35 results on '"Yejie Shi"'
Search Results
2. Omega-3 polyunsaturated fatty acids enhance cerebral angiogenesis and provide long-term protection after stroke
- Author
-
Jiayin Wang, Yejie Shi, Lili Zhang, Feng Zhang, Xiaoming Hu, Wenting Zhang, Rehana K. Leak, Yanqin Gao, Ling Chen, and Jun Chen
- Subjects
Angiogenesis ,Angiopoietin 2 ,Astrocyte ,Neuroprotection ,Omega-3 polyunsaturated fatty acids ,Stroke ,Neurosciences. Biological psychiatry. Neuropsychiatry ,RC321-571 - Abstract
Stroke is a devastating neurological disorder and one of the leading causes of death and serious disability. After cerebral ischemia, revascularization in the ischemic boundary zone provides nutritive blood flow as well as various growth factors to promote the survival and activity of neurons and neural progenitor cells. Enhancement of angiogenesis and the resulting improvement of cerebral microcirculation are key restorative mechanisms and represent an important therapeutic strategy for ischemic stroke. In the present study, we tested the hypothesis that post-stroke angiogenesis would be enhanced by omega-3 polyunsaturated fatty acids (n−3 PUFAs), a major component of dietary fish oil. To this end, we found that transgenic fat-1 mice that overproduce n−3 PUFAs exhibited long-term behavioral and histological protection against transient focal cerebral ischemia (tFCI). Importantly, fat-1 transgenic mice also exhibited robust improvements in revascularization and angiogenesis compared to wild type littermates, suggesting a potential role for n−3 fatty acids in post-stroke cerebrovascular remodeling. Mechanistically, n−3 PUFAs induced upregulation of angiopoietin 2 (Ang 2) in astrocytes after tFCI and stimulated extracellular Ang 2 release from cultured astrocytes after oxygen and glucose deprivation. Ang 2 facilitated endothelial proliferation and barrier formation in vitro by potentiating the effects of VEGF on phospholipase Cγ1 and Src signaling. Consistent with these findings, blockade of Src activity in post-stroke fat-1 mice impaired n−3 PUFA-induced angiogenesis and exacerbated long-term neurological outcomes. Taken together, our findings strongly suggest that n−3 PUFA supplementation is a potential angiogenic treatment capable of augmenting brain repair and improving long-term functional recovery after cerebral ischemia.
- Published
- 2014
- Full Text
- View/download PDF
3. Transforming Growth Factor Beta-Activated Kinase 1–Dependent Microglial and Macrophage Responses Aggravate Long-Term Outcomes After Ischemic Stroke
- Author
-
Qing Ye, Jun Chen, Ashutosh P Jadhav, Ming Jiang, Jie Chen, Yaan Liu, Sicheng Li, Jingyan Zhao, Marcelo Rocha, Yejie Shi, Hongjian Pu, Rongrong Wang, and Xiaoming Hu
- Subjects
Time Factors ,Inflammation ,Brain Ischemia ,Proinflammatory cytokine ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Macrophage ,Stroke ,030304 developmental biology ,Mice, Knockout ,Advanced and Specialized Nursing ,0303 health sciences ,Behavior, Animal ,Microglia ,biology ,Kinase ,business.industry ,Macrophages ,Transforming growth factor beta ,MAP Kinase Kinase Kinases ,medicine.disease ,medicine.anatomical_structure ,Brain Injuries ,Reperfusion Injury ,Immunology ,biology.protein ,Zearalenone ,Neurology (clinical) ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business ,030217 neurology & neurosurgery ,Tamoxifen ,medicine.drug - Abstract
Background and Purpose— Microglia/macrophages (Mi/MΦ) can profoundly influence stroke outcomes by acquiring functionally dominant phenotypes (proinflammatory or anti-inflammatory; deleterious or salutary). Identification of the molecular mechanisms that dictate the functional status of Mi/MΦ after brain ischemia/reperfusion may reveal novel therapeutic targets for stroke. We hypothesized that activation of TAK1 (transforming growth factor beta-activated kinase 1), a key MAP3K upstream of multiple inflammation-regulating pathways, drives Mi/MΦ toward a proinflammatory phenotype and potentiates ischemia/reperfusion brain injury. Methods— Young adult mice were subjected to 1 hour of middle cerebral artery occlusion (MCAO) followed by reperfusion. TAK1 was targeted by tamoxifen-induced Mi/MΦ-specific knockout or administration of a selective inhibitor 5Z-7-Oxozeaenol after MCAO. Neurobehavioral deficits and long-term gray matter and white matter injury were assessed up to 35 days after MCAO. Mi/MΦ functional status and brain inflammatory profiles were assessed 3 days after MCAO by RNA-seq, flow cytometry, and immunohistochemistry. Results— TAK1 Mi/MΦ-specific knockout markedly ameliorated neurological deficits in the rotarod and cylinder tests for at least 35 days after MCAO. Mechanistically, RNA-seq of purified brain Mi/MΦ demonstrated that proinflammatory genes and their predicted biological functions were downregulated or inhibited in microglia and macrophages from TAK1 Mi/MΦ-specific knockout mice versus WT mice 3 days after MCAO. Consistent with the anti-inflammatory phenotype of Mi/MΦ-specific knockout, oxozeaenol treatment mitigated neuroinflammation 3 days after MCAO, manifested by less Iba1 + /CD16 + proinflammatory Mi/MΦ and suppressed brain invasion of various peripheral immune cells. Oxozeaenol treatment beginning 2 hours after MCAO improved long-term sensorimotor and cognitive functions in the foot fault, rotarod, and water maze tests. Furthermore, Oxozeaenol promoted both gray matter and white matter integrity 35 days after MCAO. Conclusions— TAK1 promotes ischemia/reperfusion-induced inflammation, brain injury, and maladaptive behavior by enhancing proinflammatory and deleterious Mi/MΦ responses. Therefore, TAK1 inhibition is a promising therapy to improve long-term stroke outcomes.
- Published
- 2020
- Full Text
- View/download PDF
4. Neuroprotection against ischemic stroke requires a specific class of early responder T cells in mice
- Author
-
Wei Cai, Ligen Shi, Jingyan Zhao, Fei Xu, Connor Dufort, Qing Ye, Tuo Yang, Xuejiao Dai, Junxuan Lyu, Chenghao Jin, Hongjian Pu, Fang Yu, Sulaiman Hassan, Zeyu Sun, Wenting Zhang, T. Kevin Hitchens, Yejie Shi, Angus W. Thomson, Rehana K. Leak, Xiaoming Hu, and Jun Chen
- Subjects
Male ,Mice, Inbred C57BL ,Stroke ,Mice ,Animals ,Female ,General Medicine ,CD8-Positive T-Lymphocytes ,Neuroprotection ,Interleukin-10 ,Ischemic Stroke - Abstract
Immunomodulation holds therapeutic promise against brain injuries, but leveraging this approach requires a precise understanding of mechanisms. We report that CD8+CD122+CD49dlo T regulatory-like cells (CD8+ TRLs) are among the earliest lymphocytes to infiltrate mouse brains after ischemic stroke and temper inflammation; they also confer neuroprotection. TRL depletion worsened stroke outcomes, an effect reversed by CD8+ TRL reconstitution. The CXCR3/CXCL10 axis served as the brain-homing mechanism for CD8+ TRLs. Upon brain entry, CD8+ TRLs were reprogrammed to upregulate leukemia inhibitory factor (LIF) receptor, epidermal growth factor-like transforming growth factor (ETGF), and interleukin 10 (IL-10). LIF/LIF receptor interactions induced ETGF and IL-10 production in CD8+ TRLs. While IL-10 induction was important for the antiinflammatory effects of CD8+ TRLs, ETGF provided direct neuroprotection. Poststroke intravenous transfer of CD8+ TRLs reduced infarction, promoting long-term neurological recovery in young males or aged mice of both sexes. Thus, these unique CD8+ TRLs serve as early responders to rally defenses against stroke, offering fresh perspectives for clinical translation.
- Published
- 2021
5. Macrophages reprogram after ischemic stroke and promote efferocytosis and inflammation resolution in the mouse brain
- Author
-
Amanda D. Smith, Qing Ye, Ming Jiang, Rongrong Wang, Yejie Shi, Jingyan Zhao, Jun Chen, and Wenting Zhang
- Subjects
Male ,0301 basic medicine ,Pathology ,Apoptosis ,Monocytes ,Brain Ischemia ,Apoptotic cell clearance ,Brain ischemia ,Mice ,0302 clinical medicine ,Macrophage ,Pharmacology (medical) ,Microglia ,Brain ,Immunohistochemistry ,Stroke ,Psychiatry and Mental health ,medicine.anatomical_structure ,RNA‐seq ,Original Article ,medicine.symptom ,focal cerebral ischemia ,medicine.medical_specialty ,Ischemia ,Neuroimaging ,Inflammation ,03 medical and health sciences ,Phagocytosis ,Physiology (medical) ,medicine ,Animals ,apoptotic cell clearance ,Efferocytosis ,Pharmacology ,Whole Genome Sequencing ,Sequence Analysis, RNA ,business.industry ,Macrophages ,Monocyte ,Original Articles ,medicine.disease ,Mice, Inbred C57BL ,PPAR gamma ,030104 developmental biology ,inflammation resolution ,STAT6 Transcription Factor ,Transcriptome ,business ,030217 neurology & neurosurgery - Abstract
Aims Blood‐borne monocytes/macrophages infiltrate the brain in massive numbers after ischemic stroke, but their impact on poststroke brain injury and recovery remains elusive. This study examined the transcriptomic changes in monocytes/macrophages after ischemic stroke and the functional implications of these changes, particularly with regards to the contribution of these cells to the phagocytic clearance of dead/dying cells (efferocytosis) in the poststroke brain. Methods We performed whole‐genome RNA sequencing on the monocyte/macrophage population sorted from mouse brain and peripheral blood 5 days after permanent focal cerebral ischemia. In addition, the spatial and temporal profiles of macrophage efferocytosis were examined in vivo by immunohistochemistry 3‐7 days after brain ischemia. Results Robust transcriptomic changes occurred in monocytes/macrophages upon infiltrating the poststroke brain. Functional enrichment analysis revealed a transcriptome of brain macrophages that strongly favored efferocytic activity. A large number of efferocytosis‐related genes were upregulated in brain macrophages, the products of which are essential components involved in various steps of efferocytosis, such as chemotaxis, recognition of dead cells, engulfment, and processing of phagosomes. The efferocytic activity of brain macrophages were verified by immunohistochemistry, wherein Iba1‐labeled microglia/macrophages effectively cleared apoptotic neurons in the infarct during the subacute stage after brain ischemia. We also identified PPARγ and STAT6 as potential upstream regulators that shaped this proefferocytic and inflammation‐resolving transcriptome of macrophages in the poststroke brain. Conclusion Macrophages play a crucial role in the phagocytic clearance of dead neurons after ischemic stroke and promote the resolution of inflammation in the brain. Molecular therapies that enhance macrophage efferocytic capability may be promising treatments for ischemic stroke by facilitating inflammation resolution, brain repair, and recovery of neurological functions.
