice, STAT1 phosphorylation and nuclear translocation is enhanced, promoting apoptosis and cell death by enhanced Akt phosphorylation and caspase-3 activation (8, 74). Likewise, STAT1-/- mice are more resistant to ischemic brain injury (74), suggesting a neurodegenerative role for STAT1 during cerebral ischemia. In contrast, others reported that upregulation of STAT1 may be anti-apoptotic in other instances. For example, in human glioblastoma patients, STAT-1 expression was present via immunohistochemistry suggesting other downstream mediators regulating apoptosis (22). Pathological diseases such as infections or prion diseases also led to activation of STATs (70). In cerebellar granule neurons exposed to Clostridium difficile toxin B, upregulation of the JAK/STAT1 pathway revealed that it is pro-apoptotic in nature (42). Nevertheless, this paradigm may be beneficial in regulating cancer cells, although STAT1's involvement in neuroprotection after cerebral ischemia may be detrimental to critical neuronal function in the brain.
The role of STAT3 has also been well defined in the cortex, striatum, and hippocampus (CA1 region). STAT3 activation was found in numerous cell types in the brain including neurons, astrocytes, endothelial cells, microglia, and monocytes/macrophages (9, 31, 34, 72), impacting neuronal development (23), neuroprotection after ischemic injury (72), and regeneration after nerve injury (65). Major cellular functions such as cell proliferation, survival, and development are also regulated by STAT3 (11). Contrary to STAT1, in the MCAO model of transient focal ischemia, phosphorylated-STAT3 was observed to be co-localized with Bcl-2 (36) and not with cleaved caspase-3 (82), suggesting STAT3 is important in neuronal survival (Fig. 5). STAT3 activation has also been found in glia, which may provide indirect support to neuronal survival (69). For example, the presence of hypoxic preconditioning in cultured cortical neurons induced neuroprotection mediated by the STAT3 signaling pathway (76). Similarly, activation of STAT3-induced superoxide dismutase 2 gene expression resulted in neuroprotection (30).
FIG. 5.
Opposing effects of STAT1 and STAT3 during ischemia. Upon activation of STAT1 or STAT3 by ischemia or brain injury, Bcl-2/Bcl-x gene expression is decreased in the presence of STAT1 but enhanced in STAT3 activation. This results in decreased cell survival (pro-apoptosis) by STAT1 activation, while cell survival is enhanced via activation of STAT3 (anti-apoptosis).
Unlike STAT1, STAT3 and STAT5 induced Bcl-2 and Bcl-x gene expression resulting in anti-apoptotic events in the cell (71, 84). The opposing actions of STAT1 (pro-apoptotic) and STAT3 (anti-apoptotic) provide a balance for cellular homeostasis. Pathologically, upregulation of STAT3 after cerebral ischemia may prove beneficial by providing some degree of neuroprotection, while in cancer cells, STAT3 activation has been found to be enhanced for increased tumor growth (19). In contrast to the neuroprotection observed after cerebral ischemia and tumor cells, STAT3 mediated beta-amyloid-induced neuronal cell death in the cortex and hippocampus of mice (75). These results suggested that STAT3 activation is pro-apoptotic contrary to other reports. Thus, the general anti-apoptotic properties of STAT3 must be interpreted with caution. The role of STAT3 as an anti- or pro-apoptotic signaling pathway may depend on cell type and pathological condition and warrants further investigation.
STATs and IPC
The role of STATs and IPC has predominately been demonstrated in the heart but not in the brain, except for Kim et al, (32). For example, STAT3 activation via JAK was cardioprotective in the infarcted-reperfused heart by inducing expression of Bcl-2 or Bcl-x (49). STAT3 was overexpressed in transgenic hearts of mice, providing protection from oxidative stress via doxorubicin-induced cardiomyopathy (37). Other reports have suggested different STATs (STAT5A) are involved in preconditioning of the heart via Src kinases and JAK signaling pathways (83).
STAT activation and function as it relates to IPC have not been well defined. In fact, current literature focuses predominately on the heart. IPC enhances STAT3 expression in astrocytes in the hippocampus (34). We demonstrated that in mixed cortical neurons/astrocyte cultures, the IPC signaling cascade involves STAT3 activation mediating ischemic tolerance in vitro (32) (Fig. 6). In addition, 6 hours post-OGD, JAK-STAT signaling was detected in adult rat hippocampal slices (2) concurrent with our previous findings (32).
FIG. 6.
STAT3 activation is required for IPC-mediated neuroprotection. STAT3 serine 727 phosphorylation was determined in cells lysed immediately after 1 h of PC and at 15 min, 30 min, and 2 h of reperfusion after 1 h of PC (A). These results suggest that STAT3 is activated by IPC. (B) Histogram representing cell death measured by LDH release at 48 h of reperfusion after oxygen and glucose deprivation (OGD) (4 h) injury. Inhibition of STAT3 activity with the STAT3 inhibitory peptide (PpYLKTK, 1 and 5 mol/L) blocked IPC neuroprotection. These results indicate that STAT3 is activated by IPC and is required for neuroprotection during ischemia (modified from Ref. 32).
