101
|
Xia M, Ding Q, Zhang Z, Feng Q. Remote Limb Ischemic Preconditioning Protects Rats Against Cerebral Ischemia via HIF-1α/AMPK/HSP70 Pathway. Cell Mol Neurobiol 2017; 37:1105-1114. [PMID: 27896629 PMCID: PMC11482205 DOI: 10.1007/s10571-016-0444-2] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2016] [Accepted: 11/09/2016] [Indexed: 01/06/2023]
Abstract
Remote limb ischemic preconditioning (RIPC) is a clinically feasible strategy to protect against ischemia/reperfusion injury, but the knowledge concerning the mechanism underlying RIPC is scarce. This study was performed to examine the effect of RIPC on brain tissue suffering from ischemia challenge and explore its underlying mechanism in a rat model. The animals were divided into four groups: Sham, middle cerebral artery occlusion (MCAO), RIPC, and MCAO+RIPC. We found that previous exposure to RIPC significantly attenuated neurological dysfunction and lessened brain edema in MCAO+RIPC group. Moreover, other important events were observed in MCAO+RIPC group, including substantial decrements in the concentrations of oxidative response indicators [malondialdehyde (MDA), 8-hydroxy-2-deoxyguanosine (8-OHdG), and protein carbonyl], significant reductions in levels of inflammation mediators [myeloperoxidase (MPO), tumor necrosis factor-a (TNF-a), interleukin-1β (IL-1β), and IL-6], and significant decline in neuronal apoptosis revealed by a smaller number of TUNEL-positive cells. Interestingly, both MCAO and RIPC groups exhibited meaningful elevations in the levels of HIF-1a, HSP70, and AMP-activated protein kinase (AMPK) compared to Sham group, and previous exposure to RIPC further elevated the levels of HIF-1a, HSP70, and AMPK in MCAO+RIPC group. Furthermore, the administration of YC-1 (HIF-1 inhibitor), 8-bAMP (AMPK inhibitor), and Quercetin (HSP70 inhibitor) to MCAO+RIPC rats demonstrated that HIF-1α/AMPK/HSP70 was involved in RIPC-mediated protection against cerebral ischemia.
Collapse
Affiliation(s)
- Ming Xia
- Chinese Internal Medicine, Putuo District Central Hospital, No. 164 Lanxi Road, Putuo District, Shanghai, 200062, China
| | - Qian Ding
- Chinese Internal Medicine, Putuo District Central Hospital, No. 164 Lanxi Road, Putuo District, Shanghai, 200062, China
| | - Zhidan Zhang
- Chinese Internal Medicine, Putuo District Central Hospital, No. 164 Lanxi Road, Putuo District, Shanghai, 200062, China
| | - Qinggen Feng
- Chinese Internal Medicine, Putuo District Central Hospital, No. 164 Lanxi Road, Putuo District, Shanghai, 200062, China.
| |
Collapse
|
102
|
Geng J, Wang L, Qu M, Song Y, Lin X, Chen Y, Mamtilahun M, Chen S, Zhang Z, Wang Y, Yang GY. Endothelial progenitor cells transplantation attenuated blood-brain barrier damage after ischemia in diabetic mice via HIF-1α. Stem Cell Res Ther 2017; 8:163. [PMID: 28697748 PMCID: PMC5505148 DOI: 10.1186/s13287-017-0605-3] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Revised: 04/20/2017] [Accepted: 06/07/2017] [Indexed: 12/20/2022] Open
Abstract
Background Blood-brain barrier impairment is a major indicator of endothelial dysfunction in diabetes. Studies showed that endothelial progenitor cell (EPC) transplantation promoted angiogenesis and improved function recovery after hind limb ischemia in diabetic mice. The effect of EPC transplantation on blood-brain barrier integrity after cerebral ischemia in diabetic animals is unknown. The aim of this study is to explore the effect of EPC transplantation on the integrity of the blood-brain barrier after cerebral ischemia in diabetic mice. Methods EPCs were isolated by density gradient centrifugation and characterized by flow cytometry and immunostaining. Diabetes was induced in adult male C57BL/6 mice by a single injection of streptozotocin at 4 weeks before surgery. Diabetic mice underwent 90-minute transient middle cerebral artery occlusion surgery and received 1 × 106 EPCs transplantation immediately after reperfusion. Brain infarct volume, blood-brain barrier permeability, tight junction protein expression, and hypoxia inducible factor-1α (HIF-1α) mRNA level were examined after treatment. Results We demonstrated that neurological deficits were attenuated and brain infarct volume was reduced in EPC-transplanted diabetic mice after transient cerebral ischemia compared to the controls (p < 0.05). Blood-brain barrier leakage and tight junction protein degradation were reduced in EPC-transplanted mice (p <0.05). EPCs upregulated HIF-1α expression while HIF-1α inhibitor PX-478 abolished the beneficial effect of EPCs. Conclusions We conclude that EPCs protected blood-brain barrier integrity after focal ischemia in diabetic mice through upregulation of HIF-1α signaling.
Collapse
Affiliation(s)
- Jieli Geng
- Department of Neurology, Shanghai Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.,Department of Neurology, Shanghai Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Liping Wang
- Department of Neurology, Shanghai Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.,Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Hua Shan Road, Shanghai, 200030, China
| | - Meijie Qu
- Department of Neurology, Shanghai Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.,Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Hua Shan Road, Shanghai, 200030, China
| | - Yaying Song
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Hua Shan Road, Shanghai, 200030, China
| | - Xiaojie Lin
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Hua Shan Road, Shanghai, 200030, China
| | - Yajing Chen
- Department of Neurology, Shanghai Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China.,Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Hua Shan Road, Shanghai, 200030, China
| | - Muyassar Mamtilahun
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Hua Shan Road, Shanghai, 200030, China
| | - Shengdi Chen
- Department of Neurology, Shanghai Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China
| | - Zhijun Zhang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Hua Shan Road, Shanghai, 200030, China
| | - Yongting Wang
- Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Hua Shan Road, Shanghai, 200030, China.
| | - Guo-Yuan Yang
- Department of Neurology, Shanghai Ruijin Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200025, China. .,Neuroscience and Neuroengineering Research Center, Med-X Research Institute and School of Biomedical Engineering, Shanghai Jiao Tong University, 1954 Hua Shan Road, Shanghai, 200030, China.
| |
Collapse
|
103
|
Button EL, Bersten DC, Whitelaw ML. HIF has Biff – Crosstalk between HIF1a and the family of bHLH/PAS proteins. Exp Cell Res 2017; 356:141-145. [DOI: 10.1016/j.yexcr.2017.03.055] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2017] [Accepted: 03/27/2017] [Indexed: 12/18/2022]
|
104
|
Guan SY, Leng RX, Tao JH, Li XP, Ye DQ, Olsen N, Zheng SG, Pan HF. Hypoxia-inducible factor-1α: a promising therapeutic target for autoimmune diseases. Expert Opin Ther Targets 2017; 21:715-723. [PMID: 28553732 DOI: 10.1080/14728222.2017.1336539] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
INTRODUCTION Hypoxia-inducible factor-1α (HIF-1α) plays a crucial role in both innate and adaptive immunity. Emerging evidence indicates that HIF-1α is associated with the inflammation and pathologic activities of autoimmune diseases. Areas covered: Considering that the types of autoimmune diseases are complicated and various, this review aims to cover the typical kinds of autoimmune diseases, discuss the molecular mechanisms, biological functions and expression of HIF-1α in these diseases, and further explore its therapeutic potential. Expert opinion: Inflammation and hypoxia are interdependent. HIF-1α as a key regulator of hypoxia, exerts a crucial role in the balance between Th17 and Treg, and involves in the inflammation and pathologic activities of autoimmune diseases. Although there are many challenges remaining to be overcome, targeting HIF-1α could be a promising strategy for autoimmune diseases therapies.
Collapse
Affiliation(s)
- Shi-Yang Guan
- a Department of Epidemiology and Biostatistics, School of Public Health , Anhui Medical University , Hefei , China.,b Anhui provincial laboratory of population health & major disease screening and diagnosis , Hefei , China
| | - Rui-Xue Leng
- a Department of Epidemiology and Biostatistics, School of Public Health , Anhui Medical University , Hefei , China.,b Anhui provincial laboratory of population health & major disease screening and diagnosis , Hefei , China
| | - Jin-Hui Tao
- c Department of Rheumatology , Anhui Provincial Hospital , Hefei , China
| | - Xiang-Pei Li
- c Department of Rheumatology , Anhui Provincial Hospital , Hefei , China
| | - Dong-Qing Ye
- a Department of Epidemiology and Biostatistics, School of Public Health , Anhui Medical University , Hefei , China.,b Anhui provincial laboratory of population health & major disease screening and diagnosis , Hefei , China
| | - Nancy Olsen
- d Division of Rheumatology , Penn State University Hershey College of Medicine , Hershey , PA , USA
| | - Song Guo Zheng
- d Division of Rheumatology , Penn State University Hershey College of Medicine , Hershey , PA , USA
| | - Hai-Feng Pan
- a Department of Epidemiology and Biostatistics, School of Public Health , Anhui Medical University , Hefei , China.,b Anhui provincial laboratory of population health & major disease screening and diagnosis , Hefei , China
| |
Collapse
|
105
|
Badawi Y, Shi H. Relative Contribution of Prolyl Hydroxylase-Dependent and -Independent Degradation of HIF-1alpha by Proteasomal Pathways in Cerebral Ischemia. Front Neurosci 2017; 11:239. [PMID: 28566998 PMCID: PMC5434458 DOI: 10.3389/fnins.2017.00239] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2016] [Accepted: 04/11/2017] [Indexed: 11/16/2022] Open
Abstract
Hypoxia inducible factor-1 (HIF-1) is a key regulator in hypoxia and can determine the fate of brain cells during ischemia. However, the mechanism of HIF-1 regulation is still not fully understood in ischemic brains. We tested a hypothesis that both the 26S and the 20S proteasomal pathways were involved in HIF-1α degradation under ischemic conditions. Using in vitro ischemic model (oxygen and glucose deprivation) and a mouse model of middle cerebral artery occlusion, we tested effects of inhibitors of proteasomes and prolyl hydroxylase (PHD) on HIF-1α stability and brain injury in cerebral ischemia. We observed that 30 and 60 min of oxygen-glucose deprivation significantly increased the 20S proteasomal activity. We demonstrated that proteasome inhibitors increased HIF-1α stabilization and cell viability and were more effective than PHD inhibitors in primary cultured cortical neurons exposed to oxygen and glucose deprivation. Furthermore, the administration of the proteasome inhibitor, epoxomicin, to mice resulted in smaller infarct size and brain edema than a PHD inhibitor. Our results indicate that 20S proteasomes are involved in HIF-1α degradation in ischemic neurons and that proteasomal inhibition provides more HIF-1α stabilization and neuroprotection than PHD inhibition in cerebral ischemia.