- Published
- 2019
- Full Text
- View/download PDF
6. Post-stroke administration of omega-3 polyunsaturated fatty acids promotes neurovascular restoration after ischemic stroke in mice: Efficacy declines with aging
- Author
-
Xiaoming Hu, Jun Suenaga, Jun Chen, Hongjian Pu, Zhishuo Wei, Yejie Shi, Xiaoyan Jiang, and Amanda D. Smith
- Subjects
Male ,0301 basic medicine ,Aging ,medicine.medical_specialty ,Population ,Neovascularization, Physiologic ,Striatum ,lcsh:RC321-571 ,White matter ,Mice ,Random Allocation ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Fatty Acids, Omega-3 ,medicine ,Animals ,education ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Stroke ,chemistry.chemical_classification ,education.field_of_study ,business.industry ,Brain ,medicine.disease ,Fish oil ,Mice, Inbred C57BL ,Regimen ,030104 developmental biology ,Endocrinology ,medicine.anatomical_structure ,Neurology ,chemistry ,Docosahexaenoic acid ,Oligodendrogenesis ,lipids (amino acids, peptides, and proteins) ,Angiogenesis ,business ,030217 neurology & neurosurgery ,Polyunsaturated fatty acid - Abstract
Post-stroke treatment with omega-3 polyunsaturated fatty acids (n-3 PUFAs) may be a promising therapy in young animals but this has not been tested in aged subjects, a population at most risk of ischemic stroke. Herein we examined the therapeutic efficacy of n-3 PUFAs after distal middle cerebral artery occlusion (dMCAO) in young (10–12 weeks old) and aged (18 months old) mice. Post-ischemic mice were randomly assigned to 4 groups that received: 1) regular food with low content of n-3 PUFAs, 2) intraperitoneal docosahexaenoic acid (DHA, a major component of n-3 PUFAs) injections, 3) Fish oil (FO, containing high concentration of n-3 PUFAs) dietary supplement, or 4) combined treatment with DHA and FO dietary supplement. Long-term neurorestoration induced by n-3 PUFA post-stroke administration and its underlying mechanism(s) were analyzed up to 35 days after dMCAO. Aged mice showed more severe neurological deficits than young mice after dMCAO with histological lesions extended to the striatum. Notably, post-stroke treatment with combined DHA injections and FO dietary supplementation was more effective in reducing brain injury and improving sensorimotor function in aged mice than either treatment alone, albeit to a lesser extent than in the young mice. Unlike the improvement in spatial cognitive function observed in young mice, the combined treatment regimen failed to improve cognitive function in aged mice. The reduction in stroke-induced neurological deficits with n-3 PUFA post-treatment was associated with enhanced angiogenesis, oligodendrogenesis, neuron survival and white matter restoration. Together, these results indicate that the neurological benefits of n-3 PUFA administration after stroke extend to older animals and are associated with improved neuronal survival and brain remodeling, therefore suggesting that post-stroke administration of n-3 PUFAs is a viable clinically relevant treatment option against stroke.
- Published
- 2019
- Full Text
- View/download PDF
7. The effect of age-related risk factors and comorbidities on white matter injury and repair after ischemic stroke
- Author
-
Yanqin Gao, Mingyue Xu, Michael M. Wang, Yejie Shi, and Richard F. Keep
- Subjects
0301 basic medicine ,Aging ,medicine.medical_specialty ,Inflammation ,Comorbidity ,Article ,lcsh:RC321-571 ,White matter ,03 medical and health sciences ,0302 clinical medicine ,Risk Factors ,Internal medicine ,Diabetes mellitus ,Hyperlipidemia ,medicine ,Animals ,Humans ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Stroke ,business.industry ,White Matter Injury ,medicine.disease ,White Matter ,Diffusion tensor imaging ,030104 developmental biology ,medicine.anatomical_structure ,Neurology ,Myelin ,Hypertension ,Cardiology ,Metabolic syndrome ,medicine.symptom ,business ,030217 neurology & neurosurgery ,Diffusion MRI - Abstract
White matter injury is a crucial component of human stroke, but it has often been neglected in preclinical studies. Most human stroke is associated with one or more comorbidities, including aging, hypertension, diabetes and metabolic syndrome including hyperlipidemia. The purpose of this review is to examine how age and hypertension impact stroke-induced white matter injury as well as white matter repair in both human stroke and preclinical models. It is essential that comorbidities be examined in preclinical trials as they may impact translatability to the clinic. In addition, understanding how comorbidities impact white matter injury and repair may provide new therapeutic opportunities for patients with those conditions.
- Published
- 2019
- Full Text
- View/download PDF
8. Inhibition of TGFβ-activated kinase 1 promotes inflammation-resolving microglial/macrophage responses and recovery after stroke in ovariectomized female mice
- Author
-
Qing Ye, Yejie Shi, Hongjian Pu, Yongfang Zhao, Sicheng Li, Rongrong Wang, and Yaan Liu
- Subjects
0301 basic medicine ,Programmed cell death ,TAK1 ,Ovariectomy ,Inflammation ,Pharmacology ,MAP3K7 ,Transient focal cerebral ischemia ,Ovariectomized mice ,lcsh:RC321-571 ,Proinflammatory cytokine ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Medicine ,Animals ,Enzyme Inhibitors ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Stroke ,Neuroinflammation ,Mice, Knockout ,Microglia ,business.industry ,Macrophages ,Infarction, Middle Cerebral Artery ,Recovery of Function ,medicine.disease ,MAP Kinase Kinase Kinases ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,Neurology ,Zearalenone ,Female ,Signal transduction ,medicine.symptom ,business ,030217 neurology & neurosurgery ,5Z-7-Oxozeaenol - Abstract
TGFβ-activated kinase 1 (TAK1) is a master regulator that drives multiple cell death and proinflammatory signaling pathways, making it a promising therapeutic target to treat ischemic stroke. However, whether targeting TAK1 could improve stroke outcomes has never been tested in female subjects, hindering its potential translation into clinical use. Here we examined the therapeutic effect of 5Z-7-Oxozeaenol (OZ), a selective TAK1 inhibitor, in ovariectomized female mice after middle cerebral artery occlusion (MCAO). OZ significantly reduced neuronal cell death and axonal injury at the acute stage and mitigated neuroinflammation at the subacute stage after MCAO in ovariectomized female mice. Consistent with RNA sequencing analysis that TAK1 activation contributed to microglia/macrophage-mediated inflammatory responses in the post-stroke brain, inhibition of TAK1 with OZ caused phenotypic shift of microglia/macrophages toward an inflammation-resolving state. Furthermore, microglia/macrophage-specific TAK1 knockout (TAK1 mKO) reproduced OZ's effects, causally confirming the role of TAK1 in determining proinflammatory microglial/macrophage responses in post-stroke females. Post-stroke treatment with OZ for 5 days effectively promoted long-term neurological recovery and the integrity of both gray matter and white matter in female mice. Together, the TAK1 inhibitor OZ elicits long-lasting improvement of stroke outcomes in female mice, at least partially through enhancing beneficial microglial/macrophage responses and inflammation resolution. Given its therapeutic efficacy on both male and female rodents, TAK1 inhibitor is worth further investigation as a valid treatment to ischemic stroke.
- Published
- 2020
9. Genome-wide transcriptomic analysis of microglia reveals impaired responses in aged mice after cerebral ischemia
- Author
-
Xiaoming Hu, Wenting Zhang, Marcelo Rocha, Ligen Shi, Jun Chen, Sulaiman H Hassan, Jianhua Luo, Yejie Shi, Ming Jiang, Lawrence R. Wechsler, Maya N Adair, Liqiang Liu, Jing Xu, Qing Ye, and Sicheng Li
- Subjects
Male ,Ischemia ,Genome ,Flow cytometry ,Brain Ischemia ,Transcriptome ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Medicine ,Animals ,Humans ,Stroke ,030304 developmental biology ,Aged ,0303 health sciences ,Microglia ,medicine.diagnostic_test ,business.industry ,CLEC7A ,Sequence Analysis, RNA ,Genomics ,Original Articles ,medicine.disease ,Flow Cytometry ,Disease Models, Animal ,medicine.anatomical_structure ,Neurology ,Immunology ,Neurology (clinical) ,Cardiology and Cardiovascular Medicine ,business ,030217 neurology & neurosurgery ,Homeostasis - Abstract
Senescence-associated alterations in microglia may have profound impact on cerebral homeostasis and stroke outcomes. However, the lack of a transcriptome-wide comparison between young and aged microglia in the context of ischemia limits our understanding of aging-related mechanisms. Herein, we performed RNA sequencing analysis of microglia purified from cerebral hemispheres of young adult (10-week-old) and aged (18-month-old) mice five days after distal middle cerebral artery occlusion or after sham operation. Considerable transcriptional differences were observed between young and aged microglia in healthy brains, indicating heightened chronic inflammation in aged microglia. Following stroke, the overall transcriptional activation was more robust (>13-fold in the number of genes upregulated) in young microglia than in aged microglia. Gene clusters with functional implications in immune inflammatory responses, immune cell chemotaxis, tissue remodeling, and cell-cell interactions were markedly activated in microglia of young but not aged stroke mice. Consistent with the genomic profiling predictions, post-stroke cerebral infiltration of peripheral immune cells was markedly decreased in aged mice compared to young mice. Moreover, post-ischemic aged microglia demonstrated reduced interaction with neighboring neurons and diminished polarity toward the infarct lesion. These alterations in microglial gene response and behavior may contribute to aging-driven vulnerability and poorer recovery after ischemic stroke.
- Published
- 2020
10. Transcriptomic and functional studies reveal undermined chemotactic and angiostimulatory properties of aged microglia during stroke recovery
- Author
-
Silvia Liu, Jianhua Luo, Wei Su, Fei Xu, Yejie Shi, Hongfeng Mu, Xiaoming Hu, Zeyu Sun, Jing Xu, Rehana K. Leak, Lawrence R. Wechsler, Di Xie, Lu Jiang, and Jun Chen
- Subjects
Male ,Angiogenesis ,medicine.medical_treatment ,Biology ,Transcriptome ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Animals ,Functional studies ,030304 developmental biology ,0303 health sciences ,Microglia ,Neurodegeneration ,Chemotaxis ,Cell movement ,Original Articles ,medicine.disease ,Stroke ,Disease Models, Animal ,medicine.anatomical_structure ,Neurology ,Neurology (clinical) ,Cardiology and Cardiovascular Medicine ,Stroke recovery ,Neuroscience ,030217 neurology & neurosurgery - Abstract
Age-dependent alterations in microglia behavior have been implicated in neurodegeneration and CNS injuries. Here, we compared the transcriptional profiles of young versus aged microglia during stroke recovery. CD45intermediateCD11b+ microglia were FACS-isolated from the brains of young (10-week-old) and aged (18-month-old) male mice with sham operation or 14 days after distal middle cerebral artery occlusion and subjected to RNA-sequencing analysis. Functional groups enriched in young microglia are indicative of upregulation in cell movement, cell interactions, inflammatory responses and angiogenesis, while aged microglia exhibited a reduction or no change in these features. We confirmed reduced chemoattractive capacities of aged microglia toward ischemic brain tissue in organotypic slide co-cultures, and delayed accumulation of aged microglia around dead neurons injected into the striatum in vivo. In addition, aging is associated with an overall failure to increase the expression of microglial genes involved in cell–cell interactions, such as CXCL10. Finally, impaired upregulation of pro-angiogenic genes in aged microglia was associated with a decline in neovascularization in aged mice compared to young mice after distal middle cerebral artery occlusion. This study provides a new resource to understand the mechanisms underlying microglial alterations in the aged brain milieu and sheds light on new strategies to improve microglial functions in aged stroke victims.
