1
|
Li X, Simo L, Zhao Q, Kim EG, Ding Y, Geng X. Endothelial Cells and the Blood-Brain Barrier: Critical Determinants of Ineffective Reperfusion in Stroke. Eur J Neurosci 2025; 61:e16663. [PMID: 39935212 DOI: 10.1111/ejn.16663] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 11/05/2024] [Indexed: 02/13/2025]
Abstract
Ineffective reperfusion remains a critical challenge in neurointerventional treatment following ischemic stroke, with the integrity of the blood-brain barrier (BBB) being a key determinant of patient outcomes. This review explores the distinctive characteristics and roles of brain endothelial cells (ECs) in the context of stroke and ineffective reperfusion. We examine the unique properties of brain ECs compared to their counterparts in other tissues, focusing on their pathophysiological changes, functional impairments and the inflammatory cascades that follow stroke. Differences in gene expression between brain ECs and those in other organs offer deeper insights into their role in neuroprotective therapies. Additionally, drawing parallels between brain ECs and ECs from organs with similar ischemia-reperfusion injury profiles may inspire novel therapeutic approaches. This review highlights the critical importance of understanding the nuanced roles of ECs in BBB regulation, which ultimately impacts reperfusion outcomes.
Collapse
Affiliation(s)
- Xiang Li
- Luhe Institute of Neuroscience, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Leticia Simo
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Qianhui Zhao
- Luhe Institute of Neuroscience, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Enoch Gene Kim
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Yuchuan Ding
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, USA
| | - Xiaokun Geng
- Luhe Institute of Neuroscience, Capital Medical University, Beijing, China
- Department of Neurology, Beijing Luhe Hospital, Capital Medical University, Beijing, China
- Department of Neurosurgery, Wayne State University School of Medicine, Detroit, Michigan, USA
| |
Collapse
|
2
|
Chen H, Yuan Y, Zhang Y, Liu X, Chen Q, Liu C, Yao Q. Activation of the LKB1/AMPK/HIF-1α Pathway by Metformin to Promote Neovascularisation in Cerebral Ischaemia. Neurochem Res 2024; 49:3263-3276. [PMID: 39240424 DOI: 10.1007/s11064-024-04235-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 08/28/2024] [Accepted: 08/28/2024] [Indexed: 09/07/2024]
Abstract
As a difficult-to-treat neurological condition, cerebral ischemia is currently limited to treatments such as intravenous recombinant tissue plasminogen activator thrombolysis and thrombectomy. Metformin, a potent antidiabetic drug, has been reported to have an independent function in enhancing the prognosis of stroke patients, in addition to its glucose-lowering effects. However, the mechanism of action of metformin in this context remains unclear. In vivo, a rat model of permanent middle cerebral artery occlusion was established, and after administration of a low dose of 10.5 mg/mL metformin, infarct area was measured by TTC staining, and cortical blood flow was determined by laser Doppler imaging. In vitro, the study established human umbilical vein endothelial cells treated with cobalt chloride. Immunofluorescence, immunohistochemistry, and Western blot experiments were performed to observe the expression of angiogenic factors, tight junction proteins, and apoptotic factors. A TUNEL assay was utilized to appraise cell death by apoptosis. A tube formation assay and scratch assay were conducted to determine the endothelial neovascularization status. Animal experiments have revealed that the administration of the AMPK activator metformin significantly reduced the infarct area, promoted the expression of angiogenic factors, and maintained the stability of tight junction proteins in endothelial cells. Moreover, metformin reduces nerve cells apoptosis by affecting the expression of the apoptotic protein cleaved-caspase3 via the HIF-1α pathway. In vitro, the LKB1/AMPK signaling pathway is activated after hypoxic stimulation, attaining its peak within the early stages of hypoxia (1-12 h) and gradually weakening thereafter. The administration of AMPK pharmacological agonists (between 36 and 48 h) can enhance AMPK activity, which can lead to the expression of angiogenic factors, maintain the stability of tight-junction proteins in endothelial cells, and facilitate endothelial cell migration and vascular structure formation. Conversely, the AMPK inhibitors exert the opposite effects. The activation of the LKB1/AMPK/HIF-1α signaling pathway by metformin in cerebral ischemia contributes to angiogenesis, promotes tissue repair in the injured area, and enhances neurologically functional symptoms.
Collapse
Affiliation(s)
- Hongguang Chen
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437000, Hubei, China
| | - Yuting Yuan
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437000, Hubei, China
- School of Pharmacy, Xianning Medical College, Hubei University of Science and Technology, Xianning, Hubei, China
| | - Yue Zhang
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437000, Hubei, China
| | - Xiufen Liu
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437000, Hubei, China
| | - Qingjie Chen
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437000, Hubei, China.
| | - Chao Liu
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437000, Hubei, China.
| | - Qing Yao
- Hubei Key Laboratory of Diabetes and Angiopathy, Medicine Research Institute, Xianning Medical College, Hubei University of Science and Technology, Xianning, 437000, Hubei, China.
| |
Collapse
|
3
|
Yang J, Yang C, Yang Y, Jia N, Sun Q, Ji S. Endothelial Protection of Vasoactive Intestinal Peptide Enhances Angiogenesis Mediated by eNOS Pathway Following Focal Cerebral Ischemia in Rats. Int J Pept Res Ther 2022. [DOI: 10.1007/s10989-022-10434-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
4
|
Hu Y, Liang Y, Tian H, Xu C, Yu D, Zhang P, Ye H, Li M. Microplitis bicoloratus bracovirus regulates cyclophilin A-apoptosis-inducing factor interaction to induce cell apoptosis in the insect immunosuppressive process. ARCHIVES OF INSECT BIOCHEMISTRY AND PHYSIOLOGY 2022; 110:e21877. [PMID: 35218062 PMCID: PMC9285338 DOI: 10.1002/arch.21877] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 01/28/2022] [Accepted: 02/02/2022] [Indexed: 05/27/2023]
Abstract
Microplitis bicoloratus bracovirus (MbBV) induces apoptosis in hemocytes of the host (Spodoptera litura) via the cyclophilin A (CypA)-mediated signaling pathway. However, the mechanisms underlying CypA-mediated signaling during apoptosis remain largely unknown. Therefore, in this study, we investigated how CypA and apoptosis-inducing factor (AIF) interact during MbBV-mediated apoptosis. Our findings showed that MbBV induces apoptosis through the CypA-AIF axis of insect immune suppression. In MbBV-infected Spli221 cells, both the expression of the cypa gene and the release of AIF from the mitochondria increased the number of apoptotic cells. CypA and AIF underwent concurrent cytoplasm-nuclear translocation. Conversely, blocking of AIF release from mitochondria not only inhibited the CypA-AIF interaction but also inhibited the cytoplasmic-nuclear translocation of AIF and CypA. Importantly, the survival of the apoptotic phenotype was significantly rescued in MbBV-infected Spli221 cells. In addition, we found that the cyclosporine A-mediated inhibition of CypA did not prevent the formation of the CypA and AIF complex; rather, this only suppressed genomic DNA fragmentation. In vitro experiments revealed direct molecular interactions between recombinant CypA and AIF. Taken together, our results demonstrate that the CypA-AIF interaction plays an important role in MbBV-induced innate immune suppression. This study will help to clarify aspects of insect immunological mechanisms and will be relevant to biological pest control.
Collapse
Affiliation(s)
- Yan Hu
- School of Life SciencesYunnan UniversityKunmingChina
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and RegulationsYunnan UniversityKunmingChina
| | - Ya‐Ping Liang
- School of Life SciencesYunnan UniversityKunmingChina
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and RegulationsYunnan UniversityKunmingChina
| | - Hang‐Yu Tian
- School of Life SciencesYunnan UniversityKunmingChina
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and RegulationsYunnan UniversityKunmingChina
| | - Cui‐Xian Xu
- School of Life SciencesYunnan UniversityKunmingChina
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and RegulationsYunnan UniversityKunmingChina
| | - Dan Yu
- School of Life SciencesYunnan UniversityKunmingChina
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and RegulationsYunnan UniversityKunmingChina
| | - Pan Zhang
- School of Life SciencesYunnan UniversityKunmingChina
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and RegulationsYunnan UniversityKunmingChina
| | - Hui Ye
- School of Life SciencesYunnan UniversityKunmingChina
- School of AgricultureYunnan UniversityKunmingChina
| | - Ming Li
- School of Life SciencesYunnan UniversityKunmingChina
- Key Laboratory of the University in Yunnan Province for International Cooperation in Intercellular Communications and RegulationsYunnan UniversityKunmingChina
| |
Collapse
|
5
|
Li S, Zhang Y, Fei L, Zhang Y, Pang J, Gao W, Fan F, Xing Y, Li X. Baicalein-ameliorated cerebral ischemia-reperfusion injury dependent on calpain 1/AIF pathway. Biosci Biotechnol Biochem 2022; 86:305-312. [PMID: 34935885 DOI: 10.1093/bbb/zbab222] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Accepted: 12/13/2021] [Indexed: 11/13/2022]
Abstract
Cerebral ischemia reperfusion (CIR) has become the leading cause of death and disability. Baicalein is a natural bioactive ingredient extracted from Scutellaria baicalensis Georgi and has neuroprotective activity. In our work, baicalein was found to reduce neurological deficits, brain water content, infarct area, and neuronal death of rats induced by middle cerebral artery occlusion/reperfusion. In vitro, oxygen-glucose deprivation/reperfusion induced inordinate ROS production and apoptosis that could be reversed by baicalein. Our study revealed for the first time that baicalein has the potential to bind and inhibit the activity of calpain 1, thereby inhibiting AIF nuclear translocation. These findings demonstrated that baicalein protected against CIR injury via inhibiting AIF nuclear translocation by inhibiting calpain 1 activity.
Collapse
Affiliation(s)
- Shanshan Li
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Yaoshuai Zhang
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Lili Fei
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Yuhan Zhang
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Jinlong Pang
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Wei Gao
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Fangtian Fan
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Yadong Xing
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| | - Xian Li
- School of Pharmacy, Bengbu Medical College, Bengbu, Anhui, China
| |
Collapse
|
6
|
Scimone C, Alibrandi S, Donato L, Alafaci C, Germanò A, Vinci SL, D’Angelo R, Sidoti A. Editome landscape of CCM-derived endothelial cells. RNA Biol 2022; 19:852-865. [PMID: 35771000 PMCID: PMC9248949 DOI: 10.1080/15476286.2022.2091306] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Accepted: 06/13/2022] [Indexed: 02/03/2023] Open
Abstract
By regulating several phases of gene expression, RNA editing modifications contribute to maintaining physiological RNA expression levels. RNA editing dysregulation can affect RNA molecule half-life, coding/noncoding RNA interaction, alternative splicing, and circular RNA biogenesis. Impaired RNA editing has been observed in several pathological conditions, including cancer and Alzheimer's disease. No data has been published yet on the editome profile of endothelial cells (ECs) isolated from human cerebral cavernous malformation (CCM) lesions. Here, we describe a landscape of editome modifications in sporadic CCM-derived ECs (CCM-ECs) by comparing editing events with those observed in human brain microvascular endothelial cells (HBMECs). With a whole transcriptome-based variant calling pipeline, we identified differential edited genes in CCM-ECs that were enriched in pathways related to angiogenesis, apoptosis and cell survival, inflammation and, in particular, to thrombin signalling mediated by protease-activated receptors and non-canonical Wnt signalling. These pathways, not yet associated to CCM development, could be a novel field for further investigations on CCM molecular mechanisms. Moreover, enrichment analysis of differentially edited miRNAs suggested additional small noncoding transcripts to consider for development of targeted therapies.