Collapse
Affiliation(s)
- Yomna Badawi
- Neuroscience Program, University of KansasLawrence, KS, USA.,Department of Pharmacology and Toxicology, University of KansasLawrence, KS, USA
| | - Honglian Shi
- Neuroscience Program, University of KansasLawrence, KS, USA.,Department of Pharmacology and Toxicology, University of KansasLawrence, KS, USA
| |
Collapse
|
106
|
Endogenous regeneration: Engineering growth factors for stroke. Neurochem Int 2017; 107:57-65. [PMID: 28411103 DOI: 10.1016/j.neuint.2017.03.024] [Citation(s) in RCA: 47] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2016] [Revised: 03/30/2017] [Accepted: 03/31/2017] [Indexed: 12/31/2022]
Abstract
Despite the efforts in developing therapeutics for stroke, recombinant tissue plasminogen activator (rtPA) remains the only FDA approved drug for ischemic stroke. Regenerative medicine targeting endogenous growth factors has drawn much interest in the clinical field as it provides potential restoration for the damaged brain tissue without being limited by a narrow therapeutic window. To date, most of the translational studies using regenerative medicines have encountered problems and failures. In this review, we discuss the effects of some trophic factors which include of erythropoietin (EPO), brain derived neurotrophic factor (BDNF), granulocyte-colony stimulating factor (G-CSF), vascular endothelial growth factor (VEGF), fibroblast growth factor (FGF), epidermal growth factor (EGF) and heparin binding epidermal growth factor (HB-EGF) in experimental ischemic stroke models and elaborate the lost in translation of the candidate growth factors from bench to bedside. Several new methodologies have been developed to overcome the caveats in translational studies. This review highlights the latest bioengineering approaches including the controlled release and delivery of growth factors by hydrogel-based scaffolds and the enhancement of half-life and selectivity of growth factors by a novel approach facilitated by glycosaminoglycans.
Collapse
|
107
|
Hypoxia Inducible Factor 1α Promotes Endogenous Adaptive Response in Rat Model of Chronic Cerebral Hypoperfusion. Int J Mol Sci 2017; 18:ijms18010003. [PMID: 28106731 PMCID: PMC5297638 DOI: 10.3390/ijms18010003] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2016] [Revised: 11/16/2016] [Accepted: 12/06/2016] [Indexed: 11/17/2022] Open
Abstract
Hypoxia inducible factor 1α (HIF-1α), a pivotal regulator of gene expression in response to hypoxia and ischemia, is now considered to regulate both pro-survival and pro-death responses depending on the duration and severity of the stress. We previously showed that chronic global cerebral hypoperfusion (CCH) triggered long-lasting accumulation of HIF-1α protein in the hippocampus of rats. However, the role of the stabilized HIF-1α in CCH is obscure. Here, we knock down endogenous HIF-1α to determine whether and how HIF-1α affects the disease processes and phenotypes of CCH. Lentivirus expressing HIF-1α small hairpin RNA was injected into the bilateral hippocampus and bilateral ventricles to knock down HIF-1α gene expression in the hippocampus and other brain areas. Permanent bilateral common carotid artery occlusions, known as 2-vessel occlusions (2VOs), were used to induce CCH in rats. Angiogenesis, oxidative stress, histopathological changes of the brain, and cognitive function were tested. Knockdown of HIF-1α prior to 2VO significantly exacerbates the impairment of learning and memory after four weeks of CCH. Mechanically, reduced cerebral angiogenesis, increased oxidative damage, and increased density of astrocytes and microglia in the cortex and some subregions of hippocampus are also shown after four weeks of CCH. Furthermore, HIF-1α knockdown also disrupts upregulation of regulated downstream genes. Our findings suggest that HIF-1α-protects the brain from oxidative stress and inflammation response in the disease process of CCH. Accumulated HIF-1α during CCH mediates endogenous adaptive processes to defend against more severe hypoperfusion injury of the brain, which may provide a therapeutic benefit.
Collapse
|
108
|
Xue L, Chen H, Lu K, Huang J, Duan H, Zhao Y. Clinical significance of changes in serum neuroglobin and HIF-1α concentrations during the early-phase of acute ischemic stroke. J Neurol Sci 2017; 375:52-57. [PMID: 28320188 DOI: 10.1016/j.jns.2017.01.039] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2016] [Revised: 01/05/2017] [Accepted: 01/11/2017] [Indexed: 10/20/2022]
Abstract
BACKGROUND Neuroglobin (NGB) has been described as a neuroprotective agent in cerebral ischemia, hypoxia inducible factor (HIF) has shown an important role in modulating hypoxic and ischemic injury, and therefore they have the potential to impact outcomes after acute ischemic stroke (AIS). Thus, we investigated early changes in the concentrations of serum NGB and HIF-1α after AIS and evaluated the relations of both NGB and HIF-1α to stroke severity and prognosis. METHODS We prospectively measured the serum concentrations of NGB and HIF-1α in 40 patients with AIS at 24, 48, 72, and 96h after stroke. Correlation combined with infarct size and National Institutes of Health Stroke Scale (NIHSS) score of the patients was analyzed. Receiver operating characteristic (ROC) curve was used to appraise their value in predicting the 90-day outcome after AIS. RESULTS Serum NGB concentrations increased and peaked at 72h after AIS, whereas serum concentrations of HIF-1α increased for 48h. Peak serum NGB concentration correlated significantly with both infarct size (R2=0.484, p<0.001) and admission NIHSS score (R2=0.578, p<0.001), while serum HIF-1α concentration was only correlated to a patient's infarct size (R2=0.394, p<0.001). ROC curve analysis suggested that the serum NGB concentration had a significantly better predictive power for poor outcome. CONCLUSIONS NGB level increased in serum after AIS accompanied by increases in serum HIF-1α, and was suggested as a predictor of stroke severity and poor prognosis.
Collapse
Affiliation(s)
- Lixia Xue
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Hao Chen
- Department of Neurosurgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Kaili Lu
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Jiankang Huang
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Hao Duan
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China
| | - Yuwu Zhao
- Department of Neurology, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai, China.
| |
Collapse
|
109
|
Hirayama Y, Koizumi S. Hypoxia-independent mechanisms of HIF-1α expression in astrocytes after ischemic preconditioning. Glia 2017; 65:523-530. [PMID: 28063215 DOI: 10.1002/glia.23109] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2016] [Revised: 12/02/2016] [Accepted: 12/02/2016] [Indexed: 11/06/2022]
Abstract
We recently demonstrated that ischemic tolerance was dependent on astrocytes, for which HIF-1α had an essential role. The mild ischemia (preconditioning; PC) increased HIF-1α in a biphasic pattern, that is, a quick and transient increase in neurons, followed by a slow and sustained increase in astrocytes. However, mechanisms underlying such temporal difference in HIF-1α increase remain totally unknown. Here, we show that unlike a hypoxia-dependent mechanism in neurons, astrocytes increase HIF-1α via a novel hypoxia-independent but P2X7-dependent mechanism. Using a middle cerebral artery occlusion (MCAO) model of mice, we found that the PC (a 15-min MCAO period)-evoked increase in HIF-1α in neurons was quick and transient (from 1 to 3 days after PC), but that in astrocytes was slow-onset and long-lasting (from 3 days to at least 2 weeks after PC). The neuronal HIF-1α increase was dependent on inhibition of PHD2, an oxygen-dependent HIF-1α degrading enzyme, whereas astrocytic one was independent of PHD2. Astrocytes even do not possess this enzyme. Instead, they produced a sustained increase in P2X7 receptors, activation of which resulted in HIF-1α increase. The hypoxia-independent but P2X7-receptor-dependent mechanism could allow astrocytes to cause long-lasting HIF-1α expression, thereby leading to induction of ischemic tolerance efficiently. GLIA 2017;65:523-530.
Collapse
Affiliation(s)
- Yuri Hirayama
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Yamanashi, 409-3898, Japan.,Department of Liaison Academy, Faculty of Medicine, University of Yamanashi, Chuo, Yamanashi, 409-3898, Japan
| | - Schuichi Koizumi
- Department of Neuropharmacology, Interdisciplinary Graduate School of Medicine, University of Yamanashi, Chuo, Yamanashi, 409-3898, Japan
| |
Collapse
|
110
|
Insights into the epigenetic mechanisms involving histone lysine methylation and demethylation in ischemia induced damage and repair has therapeutic implication. Biochim Biophys Acta Mol Basis Dis 2017; 1863:152-164. [DOI: 10.1016/j.bbadis.2016.09.014] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2016] [Revised: 09/15/2016] [Accepted: 09/20/2016] [Indexed: 01/01/2023]
|
111
|
Barteczek P, Li L, Ernst AS, Böhler LI, Marti HH, Kunze R. Neuronal HIF-1α and HIF-2α deficiency improves neuronal survival and sensorimotor function in the early acute phase after ischemic stroke. J Cereb Blood Flow Metab 2017; 37:291-306. [PMID: 26746864 PMCID: PMC5363746 DOI: 10.1177/0271678x15624933] [Citation(s) in RCA: 93] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2015] [Revised: 10/30/2015] [Accepted: 12/07/2015] [Indexed: 01/01/2023]
Abstract
Hypoxia-inducible factors mediate adaptive responses to ischemia, among others, by induction of anti- and pro-survival genes. Thus, the impact of HIF on neuronal survival upon stroke is controversial. Therefore, neuron-specific knockout mice deficient for Hif1a and Hif2a were exposed to inspiratory hypoxia or ischemia-reperfusion injury. Both Hif1a- and Hif2a-deficient mice showed no altered infarct and edema size, suggesting that both HIF-α subunits might compensate for each other. Accordingly, hypoxic HIF-target gene regulation was marginally affected with exception of anti-survival Bnip3 and pro-survival erythropoietin. In the early acute stage upon stroke, Hif1a/Hif2a double knockout mice exhibited significantly reduced expression of the anti-survival Bnip3, Bnip3L, and Pmaip1 Accordingly, global cell death and edema were significantly reduced upon 24 h but not 72 h reperfusion. Behavioral assessment indicated that Hif1a/Hif2a-deficient mice initially performed better, but became significantly more impaired after 72 h accompanied by increased apoptosis and reduced angiogenesis. Our findings suggest that in neurons HIF-1 and HIF-2 have redundant functions for cellular survival under ischemic conditions. By contrast, lack of anti-survival factors in Hif1a/Hif2a-deficient mice might protect from early acute neuronal cell death and neurological impairment, indicating a benefit of HIF-pathway inhibition in neurons in the very acute phase after ischemic stroke.