- Published
- 2020
11. Abstract WMP73: TGFβ-Activated Kinase 1 (TAK1) is a Key Molecular Switch That Drives Proinflammatory Microglia/macrophage Responses and Aggravates Long-Term Stroke Outcomes
- Author
-
Qing Ye, Yejie Shi, Hongjian Pu, Jun Chen, Ming Jiang, and Rongrong Wang
- Subjects
Advanced and Specialized Nursing ,Microglia ,Kinase ,business.industry ,Inflammation ,medicine.disease ,Phenotype ,Microglia macrophages ,Proinflammatory cytokine ,Brain ischemia ,medicine.anatomical_structure ,medicine ,Cancer research ,Neurology (clinical) ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business ,Stroke - Abstract
Introduction: Microglia/macrophages (Mi/MΦ) profoundly influence stroke outcomes, but the molecular mechanisms dictating their functional phenotype after brain ischemia/reperfusion (I/R) are poorly understood. We hypothesized that activation of TAK1, a key MAPKKK upstream of multiple inflammation-regulating pathways, drives Mi/MΦ towards a proinflammatory phenotype and potentiates I/R brain injury. Methods: TAK1 was targeted in mice by tamoxifen-induced, Mi/MΦ-specific knockout (mKO) or a relatively selective inhibitor 5Z-7-oxozeaenol (OZ; 2.5 mg/kg i.p.). Sensorimotor/cognitive deficits, grey/white matter injury and brain inflammatory profiles were assessed after 1 h MCAO and reperfusion. Results: TAK1 mKO markedly ameliorated neurological deficits in the rotarod, cylinder and water maze tests (plow ) and MΦ (CD45 high ) cells revealed that multiple genes participating in pro-inflammatory responses were downregulated in both Mi and MΦ from mKO mice versus WT mice 3 d after I/R. Consistent with the phenotype of mKO, OZ mitigated neuroinflammation 3 d after I/R, reflected by less Iba1 + /CD16 + proinflammatory Mi/MΦ and less brain invasion of peripheral immune cells (neutrophils, MΦ, T cells). OZ treatment beginning 2 h after I/R effectively improved long term (35 d) sensorimotor and cognitive functions in the foot fault, rotarod and water maze tests (p Conclusions: TAK1 promotes I/R-induced inflammation, brain injury, and maladaptive behavior by enhancing neurotoxic Mi/MΦ responses. TAK1 inhibition is a promising therapy to improve long term stroke outcomes in both sexes.
- Published
- 2020
- Full Text
- View/download PDF
12. In VivoExpansion of Regulatory T Cells with IL-2/IL-2 Antibody Complex Protects against Transient Ischemic Stroke
- Author
-
Fang Yu, Peiying Li, Xiaoming Hu, Qing Ye, Yuguo Xia, Yuanyuan Yang, Zhishuo Wei, Wen Zhu, Jun Chen, Haiyue Zhang, Yejie Shi, and Angus W. Thomson
- Subjects
Genetically modified mouse ,Adoptive cell transfer ,business.industry ,General Neuroscience ,FOXP3 ,chemical and pharmacologic phenomena ,Spleen ,medicine.disease ,Neuroprotection ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Immunology ,medicine ,Systemic administration ,business ,Stroke ,Research Articles ,030217 neurology & neurosurgery ,Neuroinflammation ,030215 immunology - Abstract
Regulatory T cells (Tregs) are known to protect against ischemic stroke. However, the low frequency of Tregs restricts their clinical utility. This study investigated whether expanding the number of Tregsin vivowith the IL-2/IL-2 antibody complex (IL-2/IL-2Ab) could improve stroke outcomes and further elaborated the mechanisms of protection in male mice. C57BL/6 mice received IL-2/IL-2Ab or isotype IgG (IsoAb) intraperitoneally for 3 d before (pretreatment) or starting 2 h after (posttreatment) 60 min middle cerebral artery occlusion (MCAO). IL-2/IL-2Ab selectively increased the number of Tregs in the blood, spleen, and lymph nodes. The IL-2/IL-2Ab treatment significantly reduced infarct volume, inhibited neuroinflammation, and improved sensorimotor functions, as manifested by rotarod test and foot fault test, compared with IsoAb-treated stroke mice. Treg depletion was then achieved by diphtheria toxin (DT) injection into transgenic mice expressing the DT receptor under the control of the Foxp3 promoter (DTR mice). The depletion of Tregs completely eliminated IL-2/IL-2Ab-afforded neuroprotection. Interestingly, adoptive transfer of Tregs collected from IL-2/IL-2Ab-treated mice demonstrated more potent neuroprotection than an equal number of Tregs prepared from IsoAb-treated mice, suggesting that IL-2/IL-2Ab not only elevated Treg numbers, but also boosted their functions. Mechanistically, IL-2/IL-2Ab promoted the expression of CD39 and CD73 in expanded Tregs. CD73 deficiency diminished the protective effect of IL-2/IL-2Ab-stimulated Tregs in stroke mice. The results show that IL-2/IL-2Ab expands Tregsin vivoand boosts their immunomodulatory function. The activation of CD39/CD73 signaling in Tregs may participate as a potential mechanism underlying IL-2/IL-2Ab-afforded neuroprotection against ischemic brain injury.SIGNIFICANCE STATEMENTRegulatory T cells (Tregs) are known to protect against ischemic stroke. However, the low frequency of Tregs restricts their clinical utility. This study reported that systemic administration of the IL-2/IL-2 antibody complex (IL-2/IL-2Ab) robustly and selectively expanded the number of Tregs after stroke. IL-2/IL-2Ab pretreatment or posttreatment significantly improved stroke outcomes in a rodent model of ischemic stroke. We further discovered that IL-2/IL-2Ab not only elevated Treg numbers, but also boosted their functions and enhanced the expression of CD39 and CD73. Using CD73-deficient mice, we confirmed the importance of CD73 in the protective effect of IL-2/IL-2Ab-stimulated Tregs in stroke mice. These results shed light on IL-2/IL-2Ab as a clinically feasible immune therapy to boost endogenous Treg responses and ameliorate ischemic brain injury.
- Published
- 2018
- Full Text
- View/download PDF
13. STAT6/Arg1 promotes microglia/macrophage efferocytosis and inflammation resolution in stroke mice
- Author
-
Xiaoming Hu, Jingyan Zhao, Jie Chen, Yejie Shi, Wenting Zhang, Toshimasa Nakao, Mingyue Xu, Jun Chen, Lili Zhang, R. Anne Stetler, Julia Kofler, Wei Cai, Xuejiao Dai, Marcelo Rocha, and Boyu Yang
- Subjects
Brain Infarction ,Male ,0301 basic medicine ,Primary Cell Culture ,Inflammation ,Neuroprotection ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Phagocytosis ,Animals ,Humans ,Medicine ,Macrophage ,Efferocytosis ,Stroke ,Cells, Cultured ,STAT6 ,Aged, 80 and over ,Neurons ,Arginase ,integumentary system ,Microglia ,business.industry ,Macrophages ,Brain ,General Medicine ,medicine.disease ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,nervous system ,030220 oncology & carcinogenesis ,Cancer research ,STAT protein ,Female ,medicine.symptom ,STAT6 Transcription Factor ,business ,Research Article ,Signal Transduction - Abstract
Efferocytosis, or phagocytic clearance of dead/dying cells by brain-resident microglia and/or infiltrating macrophages, is instrumental for inflammation resolution and restoration of brain homeostasis after stroke. Here, we identify the signal transducer and activator of transcription 6/arginase1 (STAT6/Arg1) signaling axis as a potentially novel mechanism that orchestrates microglia/macrophage responses in the ischemic brain. Activation of STAT6 was observed in microglia/macrophages in the ischemic territory in a mouse model of stroke and in stroke patients. STAT6 deficiency resulted in reduced clearance of dead/dying neurons, increased inflammatory gene signature in microglia/macrophages, and enlarged infarct volume early after experimental stroke. All of these pathological changes culminated in an increased brain tissue loss and exacerbated long-term functional deficits. Combined in vivo analyses using BM chimeras and in vitro experiments using microglia/macrophage-neuron cocultures confirmed that STAT6 activation in both microglia and macrophages was essential for neuroprotection. Adoptive transfer of WT macrophages into STAT6-KO mice reduced accumulation of dead neurons in the ischemic territory and ameliorated brain infarction. Furthermore, decreased expression of Arg1 in STAT6(–/–) microglia/macrophages was responsible for impairments in efferocytosis and loss of antiinflammatory modality. Our study suggests that efferocytosis via STAT6/Arg1 modulates microglia/macrophage phenotype, accelerates inflammation resolution, and improves stroke outcomes.
- Published
- 2019
- Full Text
- View/download PDF
14. Protease-independent action of tissue plasminogen activator in brain plasticity and neurological recovery after ischemic stroke
- Author
-
Qing Ye, Hongfeng Mu, Xiaoming Hu, Na Xu, Michael V. L. Bennett, Rehana K. Leak, T. Kevin Hitchens, Shubei Ma, Hongjian Pu, Zhishuo Wei, Jun Chen, Yejie Shi, Lili Zhang, Fei Xu, Yuguo Xia, and Zhengyu Lu
- Subjects
Male ,Neurogenesis ,Ischemia ,Pharmacology ,Tissue plasminogen activator ,Brain Ischemia ,Myelin ,Mice ,Epidermal growth factor ,Neuroplasticity ,medicine ,Animals ,Epidermal growth factor receptor ,Stroke ,Neurons ,Multidisciplinary ,Neuronal Plasticity ,biology ,integumentary system ,business.industry ,Brain ,Cerebral Infarction ,Recovery of Function ,medicine.disease ,Axons ,Mice, Inbred C57BL ,Disease Models, Animal ,medicine.anatomical_structure ,PNAS Plus ,Brain Injuries ,Tissue Plasminogen Activator ,Knockout mouse ,biology.protein ,business ,medicine.drug - Abstract
Emerging evidence suggests that tissue plasminogen activator (tPA), currently the only FDA-approved medication for ischemic stroke, exerts important biological actions on the CNS besides its well-known thrombolytic effect. In this study, we investigated the role of tPA on primary neurons in culture and on brain recovery and plasticity after ischemic stroke in mice. Treatment with recombinant tPA stimulated axonal growth in culture, an effect independent of its protease activity and achieved through epidermal growth factor receptor (EGFR) signaling. After permanent focal cerebral ischemia, tPA knockout mice developed more severe sensorimotor and cognitive deficits and greater axonal and myelin injury than wild-type mice, suggesting that endogenously expressed tPA promotes long-term neurological recovery after stroke. In tPA knockout mice, intranasal administration of recombinant tPA protein 6 hours poststroke and 7 more times at 2 d intervals mitigated white matter injury, improved axonal conduction, and enhanced neurological recovery. Consistent with the proaxonal growth effects observed in vitro, exogenous tPA delivery increased poststroke axonal sprouting of corticobulbar and corticospinal tracts, which might have contributed to restoration of neurological functions. Notably, recombinant mutant tPA-S478A lacking protease activity (but retaining the EGF-like domain) was as effective as wild-type tPA in rescuing neurological functions in tPA knockout stroke mice. These findings demonstrate that tPA improves long-term functional outcomes in a clinically relevant stroke model, likely by promoting brain plasticity through EGFR signaling. Therefore, treatment with the protease-dead recombinant tPA-S478A holds particular promise as a neurorestorative therapy, as the risk for triggering intracranial hemorrhage is eliminated and tPA-S478A can be delivered intranasally hours after stroke.