We hypothesize that more subtypes of STATs are involved in IPC-induced tolerance that will confer neuroprotection in the brain after stroke, cardiac arrest, or general cerebral ischemic events that can occur in the body. Besides neuroprotection, little is known regarding STATs, as it relates to cerebral blood flow (CBF) in the presence of CBF-related deficits (stroke, cerebral ischemia, etc). Since STATs also affect brain parenchyma, it is plausible that STATs can affect vascular-associated cells, providing not only neuroprotection, but proper CBF in the presence of pathology (58).
Conclusion
STATs modulate diverse biological activities throughout the body. A variety of signal transduction pathways that are activated by IPC, such as MAPK and ɛPKC, have been demonstrated to activate STAT3 and lead to ischemic tolerance. Moreover, STAT activation affects not only signal transduction mechanisms, but also mitochondrial bioenergetics. STAT1 activity depolarizes mitochondrial membrane, leads to the production of ROS, and is linked to apoptosis. STAT3 regulates mitochondria in a neuroprotective manner by preserving efficiency of electron transport chain complexes and maintaining cellular ATP levels. STAT3-mediated neuroprotection during IPC is well characterized in cardiac models, but few reports have described this phenomenon in the brain. STAT3 activation enhances Bcl-2 and Bcl-x gene expression, resulting in anti-apoptotic events in the cell, while STAT1 decreases these genes promoting pro-apoptotic situations. The opposing affects between STAT1 and STAT3 and in particular, the role of mitochondrial localized STATs needs further clarification in the ischemic preconditioned brain. Therefore, defining the mechanisms of STAT activation via IPC can prove very beneficial in providing a new dimension of neuroprotection in the brain against ischemic challenges.
Abbreviations Used
- Akt
protein kinase B
- ATP
adenosine triphosphate
- BAD
B cell lymphoma-2-associated death promoter
- Bcl-2
B-cell lymphoma-2
- Bcl-x
B-cell lymphoma-2-associated X protein
- CBF
cerebral blood flow
- Cox-2
cyclooxygenase-2
- ɛPKC
epsilon PKC
- ERK
extracellular signal-regulated kinases
- Flk-1
VEGF receptor-2
- GSK3β
glycogen synthase kinase 3 beta
- HIF-1α
hypoxia-inducible factor 1α
- iNOS
inducible nitric oxide synthase
- IPC
ischemic preconditioning
- JAK
janus kinase
- JNK
c-Jun N-terminal protein kinases
- MAPKs
mitogen-activated protein kinases
- MCAO
middle cerebral artery occlusion
- MEK
MAPK kinase
- MEKK
MAPK kinase kinase
- MnSOD
manganese superoxide dismutase
- mPTP
mitochondrial permeability transition pore
- NAD
nicotinamide adenine dinucleotide
- NF-κB
nuclear factor kappa B
- OGD
oxygen and glucose deprivation
- PKC
protein kinases C
- ROS
reactive oxygen species
- RTK
receptor tyrosine kinase
- STATs
signal transducers and activators of transcription
- VEGF
vascular endothelial growth factor
Acknowledgments
This work was supported by National Institutes of Health Grants NS45676-01, NS054147-01, NS34773, T32-NS007459-10, and AHA-Philips 10POST4340011.