Collapse
Affiliation(s)
- Philipp Barteczek
- Institute of Physiology and Pathophysiology, University of Heidelberg, Heidelberg, Germany
| | - Lexiao Li
- Institute of Physiology and Pathophysiology, University of Heidelberg, Heidelberg, Germany
| | - Anne-Sophie Ernst
- Institute of Physiology and Pathophysiology, University of Heidelberg, Heidelberg, Germany
| | - Laura-Inés Böhler
- Institute of Physiology and Pathophysiology, University of Heidelberg, Heidelberg, Germany
| | - Hugo H Marti
- Institute of Physiology and Pathophysiology, University of Heidelberg, Heidelberg, Germany
| | - Reiner Kunze
- Institute of Physiology and Pathophysiology, University of Heidelberg, Heidelberg, Germany
| |
Collapse
|
112
|
Xu K, Ye L, Sharma K, Jin Y, Harrison MM, Caldwell T, Berthiaume JM, Luo Y, LaManna JC, Puchowicz MA. Diet-Induced Ketosis Protects Against Focal Cerebral Ischemia in Mouse. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 977:205-213. [PMID: 28685447 DOI: 10.1007/978-3-319-55231-6_28] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Over the past decade we have consistently shown that ketosis is neuroprotective against ischemic insults in rats. We reported that diet-induced ketotic rats had a significant reduction in infarct volume when subjected to middle cerebral artery occlusion (MCAO), and improved survival and recovery after cardiac arrest and resuscitation. The neuroprotective mechanisms of ketosis (via ketogenic diet; KG) include (i) ketones are alternate energy substrates that can restore energy balance when glucose metabolism is deficient and (ii) ketones modulate cell-signalling pathways that are cytoprotective. We investigated the effects of diet-induced ketosis following transient focal cerebral ischemia in mice. The correlation between levels of ketosis and hypoxic inducible factor-1alpha (HIF-1α), AKT (also known as protein kinase B or PKB) and 5' AMP-activated protein kinase (AMPK) were determined. Mice were fed with KG diet or standard lab-chow (STD) diet for 4 weeks. For the MCAO group, mice underwent 60 min of MCAO and total brain infarct volumes were evaluated 48 h after reperfusion. In a separate group of mice, brain tissue metabolites, levels of HIF-1α, phosphorylated AKT (pAKT), and AMPK were measured. After feeding a KG diet, levels of blood ketone bodies (beta-hydroxyburyrate, BHB) were increased. There was a proportional decrease in infarct volumes with increased blood BHB levels (KG vs STD; 4.2 ± 0.6 vs 7.8 ± 2.2 mm3, mean ± SEM). A positive correlation was also observed with HIF-1α and pAKT relative to blood BHB levels. Our results showed that chronic ketosis can be induced in mice by KG diet and was neuroprotective against focal cerebral ischemia in a concentration dependent manner. Potential mechanisms include upregulation of cytoprotective pathways such as those associated with HIF-1α, pAKT and AMPK.
Collapse
Affiliation(s)
- Kui Xu
- Departments of Physiology and Biophysics, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | - Lena Ye
- Departments of Physiology and Biophysics, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | - Katyayini Sharma
- Departments of Physiology and Biophysics, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | - Yongming Jin
- Neurosugery and Nutrition, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | - Matthew M Harrison
- Departments of Physiology and Biophysics, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | - Tylor Caldwell
- Departments of Physiology and Biophysics, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | - Jessica M Berthiaume
- Departments of Physiology and Biophysics, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | - Yu Luo
- Neurosugery and Nutrition, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | - Joseph C LaManna
- Departments of Physiology and Biophysics, Case Western Reserve University, School of Medicine, Cleveland, OH, USA
| | - Michelle A Puchowicz
- Nutrition, Case Western Reserve University, School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
113
|
Hsieh CH, Lin YJ, Chen WL, Huang YC, Chang CW, Cheng FC, Liu RS, Shyu WC. HIF-1α triggers long-lasting glutamate excitotoxicity via system x c- in cerebral ischaemia-reperfusion. J Pathol 2016; 241:337-349. [PMID: 27801527 DOI: 10.1002/path.4838] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2016] [Revised: 09/26/2016] [Accepted: 10/17/2016] [Indexed: 11/11/2022]
Abstract
Hypoxia-inducible factor 1α (HIF-1α) controls many genes involved in physiological and pathological processes. However, its roles in glutamatergic transmission and excitotoxicity are unclear. Here, we proposed that HIF-1α might contribute to glutamate-mediated excitotoxicity during cerebral ischaemia-reperfusion (CIR) and investigated its molecular mechanism. We showed that an HIF-1α conditional knockout mouse displayed an inhibition in CIR-induced elevation of extracellular glutamate and N-methyl-d-aspartate receptor (NMDAR) activation. By gene screening for glutamate transporters in cortical cells, we found that HIF-1α mainly regulates the cystine-glutamate transporter (system xc- ) subunit xCT by directly binding to its promoter; xCT and its function are up-regulated in the ischaemic brains of rodents and humans, and the effects lasted for several days. Genetic deletion of xCT in cortical cells of mice inhibits either oxygen glucose deprivation/reoxygenation (OGDR) or CIR-mediated glutamate excitotoxicity in vitro and in vivo. Pharmaceutical inhibition of system xc- by a clinically approved anti-cancer drug, sorafenib, improves infarct volume and functional outcome in rodents with CIR and its therapeutic window is at least 3 days. Taken together, these findings reveal that HIF-1α plays a role in CIR-induced glutamate excitotoxicity via the long-lasting activation of system xc- -dependent glutamate outflow and suggest that system xc- is a promising therapeutic target with an extended therapeutic window in stroke. Copyright © 2016 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Chia-Hung Hsieh
- Graduate Institute of Basic Medical Science, China Medical University, No 91, Hsueh-Shih Road, Taichung, Taiwan, 40402.,Department of Medical Research, China Medical University Hospital, No 2, Yuh-Der Road, Taichung, Taiwan, 40402.,Aging Medicine Program, China Medical University, No 91, Hsueh-Shih Road, Taichung, Taiwan, 40402.,Department of Biomedical Informatics, Asia University, No 500, Lioufeng Road, Taichung, Taiwan, 41354
| | - Yu-Jung Lin
- Graduate Institute of Basic Medical Science, China Medical University, No 91, Hsueh-Shih Road, Taichung, Taiwan, 40402
| | - Wei-Ling Chen
- Aging Medicine Program, China Medical University, No 91, Hsueh-Shih Road, Taichung, Taiwan, 40402
| | - Yen-Chih Huang
- Graduate Institute of Immunology, China Medical University, No 91, Hsueh-Shih Road, Taichung, Taiwan, 40402
| | - Chi-Wei Chang
- National PET/Cyclotron Center and Department of Nuclear Medicine, Taipei Veterans General Hospital, No 201, Shipai Road, Taipei, Taiwan, 11217
| | - Fu-Chou Cheng
- Stem Cell Center, Department of Medical Research, Taichung Veterans General Hospital, 1650 Taiwan Boulevard Section 4, Taichung, Taiwan, 40705
| | - Ren-Shyan Liu
- National PET/Cyclotron Center and Department of Nuclear Medicine, Taipei Veterans General Hospital, No 201, Shipai Road, Taipei, Taiwan, 11217
| | - Woei-Cherng Shyu
- Department of Neurology, Center for Neuropsychiatry, China Medical University and Hospital, No 91, Hsueh-Shih Road, Taichung, Taiwan, 40402
| |
Collapse
|
114
|
E2-25K SUMOylation inhibits proteasome for cell death during cerebral ischemia/reperfusion. Cell Death Dis 2016; 7:e2573. [PMID: 28032866 PMCID: PMC5261013 DOI: 10.1038/cddis.2016.428] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Revised: 11/14/2016] [Accepted: 11/15/2016] [Indexed: 01/12/2023]
Abstract
Cerebral ischemia/reperfusion (I/R) causes brain damage accompanied by ubiquitin accumulation and impairment of proteasome activity. In this study, we report that E2-25K, an E2-conjugating enzyme, is SUMOylated during oxidative stress and regulates cerebral I/R-induced damage. Knockdown of E2-25K expression protects against oxygen/glucose deprivation and reoxygenation (OGD/R)-induced neuronal cell death, whereas ectopic expression of E2-25K stimulates it. Compared with the control mice, cerebral infarction lesions and behavioral/neurological disorders are ameliorated in E2-25K knockout mice during middle cerebral artery occlusion and reperfusion. In particular, E2-25K is SUMOylated at Lys14 under oxidative stress, OGD/R and I/R to prompt cell death. Further, E2-25K downregulates the proteasome subunit S5a to impair proteasome complex and thus restrain proteasome activity under oxidative stress. This proteasome inhibitory activity of E2-25K is dependent on its SUMOylation. These results suggest that E2-25K has a crucial role in oxidative stress and cerebral I/R-induced damage through inhibiting proteasome via its SUMOylation.