- Published
- 2019
15. Abstract WMP77: Ischemic Preconditioning Improves Long-Term Outcomes and Preserves Blood-Brain Barrier After Ischemic Stroke via Oxidative Signaling and Nrf2 Activation
- Author
-
Leilei Mao, Tuo Yang, Yejie Shi, Jun Chen, Qianqian Li, Rehana K. Leak, Lili Zhang, Yang Sun, Meijuan Zhang, and Feng Zhang
- Subjects
Advanced and Specialized Nursing ,medicine.medical_specialty ,business.industry ,Oxidative phosphorylation ,medicine.disease ,medicine.disease_cause ,Blood–brain barrier ,Nrf2 activation ,medicine.anatomical_structure ,Internal medicine ,Ischemic stroke ,medicine ,Cardiology ,Long term outcomes ,Ischemic preconditioning ,cardiovascular diseases ,Neurology (clinical) ,Cardiology and Cardiovascular Medicine ,business ,Stroke ,Oxidative stress - Abstract
Introduction: Ischemic preconditioning (IPC) protects the brain against subsequent ischemic injuries. However, its protective mechanisms and long-term impact on stroke outcomes are unknown. IPC requires mild oxidative stress and may activate nuclear factor (erythroid-derived 2)-like 2 (Nrf2). Endogenous lipid electrophiles serve as robust Nrf2 activators. This study tests long-term effects of IPC and explores the mechanisms of IPC-mediated ischemic tolerance, focusing on lipid electrophiles and Nrf2 pathway. Methods: Wildtype (WT) and Nrf2 knockout (KO) mice were subjected to 60-min middle cerebral artery occlusion (MCAO) 3 d after IPC (12-min MCAO). Tissue loss, blood-brain barrier (BBB) damage, and neurobehavioral outcomes were assessed up to 35 d post stroke. The molecular mechanisms were explored by primary endothelial cell (EC) cultures with plasmid transfection and molecular biological approaches. Results: IPC reduced sensorimotor and cognitive deficits and tissue loss up to 35 d post stroke in WT mice, while Nrf2 KO abolished IPC-mediated protection. IPC led to mild oxidative stress, lipid electrophile generation and Nrf2 pathway activation. Prominent Nrf2 activation was seen in ECs and a selected group of astrocytes in tight association with microvessels, both of which are BBB components. As expected, IPC reduced BBB leakage 48 h post stroke and increased expressions of junctional proteins Claudin-5 and VE-Cadherin. Moreover, Nrf2 directly regulated Claudin-5 and VE-Cadherin promoter activities. Finally, a novel mechanism for electrophiles to activate Nrf2 was identified-through direct inhibition of glycogen synthase kinase 3β (GSK3β) activity via GSK3β-C199 residue. Conclusions: IPC preserves the BBB and provides long-term neuroprotection against stroke. Mechanistically, mild oxidative stress in IPC generates a pool of electrophiles, which then activates Nrf2 pathway through direct inhibition GSK3β-dependent Nrf2 degradation.
- Published
- 2019
- Full Text
- View/download PDF
16. Abstract 22: Apoptosis Signal-Regulating Kinase 1 (ASK1) is a Novel Master Molecular Switch Controlling Microglia/Macrophage Reactions That Impact Short- and Long-Term Stroke Outcomes
- Author
-
Jun Chen, Yejie Shi, Wenting Zhang, Rongrong Wang, and Rehana K. Leak
- Subjects
Advanced and Specialized Nursing ,Microglia ,business.industry ,Kinase ,Inflammation ,030204 cardiovascular system & hematology ,medicine.disease ,Microglia macrophages ,03 medical and health sciences ,0302 clinical medicine ,medicine.anatomical_structure ,Apoptosis ,medicine ,Cancer research ,ASK1 ,Functional status ,Neurology (clinical) ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business ,Stroke ,030217 neurology & neurosurgery - Abstract
Introduction: Recent studies suggest that the functional status of microglia/macrophages (Mi/MΦ) profoundly impacts stroke outcome. However, the molecular mechanisms regulating Mi/MΦ reactions after brain ischemia/reperfusion (I/R) are poorly understood. Hypothesis: Activation of ASK1, a key MAPKKK upstream of multiple inflammation-regulating pathways, critically contributes to I/R brain injury via priming Mi/MΦ towards a neurotoxic phenotype. Methods: We generated mice with cell-specific, tamoxifen-induced knockout of ASK1 in neurons (nKO) or Mi/MΦ (mKO). Brain injury, neuroinflammation, and various behavioral deficits were assessed after 1 h MCAO and reperfusion. Results: MCAO induced transient ASK1-MKK4-JNK3 activation (6-24 h) in neurons and persistent ASK1-MKK3-p38/STAT1 activation (3-5 d) in Mi/MΦ in WT, but not in ASK1 nKO or mKO mice, respectively. ASK1 nKO reduced infarct sizes by 28.5% at 2 d after MCAO (p Conclusions: ASK1-dependent activation of p38α and STAT1 promotes inflammation, I/R brain injury, and maladaptive behavior by priming Mi/MΦ towards a neurotoxic phenotype.
- Published
- 2019
- Full Text
- View/download PDF
17. A Post-stroke Therapeutic Regimen with Omega-3 Polyunsaturated Fatty Acids that Promotes White Matter Integrity and Beneficial Microglial Responses after Cerebral Ischemia
- Author
-
Jun Chen, Yanqin Gao, Xiaoyan Jiang, Yejie Shi, Hongjian Pu, Zhishuo Wei, Wenting Zhang, Dandan Hong, and Xiaoming Hu
- Subjects
Male ,0301 basic medicine ,medicine.medical_specialty ,Time Factors ,Docosahexaenoic Acids ,Ischemia ,Nerve Tissue Proteins ,Hippocampal formation ,Corpus callosum ,Article ,Corpus Callosum ,Proinflammatory cytokine ,White matter ,Mice ,03 medical and health sciences ,0302 clinical medicine ,Antigens, CD ,Leukoencephalopathies ,Internal medicine ,medicine ,Animals ,Cell Proliferation ,chemistry.chemical_classification ,Microglia ,business.industry ,General Neuroscience ,Calcium-Binding Proteins ,Microfilament Proteins ,Myelin Basic Protein ,medicine.disease ,Mice, Inbred C57BL ,Stroke ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Endocrinology ,Eicosapentaenoic Acid ,chemistry ,Ischemic Attack, Transient ,Docosahexaenoic acid ,Cerebrovascular Circulation ,Neurology (clinical) ,Nervous System Diseases ,Cardiology and Cardiovascular Medicine ,business ,030217 neurology & neurosurgery ,Polyunsaturated fatty acid - Abstract
White matter injury induced by ischemic stroke elicits sensorimotor impairments, which can be further deteriorated by persistent proinflammatory responses. We previously reported that delayed and repeated treatments with omega-3 polyunsaturated fatty acids (n-3 PUFAs) improve spatial cognitive functions and hippocampal integrity after ischemic stroke. In the present study, we report a post-stroke n-3 PUFA therapeutic regimen that not only confers protection against neuronal loss in the gray matter but also promotes white matter integrity. Beginning 2 hours after 60 minutes of middle cerebral artery occlusion (MCAO), mice were randomly assigned to receive intraperitoneal docosahexaenoic acid (DHA) injections (10 mg/kg, daily for 14 days), alone or in combination with dietary fish oil (FO) supplements starting 5 days after MCAO. Sensorimotor functions, gray and white matter injury, and microglial responses were examined up to 28 days after MCAO. Our results showed that DHA and FO combined treatment facilitated long-term sensorimotor recovery and demonstrated greater beneficial effect than DHA injections alone. Mechanistically, n-3 PUFAs not only offered direct protection on white matter components, such as oligodendrocytes, but also potentiated microglial M2 polarization, which may be important for white matter repair. Notably, the improved white matter integrity and increased M2 microglia were strongly linked to the mitigation of sensorimotor deficits after stroke upon n-3 PUFA treatments. Together, our results suggest that post-stroke DHA injections in combination with FO dietary supplement benefit white matter restoration and microglial responses, thereby dictating long-term functional improvements.