Collapse
Affiliation(s)
- Concetta Scimone
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
- therapies, I.E.ME.S.TDepartment of Biomolecular strategies, genetics, cutting-edge, Palermo, Italy
| | - Simona Alibrandi
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
- therapies, I.E.ME.S.TDepartment of Biomolecular strategies, genetics, cutting-edge, Palermo, Italy
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Messina, Italy
| | - Luigi Donato
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
- therapies, I.E.ME.S.TDepartment of Biomolecular strategies, genetics, cutting-edge, Palermo, Italy
| | - Concetta Alafaci
- Neurosurgery Unit, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Antonino Germanò
- Neurosurgery Unit, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Sergio L. Vinci
- Neuroradiology Unit, Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
| | - Rosalia D’Angelo
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
- therapies, I.E.ME.S.TDepartment of Biomolecular strategies, genetics, cutting-edge, Palermo, Italy
| | - Antonina Sidoti
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Messina, Italy
- therapies, I.E.ME.S.TDepartment of Biomolecular strategies, genetics, cutting-edge, Palermo, Italy
| |
Collapse
|
7
|
AMPK inhibitor BML-275 induces neuroprotection through decreasing cyt c and AIF expression after transient brain ischemia. Bioorg Med Chem 2021; 52:116522. [PMID: 34837819 DOI: 10.1016/j.bmc.2021.116522] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 11/09/2021] [Accepted: 11/16/2021] [Indexed: 11/23/2022]
Abstract
Stroke is a major public health problem with an imperative need for a more effective and tolerated therapy. Neuroprotective therapy may be an effective therapeutic intervention for stroke. The morbidity and mortality of stroke-induced secondary brain injury is mainly caused by neuronal apoptosis, which can be executed in a caspase-dependent or apoptosis inducing factor (AIF)-dependent manner. As apoptosis is an energy-dependent process with a relative time delay, abnormal energy metabolism could be a significant and fundamental pathophysiological basis of stroke. To our knowledge, convincible evidences that AMPK inhibition exerts neuroprotection in cerebral ischemia injury via anti-apoptosis remain to be investigated. Accordingly, the aims of this study were to investigate the protective effects of AMPK inhibitor BML-275 on cerebral ischemic/reperfusion (I/R) injury and to elucidate the underlying mechanisms. Cerebral ischemia was induced by transient middle cerebral artery occlusion (tMCAO) in male C57BL/6 mice. The therapeutic effects of BML-275 were evaluated by infarct sizes, neurological scores and the proportion of apoptotic neurons after 24 h of reperfusion. The cell apoptosis markers cyt c and AIF were also evaluated. The results showed that intraperitoneally administration of BML-275 alleviate the cerebral infarction, neurological deficit and neuronal apoptosis induced by MCAO. BML-275 simultaneously induces anti-apoptosis and decreases the expression of cyt c and AIF. This study supports the hypothesis that anti-apoptosis is one of potential neuroprotective strategies for the treatment of stroke.
Collapse
|
8
|
Zhou Y, Wei W, Shen J, Lu L, Lu T, Wang H, Xue X. Alisol A 24-acetate protects oxygen-glucose deprivation-induced brain microvascular endothelial cells against apoptosis through miR-92a-3p inhibition by targeting the B-cell lymphoma-2 gene. PHARMACEUTICAL BIOLOGY 2021; 59:513-524. [PMID: 33905668 PMCID: PMC8081307 DOI: 10.1080/13880209.2021.1912117] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
CONTEXT Alisol A 24-acetate has been used to treat vascular diseases. However, the underlying mechanisms still remain unclear. OBJECTIVE The present study evaluated the antiapoptotic effect of alisol A 24-acetate on brain microvascular endothelial cells (BMECs) and explored the underlying mechanisms. MATERIALS AND METHODS BMECs were injured through oxygen -glucose deprivation (OGD) after alisol A 24-acetate treatment. Cell viability and half-maximal inhibitory concentration (IC50) were measured using CCK-8, whereas inflammatory factors and oxidative stress indicators were measured using enzyme linked immunosorbent assay. Cell invasion and wound healing assays were detected. Cell apoptosis was assessed using flow cytometry. B-cell lymphoma-2 (Bcl-2) and Bcl-2 associated X (Bax) expression were analyzed using Western blotting. Dual-luciferase assay was applied to detect target genes of miR-92a-3p. RESULT Alisol A 24-acetate had an IC50 of 98.53 mg/L and inhibited cell viability at concentrations over 50mg/L. OGD induced apoptosis and promoted miR-92a-3p overexpression in BMECs. However, alisol A 24-acetate treatment suppressed inflammation, improved migration and invasion abilities, increased Bcl-2 expression, inhibited Bax expression, and repressed apoptosis and miR92a-3p overexpression in OGD-induced BMECs. MiR-92a-3p overexpression promoted cell apoptosis and suppressed Bcl-2 expression, whereas its inhibitor reversed the tendency. Alisol A 24-acetate treatment relieved the effects of miR-92a-3p overexpression. Dual-luciferase assay confirmed that miR-92a-3p negatively regulated the Bcl-2 expression. CONCLUSIONS These findings suggest that alisol A 24-acetate exerts antiapoptotic effects on OGD-induced BMECs through miR-92a-3p inhibition by targeting the Bcl-2 gene, indicating its potential for BMECs protection and as a novel therapeutic agent for the treatment of cerebrovascular disease.
Collapse
Affiliation(s)
- Yangjie Zhou
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Wei Wei
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Julian Shen
- Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Lu Lu
- Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Taotao Lu
- Rehabilitation Industry Institute, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Hong Wang
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
| | - Xiehua Xue
- The Affiliated Rehabilitation Hospital, Fujian University of Traditional Chinese Medicine, Fuzhou, China
- CONTACT Xiehua Xue No. 13, Hudongzhi Road, Gulou District, Fuzhou350122, China
| |
Collapse
|
9
|
Koehler RC, Dawson VL, Dawson TM. Targeting Parthanatos in Ischemic Stroke. Front Neurol 2021; 12:662034. [PMID: 34025565 PMCID: PMC8131834 DOI: 10.3389/fneur.2021.662034] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Accepted: 04/01/2021] [Indexed: 12/14/2022] Open
Abstract
Parthanatos is a cell death signaling pathway in which excessive oxidative damage to DNA leads to over-activation of poly(ADP-ribose) polymerase (PARP). PARP then generates the formation of large poly(ADP-ribose) polymers that induce the release of apoptosis-inducing factor from the outer mitochondrial membrane. In the cytosol, apoptosis-inducing factor forms a complex with macrophage migration inhibitory factor that translocates into the nucleus where it degrades DNA and produces cell death. In a review of the literature, we identified 24 publications from 13 laboratories that support a role for parthanatos in young male mice and rats subjected to transient and permanent middle cerebral artery occlusion (MCAO). Investigators base their conclusions on the use of nine different PARP inhibitors (19 studies) or PARP1-null mice (7 studies). Several studies indicate a therapeutic window of 4-6 h after MCAO. In young female rats, two studies using two different PARP inhibitors from two labs support a role for parthanatos, whereas two studies from one lab do not support a role in young female PARP1-null mice. In addition to parthanatos, a body of literature indicates that PARP inhibitors can reduce neuroinflammation by interfering with NF-κB transcription, suppressing matrix metaloproteinase-9 release, and limiting blood-brain barrier damage and hemorrhagic transformation. Overall, most of the literature strongly supports the scientific premise that a PARP inhibitor is neuroprotective, even when most did not report behavior outcomes or address the issue of randomization and treatment concealment. Several third-generation PARP inhibitors entered clinical oncology trials without major adverse effects and could be repurposed for stroke. Evaluation in aged animals or animals with comorbidities will be important before moving into clinical stroke trials.
Collapse
Affiliation(s)
- Raymond C Koehler
- Department of Anesthesiology and Critical Care Medicine, The Johns Hopkins University, Baltimore, MD, United States
| | - Valina L Dawson
- Neuroregeneration and Stem Cell Programs, The Institute of Cell Engineering, The Johns Hopkins University, Baltimore, MD, United States.,Department of Neurology, The Johns Hopkins University, Baltimore, MD, United States.,Department of Neuroscience, The Johns Hopkins University, Baltimore, MD, United States.,Department of Physiology, The Johns Hopkins University, Baltimore, MD, United States
| | - Ted M Dawson
- Neuroregeneration and Stem Cell Programs, The Institute of Cell Engineering, The Johns Hopkins University, Baltimore, MD, United States.,Department of Neurology, The Johns Hopkins University, Baltimore, MD, United States.,Department of Neuroscience, The Johns Hopkins University, Baltimore, MD, United States.,Department of Pharmacology and Molecular Sciences, The Johns Hopkins University, Baltimore, MD, United States
| |
Collapse
|
10
|
Abstract
Despite thousands of neuroprotectants demonstrating promise in preclinical trials, a neuroprotective therapeutic has yet to be approved for the treatment of acute brain injuries such as stroke or traumatic brain injury. Developing a more detailed understanding of models and populations demonstrating "neurological resilience" in spite of brain injury can give us important insights into new translational therapies. Resilience is the process of active adaptation to a stressor. In the context of neuroprotection, models of preconditioning and unique animal models of extreme physiology (such as hibernating species) reliably demonstrate resilience in the laboratory setting. In the clinical setting, resilience is observed in young patients and can be found in those with specific genetic polymorphisms. These important examples of resilience can help transform and extend the current neuroprotective framework from simply countering the injurious cascade into one that anticipates, monitors, and optimizes patients' physiological responses from the time of injury throughout the process of recovery. This review summarizes the underpinnings of key adaptations common to models of resilience and how this understanding can be applied to new neuroprotective approaches.
Collapse
Affiliation(s)
- Neel S Singhal
- Department of Neurology, University of California-San Francisco, 555 South Mission Bay Blvd, San Francisco, CA, 94158, USA.
| | - Chung-Huan Sun
- Department of Neurology, University of California-San Francisco, 555 South Mission Bay Blvd, San Francisco, CA, 94158, USA
| | - Evan M Lee
- Cardiovascular Research Institute, University of California-San Francisco, 555 South Mission Bay Blvd, San Francisco, CA, 94158, USA
- Department of Physiology, University of California-San Francisco, 555 South Mission Bay Blvd, San Francisco, CA, 94158, USA
| | - Dengke K Ma
- Cardiovascular Research Institute, University of California-San Francisco, 555 South Mission Bay Blvd, San Francisco, CA, 94158, USA
- Department of Physiology, University of California-San Francisco, 555 South Mission Bay Blvd, San Francisco, CA, 94158, USA
| |
Collapse
|
11
|
Zhao Z, Zhang X, Dai Y, Pan K, Deng Y, Meng Y, Xu T. PPAR-γ promotes p38 MAP kinase-mediated endothelial cell permeability through activating Sirt3. BMC Neurol 2019; 19:289. [PMID: 31729962 PMCID: PMC6857342 DOI: 10.1186/s12883-019-1508-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2019] [Accepted: 10/22/2019] [Indexed: 12/16/2022] Open
Abstract
Background Ischemia-reperfusion (I/R)-induced vascular dysfunction is the main factor to acute ischemic stroke. Sirt3 is one of the sirtuin family members, which plays an important role in the development of neurological diseases. Methods In this study, we constructed I/R injury model on HBMEC cells and induced the overexpression of Sirt3 in model cells. Meanwhile, the p38 activator U-46619 was used to examine the connection between Sirt3 and p38. We also examined the level of endothelial associated proteins, including occluding, ZO-1 and claudin-4 by using qRT-PCR and western blot. Results Our findings indicated that overexpression of Sirt3 decreased the permeability of model cells and promoted in the growth of endothelial cells. However, the activation of p38 could antagonize the function of Sirt3 in HBMEC cells. Moreover, Our results indicated a positive correlation between Sirt3 and inter-endothelial junction proteins. Importantly, PPAR-γ agonist and inhibitor were utilized to investigate the role of PPAR-γ in Sirt3 mediated cell function. Sirt3 was targeted by PPAR-γ in model cells. Conclusions Taken together, this research not only demonstrated PPAR-γ might benefit to the growth of endothelial cell though activating Sirt3 but also indicated its potential value in the treatment for ischemic stroke.