Collapse
|
115
|
Karuppagounder SS, Alim I, Khim SJ, Bourassa MW, Sleiman SF, John R, Thinnes CC, Yeh TL, Demetriades M, Neitemeier S, Cruz D, Gazaryan I, Killilea DW, Morgenstern L, Xi G, Keep RF, Schallert T, Tappero RV, Zhong J, Cho S, Maxfield FR, Holman TR, Culmsee C, Fong GH, Su Y, Ming GL, Song H, Cave JW, Schofield CJ, Colbourne F, Coppola G, Ratan RR. Therapeutic targeting of oxygen-sensing prolyl hydroxylases abrogates ATF4-dependent neuronal death and improves outcomes after brain hemorrhage in several rodent models. Sci Transl Med 2016; 8:328ra29. [PMID: 26936506 DOI: 10.1126/scitranslmed.aac6008] [Citation(s) in RCA: 110] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Disability or death due to intracerebral hemorrhage (ICH) is attributed to blood lysis, liberation of iron, and consequent oxidative stress. Iron chelators bind to free iron and prevent neuronal death induced by oxidative stress and disability due to ICH, but the mechanisms for this effect remain unclear. We show that the hypoxia-inducible factor prolyl hydroxylase domain (HIF-PHD) family of iron-dependent, oxygen-sensing enzymes are effectors of iron chelation. Molecular reduction of the three HIF-PHD enzyme isoforms in the mouse striatum improved functional recovery after ICH. A low-molecular-weight hydroxyquinoline inhibitor of the HIF-PHD enzymes, adaptaquin, reduced neuronal death and behavioral deficits after ICH in several rodent models without affecting total iron or zinc distribution in the brain. Unexpectedly, protection from oxidative death in vitro or from ICH in vivo by adaptaquin was associated with suppression of activity of the prodeath factor ATF4 rather than activation of an HIF-dependent prosurvival pathway. Together, these findings demonstrate that brain-specific inactivation of the HIF-PHD metalloenzymes with the blood-brain barrier-permeable inhibitor adaptaquin can improve functional outcomes after ICH in several rodent models.
Collapse
Affiliation(s)
- Saravanan S Karuppagounder
- Sperling Center for Hemorrhagic Stroke Recovery, Burke Medical Research Institute, White Plains, NY 10605, USA. Feil Family Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Ishraq Alim
- Sperling Center for Hemorrhagic Stroke Recovery, Burke Medical Research Institute, White Plains, NY 10605, USA. Feil Family Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Soah J Khim
- Sperling Center for Hemorrhagic Stroke Recovery, Burke Medical Research Institute, White Plains, NY 10605, USA. Feil Family Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Megan W Bourassa
- Sperling Center for Hemorrhagic Stroke Recovery, Burke Medical Research Institute, White Plains, NY 10605, USA. Feil Family Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Sama F Sleiman
- Sperling Center for Hemorrhagic Stroke Recovery, Burke Medical Research Institute, White Plains, NY 10605, USA. Feil Family Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Roseleen John
- Department of Psychology, University of Alberta, Edmonton, Alberta T6G 2E9, Canada
| | | | - Tzu-Lan Yeh
- Department of Chemistry, University of Oxford, OX1 3TA Oxford, UK
| | | | - Sandra Neitemeier
- Institut fuer Pharmakologie and Klinische Pharmazie, Phillips-Universitaet Marburg, D 35032 Marburg, Germany
| | - Dana Cruz
- Department of Biochemistry, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Irina Gazaryan
- Sperling Center for Hemorrhagic Stroke Recovery, Burke Medical Research Institute, White Plains, NY 10605, USA. Feil Family Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY 10065, USA
| | | | - Lewis Morgenstern
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Guohua Xi
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Richard F Keep
- Department of Neurosurgery, University of Michigan, Ann Arbor, MI 48109, USA
| | - Timothy Schallert
- Department of Psychology, University of Texas at Austin, Austin, TX 78712, USA
| | - Ryan V Tappero
- Photon Sciences Directorate, Brookhaven National Laboratory, Upton, NY 11973, USA
| | - Jian Zhong
- Sperling Center for Hemorrhagic Stroke Recovery, Burke Medical Research Institute, White Plains, NY 10605, USA. Feil Family Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Sunghee Cho
- Sperling Center for Hemorrhagic Stroke Recovery, Burke Medical Research Institute, White Plains, NY 10605, USA. Feil Family Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Frederick R Maxfield
- Department of Biochemistry, Weill Medical College of Cornell University, New York, NY 10065, USA
| | - Theodore R Holman
- Chemistry and Biochemistry, Department, University of California at Santa Cruz, Santa Cruz, CA 95064, USA
| | - Carsten Culmsee
- Institut fuer Pharmakologie and Klinische Pharmazie, Phillips-Universitaet Marburg, D 35032 Marburg, Germany
| | - Guo-Hua Fong
- Center for Vascular Biology, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Yijing Su
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Guo-li Ming
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Hongjun Song
- Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - John W Cave
- Sperling Center for Hemorrhagic Stroke Recovery, Burke Medical Research Institute, White Plains, NY 10605, USA. Feil Family Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY 10065, USA
| | | | - Frederick Colbourne
- Department of Psychology, University of Alberta, Edmonton, Alberta T6G 2E9, Canada
| | - Giovanni Coppola
- Department of Psychiatry, University of California at Los Angeles, CA 90095, USA
| | - Rajiv R Ratan
- Sperling Center for Hemorrhagic Stroke Recovery, Burke Medical Research Institute, White Plains, NY 10605, USA. Feil Family Brain and Mind Research Institute, Weill Medical College of Cornell University, New York, NY 10065, USA.
| |
Collapse
|
116
|
Chan YL, Saad S, Al-Odat I, Zaky AA, Oliver B, Pollock C, Li W, Jones NM, Chen H. Impact of maternal cigarette smoke exposure on brain and kidney health outcomes in female offspring. Clin Exp Pharmacol Physiol 2016; 43:1168-1176. [DOI: 10.1111/1440-1681.12659] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Revised: 06/17/2016] [Accepted: 07/21/2016] [Indexed: 12/11/2022]
Affiliation(s)
- Yik L Chan
- School of Life Sciences; Faculty of Science; University of Technology Sydney; Broadway NSW Australia
| | - Sonia Saad
- Kolling Institute of Medical Research; University of Sydney; St Leonards NSW Australia
| | - Ibrahim Al-Odat
- School of Life Sciences; Faculty of Science; University of Technology Sydney; Broadway NSW Australia
| | - Amgad A Zaky
- Kolling Institute of Medical Research; University of Sydney; St Leonards NSW Australia
| | - Brian Oliver
- School of Life Sciences; Faculty of Science; University of Technology Sydney; Broadway NSW Australia
| | - Carol Pollock
- Kolling Institute of Medical Research; University of Sydney; St Leonards NSW Australia
| | - Weihong Li
- Department of Science and Technology; Chengdu University of Traditional Chinese Medicine; Chengdu Sichuan China
| | - Nicole M Jones
- Department of Pharmacology; School of Medical Sciences; University of New South Wales; Broadway NSW Australia
| | - Hui Chen
- School of Life Sciences; Faculty of Science; University of Technology Sydney; Broadway NSW Australia
| |
Collapse
|
117
|
Zhang Z, Yan J, Shi H. Role of Hypoxia Inducible Factor 1 in Hyperglycemia-Exacerbated Blood-Brain Barrier Disruption in Ischemic Stroke. Neurobiol Dis 2016. [PMID: 27425889 DOI: 10.1016/j.nbd.2016.07.01] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2023] Open
Abstract
Diabetes is a major stroke risk factor and is associated with poor functional recovery after stroke. Accumulating evidence indicates that the worsened outcomes may be due to hyperglycemia-induced cerebral vascular complications, especially disruption of the blood-brain barrier (BBB). The present study tested a hypothesis that the activation of hypoxia inducible factor-1 (HIF-1) was involved in hyperglycemia-aggravated BBB disruption in an ischemic stroke model. Non-diabetic control and Streptozotocin-induced type I diabetic mice were subjected to 90min transient middle cerebral artery occlusion (MCAO) followed by reperfusion. Our results demonstrated that hyperglycemia induced higher expression of HIF-1α and vascular endothelial growth factor (VEGF) in brain microvessels after MCAO/reperfusion. Diabetic mice showed exacerbated BBB damage and tight junction disruption, increased infarct volume as well as worsened neurological deficits. Furthermore, suppressing HIF-1 activity by specific knock-out endothelial HIF-1α ameliorated BBB leakage and brain infarction in diabetic animals. Moreover, glycemic control by insulin abolished HIF-1α up-regulation in diabetic animals and reduced BBB permeability and brain infarction. These findings strongly indicate that HIF-1 plays an important role in hyperglycemia-induced exacerbation of BBB disruption in ischemic stroke. Endothelial HIF-1 inhibition warrants further investigation as a therapeutic target for the treatment of stroke patients with diabetes.
Collapse
Affiliation(s)
- Ziyan Zhang
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS 66045, United States
| | - Jingqi Yan
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS 66045, United States
| | - Honglian Shi
- Department of Pharmacology and Toxicology, University of Kansas, Lawrence, KS 66045, United States.
| |
Collapse
|
118
|
Roles of HIF-1α, VEGF, and NF-κB in Ischemic Preconditioning-Mediated Neuroprotection of Hippocampal CA1 Pyramidal Neurons Against a Subsequent Transient Cerebral Ischemia. Mol Neurobiol 2016; 54:6984-6998. [DOI: 10.1007/s12035-016-0219-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 10/12/2016] [Indexed: 10/20/2022]
|
119
|
Role of Hypoxia Inducible Factor 1 in Hyperglycemia-Exacerbated Blood-Brain Barrier Disruption in Ischemic Stroke. Neurobiol Dis 2016; 95:82-92. [PMID: 27425889 DOI: 10.1016/j.nbd.2016.07.012] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 06/16/2016] [Accepted: 07/13/2016] [Indexed: 12/18/2022] Open
Abstract
Diabetes is a major stroke risk factor and is associated with poor functional recovery after stroke. Accumulating evidence indicates that the worsened outcomes may be due to hyperglycemia-induced cerebral vascular complications, especially disruption of the blood-brain barrier (BBB). The present study tested a hypothesis that the activation of hypoxia inducible factor-1 (HIF-1) was involved in hyperglycemia-aggravated BBB disruption in an ischemic stroke model. Non-diabetic control and Streptozotocin-induced type I diabetic mice were subjected to 90min transient middle cerebral artery occlusion (MCAO) followed by reperfusion. Our results demonstrated that hyperglycemia induced higher expression of HIF-1α and vascular endothelial growth factor (VEGF) in brain microvessels after MCAO/reperfusion. Diabetic mice showed exacerbated BBB damage and tight junction disruption, increased infarct volume as well as worsened neurological deficits. Furthermore, suppressing HIF-1 activity by specific knock-out endothelial HIF-1α ameliorated BBB leakage and brain infarction in diabetic animals. Moreover, glycemic control by insulin abolished HIF-1α up-regulation in diabetic animals and reduced BBB permeability and brain infarction. These findings strongly indicate that HIF-1 plays an important role in hyperglycemia-induced exacerbation of BBB disruption in ischemic stroke. Endothelial HIF-1 inhibition warrants further investigation as a therapeutic target for the treatment of stroke patients with diabetes.