- Published
- 2016
- Full Text
- View/download PDF
18. The effect of aging on brain injury and recovery after stroke
- Author
-
Selva Baltan, Jun Chen, Yejie Shi, and Richard F. Keep
- Subjects
medicine.medical_specialty ,Aging ,business.industry ,MEDLINE ,Recovery of Function ,medicine.disease ,lcsh:RC321-571 ,Stroke ,Physical medicine and rehabilitation ,Neurology ,Brain Injuries ,Medicine ,Animals ,Humans ,business ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Introductory Journal Article - Published
- 2019
19. White matter injury and microglia/macrophage polarization are strongly linked with age-related long-term deficits in neurological function after stroke
- Author
-
Sulaiman H Hassan, Yejie Shi, Xiaoming Hu, Hongjian Pu, Yanqin Gao, Mingyue Xu, Jun Chen, R. Anne Stetler, Rehana K. Leak, and Jun Suenaga
- Subjects
Brain Infarction ,Male ,Aging ,Pathology ,medicine.medical_specialty ,Time Factors ,Macrophage polarization ,Morris water navigation task ,Cell Count ,Motor Activity ,Article ,White matter ,Mice ,Developmental Neuroscience ,Antigens, CD ,Leukoencephalopathies ,Internal medicine ,medicine.artery ,medicine ,Animals ,Muscle Strength ,Young adult ,Maze Learning ,Stroke ,Macrophages ,Cell Polarity ,medicine.disease ,Pathophysiology ,Mice, Inbred C57BL ,Disease Models, Animal ,medicine.anatomical_structure ,Neurology ,Cerebral blood flow ,Cerebrovascular Circulation ,Middle cerebral artery ,Cardiology ,Microglia ,Nervous System Diseases ,Psychology - Abstract
Most of the successes in experimental models of stroke have not translated well to the clinic. One potential reason for this failure is that stroke mainly afflicts the elderly and the majority of experimental stroke studies rely on data gathered from young adult animals. Therefore, in the present study we established a reliable, reproducible model of stroke with low mortality in aged (18month) male mice and contrasted their pathophysiological changes with those in young (2month) animals. To this end, mice were subjected to permanent tandem occlusion of the left distal middle cerebral artery (dMCAO) with ipsilateral common carotid artery occlusion (CCAO). Cerebral blood flow (CBF) was evaluated repeatedly during and after stroke. Reduction of CBF was more dramatic and sustained in aged mice. Aged mice exhibited more severe long-term sensorimotor deficits, as manifested by deterioration of performance in the Rotarod and hanging wire tests up to 35d after stroke. Aged mice also exhibited significantly worse long-term cognitive deficits after stroke, as measured by the Morris water maze test. Consistent with these behavioral observations, brain infarct size and neuronal tissue loss after dMCAO were significantly larger in aged mice at 2d and 14d, respectively. The young versus aged difference in neuronal tissue loss, however, did not persist until 35d after dMCAO. In contrast to the transient difference in neuronal tissue loss, we found significant and long lasting deterioration of white matter in aged animals, as revealed by the loss of myelin basic protein (MBP) staining in the striatum at 35d after dMCAO. We further examined the expression of M1 (CD16/CD32) and M2 (CD206) markers in Iba-1(+) microglia by double immunofluorescent staining. In both young and aged mice, the expression of M2 markers peaked around 7d after stroke whereas the expression of M1 markers peaked around 14d after stroke, suggesting a progressive M2-to-M1 phenotype shift in both groups. However, aged mice exhibited significantly reduced M2 polarization compared to young adults. Remarkably, we discovered a strong positive correlation between favorable neurological outcomes after dMCAO and MBP levels or the number of M2 microglia/macrophages. In conclusion, our studies suggest that the distal MCAO stroke model consistently results in ischemic brain injury with long-term behavioral deficits, and is therefore suitable for the evaluation of long-term stroke outcomes. Furthermore, aged mice exhibit deterioration of functional outcomes after stroke and this deterioration is linked to white matter damage and reductions in M2 microglia/macrophage polarization.
- Published
- 2015
- Full Text
- View/download PDF
20. Inhibition of WNK3 Kinase Signaling Reduces Brain Damage and Accelerates Neurological Recovery After Stroke
- Author
-
Kristopher T. Kahle, Boris E. Shmukler, Seth L. Alper, Liaoliao Li, Shaoxia Wang, Sung-Sen Yang, Yejie Shi, Dandan Sun, Hui Yuan, Gulnaz Begum, and Shih-Hua Lin
- Subjects
Male ,Mice, 129 Strain ,Mice, Transgenic ,Stimulation ,Brain damage ,Protein Serine-Threonine Kinases ,Pharmacology ,Blood–brain barrier ,medicine.disease_cause ,Article ,Mice ,Pregnancy ,medicine ,Animals ,Protein kinase A ,Stroke ,Cells, Cultured ,Mice, Knockout ,Advanced and Specialized Nursing ,business.industry ,Kinase ,Recovery of Function ,medicine.disease ,medicine.anatomical_structure ,Brain Injuries ,Anesthesia ,Female ,Neurology (clinical) ,Nervous System Diseases ,medicine.symptom ,Signal transduction ,Cardiology and Cardiovascular Medicine ,business ,Oxidative stress ,Signal Transduction - Abstract
Background and Purpose— WNK kinases, including WNK3, and the associated downstream Ste20/SPS1-related proline-alanine–rich protein kinase (SPAK) and oxidative stress responsive 1 (OSR1) kinases, comprise an important signaling cascade that regulates the cation-chloride cotransporters. Ischemia-induced stimulation of the bumetanide-sensitive Na + -K + -Cl − cotransporter (NKCC1) plays an important role in the pathophysiology of experimental stroke, but the mechanism of its regulation in this context is unknown. Here, we investigated the WNK3-SPAK/OSR1 pathway as a regulator of NKCC1 stimulation and their collective role in ischemic brain damage. Method— Wild-type WNK3 and WNK3 knockout mice were subjected to ischemic stroke via transient middle cerebral artery occlusion. Infarct volume, brain edema, blood brain barrier damage, white matter demyelination, and neurological deficits were assessed. Total and phosphorylated forms of WNK3 and SPAK/OSR1 were assayed by immunoblotting and immunostaining. In vitro ischemia studies in cultured neurons and immature oligodendrocytes were conducted using the oxygen-glucose deprivation/reoxygenation method. Results— WNK3 knockout mice exhibited significantly decreased infarct volume and axonal demyelination, less cerebral edema, and accelerated neurobehavioral recovery compared with WNK3 wild-type mice subjected to middle cerebral artery occlusion. The neuroprotective phenotypes conferred by WNK3 knockout were associated with a decrease in stimulatory hyperphosphorylations of the SPAK/OSR1 catalytic T-loop and of NKCC1 stimulatory sites Thr 203 /Thr 207 /Thr 212 , as well as with decreased cell surface expression of NKCC1. Genetic inhibition of WNK3 or small interfering RNA knockdown of SPAK/OSR1 increased the tolerance of cultured primary neurons and oligodendrocytes to in vitro ischemia. Conclusions— These data identify a novel role for the WNK3-SPAK/OSR1-NKCC1 signaling pathway in ischemic neuroglial injury and suggest the WNK3-SPAK/OSR1 kinase pathway as a therapeutic target for neuroprotection after ischemic stroke.
- Published
- 2015
- Full Text
- View/download PDF
21. Oxidative stress and DNA damage after cerebral ischemia: Potential therapeutic targets to preserve the genome and improve stroke recovery
- Author
-
Rehana K. Leak, Yan Li, Jun Chen, Michael V. L. Bennett, R. Anne Stetler, Peiying Li, Yejie Shi, and Weifeng Yu
- Subjects
0301 basic medicine ,DNA damage ,DNA repair ,medicine.medical_treatment ,Biology ,medicine.disease_cause ,Article ,Brain Ischemia ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,XRCC1 ,0302 clinical medicine ,medicine ,Animals ,Humans ,Stroke ,Pharmacology ,Genome ,Base excision repair ,Recovery of Function ,medicine.disease ,Oxidative Stress ,030104 developmental biology ,Stroke recovery ,Neuroscience ,030217 neurology & neurosurgery ,Oxidative stress ,Nucleotide excision repair ,DNA Damage - Abstract
The past two decades have witnessed remarkable advances in oxidative stress research, particularly in the context of ischemic brain injury. Oxidative stress in ischemic tissues compromises the integrity of the genome, resulting in DNA lesions, cell death in neurons, glial cells, and vascular cells, and impairments in neurological recovery after stroke. As DNA is particularly vulnerable to oxidative attack, cells have evolved the ability to induce multiple DNA repair mechanisms, including base excision repair (BER), nucleotide excision repair (NER) and non-homogenous endpoint jointing (NHEJ). Defective DNA repair is tightly correlated with worse neurological outcomes after stroke, whereas upregulation of DNA repair enzymes, such as APE1, OGG1, and XRCC1, improves long-term functional recovery following stroke. Indeed, DNA damage and repair are now known to play critical roles in fundamental aspects of stroke recovery, such as neurogenesis, white matter recovery, and neurovascular unit remodeling. Several DNA repair enzymes are essential for comprehensive neural repair mechanisms after stroke, including Polβ and NEIL3 for neurogenesis, APE1 for white matter repair, Gadd45b for axonal regeneration, and DNA-PKs for neurovascular remodeling. This review discusses the emerging role of DNA damage and repair in functional recovery after stroke and highlights the contribution of DNA repair to regenerative elements after stroke. This article is part of the Special Issue entitled 'Cerebral Ischemia'.
- Published
- 2017
22. MicroRNA-15a/16-1 antagomir ameliorates ischemic brain injury in experimental stroke
- Author
-
Ping Sun, Ke-Jie Yin, Yejie Shi, Sulaiman H Hassan, Kai Liu, R. Anne Stetler, Xinxin Yang, Jun Chen, and Xuelian Tang
- Subjects
0301 basic medicine ,Male ,medicine.medical_specialty ,Pathology ,Inflammation ,Ischemic brain injury ,Article ,Brain Ischemia ,Brain ischemia ,03 medical and health sciences ,chemistry.chemical_compound ,Mice ,0302 clinical medicine ,Internal medicine ,microRNA ,medicine ,Animals ,In patient ,Antagomir ,Stroke ,Advanced and Specialized Nursing ,Mice, Knockout ,business.industry ,Antagomirs ,Infarction, Middle Cerebral Artery ,medicine.disease ,Mice, Inbred C57BL ,Disease Models, Animal ,MicroRNAs ,030104 developmental biology ,chemistry ,Potential biomarkers ,Cardiology ,Neurology (clinical) ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business ,030217 neurology & neurosurgery - Abstract
Background and Purpose— Dysregulation of the miR-15a/16-1 cluster in plasma has been reported in patients with stroke as a potential biomarker for diagnostic and prognostic use. However, the essential role and therapeutic potential of the miR-15a/16-1 cluster in ischemic stroke are poorly understood. This study is aimed at investigating the regulatory role of the miR-15a/16-1 cluster in ischemic brain injury and insight mechanisms. Methods— Adult male miR-15a/16-1 knockout and wild-type mice, or adult male C57 BL/6J mice injected via tail vein with the miR-15a/16-1–specific inhibitor (antagomir, 30 pmol/g), were subjected to 1 hour of middle cerebral artery occlusion and 72 hours of reperfusion. The neurological scores, brain infarct volume, brain water content, and neurobehavioral tests were then evaluated and analyzed. To explore underlying signaling pathways associated with alteration of miR-15a/16-1 activity, major proinflammatory cytokines were measured by quantitative polymerase chain reaction or ELISA and antiapoptotic proteins were examined by Western blotting. Results— Genetic deletion of the miR-15a/16-1 cluster or intravenous delivery of miR-15a/16-1 antagomir significantly reduced cerebral infarct size, decreased brain water content, and improved neurological outcomes in stroke mice. Inhibition of miR-15a/16-1 significantly decreased the expression of the proinflammatory cytokines interleukin-6, monocyte chemoattractant protein-1, vascular cell adhesion molecule 1, tumor necrosis factor alpha, and increased Bcl-2 and Bcl-w levels in the ischemic brain regions. Conclusions— Our data indicate that pharmacological inhibition of the miR-15a/16-1 cluster reduces ischemic brain injury via both upregulation of antiapoptotic proteins and suppression of proinflammatory molecules. These results suggest that the miR-15a/16-1 cluster is a novel therapeutic target for ischemic stroke.