Collapse
Affiliation(s)
- Zhenzhen Zhao
- Department of Anesthesiology, Changhai hospital, Naval Medical University, Changhai Road NO.168, Shanghai City, 200433, People's Republic of China
| | - Xiaoxiu Zhang
- Department of Anesthesiology, Changhai hospital, Naval Medical University, Changhai Road NO.168, Shanghai City, 200433, People's Republic of China
| | - Yuanqiang Dai
- Department of Anesthesiology, Changhai hospital, Naval Medical University, Changhai Road NO.168, Shanghai City, 200433, People's Republic of China
| | - Ke Pan
- Department of Anesthesiology, Changhai hospital, Naval Medical University, Changhai Road NO.168, Shanghai City, 200433, People's Republic of China
| | - Yu Deng
- Department of Anesthesiology, Changhai hospital, Naval Medical University, Changhai Road NO.168, Shanghai City, 200433, People's Republic of China
| | - Yan Meng
- Department of Anesthesiology, Changhai hospital, Naval Medical University, Changhai Road NO.168, Shanghai City, 200433, People's Republic of China.
| | - Tao Xu
- Department of Anesthesiology, Changhai hospital, Naval Medical University, Changhai Road NO.168, Shanghai City, 200433, People's Republic of China.
| |
Collapse
|
12
|
Chen Y, Wang L, Zhang Y, Zhou Y, Wei W, Wan Z. The Effect of Therapeutic Mild Hypothermia on Brain Microvascular Endothelial Cells During Ischemia-Reperfusion Injury. Neurocrit Care 2019; 28:379-387. [PMID: 29327153 DOI: 10.1007/s12028-017-0486-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
BACKGROUND To determine the cerebral protective effects of mild hypothermia (MH) on cerebral microcirculation. METHODS We established ischemia-reperfusion (I/R) injury and MH treatment models with rat brain microvascular endothelial cells (RBMECs) in vitro and examined the apoptotic changes. The cultured RBMECs were randomly divided into the control group, I/R group, and MH group, which was further divided into two subgroups: intra-ischemia hypothermia (IIH) and post-ischemia hypothermia (PIH). Cell morphological changes were assessed using fluorescence microscopy. Apoptotic rates were obtained by flow cytometry. Expressions of caspase-3, Bax, and Bcl-2 were analyzed by Western blot. RESULTS I/R injury in vitro induced apoptosis of RBMECs. The apoptotic rates in the control group, I/R group, and MH group were 0.13, 19.04, and 13.13%, respectively (P < 0.01). Compared with the I/R group, the MH group showed a significant decrease in the number of apoptotic cells, mainly in stage I apoptotic cells (P < 0.0083). The caspase-3 and Bax expressions were significantly enhanced (P < 0.05) in RBMECs after I/R injury, while substantial decreases in Bcl-2 expression were noted (P < 0.05). Following MH intervention, the increase in caspase-3 and Bax expression was suppressed (P < 0.05), while Bcl-2 expression significantly increased. The apoptotic rates or protein expressions between the two subgroups were not different significantly (P > 0.05). CONCLUSIONS These results indicate that MH could inhibit RBMEC apoptosis by preventing pro-apoptotic cells and early apoptotic cells from progressing to intermediate and advanced stages. This may be due to the effect of MH on I/R-induced apoptotic gene expression changes.
Collapse
Affiliation(s)
- Yao Chen
- Department of Emergency Medicine, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu, 610041, Sichuan, China
| | - Lin Wang
- Department of Cardiology, Chengdu Shangjin Jin Nanfu Hospital, Chengdu, China
| | - Yun Zhang
- Department of Emergency, Wuxi People's Hospital, NanJing Medical University, Wuxi, China
| | - Yaxiong Zhou
- Department of Emergency Medicine, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu, 610041, Sichuan, China
| | - Wei Wei
- Department of Emergency Medicine, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu, 610041, Sichuan, China
| | - Zhi Wan
- Department of Emergency Medicine, West China Hospital, Sichuan University, 37 Guoxue Road, Chengdu, 610041, Sichuan, China.
| |
Collapse
|
13
|
Deng Y, Zhou L, Li N, Wang M, Yao L, Dong S, Zhang M, Yang P, Hao Q, Wu Y, Lyu L, Jin T, Dai Z, Kang H. Impact of four lncRNA polymorphisms (rs2151280, rs7763881, rs1136410, and rs3787016) on glioma risk and prognosis: A case-control study. Mol Carcinog 2019; 58:2218-2229. [PMID: 31489712 DOI: 10.1002/mc.23110] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 08/06/2019] [Accepted: 08/22/2019] [Indexed: 12/18/2022]
Abstract
Long noncoding RNA (lncRNA) polymorphisms are reportedly in connection with tumor susceptibility and prognosis. Glioma is one of the most aggressive and common cancers of the central nervous system. This study aimed to investigate the relationship between four lncRNA variants and glioma susceptibility and prognosis in a Chinese Han population. Sequenom Mass-ARRAY was used to genotype 605 patients with glioma and 1300 cancer-free individuals. Odds ratios or hazard ratios and related 95% confidence intervals were calculated to estimate the correlations. Logistic and Cox regression models, log-rank tests, and Kaplan-Meier curves were used for the statistical analysis. Six inheritance models showed that ANRIL rs2151280 variant genotype (A>G) was related to the susceptibility of glioma, while the other three lncRNAs showed no association. Patients treated with temozolomide or nimustine had better progression-free survival (PFS) and overall survival (OS) than those treated with platinum. Besides, patients aged older than 40 years showed a poorer OS. The Cox multivariate analysis revealed that the rs1136410 GG genotype (A>G) was beneficial for OS and PFS. The Kaplan-Meier analyses indicated that rs1136410 A>G and the rs7763881 A>C were associated with longer OS. ANRIL rs2151280 variant genotype might increase susceptibility of glioma. In addition, PARP1 rs1136410 variant genotype could be beneficial for the overall survival of patients with glioma. More research data are needed to further validate our results.
Collapse
Affiliation(s)
- Yujiao Deng
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Department of Breast Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Linghui Zhou
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Department of Breast Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Na Li
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Department of Breast Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Meng Wang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Li Yao
- Department of Neurology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Shanshan Dong
- School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, China
| | - Ming Zhang
- Department of Neurosurgery, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Pengtao Yang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Qian Hao
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Ying Wu
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Lijuan Lyu
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Tianbo Jin
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Life Sciences, Northwest University, Xi'an, China
| | - Zhijun Dai
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China.,Department of Breast Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Huafeng Kang
- Department of Oncology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
14
|
Zhao W, Li H, Hou Y, Jin Y, Zhang L. Combined Administration of Poly-ADP-Ribose Polymerase-1 and Caspase-3 Inhibitors Alleviates Neuronal Apoptosis After Spinal Cord Injury in Rats. World Neurosurg 2019; 127:e346-e352. [PMID: 30904799 DOI: 10.1016/j.wneu.2019.03.116] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2019] [Revised: 03/11/2019] [Accepted: 03/12/2019] [Indexed: 02/06/2023]
Abstract
BACKGROUND Neuronal apoptosis plays a pivotal role in spinal cord injury (SCI)-induced secondary cellular events. Caspase-dependent and -independent pathways are involved in neuronal apoptosis. Caspase-3 is the final effector of caspase-dependent apoptosis, whereas poly-ADP-ribose polymerase-1 (PARP-1) and apoptosis-inducing factor (AIF) are key executors of caspase-independent apoptosis. However, it remains unclear whether simultaneous inhibition of the 2 apoptosis pathways will be more beneficial for neuronal survival. Therefore, this study investigated the ability of coadministration of the PARP-1 inhibitor 3-aminobenzamide (3-AB) and caspase-3 inhibitor z-DEVD-fmk to attenuate apoptosis in a rat SCI model. METHODS The rats were subjected to moderate contusive SCI. Locomotor function was measured using the Basso, Beattie, and Bresnahan rating scales; neuronal apoptosis was detected using transferase-mediated deoxyuridine triphosphate-biotin nick end labeling; and immunohistochemistry and Western blotting were used to measure protein expression. RESULTS We found the locomotor function of rats was weakened within 7 days post-SCI. At day 7 post-SCI, neuronal apoptosis dramatically increased and the expression of PARP-1, AIF, and cleaved caspase-3 was significantly upregulated. Further, Bcl-2 expression was significantly downregulated. The highest locomotor function recovery was recorded after the combined administration of 3-AB and z-DEVD-fmk for 7 days post-SCI when compared with 3-AB or z-DEVD-fmk administered alone. In addition, this combination therapy significantly reduced neuronal apoptosis by preventing upregulation of PARP-1 and AIF, inhibiting caspase-3 activation, and elevating Bcl-2 expression. CONCLUSIONS These results suggest that combination therapy is beneficial for neuronal function recovery in rats with SCI. The underlying mechanism may be associated with cosuppression of caspase-dependent and caspase-independent apoptosis pathways.
Collapse
Affiliation(s)
- Wei Zhao
- Department of Histology and Embryology, Binzhou Medical University, Yantai, Shandong, China
| | - Hongxing Li
- Department of Histology and Embryology, Binzhou Medical University, Yantai, Shandong, China
| | - Yun Hou
- Department of Histology and Embryology, Binzhou Medical University, Yantai, Shandong, China
| | - Yinchuan Jin
- Department of Histology and Embryology, Binzhou Medical University, Yantai, Shandong, China
| | - Lianshuang Zhang
- Department of Histology and Embryology, Binzhou Medical University, Yantai, Shandong, China.
| |
Collapse
|
15
|
Bashir K, Sarwar R, Saeed S, Mahjabeen I, Kayani MA. Interaction among susceptibility genotypes of PARP1 SNPs in thyroid carcinoma. PLoS One 2018; 13:e0199007. [PMID: 30183716 PMCID: PMC6124699 DOI: 10.1371/journal.pone.0199007] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 04/16/2018] [Indexed: 01/21/2023] Open
Abstract
Polymorphisms in DNA repair genes may alter the repair mechanism which makes the person susceptible to DNA damage. Polymorphic variants in these DNA repair pathway genes such as Poly (ADP-ribose) polymerase- 1 (PARP1) have been associated with susceptibility of several types of cancer including thyroid. Many studies have been published on PARP1 gene polymorphisms and carcinogenesis with inconsistent results. The present study was designed to explore the link between the PARP1 polymorphisms and thyroid cancer risk. This case-control study was comprised of 456 thyroid cancer patients and 400 healthy controls. Three SNPs of PARP1 gene; rs1136410, rs1805414 and rs1805404 were analyzed using ARMS-PCR. The combined genotype and haplotype analysis were performed using haploview software 4.2. Major allele homozygote (CC) of rs1136410 and combined genotype (TT+TC) of rs180414 showed a significant association with thyroid cancer risk (OR = 1.30; 95% CI 0.99–1.77; P = 0.05) and (OR = 0.43; 95% CI = 0.27–0.67; P = 0.03). Histological subtype analysis showed the significant association of selected PARP1 SNPs with papillary, follicular and anaplastic subtypes in thyroid cancer patients. Haplotype analysis showed that TCT (p = 0.01), CTT (p = 0.02) and CTC (p = 0.03) were significantly higher in controls when compared to cases. However, TTC (p = 0.05) and TCC (p = 0.01) haplotype frequency was significantly higher in cases compared to controls. Global haplotype analysis showed that there was an overall significant difference between cases and controls (p = 0.001). Identification of these genetic risk markers may provide evidence for exploring insight into mechanisms of pathogenesis and subsequently aid in developing novel therapeutic strategies for thyroid cancer.