References
- 1.Ahn YH. Lee G. Kang SK. Molecular insights of the injured lesions of rat spinal cords: Inflammation, apoptosis, and cell survival. Biochem Biophys Res Commun. 2006;348:560–570. doi: 10.1016/j.bbrc.2006.07.105. [DOI] [PubMed] [Google Scholar]
- 2.Benardete EA. Bergold PJ. Genomic analysis of ischemic preconditioning in adult rat hippocampal slice cultures. Brain Res. 2009;1292:107–122. doi: 10.1016/j.brainres.2009.07.027. [DOI] [PubMed] [Google Scholar]
- 3.Busija DW. Gaspar T. Domoki F. Katakam PV. Bari F. Mitochondrial-mediated suppression of ROS production upon exposure of neurons to lethal stress: Mitochondrial targeted preconditioning. Adv Drug Deliv Rev. 2008;60:1471–1477. doi: 10.1016/j.addr.2008.03.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Cafè C. Torri C. Gatti S. Adinolfi D. Gaetani P. Rodriguez Y. Baena R. Marzatico F. Changes in non-synaptosomal and synaptosomal mitochondrial membrane-linked enzymatic activities after transient cerebral ischemia. Neurochem Res. 1994;19:1551–1555. doi: 10.1007/BF00969005. [DOI] [PubMed] [Google Scholar]
- 5.Caplan LR. Wong KS. Gao S. Hennerici MG. Is hypoperfusion an important cause of strokes? If so, how? Cerebrovasc Dis. 2006;21:145–153. doi: 10.1159/000090791. [DOI] [PubMed] [Google Scholar]
- 6.Charriaut–Marlangue C. Margaill I. Represa A. Popovici T. Plotkine M. Ben–Ari Y. Apoptosis and necrosis after reversible focal ischemia: An in situ DNA fragmentation analysis. J Cereb Blood Flow Metab. 1996;16:186–194. doi: 10.1097/00004647-199603000-00002. [DOI] [PubMed] [Google Scholar]
- 7.Chin YE. Kitagawa M. Su WC. You ZH. Iwamoto Y. Fu XY. Cell growth arrest and induction of cyclin-dependent kinase inhibitor p21 WAF1/CIP1 mediated by STAT1. Science. 1996;272:719–722. doi: 10.1126/science.272.5262.719. [DOI] [PubMed] [Google Scholar]
- 8.Chin YE. Kitagawa M. Kuida K. Flavell RA. Fu XY. Activation of the STAT signaling pathway can cause expression of caspase 1 and apoptosis. Mol Cell Biol. 1997;9:5328–5337. doi: 10.1128/mcb.17.9.5328. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Choi JS. Kim SY. Cha JH. Choi YS. Sung KW. Oh ST. Kim ON. Chung JW. Chun MH. Lee SB. Lee MY. Upregulation of gp130 and STAT3 activation in the rat hippocampus following transient forebrain ischemia. Glia. 2003;41:237–246. doi: 10.1002/glia.10186. [DOI] [PubMed] [Google Scholar]
- 10.Choi JS. Kim HY. Cha JH. Lee MY. Ischemic preconditioning-induced activation of ERK1/2 in the rat hippocampus. Neurosci Lett. 2006;409:187–191. doi: 10.1016/j.neulet.2006.09.053. [DOI] [PubMed] [Google Scholar]
- 11.Darnell JE., Jr STATs and gene regulation. Science. 1997;277:1630–1635. doi: 10.1126/science.277.5332.1630. [DOI] [PubMed] [Google Scholar]
- 12.Dave KR. Saul I. Prado R. Busto R. Perez–Pinzon MA. Remote organ ischemic preconditioning protect brain from ischemic damage following asphyxial cardiac arrest. Neurosci Lett. 2006;404:170–175. doi: 10.1016/j.neulet.2006.05.037. [DOI] [PubMed] [Google Scholar]
- 13.DeFazio RA. Raval AP. Lin HW. Dave KR. Della–Morte D. Perez–Pinzon MA. GABA synapses mediate neuroprotection after ischemic and epsilonPKC preconditioning in rat hippocampal slice cultures. J Cereb Blood Flow Metab. 2009;2:375–384. doi: 10.1038/jcbfm.2008.126. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Dirnagl U. Meisel A. Endogenous neuroprotection: Mitochondria as gateways to cerebral preconditioning? Neuropharmacology. 2008;55:334–344. doi: 10.1016/j.neuropharm.2008.02.017. [DOI] [PubMed] [Google Scholar]
- 15.Fiskum G. Murphy AN. Beal MF. Mitochondria in neurodegeneration: Acute ischemia and chronic neurodegenerative diseases. J Cereb Blood Flow Metab. 1999;19:351–369. doi: 10.1097/00004647-199904000-00001. [DOI] [PubMed] [Google Scholar]