- Published
- 2017
23. Abstract 49: Endothelial-targeted Overexpression of Heat Shock Protein 27 Ameliorates Rapid Blood Brain Barrier Impairment and Improves Long Term Outcomes After Ischemia and Reperfusion
- Author
-
Rehana K. Leak, Xiaoyan Jiang, Wenting Zhang, Xiaoming Hu, Michael V. L. Bennett, Yejie Shi, Richard F. Keep, Lili Zhang, Yanqin Gao, Jun Chen, and Hongjian Pu
- Subjects
Advanced and Specialized Nursing ,medicine.medical_specialty ,business.industry ,Ischemia ,Inflammation ,medicine.disease ,Blood–brain barrier ,Neurovascular bundle ,medicine.anatomical_structure ,nervous system ,Internal medicine ,Heat shock protein ,cardiovascular system ,medicine ,Long term outcomes ,Cardiology ,Neurology (clinical) ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,business ,Stroke - Abstract
Introduction: Blood brain barrier (BBB) damage resulting from ischemia/reperfusion (I/R) disrupts the neurovascular unit and leads to poor patient outcomes. We recently discovered that caveolin-1-independent subtle structural aberrations of brain microvascular endothelial cells (BMECs), such as abnormal actin polymerization, stress fiber formation and subsequent junctional protein (JP) disassembly, are a novel mechanism for rapid BBB breach after I/R. Hypothesis: Heat shock protein 27 (HSP27) reduces BBB breakdown after I/R by inhibiting actin polymerization and JP disassembly in BMECs. Methods: Neuron- or EC-specific HSP27-overexpressing mice were subjected to 1 h MCAO and reperfusion. BBB damage, tissue histology, and neurobehavioral performance were assessed up to 35 d after I/R. I/R-induced BBB damage was also simulated in BMEC cultures, where gene manipulations were achieved using lentiviral vectors. Cell-permeable TAT-HSP27 protein was injected i.v. into mice after I/R to rapidly elevate HSP27 in BMECs. Results: Targeted overexpression of HSP27 within ECs—but not neurons—was sufficient to reduce early (1-3 h) and late (24 h) BBB damage after I/R (p Conclusions: HSP27 protects against BBB disruption after I/R by inhibiting actin polymerization and JP disassembly in BMECs. HSP27 may be a therapy for ischemic stroke in conjunction with reperfusion.
- Published
- 2017
- Full Text
- View/download PDF
24. Omega-3 polyunsaturated fatty acids enhance cerebral angiogenesis and provide long-term protection after stroke
- Author
-
Rehana K. Leak, Jun Chen, Yanqin Gao, Ling Chen, Xiaoming Hu, Lili Zhang, Yejie Shi, Wengting Zhang, Jiayin Wang, and Feng Zhang
- Subjects
Fatty Acid Desaturases ,Time Factors ,Angiogenesis ,Ischemia ,Neovascularization, Physiologic ,Mice, Transgenic ,Pharmacology ,Biology ,Phospholipase ,Neuroprotection ,Article ,lcsh:RC321-571 ,Mice ,Fatty Acids, Omega-3 ,Reaction Time ,medicine ,Animals ,Caenorhabditis elegans Proteins ,Hypoxia ,Stroke ,lcsh:Neurosciences. Biological psychiatry. Neuropsychiatry ,Cells, Cultured ,Cell Proliferation ,Cerebral Cortex ,chemistry.chemical_classification ,Omega-3 polyunsaturated fatty acids ,Angiopoietin 2 ,medicine.disease ,Coculture Techniques ,Neural stem cell ,Mice, Inbred C57BL ,Disease Models, Animal ,Glucose ,Neuroprotective Agents ,medicine.anatomical_structure ,Neurology ,chemistry ,Ischemic Attack, Transient ,Nervous System Diseases ,Astrocyte ,Neuroscience ,Polyunsaturated fatty acid - Abstract
Stroke is a devastating neurological disorder and one of the leading causes of death and serious disability. After cerebral ischemia, revascularization in the ischemic boundary zone provides nutritive blood flow as well as various growth factors to promote the survival and activity of neurons and neural progenitor cells. Enhancement of angiogenesis and the resulting improvement of cerebral microcirculation are key restorative mechanisms and represent an important therapeutic strategy for ischemic stroke. In the present study, we tested the hypothesis that post-stroke angiogenesis would be enhanced by omega-3 polyunsaturated fatty acids (n-3 PUFAs), a major component of dietary fish oil. To this end, we found that transgenic fat-1 mice that overproduce n-3 PUFAs exhibited long-term behavioral and histological protection against transient focal cerebral ischemia (tFCI). Importantly, fat-1 transgenic mice also exhibited robust improvements in revascularization and angiogenesis compared to wild type littermates, suggesting a potential role for n-3 fatty acids in post-stroke cerebrovascular remodeling. Mechanistically, n-3 PUFAs induced upregulation of angiopoietin 2 (Ang 2) in astrocytes after tFCI and stimulated extracellular Ang 2 release from cultured astrocytes after oxygen and glucose deprivation. Ang 2 facilitated endothelial proliferation and barrier formation in vitro by potentiating the effects of VEGF on phospholipase Cγ1 and Src signaling. Consistent with these findings, blockade of Src activity in post-stroke fat-1 mice impaired n-3 PUFA-induced angiogenesis and exacerbated long-term neurological outcomes. Taken together, our findings strongly suggest that n-3 PUFA supplementation is a potential angiogenic treatment capable of augmenting brain repair and improving long-term functional recovery after cerebral ischemia.
- Published
- 2014
25. Proton-sensitive cation channels and ion exchangers in ischemic brain injury: New therapeutic targets for stroke?
- Author
-
Tiandong Leng, Zhi-Gang Xiong, Yejie Shi, and Dandan Sun
- Subjects
Sodium-Hydrogen Exchanger 1 ,Sodium-Hydrogen Exchangers ,business.industry ,General Neuroscience ,Glutamate receptor ,Ischemia ,TRPM Cation Channels ,medicine.disease ,Neuroprotection ,Article ,Brain Ischemia ,Acid Sensing Ion Channels ,Transient receptor potential channel ,TRPM ,TRPM7 ,Brain Injuries ,Ca2+/calmodulin-dependent protein kinase ,medicine ,Animals ,Humans ,business ,Cation Transport Proteins ,Stroke ,Neuroscience - Abstract
Ischemic brain injury results from complicated cellular mechanisms. The present therapy for acute ischemic stroke is limited to thrombolysis with the recombinant tissue plasminogen activator (rtPA) and mechanical recanalization. Therefore, a better understanding of ischemic brain injury is needed for the development of more effective therapies. Disruption of ionic homeostasis plays an important role in cell death following cerebral ischemia. Glutamate receptor-mediated ionic imbalance and neurotoxicity have been well established in cerebral ischemia after stroke. However, non-NMDA receptor-dependent mechanisms, involving acid-sensing ion channel 1a (ASIC1a), transient receptor potential melastatin 7 (TRPM7), and Na(+)/H(+) exchanger isoform 1 (NHE1), have recently emerged as important players in the dysregulation of ionic homeostasis in the CNS under ischemic conditions. These H(+)-sensitive channels and/or exchangers are expressed in the majority of cell types of the neurovascular unit. Sustained activation of these proteins causes excessive influx of cations, such as Ca(2+), Na(+), and Zn(2+), and leads to ischemic reperfusion brain injury. In this review, we summarize recent pre-clinical experimental research findings on how these channels/exchangers are regulated in both in vitro and in vivo models of cerebral ischemia. The blockade or transgenic knockdown of these proteins was shown to be neuroprotective in these ischemia models. Taken together, these non-NMDA receptor-dependent mechanisms may serve as novel therapeutic targets for stroke intervention.
- Published
- 2014
- Full Text
- View/download PDF
26. The interleukin-4/PPARγ signaling axis promotes oligodendrocyte differentiation and remyelination after brain injury
- Author
-
Jie Chen, Lesley M. Foley, Yanqin Gao, Hongfeng Mu, T. Kevin Hitchens, Rehana K. Leak, Xiaoming Hu, Wei Cai, Xuejiao Dai, Jun Chen, Fei Xu, Yejie Shi, Xiangrong Liu, Fang Yu, Wen Zhu, Ligen Shi, and Qingxiu Zhang
- Subjects
Male ,Central Nervous System ,0301 basic medicine ,Receptor expression ,Immunostaining ,Nervous System ,Vascular Medicine ,Brain Ischemia ,Diagnostic Radiology ,Mice ,Nerve Fibers ,0302 clinical medicine ,Animal Cells ,Medicine and Health Sciences ,Brain Damage ,Biology (General) ,Myelin Sheath ,Mice, Knockout ,Neurons ,Staining ,Cerebral Ischemia ,Brain Mapping ,Microglia ,Radiology and Imaging ,General Neuroscience ,Neurogenesis ,Cell Differentiation ,White Matter ,Magnetic Resonance Imaging ,Cell biology ,Stroke ,Oligodendroglia ,Diffusion Tensor Imaging ,medicine.anatomical_structure ,Neurology ,Anatomy ,Cellular Types ,medicine.symptom ,General Agricultural and Biological Sciences ,Signal Transduction ,Research Article ,Imaging Techniques ,QH301-705.5 ,Cerebrovascular Diseases ,Brain Morphometry ,Central nervous system ,Neuroimaging ,Brain damage ,Biology ,Research and Analysis Methods ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,Diagnostic Medicine ,medicine ,Animals ,Remyelination ,Ischemic Stroke ,General Immunology and Microbiology ,Oligodendrocyte differentiation ,Biology and Life Sciences ,Recovery of Function ,Cell Biology ,Axons ,Oligodendrocyte ,Nerve Regeneration ,Mice, Inbred C57BL ,PPAR gamma ,030104 developmental biology ,Specimen Preparation and Treatment ,Brain Injuries ,Cellular Neuroscience ,Interleukin-4 ,030217 neurology & neurosurgery ,Demyelinating Diseases ,Neuroscience - Abstract
The repair of white matter damage is of paramount importance for functional recovery after brain injuries. Here, we report that interleukin-4 (IL-4) promotes oligodendrocyte regeneration and remyelination. IL-4 receptor expression was detected in a variety of glial cells after ischemic brain injury, including oligodendrocyte lineage cells. IL-4 deficiency in knockout mice resulted in greater deterioration of white matter over 14 d after stroke. Consistent with these findings, intranasal delivery of IL-4 nanoparticles after stroke improved white matter integrity and attenuated long-term sensorimotor and cognitive deficits in wild-type mice, as revealed by histological immunostaining, electron microscopy, diffusion tensor imaging, and electrophysiology. The selective effect of IL-4 on remyelination was verified in an ex vivo organotypic model of demyelination. By leveraging primary oligodendrocyte progenitor cells (OPCs), microglia-depleted mice, and conditional OPC-specific peroxisome proliferator-activated receptor gamma (PPARγ) knockout mice, we discovered a direct salutary effect of IL-4 on oligodendrocyte differentiation that was mediated by the PPARγ axis. Our findings reveal a new regenerative role of IL-4 in the central nervous system (CNS), which lies beyond its known immunoregulatory functions on microglia/macrophages or peripheral lymphocytes. Therefore, intranasal IL-4 delivery may represent a novel therapeutic strategy to improve white matter integrity in stroke and other brain injuries., The repair and remyelination of white matter are of paramount importance for functional recovery after brain injuries. This study shows that interleukin-4 plays an essential role in oligodendrocyte differentiation and long-term white matter recovery, beyond its well-known immunoregulatory functions, and is mediated by the PPARγ axis.