Collapse
Affiliation(s)
- Kashif Bashir
- Department of Biosciences, COMSATS Institute of Information and Technology, Islamabad, Pakistan
| | - Romana Sarwar
- Department of Biosciences, COMSATS Institute of Information and Technology, Islamabad, Pakistan
| | - Soma Saeed
- Department of Biosciences, COMSATS Institute of Information and Technology, Islamabad, Pakistan
| | - Ishrat Mahjabeen
- Department of Biosciences, COMSATS Institute of Information and Technology, Islamabad, Pakistan
| | - Mahmood Akhtar Kayani
- Department of Biosciences, COMSATS Institute of Information and Technology, Islamabad, Pakistan
- * E-mail:
| |
Collapse
|
16
|
Abstract
Stroke is one of the major causes of mortality and morbidity worldwide, yet novel therapeutic treatments for this condition are lacking. This review focuses on the roles of the transient receptor potential melastatin 2 (TRPM2) ion channels in cellular damage following hypoxia-ischemia and their potential as a future therapeutic target for stroke. Here, we highlight the complex molecular signaling that takes place in neurons, glial cells and the blood-brain barrier following ischemic insult. We also describe the evidence of TRPM2 involvement in these processes, as shown from numerous in vitro and in vivo studies that utilize genetic and pharmacological approaches. This evidence implicates TRPM2 in a broad range of pathways that take place every stage of cerebral ischemic injury, thus making TRPM2 a promising target for drug development for stroke and other neurodegenerative conditions of the central nervous system.
Collapse
|
17
|
Wang G, Wu Y, Zhu Y. Mechanism of MALAT1 preventing apoptosis of vascular endothelial cells induced by oxygen–glucose deficiency and reoxidation. ARTIFICIAL CELLS NANOMEDICINE AND BIOTECHNOLOGY 2018; 46:798-805. [PMID: 29575939 DOI: 10.1080/21691401.2018.1436065] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Affiliation(s)
- Guoping Wang
- Department of Neurology, Anhui Provincial Hospital, Anhui Medical University, Hefei, PR China
- Department of Neurology, Anhui Provincial Hospital, The First Affiliated Hospital of University of Science and Technology of China, Hefei, PR China
| | - Yuanbo Wu
- Department of Neurology, Anhui Provincial Hospital, Anhui Medical University, Hefei, PR China
- Department of Neurology, Anhui Provincial Hospital, The First Affiliated Hospital of University of Science and Technology of China, Hefei, PR China
| | - Yuyou Zhu
- Department of Neurology, Anhui Provincial Hospital, Anhui Medical University, Hefei, PR China
- Department of Neurology, Anhui Provincial Hospital, The First Affiliated Hospital of University of Science and Technology of China, Hefei, PR China
| |
Collapse
|
18
|
Yan T, Hu G, Wang A, Sun X, Yu X, Jia J. Paris saponin VII induces cell cycle arrest and apoptosis by regulating Akt/MAPK pathway and inhibition of P-glycoprotein in K562/ADR cells. Phytother Res 2018; 32:898-907. [DOI: 10.1002/ptr.6029] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Revised: 11/30/2017] [Accepted: 12/17/2017] [Indexed: 01/06/2023]
Affiliation(s)
- Ting Yan
- School of Traditional Chinese Materia Medica; Shenyang Pharmaceutical University; Shenyang 110016 China
| | - Gaosheng Hu
- School of Traditional Chinese Materia Medica; Shenyang Pharmaceutical University; Shenyang 110016 China
| | - Anhua Wang
- School of Traditional Chinese Materia Medica; Shenyang Pharmaceutical University; Shenyang 110016 China
| | - Xianduo Sun
- School of Traditional Chinese Medicines; Guangdong Pharmaceutical University; Guangzhou 510006 China
| | - Xiangyong Yu
- School of Traditional Chinese Materia Medica; Shenyang Pharmaceutical University; Shenyang 110016 China
| | - Jingming Jia
- School of Traditional Chinese Materia Medica; Shenyang Pharmaceutical University; Shenyang 110016 China
| |
Collapse
|
19
|
Oxidative stress and DNA damage after cerebral ischemia: Potential therapeutic targets to repair the genome and improve stroke recovery. Neuropharmacology 2017; 134:208-217. [PMID: 29128308 DOI: 10.1016/j.neuropharm.2017.11.011] [Citation(s) in RCA: 200] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Revised: 11/02/2017] [Accepted: 11/05/2017] [Indexed: 12/12/2022]
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'.
Collapse
|
20
|
Xu M, Yang X, Zeng Q, He H, Lu P, Huang G. BIRC5 is a novel target of peroxisome proliferator‑activated receptor γ in brain microvascular endothelium cells during cerebral ischemia. Mol Med Rep 2017; 16:8882-8890. [PMID: 29039513 PMCID: PMC5779969 DOI: 10.3892/mmr.2017.7750] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2017] [Accepted: 08/16/2017] [Indexed: 11/23/2022] Open
Abstract
Cerebral ischemia is a leading cause of ischemic stroke, which may lead to severe disability and mortality worldwide. There are some key factors concerned in cardioprotection, such as peroxisome proliferator-activated receptor γ (PPARγ), a ligand binding transcription factor involved in various biological functions including atherosclerosis, vascular dysfunction and hypertension, and baculoviral IAP repeat-containing 5 (BIRC5), which may protect human brain endothelial cells from ischemia-induced apoptosis. To determine the potential roles of PPARγ in brain microvascular endothelial (bEnd.3) cells during cerebral ischemia and the relationship between PPARγ and BIRC5, a cerebral ischemia model was established with bEnd.3 cells cells by oxygen-glucose deprivation (OGD) treatment. OGD treatment reduced proliferation and enhanced apoptosis of bEnd.3 cells in a time-dependent manner. PPARγ expression levels were decreased in bEnd.3 cells following OGD treatment. Upregulation of PPARγ expression protected bEnd.3 cells from ischemia injury and also upregulated BIRC5 expression. PPARγ-specific binding sites in the BIRC5 promoter were predicted bioinformatically and verified by luciferase reporter experiments. Results from electrophoretic mobility shift/supershift and chromatin immunoprecipitation assays suggested that BIRC5 may be a novel target of PPARγ transcriptional regulation during ischemic injury. The present results indicated that PPARγ may serve a protective role on bEnd.3 cells and that BIRC5 may be a downstream target of PPARγ regulation during cerebral ischemia.
Collapse
Affiliation(s)
- Mingjing Xu
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Xianli Yang
- Department of Medical Quality Management, Guizhou Provincial People's Hospital, Guiyang, Guizhou 550002, P.R. China
| | - Qing Zeng
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - He He
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Pengcheng Lu
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| | - Guozhi Huang
- Department of Rehabilitation Medicine, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong 510282, P.R. China
| |
Collapse
|
21
|
Zhou Y, Zhou B, Tu H, Tang Y, Xu C, Chen Y, Zhao Z, Miao Z. The degradation of mixed lineage kinase domain-like protein promotes neuroprotection after ischemic brain injury. Oncotarget 2017; 8:68393-68401. [PMID: 28978125 PMCID: PMC5620265 DOI: 10.18632/oncotarget.19416] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 06/05/2017] [Indexed: 12/20/2022] Open
Abstract
Mixed lineage kinase domain-like (MLKL) protein was recently found to play a critical role in necrotic cell death. To explore its role in neurological diseases, we measured MLKL protein expression after ischemia injury in a mouse model. We found that MLKL expression significantly increased 12 h after ischemia/reperfusion (I/R) injury with peak levels at 48 h. Inhibition of MLKL by intraperitoneal administration of NSA significantly reduced infarct volume and improved neurological deficits after 75 min of ischemia and 24 h of reperfusion. Further, we found NSA reduced MLKL levels via the ubiquitination proteasome pathway, but not by inhibiting RNA transcription. Interestingly, NSA administration increased cleaved PARP-1 levels, indicating the protective effects of MLKL inhibition is not related to apoptosis. These findings suggest MLKL is a new therapeutic target for neurological pathologies like stroke. Therefore, promoting degradation of MLKL may be a novel avenue to reduce necrotic cell death after ischemic brain injury.
Collapse
Affiliation(s)
- Yanlong Zhou
- Institute of Neuroscience, Soochow University, Suzhou City, Jiangsu Province, China.,Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Beiqun Zhou
- Institute of Neuroscience, Soochow University, Suzhou City, Jiangsu Province, China.,Department of Neurology, The Second Affiliated Hospital of Jiaxing University, Jiaxing City, Zhejiang Province, China
| | - Hui Tu
- Department of Anesthesia, The Second Affiliated Hospital of Soochow University, Suzhou City, Jiangsu Province, China
| | - Yan Tang
- Institute of Neuroscience, Soochow University, Suzhou City, Jiangsu Province, China
| | - Chen Xu
- Institute of Neuroscience, Soochow University, Suzhou City, Jiangsu Province, China.,Department of Neurology, The Affiliated Suzhou Hospital, Nanjing Medical University, Suzhou City, Jiangsu Province, China
| | - Yanbo Chen
- Department of Neurology, The Affiliated Hospital of Xuzhou Medical University, Xuzhou City, Jiangsu Province, China
| | - Zhong Zhao
- Department of Neurology, The Affiliated Suzhou Hospital, Nanjing Medical University, Suzhou City, Jiangsu Province, China
| | - Zhigang Miao
- Institute of Neuroscience, Soochow University, Suzhou City, Jiangsu Province, China
| |
Collapse
|
22
|
Xin JW, Jiang YG. Long noncoding RNA MALAT1 inhibits apoptosis induced by oxygen-glucose deprivation and reoxygenation in human brain microvascular endothelial cells. Exp Ther Med 2017; 13:1225-1234. [PMID: 28413461 PMCID: PMC5377418 DOI: 10.3892/etm.2017.4095] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2015] [Accepted: 07/07/2016] [Indexed: 12/15/2022] Open
Abstract
Cerebral ischemia/reperfusion (I/R) injury leads to brain vascular dysfunction, which is characterized by endothelial cell injury or death. Long noncoding (lnc) RNA metastasis-associated lung adenocarcinoma transcript 1 (MALAT1) is reportedly associated with endothelial cell functions and dysfunctions. In the present study, the role of MALAT1 in I/R-induced cerebral vascular endothelial cell apoptosis was explored using oxygen-glucose deprivation and reoxygenation (OGD-R) as an in vitro I/R injury model. Primary human brain microvascular endothelial cells were cultured under OGD-R, and the expression levels of MALAT1 and cell apoptosis were measured at 6, 9, 12, 24 and 36 h post-reoxygenation. The expression levels of MALAT1 and the apoptotic rate of cells exposed to OGD-R exhibited contrasting trends following reoxygenation. Following OGD-R, lentiviral overexpression of MALAT1 increased phosphatidylinositol 3-kinase (PI3K) activities and the activation of Akt phosphorylation, and decreased cell apoptosis and caspase 3 activities, which were successfully abolished by treatment with a PI3K inhibitor, Wortmannin. Conversely, lentiviral knockdown of MALAT1 decreased PI3K activities and the activation of Akt phosphorylation, and increased cell apoptosis and caspase 3 activity. Overexpression and knockdown of MALAT1 exhibited no significant effects on OGD-R-induced reactive oxygen species (ROS) production. In conclusion, to the best of our knowledge, the present study was the first to suggest that lncRNA MALAT1 may protect human brain vascular endothelial cells from OGD-R-induced apoptosis via a PI3K-dependent mechanism. These findings suggest that MALAT1 may be a potential novel therapeutic target for cerebral I/R injury.