- 16.Gough DJ. Corlett A. Schlessinger K. Wegrzyn J. Larner AC. Levy DE. Mitochondrial STAT3 supports Ras-dependent oncogenic transformation. Science. 2009;324:1713–1716. doi: 10.1126/science.1171721. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Grabb MC. Choi DW. Ischemic tolerance in murine cortical cell culture: critical role for NMDA receptors. J Neurosci. 1999;19:1657–1662. doi: 10.1523/JNEUROSCI.19-05-01657.1999. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Greenamyre JT. The role of glutamate in neurotransmission and in neurologic disease. Arch Neurol. 1986;43:1058–1063. doi: 10.1001/archneur.1986.00520100062016. [DOI] [PubMed] [Google Scholar]
- 19.Gritsko T. Williams A. Turkson J. Kaneko S. Bowman T. Huang M. Nam S. Eweis I. Diaz N. Sullivan D. Yoder S. Enkemann S. Eschrich S. Lee JH. Beam CA. Cheng J. Minton S. Muro–Cacho CA. Jove R. Persistent activation of stat3 signaling induces survivin gene expression and confers resistance to apoptosis in human breast cancer cells. Clin Cancer Res. 2006;12:11–19. doi: 10.1158/1078-0432.CCR-04-1752. [DOI] [PubMed] [Google Scholar]
- 20.Gu Z. Jiang Q. Zhang G. Extracellular signal-regulated kinase and c-Jun N-terminal protein kinase in ischemic tolerance. Neuroreport. 2001;12:3487–3491. doi: 10.1097/00001756-200111160-00023. [DOI] [PubMed] [Google Scholar]
- 21.Hansen AJ. Effect of anoxia on ion distribution in the brain. Physiol Rev. 1985;65:101–148. doi: 10.1152/physrev.1985.65.1.101. [DOI] [PubMed] [Google Scholar]
- 22.Haybaeck J. Obrist P. Schindler CU. Spizzo G. Doppler W. STAT-1 expression in human glioblastoma and peritumoral tissue. Anticancer Res. 2007;27:3829–3835. [PubMed] [Google Scholar]
- 23.He JC. Gomes I. Nguyen T. Jayaram G. Ram PT. Devi LA. Iyengar R. The G alpha(o/i)-coupled cannabinoid receptor-mediated neurite outgrowth involves Rap regulation of Src and Stat3. J Biol Chem. 2005;280:33426–33434. doi: 10.1074/jbc.M502812200. [DOI] [PubMed] [Google Scholar]
- 24.Ho HH. Ivashkiv LB. Role of STAT3 in type I interferon responses. Negative regulation of STAT1-dependent inflammatory gene activation. J Biol Chem. 2006;281:14111–14118. doi: 10.1074/jbc.M511797200. [DOI] [PubMed] [Google Scholar]
- 25.Hossmann KA. Reperfusion of the brain after global ischemia: Hemodynamic disturbances. Shock. 1997;8:95–101. doi: 10.1097/00024382-199708000-00004. [DOI] [PubMed] [Google Scholar]
- 26.Irving EA. Bamford M. Role of mitogen- and stress-activated kinases in ischemic injury. J Cereb Blood Flow Metab. 2002;22:631–647. doi: 10.1097/00004647-200206000-00001. [DOI] [PubMed] [Google Scholar]
- 27.Jang JY. Lee CE. Mitochondrial adenine nucleotide translocator 3 is regulated by IL-4 and IFN-gamma via STAT-dependent pathways. Cell Immunol. 2003;226:11–19. doi: 10.1016/j.cellimm.2003.11.004. [DOI] [PubMed] [Google Scholar]
- 28.Jones NM. Bergeron M. Hypoxic preconditioning induces changes in HIF-1 target genes in neonatal rat brain. J Cereb Blood Flow Metab. 2001;21:1105–1114. doi: 10.1097/00004647-200109000-00008. [DOI] [PubMed] [Google Scholar]
- 29.Jung JE. Kim GS. Narasimhan P. Song YS. Chan PH. Regulation of Mn-superoxide dismutase activity and neuroprotection by STAT3 in mice after cerebral ischemia. J Neurosci. 2009;29:7003–7014. doi: 10.1523/JNEUROSCI.1110-09.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Jung JE. Kim GS. Chen H. Maier CM. Narasimhan P. Song YS. Niizuma K. Katsu M. Okami N. Yoshioka H. Sakata H. Goeders CE. Chan PH. Reperfusion and neurovascular dysfunction in stroke: From basic mechanisms to potential strategies for neuroprotection. Mol Neurobiol. 2010;41:172–179. doi: 10.1007/s12035-010-8102-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Justicia C. Gabriel C. Planas AM. Activation of the JAK/STAT pathway following transient focal cerebral ischemia: signaling through Jak1 and Stat3 in astrocytes. Glia. 2000;30:253–2570. doi: 10.1002/(sici)1098-1136(200005)30:3<253::aid-glia5>3.0.co;2-o. [DOI] [PubMed] [Google Scholar]