- Published
- 2019
- Full Text
- View/download PDF
27. Corrigendum to 'Brain ischemic preconditioning protects against ischemic injury and preserves the blood-brain barrier via oxidative signaling and Nrf2 activation' [Redox Biol. 17 (2018) 323–337]
- Author
-
Tuo Yang, Qianqian Li, Rehana K. Leak, Yejie Shi, Jun Chen, Lili Zhang, Yang Sun, Leilei Mao, Meijuan Zhang, and Feng Zhang
- Subjects
NF-E2-Related Factor 2 ,Clinical Biochemistry ,Oxidative phosphorylation ,Pharmacology ,Blood–brain barrier ,Biochemistry ,Redox ,Brain Ischemia ,Nrf2 activation ,Mice ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Humans ,Ischemic Preconditioning ,030304 developmental biology ,0303 health sciences ,Glycogen Synthase Kinase 3 beta ,Kelch-Like ECH-Associated Protein 1 ,business.industry ,Organic Chemistry ,Ischemic injury ,Stroke ,Disease Models, Animal ,medicine.anatomical_structure ,Gene Expression Regulation ,Blood-Brain Barrier ,Proteolysis ,Ischemic preconditioning ,Lipid Peroxidation ,Corrigendum ,business ,030217 neurology & neurosurgery - Abstract
Brain ischemic preconditioning (IPC) with mild ischemic episodes is well known to protect the brain against subsequent ischemic challenges. However, the underlying mechanisms are poorly understood. Here we demonstrate the critical role of the master redox transcription factor, nuclear factor (erythroid-derived 2)-like 2 (Nrf2), in IPC-mediated neuroprotection and blood-brain barrier (BBB) preservation. We report that IPC causes generation of endogenous lipid electrophiles, including 4-hydroxy-2-nonenal (4-HNE), which release Nrf2 from inhibition by Keap1 (via Keap1-C288) and inhibition by glycogen synthase kinase 3β (via GSK3β-C199). Nrf2 then induces expression of its target genes, including a new target, cadherin 5, a key component of adherens junctions of the BBB. These effects culminate in mitigation of BBB leakage and of neurological deficits after stroke. Collectively, these studies are the first to demonstrate that IPC protects the BBB against ischemic injury by generation of endogenous electrophiles and activation of the Nrf2 pathway through inhibition of Keap1- and GSK3β-dependent Nrf2 degradation.
- Published
- 2019
- Full Text
- View/download PDF
28. Blood-brain barrier dysfunction and recovery after ischemic stroke
- Author
-
Jun Chen, Michael V. L. Bennett, Tuo Yang, Xiaoyan Jiang, Richard F. Keep, Anuska V. Andjelkovic, Ling Zhu, and Yejie Shi
- Subjects
0301 basic medicine ,Inflammation ,Blood–brain barrier ,HMGB1 ,Article ,Brain Ischemia ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Humans ,PI3K/AKT/mTOR pathway ,biology ,General Neuroscience ,Neurovascular bundle ,Extravasation ,Stroke ,030104 developmental biology ,medicine.anatomical_structure ,nervous system ,Blood-Brain Barrier ,biology.protein ,cardiovascular system ,medicine.symptom ,Psychology ,Neuroscience ,030217 neurology & neurosurgery ,Homeostasis - Abstract
The blood-brain barrier (BBB) plays a vital role in regulating the trafficking of fluid, solutes and cells at the blood-brain interface and maintaining the homeostatic microenvironment of the CNS. Under pathological conditions, such as ischemic stroke, the BBB can be disrupted, followed by the extravasation of blood components into the brain and compromise of normal neuronal function. This article reviews recent advances in our knowledge of the mechanisms underlying BBB dysfunction and recovery after ischemic stroke. CNS cells in the neurovascular unit, as well as blood-borne peripheral cells constantly modulate the BBB and influence its breakdown and repair after ischemic stroke. The involvement of stroke risk factors and comorbid conditions further complicate the pathogenesis of neurovascular injury by predisposing the BBB to anatomical and functional changes that can exacerbate BBB dysfunction. Emphasis is also given to the process of long-term structural and functional restoration of the BBB after ischemic injury. With the development of novel research tools, future research on the BBB is likely to reveal promising potential therapeutic targets for protecting the BBB and improving patient outcome after ischemic stroke.
- Published
- 2016
29. APE1/Ref-1 facilitates recovery of gray and white matter and neurological function after mild stroke injury
- Author
-
Yanqin Gao, Yejie Shi, Feng Zhang, Gregg E. Homanics, Michael V. L. Bennett, Jun Chen, Hongjian Pu, Rehana K. Leak, Sulaiman H Hassan, Guodong Cao, Xiaoming Hu, Zhongfang Weng, Carolyn Ferguson, R. Anne Stetler, and Lili Zhang
- Subjects
0301 basic medicine ,Central nervous system ,Ischemia ,Cre recombinase ,Biology ,White matter ,03 medical and health sciences ,Mice ,0302 clinical medicine ,Conditional gene knockout ,medicine ,DNA-(Apurinic or Apyrimidinic Site) Lyase ,Animals ,Gray Matter ,Mice, Knockout ,Multidisciplinary ,Behavior, Animal ,Neurodegeneration ,medicine.disease ,DNA-(apurinic or apyrimidinic site) lyase ,White Matter ,Oligodendrocyte ,Mice, Inbred C57BL ,Stroke ,030104 developmental biology ,medicine.anatomical_structure ,PNAS Plus ,Neuroscience ,030217 neurology & neurosurgery - Abstract
A major hallmark of oxidative DNA damage after stroke is the induction of apurinic/apyrimidinic (AP) sites and strand breaks. To mitigate cell loss after oxidative DNA damage, ischemic cells rapidly engage the base excision-repair proteins, such as the AP site-repairing enzyme AP endonuclease-1 (APE1), also named redox effector factor-1 (Ref-1). Although forced overexpression of APE1 is known to protect against oxidative stress-induced neurodegeneration, there is no concrete evidence demonstrating a role for endogenous APE1 in the long-term recovery of gray and white matter following ischemic injury. To address this gap, we generated, to our knowledge, the first APE1 conditional knockout (cKO) mouse line under control of tamoxifen-dependent Cre recombinase. Using a well-established model of transient focal cerebral ischemia (tFCI), we show that induced deletion of APE1 dramatically enlarged infarct volume and impaired the recovery of sensorimotor and cognitive deficits. APE1 cKO markedly increased postischemic neuronal and oligodendrocyte degeneration, demonstrating that endogenous APE1 preserves both gray and white matter after tFCI. Because white matter repair is instrumental in behavioral recovery after stroke, we also examined the impact of APE1 cKO on demyelination and axonal conduction and discovered that APE1 cKO aggravated myelin loss and impaired neuronal communication following tFCI. Furthermore, APE1 cKO increased AP sites and activated the prodeath signaling proteins, PUMA and PARP1, after tFCI in topographically distinct manners. Our findings provide evidence that endogenous APE1 protects against ischemic infarction in both gray and white matter and facilitates the functional recovery of the central nervous system after mild stroke injury.
- Published
- 2016
30. Microglia: A Double-Sided Sword in Stroke
- Author
-
Xiaoming Hu, Hong Shi, Yejie Shi, Jun Chen, Yanqin Gao, and Mingyue Xu
- Subjects
Microglia ,business.industry ,Angiogenesis ,Neurogenesis ,Central nervous system ,medicine.disease ,medicine.anatomical_structure ,Immune system ,medicine ,Signal transduction ,business ,Receptor ,Neuroscience ,Stroke - Abstract
Microglia are the residential immune cells in the central nervous system (CNS). They serve as the first line of defense against CNS injuries such as ischemic stroke. Microglia express a wide range of surface receptors, which control the “On” or “Off” responses of microglia and maintain their functional homeostasis. Upon activation, these highly plastic cells may assume diverse phenotypes and play dualistic roles in brain injury and recovery. In this review, we describe the main surface receptors that involve in microglial activation after stroke, with a focus on their engagement of distinct functional programs. We also discuss the different roles of activated microglia in ischemic brain injury and post-injury brain repair. Further identification of the microglial receptors and/or signaling pathways that are in charge of functional phenotype switch is essential for the research in the stroke field and for the identification of therapeutic targets.
- Published
- 2016
- Full Text
- View/download PDF
31. Translational Stroke Research on Blood-Brain Barrier Damage: Challenges, Perspectives, and Goals
- Author
-
Jun Chen, Yejie Shi, Richard F. Keep, and Rehana K. Leak
- Subjects
0301 basic medicine ,medicine.medical_specialty ,Neurology ,Heart disease ,Population ,Ischemia ,Article ,Translational Research, Biomedical ,03 medical and health sciences ,0302 clinical medicine ,Internal medicine ,Diabetes mellitus ,medicine ,Animals ,Humans ,education ,Stroke ,Cause of death ,Intracerebral hemorrhage ,education.field_of_study ,business.industry ,General Neuroscience ,medicine.disease ,030104 developmental biology ,Blood-Brain Barrier ,Cardiology ,Neurology (clinical) ,Cardiology and Cardiovascular Medicine ,business ,030217 neurology & neurosurgery - Abstract
Over the past few decades, basic and clinical research has identified numerous risk factors for the development of stroke and led to major improvements in health management in the USA. As a result of these efforts, the relative rate of stroke death dropped by 33.7 %, and the actual occurrence of stroke deaths fell by 18.2 % in the decade spanning from 2003 to 2013, according to the American Heart Association. Thus, stroke fell from the fourth to the fifth leading cause of death in 2013, behind heart disease, cancer, chronic lower respiratory diseases, and unintentional injuries. These improvements are largely attributed to superior control of hypertension, diabetes mellitus, high cholesterol, and tobacco use [1]. To date, the treatment of acute ischemic stroke is largely dependent on recanalization using recombinant tissue-type plasminogen activator (tPA) in the appropriate patient population [2, 3]. Encouragingly, recent clinical trials have demonstrated significant benefits for intra-arterial thrombectomy in a subset of acute stroke patients with intracranial large artery occlusion [4]. Despite these improvements in population health and stroke treatment, stroke still remains a leading cause of longterm disability and approximately 795,000 people experience a new or recurrent stroke every year [1]. Thus, basic and clinical investigations of the mechanisms underlying ischemic brain injurymust remain an urgent priority in order to promote the discovery of novel therapeutic targets and improve the safety and efficacy of current tPA and thrombectomy treatments. During and after ischemic stroke, loss of blood-brain barrier (BBB) integrity is a prominent pathological event that contributes to further evolution of the injury. BBB dysfunction is also a hallmark of intracerebral hemorrhage [5, 6]. Despite its obvious clinical relevance, BBB protection has received much less attention than is warranted. An impaired BBB not only facilitates the development of brain edema and neuroinflammation, but also increases the risk of lethal hemorrhagic transformation during thrombolysis, thereby limiting the use of tPA and leading to poor patient outcomes [7, 8]. Recent advances in stroke telemedicine provide an effective and promising method to increase the use of tPA therapy [9], which, together with the growing application of thrombectomy, is likely to improve post-ischemia reperfusion in a larger population of stroke patients in the near future. As this treatment method works better when the BBB remains intact, therapeutic strategies aimed at neurovascular unit protection and prevention of BBB damage after ischemia/reperfusion (I/R) need to be better prioritized in stroke research. In earlier reports, it was widely held that all forms of BBB rupture after I/R were the direct consequence of matrix metalloproteinase (MMP)-mediated degradation of endothelial intercellular junctions and basal lamina [10–14]. However, recent animal models of stroke have revealed a complex, biphasic temporal profile of BBB breakdown, with an immediate phase of early BBB hyperpermeability 4–6 h after stroke, followed by a delayed opening of the BBB 48–72 h after stroke. In recent years, the availability of advanced imaging techniques and novel transgenic animal models have greatly facilitated research on BBB dysfunction after stroke, with * Jun Chen chenj2@upmc.edu
- Published
- 2015
32. Protease-independent action of tissue plasminogen activator in brain plasticity and neurological recovery after ischemic stroke.