Collapse
Affiliation(s)
- Jia-Wei Xin
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| | - Yu-Gang Jiang
- Department of Neurosurgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan 410011, P.R. China
| |
Collapse
|
23
|
Hu K, Yang M, Xu YY, Wei K, Ren J. Cell Cycle Arrest, Apoptosis, and Autophagy Induced by Chabamide in Human Leukemia Cells. CHINESE HERBAL MEDICINES 2016. [DOI: 10.1016/s1674-6384(16)60005-9] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023] Open
|
24
|
Chabamide induces cell cycle arrest and apoptosis by the Akt/MAPK pathway and inhibition of P-glycoprotein in K562/ADR cells. Anticancer Drugs 2015; 26:498-507. [PMID: 25714087 DOI: 10.1097/cad.0000000000000209] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
One of the major mechanisms of multidrug resistance in cancer therapy is the overexpression of P-glycoprotein (P-gp). Chabamide, a dimeric alkaloid isolated from Piper chaba Hunter, shows antimalarial, antituberculosis, and cytotoxic activities. However, its mechanism of action has not been elucidated. In this study, the molecular mechanism underlying the cytotoxicity and downregulation of P-gp expression by chabamide in adriamycin-resistant human leukemia cells (K562/ADR) was clarified. Results show that chabamide inhibited the growth of K562/ADR cells in a dose-dependent and time-dependent manner, and significantly inhibited cell proliferation by cell cycle arrest in the G0/G1 phase, which was associated with an obvious increase in p21 and decrease in cyclin D1 and CDK2/4/6 protein expression. Moreover, chabamide could regulate the changes in the mitochondrial membrane potential, increase the expression of apoptosis-related proteins, such as Bax and cytochrome c, and decrease the protein expression levels of Bcl-2, caspase-9, caspase-3, PARP-1, and p-Akt. In addition, we found that JNK, ERK1/2, and p38 were regulated by chabamide in K562/ADR cells. Further studies indicated that the decrease in the reactive oxygen species level inhibited intrinsic P-gp expression. Therefore, chabamide-induced apoptosis in K562/ADR cells was associated with Akt/MAPK and the inhibition of P-gp. These results provide a biochemical basis for possible clinical applications of chabamide in the treatment of leukemia.
Collapse
|
25
|
Wang J, Hao L, Wang Y, Qin W, Wang X, Zhao T, Liu Y, Sheng L, Du Y, Zhang M, Lu Q. Inhibition of poly (ADP-ribose) polymerase and inducible nitric oxide synthase protects against ischemic myocardial damage by reduction of apoptosis. Mol Med Rep 2014; 11:1768-76. [PMID: 25412407 PMCID: PMC4270331 DOI: 10.3892/mmr.2014.2977] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2014] [Accepted: 09/19/2014] [Indexed: 01/12/2023] Open
Abstract
Myocardial infarction (MI) is defined as the deprivation of the myocardial tissue of oxygen and nutrients, resulting in the induction of inflammation and apoptosis of the cardiomyocytes. Poly (ADP-ribose) polymerase 1 (PARP1) is a nuclear enzyme closely associated with MI, that can be activated by DNA damage. Inducible nitric oxide synthase (iNOS) is a critical enzyme among the inflammatory cytokines. The present study aimed to investigate the underlying mechanism of the protective effects of PARP1 and iNOS inhibitor against MI, in rats. A total of 40 male Wistar rats were divided into four groups. The rats were anesthetized with sodium pentobarbital (50 mg/kg), and the left anterior descending coronary artery was occluded by ligation, using a 6-0 polypropylene monofilament suture, at the left atrial apex, in order to induce MI. The rats from each group received an abdominal injection of either dimethylsulfoxide (100 μl, for MI group); PARP-1 inhibitor, 3,4-dihydro-5-[4-(1-piperidinyl)butoxy]-1(2H)-isoquinolinone (DPQ; 10 mg/kg); or iNOS inhibitor, N-(1-naphthyl)ethylenediamine dihydrochloride (1400W; 10 mg/kg). The hearts were harvested from the rats after four weeks. Inhibition of PARP and iNOS activity improved heart function, as determined by serial echocardiography. The rate of apoptosis, as determined by a terminal deoxynucleotidyl-transferase-mediated dUTP nick end labeling assay, was reduced by 39.71 and 39.00% in the DPQ and 1400W groups, respectively, and this was accompanied by the downregulated expression of cleaved caspase-3 and PARP1. Effective inhibition of PARP and iNOS, by DPQ and 1400W, was detected by western blotting and immunofluorescence, and was shown to repress O2− and nitrotyrosine levels, following MI. The present study confirmed that inhibition of PARP1 and iNOS was able to protect against ischemic myocardial damage, by reducing the levels of apoptosis.
Collapse
Affiliation(s)
- Juan Wang
- Department of Cardiovascular Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Lin Hao
- Department of Cardiovascular Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Yan Wang
- Department of Anesthesiology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250062, P.R. China
| | - Weidong Qin
- Department of Intensive Care Unit, Qilu Hospital Affiliated to Shandong University, Jinan, Shandong 250012, P.R. China
| | - Xin Wang
- Department of Cardiovascular Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Tong Zhao
- Department of Cardiovascular Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Yusheng Liu
- Department of Cardiovascular Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Lin Sheng
- Department of Cardiovascular Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Yimeng Du
- Department of Cardiovascular Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| | - Mengyuan Zhang
- Department of Anesthesiology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250062, P.R. China
| | - Qinghua Lu
- Department of Cardiovascular Medicine, The Second Hospital of Shandong University, Jinan, Shandong 250033, P.R. China
| |
Collapse
|
26
|
Chimaphilin induces apoptosis in human breast cancer MCF-7 cells through a ROS-mediated mitochondrial pathway. Food Chem Toxicol 2014; 70:1-8. [DOI: 10.1016/j.fct.2014.04.014] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Revised: 03/21/2014] [Accepted: 04/08/2014] [Indexed: 12/18/2022]
|
27
|
Wang T, Li Y, Wang Y, Zhou R, Ma L, Hao Y, Jin S, Du J, Zhao C, Sun T, Yu J. Lycium barbarum polysaccharide prevents focal cerebral ischemic injury by inhibiting neuronal apoptosis in mice. PLoS One 2014; 9:e90780. [PMID: 24595452 PMCID: PMC3940937 DOI: 10.1371/journal.pone.0090780] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 02/04/2014] [Indexed: 01/06/2023] Open
Abstract
AIMS OF THE STUDY To investigate the neuroprotective effect of Lycium barbarum polysaccharide (LBP) on focal cerebral ischemic injury in mice and to explore its possible mechanism. MATERIALS AND METHODS Male ICR mice were used to make the model of middle cerebral artery occlusion (MCAO) after intragastric administration with LBP (10, 20 and 40 mg/kg) and Nimodipine (0.4 mg/kg) for seven successive days. After 24 h of reperfusion, neurological scores were estimated and infarct volumes were measured by 2, 3, 5-triphenyltetrazolium chloride (TTC) staining. Morphological changes in ischemic brains were performed for hematoxylin-eosin (HE) staining. The number of apoptotic neurons was detected by TUNEL staining. The Bax, Bcl-2 protein expression and CytC, Caspase-3, -9 and cleaved PARP-1 activation were investigated by immunofluorescence and western-blot analysis. RESULTS LBP (10, 20 and 40 mg/kg) treatment groups significantly reduced infract volume and neurological deficit scores. LBP also relieved neuronal morphological damage and attenuated the neuronal apoptosis. LBP at the dose of 40 mg/kg significantly suppressed overexpression of Bax, CytC, Caspase-3, -9 and cleaved PARP-1, and inhibited the reduction of Bcl-2 expression. CONCLUSIONS Based on these findings we propose that LBP protects against focal cerebral ischemic injury by attenuating the mitochondrial apoptosis pathway.
Collapse
Affiliation(s)
- Tengfei Wang
- Department of Pharmacology, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Yuxiang Li
- College of Nursing, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Yongsheng Wang
- Department of Pharmacology, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Ru Zhou
- Department of Pharmacology, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Lin Ma
- Key Lab of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Yinju Hao
- Department of Pharmacology, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Shaoju Jin
- Department of Pharmacology, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Juan Du
- Department of Pharmacology, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Chengjun Zhao
- Key Laboratory of Fertility Preservation and Maintenance, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Tao Sun
- Key Lab of Craniocerebral Diseases of Ningxia Hui Autonomous Region, Ningxia Medical University, Yinchuan, People's Republic of China
| | - Jianqiang Yu
- Department of Pharmacology, Ningxia Medical University, Yinchuan, People's Republic of China
- Ningxia Hui Medicine Modern Engineering Research Center, Ningxia Medical University, Yinchuan, People's Republic of China
- * E-mail:
| |
Collapse
|
28
|
Dos Santos Pereira RT, Porto CS, Abdalla FMF. Ovariectomy and 17β-estradiol replacement play a role on the expression of Endonuclease-G and phosphorylated cyclic AMP response element-binding (CREB) protein in hippocampus. Mol Cell Endocrinol 2014; 382:227-233. [PMID: 24121025 DOI: 10.1016/j.mce.2013.09.037] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2013] [Revised: 09/16/2013] [Accepted: 09/30/2013] [Indexed: 11/19/2022]
Abstract
The aim of the present study was to investigate the effects of different periods of ovariectomy and 17β-estradiol (E2) replacement on the expression of Cytochrome C, apoptosis inducing factor (AIF) and Endonuclease-G (Endo-G) in mitochondrial and cytosolic fractions obtained from hippocampus of the adult female rats. In addition, the expression of phosphorylated CREB (phospho-CREB) was also analyzed in hippocampus. Ovariectomy or E2 treatment did not change the expression of Cytochrome C and AIF. Ovariectomy (15, 21 and 36 days) decreased the expression of Endo-G in the mitochondrial fractions and increased it in the cytosolic fractions obtained from hippocampus. The treatment with E2 after 15 days of ovariectomy for 7 days or 21 days, and throughout the post-ovariectomy period prevented the effects of ovariectomy on Endo-G expression. Our results suggest that ovariectomy-induced apoptotic cell death in hippocampal tissue could be mediated by Endo-G, but not by AIF, via a caspase-independent apoptotic pathway. Furthermore, ovariectomy decreased the expression of phospho-CREB and the treatment with E2 prevented these effects. In conclusion, E2 may help maintain long-term neuronal viability by regulating the expression of members of the Bcl-2 family. Regulation of Endo-G released from mitochondria, but not of Cytochrome C and AIF, is also involved in the neuroprotective actions of E2. Furthermore, CREB may be involved in the expression of Bcl-2. These data provide new understanding into the mechanisms involved in the neuroprotective role of estrogen.