- 32.Kim EJ. Raval AP. Perez–Pinzon MA. Preconditioning mediated by sublethal oxygen-glucose deprivation-induced cyclooxygenase-2 expression via the signal transducers and activators of transcription 3 phosphorylation. J Cereb Blood Flow Metab. 2008;28:1329–1340. doi: 10.1038/jcbfm.2008.26. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Kim JK. Raval AP. Hirsch N. Perez–Pinzon MA. Ischemic preconditioning mediates cyclooxygenase-2 expression via nuclear factor-kappa B activation in mixed cortical neuronal cultures. Transl Stroke Res. 2010;1:40–47. doi: 10.1007/s12975-009-0006-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Kim SY. Park HJ. Choi JS. Lee JE. Cha JH. Choi YS. Cho KO. Chun MH. Lee MY. Ischemic preconditioning-induced expression of gp130 and STAT3 in astrocytes of the rat hippocampus. Brain Res Mol Brain Res. 2004;129:96–103. doi: 10.1016/j.molbrainres.2004.06.025. [DOI] [PubMed] [Google Scholar]
- 35.Kim TK. Maniatis T. Regulation of interferon-gamma-activated STAT1 by the ubiquitin-proteasome pathway. Science. 1996;273:1717–1719. doi: 10.1126/science.273.5282.1717. [DOI] [PubMed] [Google Scholar]
- 36.Komine–Kobayashi M. Zhang N. Liu M. Tanaka R. Hara H. Osaka A. Mochizuki H. Mizuno Y. Urabe T. Neuroprotective effect of recombinant human granulocyte colony-stimulating factor in transient focal ischemia of mice. J Cereb Blood Flow Metab. 2006;26:402–413. doi: 10.1038/sj.jcbfm.9600195. [DOI] [PubMed] [Google Scholar]
- 37.Kunisada K. Negoro S. Tone E. Funamoto M. Osugi T. Yamada S. Okabe M. Kishimoto T. Yamauchi–Takihara K. Signal transducer and activator of transcription 3 in the heart transduces not only a hypertrophic signal but a protective signal against doxorubicin-induced cardiomyopathy. Proc Natl Acad Sci USA. 2000;97:315–319. doi: 10.1073/pnas.97.1.315. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Lange–Asschenfeldt C. Raval AP. Dave KR. Mochly–Rosen D. Sick TJ. Pérez–Pinzón MA. Epsilon protein kinase C mediated ischemic tolerance requires activation of the extracellular regulated kinase pathway in the organotypic hippocampal slice. J Cereb Blood Flow Metab. 2004;24:636–645. doi: 10.1097/01.WCB.0000121235.42748.BF. [DOI] [PubMed] [Google Scholar]
- 39.Lee HJ. Oh YK. Rhee M. Lim JY. Hwang JY. Park YS. Kwon Y. Choi KH. Jo I. Park SI. Gao B. Kim WH. The role of STAT1/IRF-1 on synergistic ROS production and loss of mitochondrial transmembrane potential during hepatic cell death induced by LPS/d-GalN. J Mol Biol. 2007;369:967–984. doi: 10.1016/j.jmb.2007.03.072. [DOI] [PubMed] [Google Scholar]
- 40.Lin HY. Huang CC. Chang KF. Lipopolysaccharide preconditioning reduces neuroinflammation against hypoxic ischemia and provides long-term outcome of neuroprotection in neonatal rat. Pediatr Res. 2009;66:254–259. doi: 10.1203/PDR.0b013e3181b0d336. [DOI] [PubMed] [Google Scholar]
- 41.Liu J. Ginis I. Spatz M. Hallenbeck JM. Hypoxic preconditioning protects cultured neurons against hypoxic stress via TNF-alpha and ceramide. Am J Physiol Cell Physiol. 2000;278:C144–153. doi: 10.1152/ajpcell.2000.278.1.C144. [DOI] [PubMed] [Google Scholar]
- 42.Loucks FA. Le SS. Zimmermann AK. Ryan KR. Barth H. Aktories K. Linseman DA. Rho family GTPase inhibition reveals opposing effects of mitogen-activated protein kinase kinase/extracellular signal-regulated kinase and Janus kinase/signal transducer and activator of transcription signaling cascades on neuronal survival. J Neurochem. 2006;97:957–967. doi: 10.1111/j.1471-4159.2006.03802.x. [DOI] [PubMed] [Google Scholar]
- 43.Lowry OH. Passonneau JV. Hasselberger FX. Schulz DW. Effect of ischemia on known substrates and cofactors of the glycolytic pathway in the brain. J Biol Chem. 1964;239:18–30. [PubMed] [Google Scholar]
- 44.Marzo I. Brenner C. Zamzami N. Jürgensmeier JM. Susin SA. Vieira HL. Prévost MC. Xie Z. Matsuyama S. Reed JC. Kroemer G. Bax and adenine nucleotide translocator cooperate in the mitochondrial control of apoptosis. Science. 1998;281:2027–2031. doi: 10.1126/science.281.5385.2027. [DOI] [PubMed] [Google Scholar]
- 45.Mascareno E. El–Shafei M. Maulik N. Sato M. Guo Y. Das DK. Siddiqui MA. JAK/STAT signaling is associated with cardiac dysfunction during ischemia and reperfusion. Circulation. 2001;104:325–329. doi: 10.1161/01.cir.104.3.325. [DOI] [PubMed] [Google Scholar]