Collapse
|
120
|
Chang YK, Choi H, Jeong JY, Na KR, Lee KW, Lim BJ, Choi DE. Dapagliflozin, SGLT2 Inhibitor, Attenuates Renal Ischemia-Reperfusion Injury. PLoS One 2016; 11:e0158810. [PMID: 27391020 PMCID: PMC4938401 DOI: 10.1371/journal.pone.0158810] [Citation(s) in RCA: 112] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2016] [Accepted: 06/22/2016] [Indexed: 01/24/2023] Open
Abstract
Dapagliflozin, a new type of drug used to treat diabetes mellitus (DM), is a sodium/glucose cotransporter 2 (SGLT2) inhibitor. Although some studies showed that SGLT2 inhibition attenuated reactive oxygen generation in diabetic kidney the role of SGLT2 inhibition is unknown. We evaluated whether SLT2 inhibition has renoprotective effects in ischemia-reperfusion (IR) models. We evaluated whether dapagliflozin reduces renal damage in IR mice model. In addition, hypoxic HK2 cells were treated with or without SGLT2 inhibitor to investigate cell survival, the apoptosis signal pathway, and the induction of hypoxia-inducible factor 1 (HIF1) and associated proteins. Dapagliflozin improved renal function. Dapagliflozin reduced renal expression of Bax, renal tubule injury and TUNEL-positive cells and increased renal expression of HIF1 in IR-injured mice. HIF1 inhibition by albendazole negated the renoprotective effects of dapagliflozin treatment in IR-injured mice. In vitro, dapagliflozin increased the expression of HIF1, AMP-activated protein kinase (AMPK), and ERK and increased cell survival of hypoxic HK2 cells in a dose-dependent manner. In conclusion, dapagliflozin attenuates renal IR injury. HIF1 induction by dapagliflozin may play a role in renoprotection against renal IR injury.
Collapse
Affiliation(s)
- Yoon-Kyung Chang
- Department of Nephrology, Daejeon St. Mary Hospital, Daejeon, South Korea
- Department of Nephrology, Catholic University of Korea, Seoul, South Korea
| | - Hyunsu Choi
- Clinical Research Institute, Daejeon St. Mary Hospital, Daejeon, South Korea
| | - Jin Young Jeong
- Department of Nephrology, School of medicine, Chungnam National University, Daejeon, South Korea
- Department of Medical Science, School of medicine, Chungnam National University, Daejeon, South Korea
| | - Ki-Ryang Na
- Department of Nephrology, School of medicine, Chungnam National University, Daejeon, South Korea
| | - Kang Wook Lee
- Department of Nephrology, School of medicine, Chungnam National University, Daejeon, South Korea
| | - Beom Jin Lim
- Department of pathology, College of medicine, Yeonse University, Seoul, South Korea
- * E-mail: (DEC); (BJL)
| | - Dae Eun Choi
- Department of Nephrology, School of medicine, Chungnam National University, Daejeon, South Korea
- * E-mail: (DEC); (BJL)
| |
Collapse
|
121
|
Kopach O, Maistrenko A, Lushnikova I, Belan P, Skibo G, Voitenko N. HIF-1α-mediated upregulation of SERCA2b: The endogenous mechanism for alleviating the ischemia-induced intracellular Ca2+ store dysfunction in CA1 and CA3 hippocampal neurons. Cell Calcium 2016; 59:251-61. [DOI: 10.1016/j.ceca.2016.02.014] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2015] [Revised: 02/11/2016] [Accepted: 02/22/2016] [Indexed: 01/27/2023]
|
122
|
Huang X, Yang K, Zhang Y, Wang Q, Li Y. Quinolinic acid induces cell apoptosis in PC12 cells through HIF-1-dependent RTP801 activation. Metab Brain Dis 2016; 31:435-44. [PMID: 26738727 DOI: 10.1007/s11011-015-9782-x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2015] [Accepted: 12/22/2015] [Indexed: 02/01/2023]
Abstract
Neurological disease comprises a series of disorders featuring brain dysfunction and neuronal cell death. Among the factors contributing to neuronal death, excitotoxicity induced by excitatory amino acids, such as glutamate, plays a critical role. However, the mechanisms about how the excitatory amino acids induce neuronal death remain elucidated. In this study, we investigated the role of HIF-1α (hypoxia inducible factor-1α) and RTP801 in cell apoptosis induced by quinolinic acid (QUIN), a glutamatergic agonist, in PC12 cells. We found that QUIN at 5 μM increased the expression of HIF-1α significantly with a peak at 24 h. After the treatment with QUIN (5-20 μM) for 24 h, the cells exhibited decreased viability and cell apoptosis with a concomitant increased expression of apoptosis related proteins. QUIN treatment also induced the generation of intracellular reactive oxygen species and RTP801 up-regulation in a HIF-1α-dependent manner that were inhibited by 2-methoxyestradiol, a HIF-1α inhibitor. Importantly, HIF-1 or RTP801 invalidation by siRNA rescued the cell apoptosis induced by QUIN or cobalt chloride, a chemical inducer of HIF-1. Taken together, these findings support the concept that neurotoxicity induced by QUIN is associated with HIF-1-dependent RTP801 activation and provide insight into the potential of RTP801 inhibitor in treatment of neurological disorders.
Collapse
Affiliation(s)
- Xiaojia Huang
- Department of Pharmacology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China.
| | - Kaiyong Yang
- Department of Pharmacology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Yi Zhang
- Department of Pharmacology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Qiang Wang
- Department of Preventive Medicine, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| | - Yongjin Li
- Department of Pharmacology, School of Medicine, Jiangsu University, Zhenjiang, Jiangsu, 212013, China
| |
Collapse
|
123
|
Heyman SN, Leibowitz D, Mor-Yosef Levi I, Liberman A, Eisenkraft A, Alcalai R, Khamaisi M, Rosenberger C. Adaptive response to hypoxia and remote ischaemia pre-conditioning: a new hypoxia-inducible factors era in clinical medicine. Acta Physiol (Oxf) 2016; 216:395-406. [PMID: 26449466 DOI: 10.1111/apha.12613] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Revised: 07/01/2015] [Accepted: 10/02/2015] [Indexed: 01/30/2023]
Abstract
Transient ischaemia leads to tolerance to subsequent protracted ischaemia. This 'ischaemia pre-conditioning' results from the induction of numerous protective genes, involved in cell metabolism, proliferation and survival, in antioxidant capacity, angiogenesis, vascular tone and erythropoiesis. Hypoxia-inducible factors (HIF) play a pivotal role in this transcriptional adaptive response. HIF prolyl hydroxylases (PHDs), serving as oxygen sensors, control HIFα degradation. HIF-mediated ischaemic pre-conditioning can be achieved with the administration of PHD inhibitors, with the attenuation of organ injury under various hypoxic and toxic insults. Clinical trials are currently under way, evaluating PHD inhibitors as inducers of erythropoietin. Once their safety is established, their potential use might be further tested in clinical trials in various forms of acute ischaemic and toxic organ damage. Repeated transient limb ischaemia was also found to attenuate ischaemic injury in remote organs. This 'remote ischaemic pre-conditioning' phenomenon (RIP) has been extensively studied recently in small clinical trials, preceding, or in parallel with an abrupt insult, such as myocardial infarction, cardiac surgery or radiocontrast administration. Initial results are promising, suggesting organ protection. Large-scale multi-centre studies are currently under way, evaluating the protective potential of RIP in cardiac surgery, in the management of myocardial infarction and in organ transplantation. The mechanisms of organ protection provided by RIP are poorly understood, but HIF seemingly play a role as well. Thus, Inhibition of HIF degradation with PHD inhibitors, as well as RIP (in part through HIF), might develop into novel clinical interventions in organ protection in the near future.