Collapse
Affiliation(s)
| | - Catarina Segreti Porto
- Section of Experimental Endocrinology, Department of Pharmacology, Escola Paulista de Medicina-Universidade Federal de São Paulo, São Paulo, Brazil
| | | |
Collapse
|
29
|
New role of silent information regulator 1 in cerebral ischemia. Neurobiol Aging 2013; 34:2879-88. [DOI: 10.1016/j.neurobiolaging.2013.06.008] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 06/06/2013] [Accepted: 06/14/2013] [Indexed: 01/23/2023]
|
30
|
|
31
|
Geng X, Parmar S, Li X, Peng C, Ji X, Chakraborty T, Li WA, Du H, Tan X, Ling F, Guthikonda M, Rafols JA, Ding Y. Reduced apoptosis by combining normobaric oxygenation with ethanol in transient ischemic stroke. Brain Res 2013; 1531:17-24. [PMID: 23920008 DOI: 10.1016/j.brainres.2013.07.051] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 07/22/2013] [Accepted: 07/29/2013] [Indexed: 10/26/2022]
Abstract
BACKGROUND AND PURPOSE The effect of normobaric oxygen (NBO) on apoptosis remains controversial. The present study evaluated the effect of NBO on ischemia-induced apoptosis and assessed the potential for improved outcomes by combining NBO administration with another neuroprotective agent, ethanol, in a rat stroke model. METHODS Rats were subjected to right middle cerebral artery occlusion (MCAO) for 2h. At the onset of reperfusion, ischemic animals received either NBO (2h duration), an intraperitoneal injection of ethanol (1.0g/kg), or both NBO and ethanol. Extent of brain injury was determined by infarct volume, neurological deficit, and apoptotic cell death. Expression of pro- and anti-apoptotic proteins was evaluated through Western immunoblotting. RESULTS Given alone, NBO and ethanol each slightly (p<0.05) reduced infarct volume to 38% and 37%, respectively, as compared to the impressive reduction of 51% (p<0.01) seen with combined NBO-ethanol administration. Neurologic deficits were also significantly reduced by 48% with combined NBO-ethanol therapy, as compared to lesser reductions of 24% and 23% with NBO or ethanol, respectively. Combined NBO-ethanol therapy decreased apoptotic cell death by 49%, as compared to 31% with NBO and 30% with ethanol. Similarly, combination therapy significantly increased expression of anti-apoptotic factors (Bcl-2 and Bcl-xL) and significantly reduced expression of pro-apoptotic proteins (BAX, Caspase-3, and AIF), as compared to the minimal or nil protein expression changes elicited by NBO or ethanol alone. CONCLUSIONS In rats subjected to ischemic stroke, NBO administration salvages ischemic brain tissue through evidenced decrease in apoptotic cell death. Combined NBO therapy with ethanol administration greatly improves both degree of neuroprotection and associated apoptosis.
Collapse
Affiliation(s)
- Xiaokun Geng
- China-America Institute of Neuroscience, Luhe Hospital, Capital Medical University, Beijing, China
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Zhang P, Hou J, Fu J, Li D, Zhang C, Liu J. Baicalin protects rat brain microvascular endothelial cells injured by oxygen-glucose deprivation via anti-inflammation. Brain Res Bull 2013; 97:8-15. [DOI: 10.1016/j.brainresbull.2013.05.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2013] [Revised: 05/10/2013] [Accepted: 05/13/2013] [Indexed: 11/30/2022]
|
33
|
Xu A, Szczepanek K, Hu Y, Lesnefsky EJ, Chen Q. Cardioprotection by modulation of mitochondrial respiration during ischemia-reperfusion: role of apoptosis-inducing factor. Biochem Biophys Res Commun 2013; 435:627-33. [PMID: 23685150 DOI: 10.1016/j.bbrc.2013.05.033] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2013] [Accepted: 05/08/2013] [Indexed: 10/26/2022]
Abstract
The transient, reversible blockade of electron transport (BET) during ischemia or at the onset of reperfusion protects mitochondria and decreases cardiac injury. Apoptosis inducing factor (AIF) is located within the mitochondrial intermembrane space. A release of AIF from mitochondria into cytosol and nucleus triggers caspase-independent cell death. We asked if BET prevents the loss of AIF from mitochondria as a mechanism of protection in the buffer perfused heart. BET during ischemia with amobarbital, a rapidly reversible inhibitor of mitochondrial complex I, attenuated a release of AIF from mitochondria into cytosol, in turn decreasing the formation of cleaved and activated PARP-1. These results suggest that BET-mediated protection may occur through prevention of the loss of AIF from mitochondria during ischemia-reperfusion. In order to further clarify the role of mitochondrial AIF in BET-mediated protection, Harlequin (Hq) mice, a genetic model with mitochondrial AIF deficiency, were used to test whether BET could still decrease cell injury in Hq mouse hearts during reperfusion. BET during ischemia protected Hq mouse hearts against ischemia-reperfusion injury and improved mitochondrial function in these hearts during reperfusion. Thus, cardiac injury can still be decreased in the presence of down-regulated mitochondrial AIF content. Taken together, BET during ischemia protects both hearts with normal mitochondrial AIF content and hearts with mitochondrial AIF deficiency. Although preservation of mitochondrial AIF content plays a key role in reducing cell injury during reperfusion, the protection derived from the BET is not fully dependent on AIF-driven mechanisms.
Collapse
Affiliation(s)
- Aijun Xu
- Department of Internal Medicine (Division of Cardiology), Virginia Commonwealth University, Richmond, VA 23298, USA
| | | | | | | | | |
Collapse
|
34
|
Chong ZZ, Yao Q, Li HH. The rationale of targeting mammalian target of rapamycin for ischemic stroke. Cell Signal 2013; 25:1598-607. [PMID: 23563259 DOI: 10.1016/j.cellsig.2013.03.017] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2013] [Accepted: 03/28/2013] [Indexed: 02/06/2023]
Abstract
Given the current limitation of therapeutic approach for ischemic stroke, a leading cause of disability and mortality in the developed countries, to develop new therapeutic strategies for this devastating disease is urgently necessary. As a serine/threonine kinase, mammalian target of rapamycin (mTOR) activation can mediate broad biological activities that include protein synthesis, cytoskeleton organization, and cell survival. mTOR functions through mTORC1 and mTORC2 complexes and their multiple downstream substrates, such as eukaryotic initiation factor 4E-binding protein 1, p70 ribosomal S6 kinase, sterol regulatory element-binding protein 1, hypoxia inducible factor-1, and signal transducer and activator transcription 3, Yin Ying 1, Akt, protein kinase c-alpha, Rho GTPase, serum-and gucocorticoid-induced protein kinase 1, etc. Specially, the role of mTOR in the central nervous system has been attracting considerable attention. Based on the ability of mTOR to prevent neuronal apoptosis, inhibit autophagic cell death, promote neurogenesis, and improve angiogenesis, mTOR may acquire the capability of limiting the ischemic neuronal death and promoting the neurological recovery. Consequently, to regulate the activity of mTOR holds a potential as a novel therapeutic strategy for ischemic stroke.
Collapse
Affiliation(s)
- Zhao Zhong Chong
- Department of Neurology and Neurosciences, Cancer Center, New Jersey Medical School, University of Medicine and Dentistry of New Jersey, Newark, NJ 07101, USA.
| | | | | |
Collapse
|
35
|
Fu P, Peng C, Ding JY, Asmaro K, Sullivan JM, Guthikonda M, Ding Y. Acute administration of ethanol reduces apoptosis following ischemic stroke in rats. Neurosci Res 2013; 76:93-7. [PMID: 23511554 DOI: 10.1016/j.neures.2013.02.011] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Revised: 02/01/2013] [Accepted: 02/19/2013] [Indexed: 01/23/2023]
Abstract
In recent studies, acute ethanol administration appears to play a neuroprotective role during ischemic stroke. We sought to confirm these findings by identifying if ethanol-derived neuroprotection is associated with a reduction in apoptosis. Ethanol at 0.5 and 1.5 g/kg doses was given by intraperitoneal injections to Sprague-Dawley rats after 2h of middle cerebral artery (MCA) occlusion, followed by reperfusion. We quantified apoptotic cell death in each of the treatment groups with ELISA, and measured pro- and anti-apoptotic protein expression with Western blot analysis. Cell death was significantly increased in rats after ischemia and was subsequently significantly reduced by the administration of 1.5 g/kg of ethanol. We found that the 1.5 g/kg dose promoted the expression of pro-survival factors and decreased the expression of apoptotic proteins at 3h after reperfusion. This effect was maintained at 24h for Caspase-3 and apoptosis-inducing factor (AIF), although not for Bcl-2, Bcl-xL, and Bcl-2-associated X (Bax). Administration of 0.5 g/kg of ethanol was not as effective in regulating protein expression as the 1.5 g/kg dose. Our study suggests that administration of ethanol at a dose of 1.5 g/kg after stroke - which provides rat blood alcohol levels equivalent to the legal driving limit - produces a differential protein profile, with increased expression of anti-apoptotic proteins and decrease in pro-apoptotic factors. This results in a significant reduction of neuronal apoptosis and is neuroprotective in ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Paul Fu
- Department of Neurological Surgery, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | | | | | | | | | | | | |
Collapse
|
36
|
Poly(ADP-ribose) polymerase inhibition protects epileptic hippocampal neurons from apoptosis via suppressing Akt-mediated apoptosis-inducing factor translocation in vitro. Neuroscience 2013. [DOI: 10.1016/j.neuroscience.2012.11.009] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
|
37
|
Moroni F, Cozzi A, Chiarugi A, Formentini L, Camaioni E, Pellegrini-Giampietro DE, Chen Y, Liang S, Zaleska MM, Gonzales C, Wood A, Pellicciari R. Long-lasting neuroprotection and neurological improvement in stroke models with new, potent and brain permeable inhibitors of poly(ADP-ribose) polymerase. Br J Pharmacol 2012; 165:1487-500. [PMID: 21913897 DOI: 10.1111/j.1476-5381.2011.01666.x] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND AND PURPOSES Thienyl-isoquinolone (TIQ-A) is a relatively potent PARP inhibitor able to reduce post-ischaemic neuronal death in vitro. Here we have studied, in different stroke models in vivo, the neuroprotective properties of DAMTIQ and HYDAMTIQ, two TIQ-A derivatives able to reach the brain and to inhibit PARP-1 and PARP-2. EXPERIMENTAL APPROACH Studies were carried out in (i) transient (2 h) middle cerebral artery occlusion (tMCAO), (ii) permanent MCAO (pMCAO) and (iii) electrocoagulation of the distal portion of MCA in conjunction with transient (90 min) bilateral carotid occlusion (focal cortical ischaemia). KEY RESULTS In male rats with tMCAO, HYDAMTIQ (0.1-10 mg·kg(-1)) injected i.p. three times, starting 4 h after MCAO, reduced infarct volumes by up to 70%, reduced the loss of body weight by up to 60% and attenuated the neurological impairment by up to 40%. In age-matched female rats, HYDAMTIQ also reduced brain damage. Protection, however, was less pronounced than in the male rats. In animals with pMCAO, HYDAMTIQ administered 30 min after MCAO reduced infarct volumes by approximately 40%. In animals with focal cortical ischaemia, HYDAMTIQ treatment decreased post-ischaemic accumulation of PAR (the product of PARP activity) and the presence of OX42-positive inflammatory cells in the ischaemic cortex. It also reduced sensorimotor deficits for up to 90 days after MCAO. CONCLUSION AND IMPLICATIONS Our results show that HYDAMTIQ is a potent PARP inhibitor that conferred robust neuroprotection and long-lasting improvement of post-stroke neurological deficits.
Collapse
Affiliation(s)
- F Moroni
- Department of Preclinical and Clinical Pharmacology, University of Florence, Florence, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
38
|
Kim GS, Jung JE, Narasimhan P, Sakata H, Yoshioka H, Song YS, Okami N, Chan PH. Release of mitochondrial apoptogenic factors and cell death are mediated by CK2 and NADPH oxidase. J Cereb Blood Flow Metab 2012; 32:720-30. [PMID: 22146192 PMCID: PMC3318149 DOI: 10.1038/jcbfm.2011.176] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/18/2011] [Revised: 10/28/2011] [Accepted: 11/09/2011] [Indexed: 11/09/2022]
Abstract
Activation of the NADPH oxidase subunit, NOX2, and increased oxidative stress are associated with neuronal death after cerebral ischemia and reperfusion. Inhibition of NOX2 by casein kinase 2 (CK2) leads to neuronal survival, but the mechanism is unknown. In this study, we show that in copper/zinc-superoxide dismutase transgenic (SOD1 Tg) mice, degradation of CK2α and CK2α' and dephosphorylation of CK2β against oxidative stress were markedly reduced compared with wild-type (WT) mice that underwent middle cerebral artery occlusion. Inhibition of CK2 pharmacologically or by ischemic reperfusion facilitated accumulation of poly(ADP-ribose) polymers, the translocation of apoptosis-inducing factor (AIF), and cytochrome c release from mitochondria after ischemic injury. The eventual enhancement of CK2 inhibition under ischemic injury strongly increased 8-hydroxy-2'-deoxyguanosine and phosphorylation of H2A.X. Furthermore, CK2 inhibition by tetrabromocinnamic acid (TBCA) in SOD1 Tg and gp91 knockout (KO) mice after ischemia reperfusion induced less release of AIF and cytochrome c than in TBCA-treated WT mice. Inhibition of CK2 in gp91 KO mice subjected to ischemia reperfusion did not increase brain infarction compared with TBCA-treated WT mice. These results strongly suggest that NOX2 activation releases reactive oxygen species after CK2 inhibition, triggering release of apoptogenic factors from mitochondria and inducing DNA damage after ischemic brain injury.