- 46.Murriel CL. Mochly–Rosen D. Opposing roles of delta and epsilonPKC in cardiac ischemia and reperfusion: Targeting the apoptotic machinery. Arch Biochem Biophys. 2003;420:246–254. doi: 10.1016/j.abb.2003.08.038. [DOI] [PubMed] [Google Scholar]
- 47.Murry CE. Jennings RB. Reimer KA. Preconditioning with ischemia: A delay of lethal cell injury in ischemic myocardium. Circulation. 1986;74:1124–1136. doi: 10.1161/01.cir.74.5.1124. [DOI] [PubMed] [Google Scholar]
- 48.Nakajima T. Iwabuchi S. Miyazaki H. Okuma Y. Kuwabara M. Nomura Y. Kawahara K. Preconditioning prevents ischemia-induced neuronal death through persistent Akt activation in the penumbra region of the rat brain. J Vet Med Sci. 2004;66:521–527. doi: 10.1292/jvms.66.521. [DOI] [PubMed] [Google Scholar]
- 49.Negoro S. Kunisada K. Tone E. Funamoto M. Oh H. Kishimoto T. Yamauchi–Takihara K. Activation of JAK/STAT pathway transduces cytoprotective signal in rat acute myocardial infarction. Cardiovasc Res. 2000;47:797–805. doi: 10.1016/s0008-6363(00)00138-3. [DOI] [PubMed] [Google Scholar]
- 50.Pérez–Pinzón MA. Mumford PL. Rosenthal M. Sick TJ. Antioxidants, mitochondrial hyperoxidation and electrical recovery after anoxia in hippocampal slices. Brain Res. 1997;754:163–70. doi: 10.1016/s0006-8993(97)00066-8. [DOI] [PubMed] [Google Scholar]
- 51.Pérez–Pinzón MA. Born JG. Rapid preconditioning neuroprotection following anoxia in hippocampal slices: Role of the K+ ATP channel and protein kinase C. Neuroscience. 1999;89:453–459. doi: 10.1016/s0306-4522(98)00560-0. [DOI] [PubMed] [Google Scholar]
- 52.Pérez–Pinzón MA. Neuroprotective effects of ischemic preconditioning in brain mitochondria following cerebral ischemia. J Bioenerg Biomembr. 2004;36:323–327. doi: 10.1023/B:JOBB.0000041762.47544.ff. [DOI] [PubMed] [Google Scholar]
- 53.Planas AM. Justicia C. Ferrer I. Stat1 in developing and adult rat brain. Induction after transient focal ischemia. Neuroreport. 1997;8:1359–1362. doi: 10.1097/00001756-199704140-00008. [DOI] [PubMed] [Google Scholar]
- 54.Potla R. Koeck T. Wegrzyn J. Cherukuri S. Shimoda K. Baker DP. Wolfman J. Planchon SM. Esposito C. Hoit B. Dulak J. Wolfman A. Stuehr D. Larner AC. Tyk2 tyrosine kinase expression is required for the maintenance of mitochondrial respiration in primary pro-B lymphocytes. Mol Cell Biol. 2006;26:8562–8571. doi: 10.1128/MCB.00497-06. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55.Ran R. Xu H. Lu A. Bernaudin M. Sharp FR. Hypoxia preconditioning in the brain. Dev Neurosci. 2005;27:87–92. doi: 10.1159/000085979. [DOI] [PubMed] [Google Scholar]
- 56.Raval AP. Dave KR. Mochly–Rosen D. Sick TJ. Pérez–Pinzón MA. Epsilon PKC is required for the induction of tolerance by ischemic and NMDA-mediated preconditioning in the organotypic hippocampal slice. J Neurosci. 2003;23:384–391. doi: 10.1523/JNEUROSCI.23-02-00384.2003. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57.Reich NC. STAT3 revs up the powerhouse. Sci Signal. 2009;2:60–61. doi: 10.1126/scisignal.290pe61. [DOI] [PubMed] [Google Scholar]
- 58.Rivest S. Lacroix S. Vallières L. Nadeau S. Zhang J. Laflamme N. How the blood talks to the brain parenchyma and the paraventricular nucleus of the hypothalamus during systemic inflammatory and infectious stimuli. Proc Soc Exp Biol Med. 2000;223:22–38. doi: 10.1046/j.1525-1373.2000.22304.x. [DOI] [PubMed] [Google Scholar]
- 59.Rosenthal M. Mumford PL. Sick TJ. Pérez–Pinzón MA. Mitochondrial hyperoxidation after cerebral anoxia/ischemia. Epiphenomenon or precursor to residual damage? Adv Exp Med Biol. 1997;428:189–195. [PubMed] [Google Scholar]