Collapse
Affiliation(s)
- S. N. Heyman
- Department of Medicine; Hadassah Hebrew University Hospitals; Jerusalem Israel
| | - D. Leibowitz
- Department of Medicine; Hadassah Hebrew University Hospitals; Jerusalem Israel
- Department of Cardiology; Hadassah Hebrew University Hospitals; Jerusalem Israel
| | - I. Mor-Yosef Levi
- Department of Nephrology; Hadassah Hebrew University Hospitals; Jerusalem Israel
| | - A. Liberman
- Department of Neurology; Hadassah Hebrew University Hospitals; Jerusalem Israel
| | - A. Eisenkraft
- The Research Institute for Military Medicine; The Hebrew University Medical School and the Israeli Defense Force Medical Corps; Jerusalem Israel
| | - R. Alcalai
- Department of Medicine; Hadassah Hebrew University Hospitals; Jerusalem Israel
- Department of Cardiology; Hadassah Hebrew University Hospitals; Jerusalem Israel
| | | | | |
Collapse
|
124
|
Neuronal deficiency of HIF prolyl 4-hydroxylase 2 in mice improves ischemic stroke recovery in an HIF dependent manner. Neurobiol Dis 2016; 91:221-35. [PMID: 27001147 DOI: 10.1016/j.nbd.2016.03.018] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2015] [Revised: 03/09/2016] [Accepted: 03/17/2016] [Indexed: 12/30/2022] Open
Abstract
Hypoxia inducible factors (HIFs) mediate the endogenous adaptive responses to hypoxia. HIF prolyl 4-hydroxylase domain proteins (PHD) are important suppressors of the HIF pathway. Recently, we demonstrated that neuron-specific deletion of Phd2 reduces cerebral tissue damage in the very acute phase of ischemic stroke. In the present study, we investigated whether neuronal Phd2 ablation is likewise beneficial for stroke recovery, and aimed to identify underlying cellular mechanisms. Mice underwent permanent occlusion of the distal middle cerebral artery (pdMCAO) for either 7days (sub-acute stage) or 30days (chronic stage). One week after pdMCAO the infarct size of Phd2-deficient mice was significantly reduced as compared to wild-type (WT) mice. Accordingly, Phd2-deficient animals showed less impaired sensorimotor function. Neuronal loss of Phd2 upregulated vascular endothelial growth factor (VEGF) and significantly increased microvascular density along the infarct border in the sub-acute stage of stroke. Phd2-deficient mice showed reduced expression of pro-inflammatory cytokines and increased numbers of resting microglia/macrophages and reactive astrocytes within peri-infarct regions in comparison to WT littermates. Finally, brain tissue protection and increased angiogenesis upon sub-acute ischemic stroke was completely absent in Phd2 knockout mice that were additionally deficient for both Hif1a and Hif2a. Our findings suggest that lack of PHD2 in neurons improves histological and functional long-term outcome from ischemic stroke at least partly by amplifying endogenous adaptive neovascularization through activation of the HIF-VEGF axis.
Collapse
|
125
|
Rosafio K, Castillo X, Hirt L, Pellerin L. Cell-specific modulation of monocarboxylate transporter expression contributes to the metabolic reprograming taking place following cerebral ischemia. Neuroscience 2016; 317:108-20. [DOI: 10.1016/j.neuroscience.2015.12.052] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2015] [Revised: 12/17/2015] [Accepted: 12/29/2015] [Indexed: 01/23/2023]
|
126
|
Mordel J, Sheikh A, Tsohataridis S, Kanold PO, Zehendner CM, Luhmann HJ. Mild systemic inflammation and moderate hypoxia transiently alter neuronal excitability in mouse somatosensory cortex. Neurobiol Dis 2016; 88:29-43. [PMID: 26763603 DOI: 10.1016/j.nbd.2015.12.019] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2015] [Revised: 12/11/2015] [Accepted: 12/29/2015] [Indexed: 11/20/2022] Open
Abstract
During the perinatal period, the brain is highly vulnerable to hypoxia and inflammation, which often cause white matter injury and long-term neuronal dysfunction such as motor and cognitive deficits or epileptic seizures. We studied the effects of moderate hypoxia (HYPO), mild systemic inflammation (INFL), or the combination of both (HYPO+INFL) in mouse somatosensory cortex induced during the first postnatal week on network activity and compared it to activity in SHAM control animals. By performing in vitro electrophysiological recordings with multi-electrode arrays from slices prepared directly after injury (P8-10), one week after injury (P13-16), or in young adults (P28-30), we investigated how the neocortical network developed following these insults. No significant difference was observed between the four groups in an extracellular solution close to physiological conditions. In extracellular 8mM potassium solution, slices from the HYPO, INFL, and HYPO+INFL group were more excitable than SHAM at P8-10 and P13-16. In these two age groups, the number and frequency of spontaneous epileptiform events were significantly increased compared to SHAM. The frequency of epileptiform events was significantly reduced by the NMDA antagonist D-APV in HYPO, INFL, and HYPO+INFL, but not in SHAM, indicating a contribution of NMDA receptors to this pathophysiological activity. In addition, the AMPA/kainate receptor antagonist CNQX suppressed the remaining epileptiform activity. Electrical stimulation evoked prominent epileptiform activity in slices from HYPO, INFL and HYPO+INFL animals. Stimulation threshold to elicit epileptiform events was lower in these groups than in SHAM. Evoked events spread over larger areas and lasted longer in treated animals than in SHAM. In addition, the evoked epileptiform activity was reduced in the older (P28-30) group indicating that cortical dysfunction induced by hypoxia and inflammation was transient and compensated during early development.
Collapse
Affiliation(s)
- Jérôme Mordel
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University, 55128 Mainz, Germany
| | - Aminah Sheikh
- Department of Biology, University of Maryland, College Park, MD 20742, USA
| | - Simeon Tsohataridis
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University, 55128 Mainz, Germany
| | - Patrick O Kanold
- Department of Biology, University of Maryland, College Park, MD 20742, USA
| | - Christoph M Zehendner
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University, 55128 Mainz, Germany; ZIM III, Department of Cardiology, Institute for Cardiovascular Regeneration, Goethe University Frankfurt, 60590 Frankfurt am Main, Germany
| | - Heiko J Luhmann
- Institute of Physiology, University Medical Center of the Johannes Gutenberg University, 55128 Mainz, Germany.
| |
Collapse
|
127
|
Wu K, Zhou K, Wang Y, Zhou Y, Tian N, Wu Y, Chen D, Zhang D, Wang X, Xu H, Zhang X. Stabilization of HIF-1α by FG-4592 promotes functional recovery and neural protection in experimental spinal cord injury. Brain Res 2015; 1632:19-26. [PMID: 26707978 DOI: 10.1016/j.brainres.2015.12.017] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2015] [Revised: 12/04/2015] [Accepted: 12/11/2015] [Indexed: 11/27/2022]
Abstract
Previous studies have shown that inhibition of prolyl hydroxylase(PHD) stabilizes Hypoxia-inducible factor 1, alpha subunit(HIF-1α), increases tolerance to hypoxia, and improves the prognosis of many diseases. However, the role of PHD inhibitor (PHDI) in the recovery of spinal cord injury remains controversial. In this study, we investigated the protective role of a novel PHDI FG-4592 both in vivo and in vitro. FG-4592 treatment stabilized HIF1α expression both in PC12 cells and in spinal cord. FG-4592 treatment significantly inhibited tert-Butyl hydroperoxide(TBHP)-induced apoptosis and increases the survival of neuronal PC-12 cells. FG-4592 administration also improved recovery and increased the survival of neurons in spinal cord lesions in the mice model. Combination therapy including the specific HIF-1α blocker YC-1 down-regulated the HIF-1α expression and partially abolished the protective effect of FG-4592. Taken together, our results revealed that the role of FG-4592 in SCI recovery is related to the stabilization of HIF-1α and inhibition of apoptosis. Overall, our study suggests that PHDIs may be feasible candidates for therapeutic intervention after SCI and central nervous system disorders in humans.
Collapse
Affiliation(s)
- Kai Wu
- Department of Orthopaedics, The Second affiliated hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key laboratory of orthopaedics, Wenzhou, Zhejiang Province, China
| | - Kailiang Zhou
- Department of Orthopaedics, The Second affiliated hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key laboratory of orthopaedics, Wenzhou, Zhejiang Province, China
| | - Yongli Wang
- Department of Orthopaedics, The Second affiliated hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key laboratory of orthopaedics, Wenzhou, Zhejiang Province, China
| | - Yifei Zhou
- Department of Orthopaedics, The Second affiliated hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key laboratory of orthopaedics, Wenzhou, Zhejiang Province, China
| | - Naifeng Tian
- Department of Orthopaedics, The Second affiliated hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key laboratory of orthopaedics, Wenzhou, Zhejiang Province, China
| | - Yaosen Wu
- Department of Orthopaedics, The Second affiliated hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key laboratory of orthopaedics, Wenzhou, Zhejiang Province, China
| | - Deheng Chen
- Department of Orthopaedics, The Second affiliated hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key laboratory of orthopaedics, Wenzhou, Zhejiang Province, China
| | - Di Zhang
- Department of Orthopaedics, The Second affiliated hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key laboratory of orthopaedics, Wenzhou, Zhejiang Province, China
| | - Xiangyang Wang
- Department of Orthopaedics, The Second affiliated hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key laboratory of orthopaedics, Wenzhou, Zhejiang Province, China
| | - Huazi Xu
- Department of Orthopaedics, The Second affiliated hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key laboratory of orthopaedics, Wenzhou, Zhejiang Province, China.
| | - Xiaolei Zhang
- Department of Orthopaedics, The Second affiliated hospital of Wenzhou Medical University, Wenzhou, Zhejiang Province, China; Zhejiang Provincial Key laboratory of orthopaedics, Wenzhou, Zhejiang Province, China.
| |
Collapse
|
128
|
Recent Advances in Developing Inhibitors for Hypoxia-Inducible Factor Prolyl Hydroxylases and Their Therapeutic Implications. Molecules 2015; 20:20551-68. [PMID: 26610437 PMCID: PMC6332328 DOI: 10.3390/molecules201119717] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2015] [Revised: 11/10/2015] [Accepted: 11/11/2015] [Indexed: 12/13/2022] Open
Abstract
Hypoxia-inducible factor (HIF) prolyl hydroxylases (PHDs) are members of the 2-oxoglutarate dependent non-heme iron dioxygenases. Due to their physiological roles in regulation of HIF-1α stability, many efforts have been focused on searching for selective PHD inhibitors to control HIF-1α levels for therapeutic applications. In this review, we first describe the structure of PHD2 as a molecular basis for structure-based drug design (SBDD) and various experimental methods developed for measuring PHD activity. We further discuss the current status of the development of PHD inhibitors enabled by combining SBDD approaches with high-throughput screening. Finally, we highlight the clinical implications of small molecule PHD inhibitors.