Collapse
Affiliation(s)
- Gab Seok Kim
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Program in Neurosciences, Stanford University School of Medicine, Stanford, California, USA
| | - Joo Eun Jung
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Program in Neurosciences, Stanford University School of Medicine, Stanford, California, USA
| | - Purnima Narasimhan
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Program in Neurosciences, Stanford University School of Medicine, Stanford, California, USA
| | - Hiroyuki Sakata
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Program in Neurosciences, Stanford University School of Medicine, Stanford, California, USA
| | - Hideyuki Yoshioka
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Program in Neurosciences, Stanford University School of Medicine, Stanford, California, USA
| | - Yun Seon Song
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Program in Neurosciences, Stanford University School of Medicine, Stanford, California, USA
| | - Nobuya Okami
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Program in Neurosciences, Stanford University School of Medicine, Stanford, California, USA
| | - Pak H Chan
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California, USA
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, California, USA
- Program in Neurosciences, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
39
|
Zhang F, Wang S, Gan L, Vosler PS, Gao Y, Zigmond MJ, Chen J. Protective effects and mechanisms of sirtuins in the nervous system. Prog Neurobiol 2011; 95:373-95. [PMID: 21930182 DOI: 10.1016/j.pneurobio.2011.09.001] [Citation(s) in RCA: 161] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2011] [Revised: 08/29/2011] [Accepted: 09/01/2011] [Indexed: 12/13/2022]
Abstract
Silent information regulator two proteins (sirtuins or SIRTs) are a group of histone deacetylases whose activities are dependent on and regulated by nicotinamide adenine dinucleotide (NAD(+)). They suppress genome-wide transcription, yet upregulate a select set of proteins related to energy metabolism and pro-survival mechanisms, and therefore play a key role in the longevity effects elicited by calorie restriction. Recently, a neuroprotective effect of sirtuins has been reported for both acute and chronic neurological diseases. The focus of this review is to summarize the latest progress regarding the protective effects of sirtuins, with a focus on SIRT1. We first introduce the distribution of sirtuins in the brain and how their expression and activity are regulated. We then highlight their protective effects against common neurological disorders, such as cerebral ischemia, axonal injury, Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and multiple sclerosis. Finally, we analyze the mechanisms underlying sirtuin-mediated neuroprotection, centering on their non-histone substrates such as DNA repair enzymes, protein kinases, transcription factors, and coactivators. Collectively, the information compiled here will serve as a comprehensive reference for the actions of sirtuins in the nervous system to date, and will hopefully help to design further experimental research and expand sirtuins as therapeutic targets in the future.
Collapse
Affiliation(s)
- Feng Zhang
- State Key Laboratory of Medical Neurobiology and Institute of Brain Science, Fudan University, Shanghai 200032, China.
| | | | | | | | | | | | | |
Collapse
|
40
|
Choudhury S, Bae S, Ke Q, Lee JY, Kim J, Kang PM. Mitochondria to nucleus translocation of AIF in mice lacking Hsp70 during ischemia/reperfusion. Basic Res Cardiol 2011; 106:397-407. [PMID: 21387140 PMCID: PMC3205442 DOI: 10.1007/s00395-011-0164-1] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/29/2010] [Revised: 02/02/2011] [Accepted: 02/21/2011] [Indexed: 11/25/2022]
Abstract
Heat shock protein 70 (Hsp70) has been shown to have an anti-apoptotic function, but its mechanism is not clear in heart. In this study, we examined the effect of Hsp70 deletion on AIF-induced apoptosis during ischemia/reperfusion (I/R) in vivo. Although Hsp70 KO and WT mice demonstrated similar amounts of AIF released from mitochondria after I/R surgery, Hsp70 KO mice showed a significantly greater increase in apoptosis, larger infarct size, and decreased cardiac output. There was also a significant fourfold increase in the nuclear accumulation of AIF in Hsp70 KO mice compared with WT mice. Treatment with 4-AN (4-amino-1,8-napthalimide, 3 mg/kg), a potent inhibitor of PARP-1, which is a critical regulator of AIF-induced apoptosis, significantly blocked the release of AIF from mitochondria and the translocation of AIF into the nuclei after I/R in both WT and Hsp70 KO mice. In addition, 4-AN treatment resulted in a significant inhibition of apoptosis, a reduction of infarct size, and attenuated cardiac dysfunction in both WT and Hsp70 KO mice after I/R. The anti-apoptotic function of Hsp70 occurs through the inhibition of AIF-induced apoptosis by blocking the mitochondria to nucleus translocation of AIF. PARP-1 inhibition improves cardiac function by blocking AIF-induced apoptosis.
Collapse
Affiliation(s)
- Sangita Choudhury
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Soochan Bae
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Qingen Ke
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Ji Yoo Lee
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Jacob Kim
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Peter M. Kang
- Cardiovascular Institute, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA, Department of BIN Fusion Technology, Chonbuk National University, Jeonju, South Korea, Cardiovascular Institute, Beth Israel Deaconess Medical Center, 3 Blackfan Circle, CLS-910, Boston, MA 02215, USA,
| |
Collapse
|
41
|
Feng J, Zhang D, Chen B. Endothelial mechanisms of endothelial dysfunction in patients with obstructive sleep apnea. Sleep Breath 2011; 16:283-94. [PMID: 21479903 DOI: 10.1007/s11325-011-0519-8] [Citation(s) in RCA: 61] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2011] [Revised: 03/23/2011] [Accepted: 03/30/2011] [Indexed: 12/12/2022]
Abstract
BACKGROUND Obstructive sleep apnea (OSA) occurs in 2% of middle-aged women and 4% of middle-aged men in the general population and the prevalence is much higher in specific patient groups. Intermittent hypoxia (IH, oxygen desaturation and re-oxygenation) cycle, a major pathophysiologic character of OSA, and the physiological responses this evokes are thought to be responsible for its association with increased cardiovascular morbidity and mortality. Endothelial dysfunction, resulting from IH and as a key early event in atherosclerosis, was demonstrated repeatedly in patients with OSA and in animal models of IH, providing an important mechanistic link between the acute cyclical IH during sleep and the increased prevalence of chronic vascular diseases. CONCLUSIONS From this work, we conclude that IH from OSA may result in endothelial dysfunction, as a potential promoter of atherosclerosis, through nitric oxide unavailability, oxidative stress and inflammation, cell apoptosis, the crosstalk between endothelial cells and circulating inflammatory cells, microparticles, and damage repairing process. Though effective continuous positive airway pressure (CPAP) may specifically improve endothelial function, more controlled larger interventional trials that will include multiple centers and randomized allocation of CPAP therapy are needed to see if such changes are reversible before cause and effect can be implied finally, while further studies on cellular and animal level are also needed to elucidate molecular biologic/pathologic pathways.
Collapse
Affiliation(s)
- Jing Feng
- Respiratory Department of Tianjin Medical University General Hospital, Tianjin, 300052, China
| | | | | |
Collapse
|
42
|
Strosznajder RP, Czubowicz K, Jesko H, Strosznajder JB. Poly(ADP-ribose) metabolism in brain and its role in ischemia pathology. Mol Neurobiol 2010; 41:187-96. [PMID: 20411356 DOI: 10.1007/s12035-010-8124-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2009] [Accepted: 03/23/2010] [Indexed: 11/24/2022]
Abstract
The biological roles of poly(ADP-ribose) polymers (PAR) and poly(ADP-ribosyl)ation of proteins in the central nervous system are diverse. The homeostasis of PAR orchestrated by poly(ADP-ribose) polymerase-1 (PARP-1) and poly(ADP-ribose) glycohydrolase (PARG) is crucial for cell physiology and pathology. Both enzymes are ubiquitously distributed in neurons and glia; however, they are segregated at the subcellular level. PARP-1 serves as a "nick sensor" for single- or double-stranded breaks in DNA and is involved in long and short patch base-excision repair, while PARG breaks down PAR. The stimulation of PARP-1 and PAR formation can activate proinflammatory transcription factors, including nuclear factor kappa B. However, hyperactivation of PARP-1 can result in depletion of NAD/ATP, and in PAR-dependent mitochondrial pore formation leading to release of apoptosis inducing factor and cell death. The role of PAR as a death signaling molecule in brain ischemia-reperfusion and inflammation as well as the effect of gender and aging is presented in this review. Modulating the PAR level through pharmacological or genetic intervention on PARP-1/PARG activity and gene expression should be a valuable way for neuroprotective strategy.
Collapse
Affiliation(s)
- Robert Piotr Strosznajder
- Department of Neurosurgery, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawinskiego St., 02-106, Warsaw, Poland.
| | | | | | | |
Collapse
|
43
|
Choudhury S, Bae S, Kumar SR, Ke Q, Yalamarti B, Choi JH, Kirshenbaum LA, Kang PM. Role of AIF in cardiac apoptosis in hypertrophic cardiomyocytes from Dahl salt-sensitive rats. Cardiovasc Res 2010; 85:28-37. [PMID: 19633014 DOI: 10.1093/cvr/cvp261] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
AIMS The caspases are thought to be central mediators of the apoptotic program, but recent data indicate that apoptosis may also be mediated by caspase-independent mechanisms such as apoptosis-inducing factor (AIF). The role of AIF-induced apoptosis in heart, however, is currently not well understood. The aim of this study was to investigate the presence of and conditions for AIF-induced cardiac apoptosis in vitro. METHODS AND RESULTS Hypertrophic cardiomyocyte (H-CM) cultures were prepared from the hearts of Dahl salt-sensitive rats fed a high salt diet. Apoptotic stimulation induced by hypoxia/reoxygenation or staurosporine (1 microM) enhanced AIF release in H-CMs compared with non-hypertrophic cardiomyocytes (N-CMs). Caspase inhibition using zVAD.fmk (25 microM) or overexpression of CrmA using recombinant adenovirus only partially protected N-CMs from apoptosis (63 +/- 0.93%) and provided no significant protection against apoptosis in hypertrophic cells (23 +/- 1.03%). On the other hand, poly-ADP-ribose polymerase inhibition using 4-AN (20 microM) during apoptotic stimulation blocked the release of AIF from mitochondria and significantly improved cell viability in hypertrophied cardiomyocytes (74 +/- 1.18%). CONCLUSION A caspase-dependent, apoptotic pathway is important for N-CM death, whereas a caspase-independent, AIF-mediated pathway plays a critical role in H-CMs.