- 60.Sarafian TA. Montes C. Imura T. Qi J. Coppola G. Geschwind DH. Sofroniew MV. Disruption of astrocyte STAT3 signaling decreases mitochondrial function and increases oxidative stress in vitro. PLoS One. 2010;5:e9532. doi: 10.1371/journal.pone.0009532. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Saurin AT. Pennington DJ. Raat NJ. Latchman DS. Owen MJ. Marber MS. Targeted disruption of the protein kinase C epsilon gene abolishes the infarct size reduction that follows ischaemic preconditioning of isolated buffer-perfused mouse hearts. Cardiovasc. 2002;55:672–680. doi: 10.1016/s0008-6363(02)00325-5. [DOI] [PubMed] [Google Scholar]
- 62.Schindler C. Darnell JE., Jr Transcriptional responses to polypeptide ligands: The JAK-STAT pathway. Annu Rev Biochem. 1995;64:621–651. doi: 10.1146/annurev.bi.64.070195.003201. [DOI] [PubMed] [Google Scholar]
- 63.Schindler C. Levy DE. Decker T. JAK-STAT signaling: From interferons to cytokines. J Biol Chem. 2007;282:20059–20063. doi: 10.1074/jbc.R700016200. [DOI] [PubMed] [Google Scholar]
- 64.Schurr A. Reid KH. Tseng MT. West C. Rigor BM. Adaptation of adult brain tissue to anoxia and hypoxia in vitro. Brain Res. 1986;374:244–248. doi: 10.1016/0006-8993(86)90418-x. [DOI] [PubMed] [Google Scholar]
- 65.Schweizer U. Gunnersen J. Karch C. Wiese S. Holtmann B. Takeda K. Akira S. Sendtner M. Conditional gene ablation of Stat3 reveals differential signaling requirements for survival of motoneurons during development and after nerve injury in the adult. J Cell Biol. 2002;156:287–297. doi: 10.1083/jcb.200107009. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Sciamanna MA. Zinkel J. Fabi AY. Lee CP. Ischemic injury to rat forebrain mitochondria and cellular calcium homeostasis. Biochim Biophys Acta. 1992;1134:223–232. doi: 10.1016/0167-4889(92)90180-j. [DOI] [PubMed] [Google Scholar]
- 67.Sims NR. Pulsinelli WA. Altered mitochondrial respiration in selectively vulnerable brain subregions following transient forebrain ischemia in the rat. J Neurochem. 1987;49:1367–1374. doi: 10.1111/j.1471-4159.1987.tb01001.x. [DOI] [PubMed] [Google Scholar]
- 68.Smith RM. Suleman N. Lacerda L. Opie LH. Akira S. Chien KR. Sack MN. Genetic depletion of cardiac myocyte STAT-3 abolishes classical preconditioning. Cardiovasc Res. 2004;63:611–616. doi: 10.1016/j.cardiores.2004.06.019. [DOI] [PubMed] [Google Scholar]
- 69.Solaroglu I. Tsubokawa T. Cahill J. Zhang JH. Anti-apoptotic effect of granulocyte-colony stimulating factor after focal cerebral ischemia in the rat. Neuroscience. 2006;143:965–974. doi: 10.1016/j.neuroscience.2006.09.014. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Spudich A. Frigg R. Kilic E. Kilic U. Oesch B. Raeber A. Bassetti CL. Hermann DM. Aggravation of ischemic brain injury by prion protein deficiency: Role of ERK-1/-2 and STAT-1. Neurobiol Dis. 2005;20:442–449. doi: 10.1016/j.nbd.2005.04.002. [DOI] [PubMed] [Google Scholar]
- 71.Stephanou A. Brar BK. Knight RA. Latchman DS. Opposing actions of STAT-1 and STAT-3 on the Bcl-2 and Bcl-x promoters. Cell Death Differ. 2000;7:329–330. doi: 10.1038/sj.cdd.4400656. [DOI] [PubMed] [Google Scholar]
- 72.Suzuki S. Tanaka K. Nogawa S. Dembo T. Kosakai A. Fukuuchi Y. Phosphorylation of signal transducer and activator of transcription-3 (Stat3) after focal cerebral ischemia in rats. Exp Neurol. 2001;170:63–71. doi: 10.1006/exnr.2001.7701. [DOI] [PubMed] [Google Scholar]
- 73.Taie S. Ono J. Iwanaga Y. Tomita S. Asaga T. Chujo K. Ueki M. Hypoxia-inducible factor-1 alpha has a key role in hypoxic preconditioning. J Clin Neurosci. 2009;16:1056–1060. doi: 10.1016/j.jocn.2008.09.024. [DOI] [PubMed] [Google Scholar]
- 74.Takagi Y. Harada J. Chiarugi A. Moskowitz MA. STAT1 is activated in neurons after ischemia and contributes to ischemic brain injury. J Cereb Blood Flow Meta. 2002;22:1311–1318. doi: 10.1097/01.WCB.0000034148.72481.F4. [DOI] [PubMed] [Google Scholar]
- 75.Wan J. Fu AK. Ip FC. Ng HK. Hugon J. Page G. Wang JH. Lai KO. Wu Z. Ip NY. Tyk2/STAT3 signaling mediates beta-amyloid-induced neuronal cell death: Implications in Alzheimer's disease. J Neurosci. 2010;30:6873–6881. doi: 10.1523/JNEUROSCI.0519-10.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Wang G. Zhou D. Wang C. Gao Y. Zhou Q. Qian G. Decoster MA. Hypoxic preconditioning suppresses group III secreted phospholipase A2-induced apoptosis via JAK2-STAT3 activation in cortical neurons. J Neurochem. 2010;114:1039–1048. doi: 10.1111/j.1471-4159.2010.06817.x. [DOI] [PubMed] [Google Scholar]