Collapse
|
129
|
Kapitsinou PP, Haase VH. Molecular mechanisms of ischemic preconditioning in the kidney. Am J Physiol Renal Physiol 2015; 309:F821-34. [DOI: 10.1152/ajprenal.00224.2015] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Accepted: 08/21/2015] [Indexed: 12/26/2022] Open
Abstract
More effective therapeutic strategies for the prevention and treatment of acute kidney injury (AKI) are needed to improve the high morbidity and mortality associated with this frequently encountered clinical condition. Ischemic and/or hypoxic preconditioning attenuates susceptibility to ischemic injury, which results from both oxygen and nutrient deprivation and accounts for most cases of AKI. While multiple signaling pathways have been implicated in renoprotection, this review will focus on oxygen-regulated cellular and molecular responses that enhance the kidney's tolerance to ischemia and promote renal repair. Central mediators of cellular adaptation to hypoxia are hypoxia-inducible factors (HIFs). HIFs play a crucial role in ischemic/hypoxic preconditioning through the reprogramming of cellular energy metabolism, and by coordinating adenosine and nitric oxide signaling with antiapoptotic, oxidative stress, and immune responses. The therapeutic potential of HIF activation for the treatment and prevention of ischemic injuries will be critically examined in this review.
Collapse
Affiliation(s)
- Pinelopi P. Kapitsinou
- Departments of Medicine, Anatomy and Cell Biology, and the Kidney Institute, University of Kansas Medical Center, Kansas City, Kansas
| | - Volker H. Haase
- Departments of Medicine, Cancer Biology, and Molecular Physiology and Biophysics, Vanderbilt University School of Medicine, Nashville, Tennessee; and
- Medicine and Research Services, Department of Veterans Affairs Hospital, Tennessee Valley Healthcare System, Nashville, Tennessee
| |
Collapse
|
130
|
Cho Y, Shin JE, Ewan EE, Oh YM, Pita-Thomas W, Cavalli V. Activating Injury-Responsive Genes with Hypoxia Enhances Axon Regeneration through Neuronal HIF-1α. Neuron 2015; 88:720-34. [PMID: 26526390 DOI: 10.1016/j.neuron.2015.09.050] [Citation(s) in RCA: 113] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2015] [Revised: 08/24/2015] [Accepted: 09/22/2015] [Indexed: 02/07/2023]
Abstract
Injured peripheral neurons successfully activate a proregenerative transcriptional program to enable axon regeneration and functional recovery. How transcriptional regulators coordinate the expression of such program remains unclear. Here we show that hypoxia-inducible factor 1α (HIF-1α) controls multiple injury-induced genes in sensory neurons and contribute to the preconditioning lesion effect. Knockdown of HIF-1α in vitro or conditional knock out in vivo impairs sensory axon regeneration. The HIF-1α target gene Vascular Endothelial Growth Factor A (VEGFA) is expressed in injured neurons and contributes to stimulate axon regeneration. Induction of HIF-1α using hypoxia enhances axon regeneration in vitro and in vivo in sensory neurons. Hypoxia also stimulates motor neuron regeneration and accelerates neuromuscular junction re-innervation. This study demonstrates that HIF-1α represents a critical transcriptional regulator in regenerating neurons and suggests hypoxia as a tool to stimulate axon regeneration.
Collapse
Affiliation(s)
- Yongcheol Cho
- Department of Anatomy and Neurobiology, School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Jung Eun Shin
- Department of Developmental Biology, School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Eric Edward Ewan
- Department of Anatomy and Neurobiology, School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Young Mi Oh
- Department of Anatomy and Neurobiology, School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Wolfgang Pita-Thomas
- Department of Anatomy and Neurobiology, School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Valeria Cavalli
- Department of Anatomy and Neurobiology, School of Medicine, Washington University in St. Louis, St. Louis, MO 63110, USA.
| |
Collapse
|
131
|
Li G, Zhao Y, Li Y, Lu J. Up-Regulation of Neuronal Nitric Oxide Synthase Expression by Cobalt Chloride Through a HIF-1α Mechanism in Neuroblastoma Cells. Neuromolecular Med 2015; 17:443-53. [PMID: 26458913 DOI: 10.1007/s12017-015-8373-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2015] [Accepted: 10/03/2015] [Indexed: 10/22/2022]
Abstract
Nitric oxide (NO) plays a dual role in response to neural hypoxia. NO is synthesized by three isoforms of nitric oxide synthase (NOS), among which the neuronal NOS (nNOS) is predominant in the nervous system. Hypoxia-inducible factor-1α (HIF-1α) is a transcription factor that is induced under hypoxic conditions, but its correlation with nNOS remains unclear. In the present study, we aimed at clarifying the regulation pattern of the nNOS expression in response to cobalt chloride (CoCl2), a widely used chemical mimic of hypoxia, and the role of HIF-1α in this process in neuroblastoma cells. We found CoCl2 evidently increased the nNOS expression and NO production in human neuroblastoma SK-N-SH cells, but the effect of CoCl2 on NO was partially abrogated by 7-nitroindazole, a selective inhibitor for nNOS. Importantly, we identified a hypoxia response element (HRE) within the nNOS promoter, to which HIF-1α may bind, and CoCl2 greatly enhanced the HIF-1α expression and its binding to the HRE. Meanwhile, we demonstrated that this HRE was functionally important for the activation of the nNOS transcription, and CoCl2 increased the transcriptional activity of the nNOS promoter through this HRE. Taken together, our study shows that CoCl2 may induce the nNOS expression and NO production through a HIF-1α mechanism in neuroblastoma cells, which may provide a potential target for the treatment of neurological hypoxic disorders caused by NO dysregulation.
Collapse
Affiliation(s)
- Guangyu Li
- Department of Neurosurgery, The First Affiliated Hospital, China Medical University, No. 155, Nanjing Street, Heping District, Shenyang, 110001, People's Republic of China.
| | - Yanyan Zhao
- Department of Medical Genetics, College of Basic Medical Science, China Medical University, No. 92, Bei Er Road, Heping District, Shenyang, 110001, People's Republic of China.
| | - Yinghui Li
- Department of Medical Genetics, College of Basic Medical Science, China Medical University, No. 92, Bei Er Road, Heping District, Shenyang, 110001, People's Republic of China
| | - Jingyu Lu
- Department of Medical Genetics, College of Basic Medical Science, China Medical University, No. 92, Bei Er Road, Heping District, Shenyang, 110001, People's Republic of China
| |
Collapse
|
132
|
Jiang G, Zhou R, He X, Shi Z, Huang M, Yu J, Wang X. Expression levels of microRNA-199 and hypoxia-inducible factor-1 alpha in brain tissue of patients with intractable epilepsy. Int J Neurosci 2015; 126:326-34. [PMID: 25539181 DOI: 10.3109/00207454.2014.994209] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
OBJECTIVES During the last decade, experimental evidence has demonstrated an important role of hypoxia, which leads to neuronal cell death and angiogenesis, in the mechanisms of seizure precipitation and recurrence. MicroRNA-199 targets hypoxia-inducible factor-1alpha (HIF-1α), which has recently been implicated in the pathophysiology of the hypoxic state and brain injury. However, little is known about the roles of MicroRNA-199 and HIF-1α in the human epileptogenic process. DESIGN AND METHODS In this study, we investigated the expression of miR-199a-5p, miR-199b-5p and HIF-1α using real-time PCR, immunohistochemistry and western blots in the temporal neocortex of twenty four patients with intractable epilepsy and twelve control subjects. RESULTS Compared with the control group, the expression of miR-199a-5p and miR-199b-5p was significantly lower in epileptic brain tissues (p < 0.05). The levels of HIF-1α mRNA and protein were highly up-regulated in epileptic brain tissues compared with those of control subjects (p < 0.05). CONCLUSION These data suggest that the abnormal expression of miR-199 and HIF-1α in epileptic brain tissue may be involved in the pathophysiology of human epilepsy and that the expression of HIF-1α may be regulated by miR-199. These findings may provide new insights into the treatment of epilepsy.
Collapse
Affiliation(s)
- Guohui Jiang
- a 1 Department of Neurology, Institute of Neurology, Affiliated Hospital of North Sichuan Medical College , Wen Hua Road, Nanchong 637000 , China
| | - Ruijiao Zhou
- a 1 Department of Neurology, Institute of Neurology, Affiliated Hospital of North Sichuan Medical College , Wen Hua Road, Nanchong 637000 , China
| | - Xuzhi He
- b 2 Department of Neurosurgery, Daping Hospital and Institute of Surgery Research, Third Military Medical University , Chongqing 400042 , China
| | - Zhiqing Shi
- c 3 Diagnosis Department of Hebei Medical University , Shijiazhuang 050000 , China
| | - Min Huang
- a 1 Department of Neurology, Institute of Neurology, Affiliated Hospital of North Sichuan Medical College , Wen Hua Road, Nanchong 637000 , China
| | - Juming Yu
- a 1 Department of Neurology, Institute of Neurology, Affiliated Hospital of North Sichuan Medical College , Wen Hua Road, Nanchong 637000 , China
| | - Xiaoming Wang
- a 1 Department of Neurology, Institute of Neurology, Affiliated Hospital of North Sichuan Medical College , Wen Hua Road, Nanchong 637000 , China
| |
Collapse
|
133
|
|
134
|
Inhibition of the oxygen sensor PHD2 in the liver improves survival in lactic acidosis by activating the Cori cycle. Proc Natl Acad Sci U S A 2015; 112:11642-7. [PMID: 26324945 DOI: 10.1073/pnas.1515872112] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Loss of prolyl hydroxylase 2 (PHD2) activates the hypoxia-inducible factor-dependent hypoxic response, including anaerobic glycolysis, which causes large amounts of lactate to be released from cells into the circulation. We found that Phd2-null mouse embryonic fibroblasts (MEFs) produced more lactate than wild-type MEFs, as expected, whereas systemic inactivation of PHD2 in mice did not cause hyperlacticacidemia. This unexpected observation led us to hypothesize that the hypoxic response activated in the liver enhances the Cori cycle, a lactate-glucose carbon recycling system between muscle and liver, and thereby decreases circulating lactate. Consistent with this hypothesis, blood lactate levels measured after a treadmill or lactate tolerance test were significantly lower in Phd2-liver-specific knockout (Phd2-LKO) mice than in control mice. An in vivo (13)C-labeled lactate incorporation assay revealed that the livers of Phd2-LKO mice produce significantly more glucose derived from (13)C-labeled lactate than control mice, suggesting that blockade of PHD2 in the liver ameliorates lactic acidosis by activating gluconeogenesis from lactate. Phd2-LKO mice were resistant to lactic acidosis induced by injection of a lethal dose of lactate, displaying a significant elongation of survival. Moreover, oral administration of a PHD inhibitor improved survival in an endotoxin shock mice model. These data suggest that PHD2 is a potentially novel drug target for the treatment of lactic acidosis, which is a serious and often fatal complication observed in some critically ill patients.