Collapse
|
44
|
Andresen JH, Løberg EM, Wright M, Goverud IL, Stray-Pedersen B, Saugstad OD. Nicotine affects the expression of brain-derived neurotrophic factor mRNA and protein in the hippocampus of hypoxic newborn piglets. J Perinat Med 2010; 37:553-60. [PMID: 19492919 DOI: 10.1515/jpm.2009.081] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Brain-derived neurotrophic factor (BDNF) is highly expressed in the developing brain. It has anti-apoptotic abilities, and protects the neonatal brain. In experimental settings in adult animals, pre-treatment with nicotine has shown increased BDNF levels, indicating a possible contribution to nicotine's anti-apoptotic effect. Apoptosis contributes to the development of brain damage in perinatal asphyxia. We examined the effects of nicotine on apoptosis-inducing factor (AIF), caspase-3 and BDNF in the hippocampus of a neonatal piglet model of global hypoxia. Forty-one anesthetized newborn piglets were randomized to one of four groups receiving different infusions after hypoxia (1) nicotine 130 microg/kg/h, 2) 260 microg/kg/h, 3) adrenaline, and 4) saline, all 2.6 mL/kg/h. Four hours after hypoxia they were euthanized. The left hemisphere/hippocampus was examined by histopathology and immunohistochemistry; the right hippocampus was analyzed using real time PCR. There was a significantly higher expression of BDNF mRNA and protein in the animals treated with nicotine 130 microg/kg/h vs. the saline treated group (mRNA P=0.038; protein P=0.009). There were no differences regarding AIF or caspase-3. We conclude that nicotine (130 microg/kg/h), infused over 1 h after global hypoxia in neonatal piglets, increases levels of both BDNF mRNA and protein in the hippocampus. This might imply neuroprotective effects of nicotine in asphyxiated neonates.
Collapse
Affiliation(s)
- Jannicke Hanne Andresen
- Department of Pediatric Research, Institute of Surgical Research, Medical Faculty, University of Oslo, Rikshospitalet University Hospital, Oslo, Norway.
| | | | | | | | | | | |
Collapse
|
45
|
Jelic S, Lederer DJ, Adams T, Padeletti M, Colombo PC, Factor P, Le Jemtel TH. Endothelial repair capacity and apoptosis are inversely related in obstructive sleep apnea. Vasc Health Risk Manag 2009; 5:909-20. [PMID: 19997572 PMCID: PMC2788596 DOI: 10.2147/vhrm.s8123] [Citation(s) in RCA: 53] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Purpose: To investigate the impact of obstructive sleep apnea (OSA) on endothelial repair capacity and apoptosis in the absence of potentially confounding factors including obesity. Patients and methods: Sixteen patients with a body mass index <30 and newly diagnosed OSA and 16 controls were studied. Circulating levels of endothelial progenitor cells, a marker of endothelial repair capacity, and endothelial microparticles, a marker of endothelial apoptosis, were quantified before and after four-week therapy with continuous positive airway pressure (CPAP). Endothelial cell apoptotic rate was also quantified in freshly harvested venous endothelial cells. Vascular reactivity was measured by flow-mediated dilation. Results: Before treatment, endothelial microparticle levels were greater and endothelial progenitor cell levels were lower in patients with OSA than in controls (P < 0.001 for both). Levels of endothelial microparticles and progenitors cells were inversely related (r = −0.67, P < 0.001). Endothelial progenitor cell levels increased after effective treatment (P = 0.036). Conclusions: In the absence of any co-morbid conditions including obesity, OSA alone impairs endothelial repair capacity and promotes endothelial apoptosis. These early endothelial alterations may underlie accelerated atherosclerosis and increased cardiovascular risk in OSA.
Collapse
Affiliation(s)
- Sanja Jelic
- Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA.
| | | | | | | | | | | | | |
Collapse
|
46
|
Xing C, Lee S, Kim WJ, Wang H, Yang YG, Ning M, Wang X, Lo EH. Neurovascular effects of CD47 signaling: promotion of cell death, inflammation, and suppression of angiogenesis in brain endothelial cells in vitro. J Neurosci Res 2009; 87:2571-7. [PMID: 19360900 DOI: 10.1002/jnr.22076] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
The concept of the neurovascular unit emphasizes that common signals and substrates underlie the physiology and pathophysiology of neuronal and endothelial compartments in brain. Recent data suggest that activation of the integrin-associated protein CD47 promotes neuronal cell death. Is it possible that CD47 may also negatively affect cerebral endothelial cells? Exposure of wild-type primary mouse cerebral endothelial cells to the CD47 ligand thrombospondin 1 (TSP-1) induced an increasing amount of cell death, whereas cytotoxicity was significantly decreased in cerebral endothelial cells derived from CD47 knockout mice. The specific CD47-activating peptide, 4N1K, similarly induced cell death in human brain microvascular endothelial cells. Promotion of inflammation was also involved because lower TSP-1 was able to up-regulate the adhesion molecules intercellular adhesion molecule-1 and vascular cell adhesion molecule-1. Finally, CD47 signaling may suppress angiogenesis because 4N1K significantly inhibited endothelial cell migration and tube formation in vitro. We conclude that CD47 signaling can negatively affect the viability and function of cerebral endothelial cells, further supporting the notion that CD47 may be a potential neurovascular target for stroke and brain injury.
Collapse
Affiliation(s)
- Changhong Xing
- Neuroprotection Research Laboratory, Department of Radiology and Neurology, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02129, USA
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
Endothelial activation and inflammation are important mediators of accelerated atherogenesis and consequent increased cardiovascular morbidity in obstructive sleep apnea (OSA). Repetitive episodes of hypoxia/reoxygenation associated with transient cessation of breathing during sleep in OSA resemble ischemia/reperfusion injury and may be the main culprit underlying endothelial dysfunction in OSA. Additional factors such as repetitive arousals resulting in sleep fragmentation and deprivation and individual genetic suseptibility to vascular manifestations of OSA contribute to impaired endothelial function in OSA. The present review focuses on possible mechanisms that underlie endothelial activation and inflammation in OSA.
Collapse
Affiliation(s)
- Amy Atkeson
- Division of Pulmonary, Allergy, and Critical Care Medicine, Columbia University College of Physicians and Surgeons, 630 West 168th Street, New York, NY 10032, USA
| | | |
Collapse
|
48
|
Yamada T, Ichimura K, Kanekatsu M, van Doorn WG. Homologs of genes associated with programmed cell death in animal cells are differentially expressed during senescence of Ipomoea nil petals. PLANT & CELL PHYSIOLOGY 2009; 50:610-25. [PMID: 19182226 DOI: 10.1093/pcp/pcp019] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
In senescent petals of Ipomoea nil, we investigated the expression of genes showing homology to genes involved in animal programmed cell death (PCD). Three encoded proteins were homologous to apoptotic proteins in animals: Bax inhibitor-1 (BI-1), a vacuolar processing enzyme (VPE; homologous to caspases) and a monodehydroascorbate reductase [MDAR; homologous to apoptosis-inducing factor (AIF)]. AIFs harbor an oxidoreductase domain and an apoptotic domain. MDARs exhibit homology to the AIF oxidoreductase domain, not to the apoptotic domain. The three other genes studied relate to autophagy. They encode homologs to vacuolar protein sorting 34 (VPS34) and to the Arabidopsis autophagy-related proteins 4b and 8a (ATG4b and ATG8a). The transcript abundance of MDAR decreased continuously, whereas that of the other genes studies exhibited a transient increase, except ATG4b whose abundance stayed high after an increase. Treatment with ethylene advanced the time to visible petal senescence, and hastened the changes in expression of each of the genes studied. In order to assess the role of VPS34 in petal senescence, we studied the effect of its inhibitor 3-methyladenine (3-MA). 3-MA reduced the time to visible petal senescence, and also accelerated the time to DNA degradation. Remarkably, 3-MA increased the time to nuclear fragmentation, indicating that the time to visible petal senescence was independent of nuclear fragmentation. The data on 3-MA might suggest the idea that autophagy is not a cause of PCD, but part of the remobilization process.
Collapse
Affiliation(s)
- Tetsuya Yamada
- National Institute of Floricultural Science, Tsukuba 305-8519, Japan.
| | | | | | | |
Collapse
|
49
|
Hua Q, Zhu XL, Li PT, Liu Y, Zhang N, Xu Y, Jia X. The inhibitory effects of cholalic acid and hyodeoxycholalic acid on the expression of TNFalpha and IL-1beta after cerebral ischemia in rats. Arch Pharm Res 2009; 32:65-73. [PMID: 19183878 DOI: 10.1007/s12272-009-1119-z] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2008] [Revised: 12/04/2008] [Accepted: 12/05/2008] [Indexed: 11/26/2022]
Abstract
Previous studies have shown that Qing Kai Ling, a traditional Chinese medicine, was able to effectively prevent the inflammation from cerebral ischemia (Chen et al., 2002). The cholalic acid and hyodeoxycholalic acid (cholalic acid mixture) was major active components in Qing Kai Ling. To study the effects of cholalic acid mixture on the damage cascade of cerebral ischemia, rat model of focal cerebral ischemia was established by permanent occlusion of left middle cerebral artery. We found that the administration of cholalic acid mixture could reduce the ischemic infarct size after 24 h of ischemia, and cholalic acid mixture could be detected in cerebrospinal fluid after 2h of administration. We also found that the concentrations of tumor necrosis factor-alpha and interlukin-1beta in rat brain were significantly lower when compared to the untreated animals after 12 h and 24 h of ischemia. The concentrations of von Willebrand factor and neuron specific enolase in the plasma were remarkably decreased in cholalic acid mixture treated animals than in the untreated ones after 12h of ischemia. Our results suggested that cholalic acid mixture is able to decrease the expression of inflammation factors including tumor necrosis factor-alpha and interlukin-1beta after focal cerebral ischemia.
Collapse
Affiliation(s)
- Qian Hua
- School of Preclinical Medicine, Beijing University of Chinese Medicine, Chaoyang District, Beijing, 100029, China
| | | | | | | | | | | | | |
Collapse
|
50
|
High glucose and oxidative/nitrosative stress conditions induce apoptosis in retinal endothelial cells by a caspase-independent pathway. Exp Eye Res 2009; 88:983-91. [PMID: 19146853 DOI: 10.1016/j.exer.2008.12.010] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2008] [Revised: 11/29/2008] [Accepted: 12/19/2008] [Indexed: 11/21/2022]
Abstract
Diabetic retinopathy (DR) is a leading cause of vision loss among working-age adults. Retinal endothelial cell apoptosis is an early event in DR, and oxidative stress is known to play an important role in this pathology. Recently, we found that high glucose induces apoptosis in retinal neural cells by a caspase-independent mechanism. Here, we investigated the mechanisms underlying retinal endothelial cell apoptosis induced by high glucose and oxidative/nitrosative stress conditions. Endothelial cells (TR-iBRB2 rat retinal endothelial cell line) were exposed to high glucose (long-term exposure, 7 days), or to NOC-18 (nitric oxide donor; 250microM) or H(2)O(2) (100microM) for 24h. Cell viability was assessed by the MTT assay and cell proliferation by [methyl-(3)H]-thymidine incorporation into DNA. Apoptotic cells were detected with Hoechst or Annexin V staining. Active caspases were detected by an apoptosis detection kit. Active caspase-3 and apoptosis-inducing factor (AIF) protein levels were assessed by Western blot or immunohistochemistry. High glucose, NOC-18 and H(2)O(2) increased apoptosis in retinal endothelial cells. High glucose and mannitol decreased cell proliferation, but mannitol did not induce apoptosis. Caspase activation did not increase in high glucose- or NOC-18-treated cells, but it increased in cells exposed to H(2)O(2). However, the protein levels of AIF decreased in mitochondrial fractions and increased in nuclear fractions, in all conditions. These results are the first demonstrating that retinal endothelial cell apoptosis induced by high glucose is independent of caspase activation, and is correlated with AIF translocation to the nucleus. NOC-18 and H(2)O(2) also activate a caspase-independent apoptotic pathway, although H(2)O(2) can also induce caspase-mediated apoptosis.
Collapse
|