- 77.Watson BD. Ginsberg MD. Ischemic injury in the brain. Role of oxygen radical-mediated processes. Ann NY Acad Sci. 1989;559:269–281. doi: 10.1111/j.1749-6632.1989.tb22615.x. [DOI] [PubMed] [Google Scholar]
- 78.Wegener S. Gottschalk B. Jovanovic V. Knab R. Fiebach JB. Schellinger PD. Kucinski T. Jungehülsing GJ. Brunecker P. Müller B. Banasik A. Amberger N. Wernecke KD. Siebler M. Röther J. Villringer A. Weih M. MRI in Acute Stroke Study Group of the German Competence Network Stroke. Transient ischemic attacks before ischemic stroke: Preconditioning the human brain? A multicenter magnetic resonance imaging study. Stroke. 2004;35:616–621. doi: 10.1161/01.STR.0000115767.17923.6A. [DOI] [PubMed] [Google Scholar]
- 79.Wegrzyn J. Potla R. Chwae YJ. Sepuri NB. Zhang Q. Koeck T. Derecka M. Szczepanek K. Szelag M. Gornicka A. Moh A. Moghaddas S. Chen Q. Bobbili S. Cichy J. Dulak J. Baker DP. Wolfman A. Stuehr D. Hassan MO. Fu XY. Avadhani N. Drake JI. Fawcett P. Lesnefsky EJ. Larner AC. Function of mitochondrial Stat3 in cellular respiration. Science. 2009;323:793–797. doi: 10.1126/science.1164551. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Wick A. Wick W. Waltenberger J. Weller M. Dichgans J. Schulz JB. Neuroprotection by hypoxic preconditioning requires sequential activation of vascular endothelial growth factor receptor and Akt. J Neurosci. 2002;22:6401–6407. doi: 10.1523/JNEUROSCI.22-15-06401.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81.Xuan YT. Guo Y. Han H. Zhu Y. Bolli R. An essential role of the JAK-STAT pathway in ischemic preconditioning. Proc Natl Acad Sci USA. 2001;98:9050–9055. doi: 10.1073/pnas.161283798. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Yamashita T. Sawamoto K. Suzuki S. Suzuki N. Adachi K. Kawase T. Mihara M. Ohsugi Y. Abe K. Okano H. Blockade of interleukin-6 signaling aggravates ischemic cerebral damage in mice: Possible involvement of Stat3 activation in the protection of neurons. J Neurochem. 2005;94:459–468. doi: 10.1111/j.1471-4159.2005.03227.x. [DOI] [PubMed] [Google Scholar]
- 83.Yamaura G. Turoczi T. Yamamoto F. Siddqui MA. Maulik N. Das DK. STAT signalingmin ischemic heart: A role of STAT5A in ischemic preconditioning. Am J Physiol Heart Circ Physiol. 2003;285:H476–482. doi: 10.1152/ajpheart.00079.2003. [DOI] [PubMed] [Google Scholar]
- 84.Zhang F. Wang S. Cao G. Gao Y. Chen J. Signal transducers and activators of transcription 5 contributes to erythropoietin-mediated neuroprotection against hippocampal neuronal death after transient global cerebral ischemia. Neurobiol Dis. 2007;25:45–53. doi: 10.1016/j.nbd.2006.08.007. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 85.Zhang HX. Du GH. Zhang JT. Ischemic pre-conditioning preserves brain mitochondrial functions during the middle cerebral artery occlusion in rat. Neurol Res. 2003;25:471–476. doi: 10.1179/016164103101201878. [DOI] [PubMed] [Google Scholar]
- 86.Zhang QG. Wang RM. Han D. Yang LC. Li J. Brann DW. Preconditioning neuroprotection in global cerebral ischemia involves NMDA receptor-mediated ERK-JNK3 crosstalk. Neurosci Res. 2009;63:205–212. doi: 10.1016/j.neures.2008.12.010. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Zhao H. Sapolsky RM. Steinberg GK. Interrupting reperfusion as a stroke therapy: Ischemic postconditioning reduces infarct size after focal ischemia in rats. J Cereb Blood Flow Metab. 2006;26:1114–1121. doi: 10.1038/sj.jcbfm.9600348. [DOI] [PubMed] [Google Scholar]
- 88.Zhou AM. Li WB. Li QJ. Liu HQ. Feng RF. Zhao HG. A short cerebral ischemic preconditioning up-regulates adenosine receptors in the hippocampal CA1 region of rats. Neurosci Res. 2004;48:397–404. doi: 10.1016/j.neures.2003.12.010. [DOI] [PubMed] [Google Scholar]