- Author
-
Hongjian Pu, Yejie Shi, Lili Zhang, Zhengyu Lu, Qing Ye, Leak, Rehana K., Fei Xu, Shubei Ma, Hongfeng Mu, Zhishuo Wei, Na Xu, Yuguo Xia, Xiaoming Hu, Hitchens, T. Kevin, Bennett, Michael V. L., and Jun Chen
- Subjects
- *
PROTEOLYTIC enzymes , *TISSUE plasminogen activator , *CIRCULATING anticoagulants , *ISCHEMIA , *STROKE , *WHITE matter (Nerve tissue) , *LABORATORY mice - Abstract
Emerging evidence suggests that tissue plasminogen activator (tPA), currently the only FDA-approved medication for ischemic stroke, exerts important biological actions on the CNS besides its well-known thrombolytic effect. In this study, we investigated the role of tPA on primary neurons in culture and on brain recovery and plasticity after ischemic stroke in mice. Treatment with recombinant tPA stimulated axonal growth in culture, an effect independent of its protease activity and achieved through epidermal growth factor receptor (EGFR) signaling. After permanent focal cerebral ischemia, tPA knockout mice developed more severe sensorimotor and cognitive deficits and greater axonal and myelin injury than wildtype mice, suggesting that endogenously expressed tPA promotes longterm neurological recovery after stroke. In tPA knockoutmice, intranasal administration of recombinant tPA protein 6 hours poststroke and 7 more times at 2 d intervals mitigated white matter injury, improved axonal conduction, and enhanced neurological recovery. Consistent with the proaxonal growth effects observed in vitro, exogenous tPA delivery increased poststroke axonal sprouting of corticobulbar and corticospinal tracts, which might have contributed to restoration of neurological functions. Notably, recombinant mutant tPA-S478A lacking protease activity (but retaining the EGF-like domain) was as effective aswild-type tPA in rescuing neurological functions in tPA knockout stroke mice. These findings demonstrate that tPA improves long-term functional outcomes in a clinically relevant stroke model, likely by promoting brain plasticity through EGFR signaling. Therefore, treatment with the protease-dead recombinant tPA-S478A holds particular promise as a neurorestorative therapy, as the risk for triggering intracranial hemorrhage is eliminated and tPA-S478A can be delivered intranasally hours after stroke. [ABSTRACT FROM AUTHOR]
- Published
- 2019
- Full Text
- View/download PDF
33. Intracellular pH reduction prevents excitotoxic and ischemic neuronal death by inhibiting NADPH oxidase
- Author
-
Dandan Sun, Angela M. Brennan-Minnella, Colleen Hefner, Yejie Shi, Yiguo Shen, Seok Joon Won, Raymond A. Swanson, and Tina I. Lam
- Subjects
Intracellular Fluid ,Sodium-Hydrogen Exchangers ,Intracellular pH ,Cells ,Real-Time Polymerase Chain Reaction ,Receptors, N-Methyl-D-Aspartate ,Fluorescence ,chemistry.chemical_compound ,Mice ,NOX2 ,Superoxides ,Receptors ,Animals ,Cation Transport Proteins ,Cells, Cultured ,DNA Primers ,Neurons ,cariporide ,Analysis of Variance ,Multidisciplinary ,NADPH oxidase ,Cultured ,Sodium-Hydrogen Exchanger 1 ,biology ,Cariporide ,Cell Death ,Superoxide ,Intracellular pH reduction ,Glutamate receptor ,Neurosciences ,Brain ,NADPH Oxidases ,Hydrogen-Ion Concentration ,Cell biology ,Brain Disorders ,Stroke ,PNAS Plus ,nervous system ,chemistry ,Neurological ,biology.protein ,NMDA receptor ,acidosis ,Intracellular ,Hv1 ,N-Methyl-D-Aspartate - Abstract
Sustained activation of N-methyl-d-aspartate (NMDA) -type glutamate receptors leads to excitotoxic neuronal death in stroke, brain trauma, and neurodegenerative disorders. Superoxide production by NADPH oxidase is a requisite event in the process leading from NMDA receptor activation to excitotoxic death. NADPH oxidase generates intracellular H(+) along with extracellular superoxide, and the intracellular H(+) must be released or neutralized to permit continued NADPH oxidase function. In cultured neurons, NMDA-induced superoxide production and neuronal death were prevented by intracellular acidification by as little as 0.2 pH units, induced by either lowered medium pH or by inhibiting Na(+)/H(+) exchange. In mouse brain, superoxide production induced by NMDA injections or ischemia-reperfusion was likewise prevented by inhibiting Na(+)/H(+) exchange and by reduced expression of the Na(+)/H(+) exchanger-1 (NHE1). Neuronal intracellular pH and neuronal Na(+)/H(+) exchange are thus potent regulators of excitotoxic superoxide production. These findings identify a mechanism by which cell metabolism can influence coupling between NMDA receptor activation and superoxide production.
- Published
- 2013
34. In Vivo Expansion of Regulatory T Cells with IL-2/IL-2 Antibody Complex Protects against Transient Ischemic Stroke.
- Author
-
Haiyue Zhang, Yuguo Xia, Qing Ye, Fang Yu, Wen Zhu, Peiying Li, Zhishuo Wei, Yuanyuan Yang, Yejie Shi, Thomson, Angus W., Jun Chen, and Xiaoming Hu
- Subjects
T cells ,CORONARY disease ,IMMUNOGLOBULINS ,STROKE ,ARTERIAL occlusions - Abstract
Regulatory T cells (Tregs) are known to protect against ischemic stroke. However, the low frequency of Tregs restricts their clinical utility. This study investigated whether expanding the number of Tregs in vivo with the IL-2/IL-2 antibody complex (IL-2/IL-2Ab) could improve stroke outcomes and further elaborated the mechanisms of protection in male mice. C57BL/6 mice received IL-2/IL-2Ab or isotype IgG (IsoAb) intraperitoneally for 3 d before (pretreatment) or starting 2 h after (posttreatment) 60 min middle cerebral artery occlusion (MCAO). IL-2/IL-2Ab selectively increased the number of Tregs in the blood, spleen, and lymph nodes. The IL-2/IL-2Ab treatment significantly reduced infarct volume, inhibited neuroinflammation, and improved sensorimotor functions, as manifested by rotarod test and foot fault test, compared with IsoAb-treated stroke mice. Treg depletion was then achieved by diphtheria toxin (DT) injection into transgenic mice expressing the DT receptor under the control of the Foxp3 promoter (DTR mice). The depletion of Tregs completely eliminated IL-2/IL-2Ab-afforded neuroprotection. Interestingly, adoptive transfer of Tregs collected from IL-2/IL-2Ab-treated mice demonstrated more potent neuroprotection than an equal number of Tregs prepared from IsoAb-treated mice, suggesting that IL-2/IL- 2Ab not only elevated Treg numbers, but also boosted their functions. Mechanistically, IL-2/IL-2Ab promoted the expression of CD39 and CD73 in expanded Tregs. CD73 deficiency diminished the protective effect of IL-2/IL-2Ab-stimulated Tregs in stroke mice. The results show that IL-2/IL-2Ab expands Tregs in vivo and boosts their immunomodulatory function. The activation of CD39/CD73 signaling in Tregs may participate as a potential mechanism underlying IL-2/IL-2Ab-afforded neuroprotection against ischemic brain injury. [ABSTRACT FROM AUTHOR]
- Published
- 2018
- Full Text
- View/download PDF
35. APE1/Ref-1 facilitates recovery of gray and white matter and neurological function after mild stroke injury.
- Author
-
Stetler, R. Anne, Yanqin Gao, Leak, Rehana K., Zhongfang Weng, Yejie Shi, Lili Zhang, Hongjian Pu, Feng Zhang, Xiaoming Hu, Hassan, Sulaiman, Ferguson, Carolyn, Homanics, Gregg E., Guodong Cao, Bennett, Michael V. L., and Jun Chen
- Subjects
STROKE ,DNA damage ,ENDONUCLEASES ,NEURODEGENERATION ,TAMOXIFEN ,CENTRAL nervous system - Abstract
A major hallmark of oxidative DNA damage after stroke is the induction of apurinic/apyrimidinic (AP) sites and strand breaks. To mitigate cell loss after oxidative DNA damage, ischemic cells rapidly engage the base excision-repair proteins, such as the AP site-repairing enzyme AP endonuclease-1 (APE1), also named redox effector factor-1 (Ref-1). Although forced overexpression of APE1 is known to protect against oxidative stress-induced neurodegeneration, there is no concrete evidence demonstrating a role for endogenous APE1 in the long-term recovery of gray and white matter following ischemic injury. To address this gap, we generated, to our knowledge, the first APE1 conditional knockout (cKO) mouse line under control of tamoxifen-dependent Cre recombinase. Using a well-established model of transient focal cerebral ischemia (tFCI), we show that induced deletion of APE1 dramatically enlarged infarct volume and impaired the recovery of sensorimotor and cognitive deficits. APE1 cKO markedly increased postischemic neuronal and oligodendrocyte degeneration, demonstrating that endogenous APE1 preserves both gray and white matter after tFCI. Because white matter repair is instrumental in behavioral recovery after stroke, we also examined the impact of APE1 cKO on demyelination and axonal conduction and discovered that APE1 cKO aggravated myelin loss and impaired neuronal communication following tFCI. Furthermore, APE1 cKO increased AP sites and activated the prodeath signaling proteins, PUMA and PARP1, after tFCI in topographically distinct manners. Our findings provide evidence that endogenous APE1 protects against ischemic infarction in both gray and white matter and facilitates the functional recovery of the central nervous system after mild stroke injury. [ABSTRACT FROM AUTHOR]
- Published
- 2016
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.