Collapse
|
135
|
Min JW, Hu JJ, He M, Sanchez RM, Huang WX, Liu YQ, Bsoul NB, Han S, Yin J, Liu WH, He XH, Peng BW. Vitexin reduces hypoxia-ischemia neonatal brain injury by the inhibition of HIF-1alpha in a rat pup model. Neuropharmacology 2015; 99:38-50. [PMID: 26187393 DOI: 10.1016/j.neuropharm.2015.07.007] [Citation(s) in RCA: 70] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2015] [Revised: 06/16/2015] [Accepted: 07/07/2015] [Indexed: 01/25/2023]
Abstract
Previous studies have demonstrated that the early suppression of HIF-1α after hypoxia-ischemia (HI) injury provides neuroprotection. Vitexin (5, 7, 4-trihydroxyflavone-8-glucoside), an HIF-1α inhibitor, is a c-glycosylated flavone that has been identified in medicinal plants. Therefore, we hypothesized that treatment with vitexin would protect against HI brain injury. Newborn rat pups were subjected to unilateral carotid artery ligation followed by 2.5 h of hypoxia (8% O2 at 37 °C). Vitexin (30, 45 or 60 mg/kg) was administered intraperitoneally at 5 min or 3 h after HI. Vitexin, administered 5 min after HI, was neuroprotective as seen by decreased infarct volume evaluated at 48 h post-HI. This neuroprotection was removed when vitexin was administered 3 h after HI. Neuronal cell death, blood-brain barrier (BBB) integrity, brain edema, HIF-1α and VEGF protein levels were evaluated using a combination of Nissl staining, IgG staining, brain water content, immunohistochemistry and Western blot at 24 and 48 h after HI. The long-term effects of vitexin were evaluated by brain atrophy measurement, Nissl staining and neurobehavioral tests. Vitexin (45 mg/kg) ameliorated brain edema, BBB disruption and neuronal cell death; Upregulation of HIF-1α by dimethyloxalylglycine (DMOG) increased the BBB permeability and brain edema compared to HI alone. Vitexin attenuated the increase in HIF-1α and VEGF. Vitexin also had long-term effects of protecting against the loss of ipsilateral brain and improveing neurobehavioral outcomes. In conclusion, our data indicate early HIF-1α inhibition with vitexin provides both acute and long-term neuroprotection in the developing brain after neonatal HI injury.
Collapse
Affiliation(s)
- Jia-Wei Min
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Jiang-Jian Hu
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Miao He
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Russell M Sanchez
- Department of Surgery, College of Medicine, Texas A&M Health Science Center, Neuroscience Institute, Scott & White Hospital, & Central Texas Veterans Health Care System, Temple, TX, USA
| | - Wen-Xian Huang
- Department of Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Yu-Qiang Liu
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Najeeb Bassam Bsoul
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Song Han
- Department of Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Jun Yin
- Department of Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Wan-Hong Liu
- Department of Immunology, School of Basic Medical Sciences, Wuhan University, Wuhan, China
| | - Xiao-Hua He
- Department of Pathophysiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China.
| | - Bi-Wen Peng
- Department of Physiology, Hubei Provincial Key Laboratory of Developmentally Originated Disorder, School of Basic Medical Sciences, Wuhan University, Wuhan, China.
| |
Collapse
|
136
|
Aune SE, Herr DJ, Kutz CJ, Menick DR. Histone Deacetylases Exert Class-Specific Roles in Conditioning the Brain and Heart Against Acute Ischemic Injury. Front Neurol 2015; 6:145. [PMID: 26175715 PMCID: PMC4485035 DOI: 10.3389/fneur.2015.00145] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Accepted: 06/15/2015] [Indexed: 12/12/2022] Open
Abstract
Ischemia-reperfusion (IR) injury comprises a significant portion of morbidity and mortality from heart and brain diseases worldwide. This enduring clinical problem has inspired myriad reports in the scientific literature of experimental interventions seeking to elucidate the pathology of IR injury. Elective cardiac surgery presents perhaps the most viable scenario for protecting the heart and brain from IR injury due to the opportunity to condition the organs prior to insult. The physiological parameters for the preconditioning of vital organs prior to insult through mechanical and pharmacological maneuvers have been heavily examined. These investigations have revealed new insights into how preconditioning alters cellular responses to IR injury. However, the promise of preconditioning remains unfulfilled at the clinical level, and research seeking to implicate cell signals essential to this protection continues. Recent discoveries in molecular biology have revealed that gene expression can be controlled through posttranslational modifications, without altering the chemical structure of the genetic code. In this scenario, gene expression is repressed by enzymes that cause chromatin compaction through catalytic removal of acetyl moieties from lysine residues on histones. These enzymes, called histone deacetylases (HDACs), can be inhibited pharmacologically, leading to the de-repression of protective genes. The discovery that HDACs can also alter the function of non-histone proteins through posttranslational deacetylation has expanded the potential impact of HDAC inhibitors for the treatment of human disease. HDAC inhibitors have been applied in a very small number of experimental models of IR. However, the scientific literature contains an increasing number of reports demonstrating that HDACs converge on preconditioning signals in the cell. This review will describe the influence of HDACs on major preconditioning signaling pathways in the heart and brain.
Collapse
Affiliation(s)
- Sverre E Aune
- Gazes Cardiac Research Institute, Medical University of South Carolina , Charleston, SC , USA
| | - Daniel J Herr
- Gazes Cardiac Research Institute, Medical University of South Carolina , Charleston, SC , USA
| | - Craig J Kutz
- Gazes Cardiac Research Institute, Medical University of South Carolina , Charleston, SC , USA
| | - Donald R Menick
- Gazes Cardiac Research Institute, Medical University of South Carolina , Charleston, SC , USA
| |
Collapse
|
137
|
Chu HX, Jones NM. Changes in Hypoxia-Inducible Factor-1 (HIF-1) and Regulatory Prolyl Hydroxylase (PHD) Enzymes Following Hypoxic–Ischemic Injury in the Neonatal Rat. Neurochem Res 2015; 41:515-22. [DOI: 10.1007/s11064-015-1641-y] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2015] [Revised: 06/12/2015] [Accepted: 06/15/2015] [Indexed: 01/31/2023]
|
138
|
Huang W, Lv B, Zeng H, Shi D, Liu Y, Chen F, Li F, Liu X, Zhu R, Yu L, Jiang X. Paracrine Factors Secreted by MSCs Promote Astrocyte Survival Associated With GFAP Downregulation After Ischemic Stroke via p38 MAPK and JNK. J Cell Physiol 2015; 230:2461-75. [DOI: 10.1002/jcp.24981] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2015] [Revised: 02/23/2015] [Accepted: 03/02/2015] [Indexed: 12/16/2022]
Affiliation(s)
- Weiyi Huang
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Bingke Lv
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Huijun Zeng
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Dandan Shi
- Department of Anatomy; Key Laboratory of Construction and Detection of Guangdong Province; Southern Medical University; Guangzhou China
| | - Yi Liu
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Fanfan Chen
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Feng Li
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| | - Xinghui Liu
- Department of Anatomy; Key Laboratory of Construction and Detection of Guangdong Province; Southern Medical University; Guangzhou China
| | - Rong Zhu
- Department of Anatomy; Key Laboratory of Construction and Detection of Guangdong Province; Southern Medical University; Guangzhou China
| | - Lei Yu
- Department of Anatomy; Key Laboratory of Construction and Detection of Guangdong Province; Southern Medical University; Guangzhou China
| | - Xiaodan Jiang
- The National Key Clinic Specialty; The Neurosurgery Institute of Guangdong Province; Guangdong Provincial Key Laboratory on Brain Function Repair and Regeneration; Department of Neurosurgery, Zhujiang Hospital; Southern Medical University; Guangzhou China
| |
Collapse
|
139
|
Liu FJ, Kaur P, Karolina DS, Sepramaniam S, Armugam A, Wong PTH, Jeyaseelan K. MiR-335 Regulates Hif-1α to Reduce Cell Death in Both Mouse Cell Line and Rat Ischemic Models. PLoS One 2015; 10:e0128432. [PMID: 26030758 PMCID: PMC4452242 DOI: 10.1371/journal.pone.0128432] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2015] [Accepted: 04/27/2015] [Indexed: 01/25/2023] Open
Abstract
Hypoxia inducible factor-1α facilitates cellular adaptation to hypoxic conditions. Hence its tight regulation is crucial in hypoxia related diseases such as cerebral ischemia. Changes in hypoxia inducible factor-1α expression upon cerebral ischemia influence the expression of its downstream genes which eventually determines the extent of cellular damage. MicroRNAs are endogenous regulators of gene expression that have rapidly emerged as promising therapeutic targets in several diseases. In this study, we have identified miR-335 as a direct regulator of hypoxia inducible factor-1α and as a potential therapeutic target in cerebral ischemia. MiR-335 and hypoxia inducible factor-1α mRNA showed an inverse expression profile, both in vivo and in vitro ischemic conditions. Given the biphasic nature of hypoxia inducible factor-1α expression during cerebral ischemia, miR-335 mimic was found to reduce infarct volume in the early time (immediately after middle cerebral artery occlusion) of embolic stroke animal models while the miR-335 inhibitor appears to be beneficial at the late time of stroke (24 hrs after middle cerebral artery occlusion). Modulation of hypoxia inducible factor-1α expression by miR-335 also influenced the expression of crucial genes implicated in neurovascular permeability, cell death and maintenance of the blood brain barrier. These concerted effects, resulting in a reduction in infarct volume bring about a beneficial outcome in ischemic stroke.
Collapse
Affiliation(s)
- Fu Jia Liu
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, 8 Medical Drive, 117597, Singapore, Singapore
| | - Prameet Kaur
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, 8 Medical Drive, 117597, Singapore, Singapore
| | - Dwi S. Karolina
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, 8 Medical Drive, 117597, Singapore, Singapore
| | - Sugunavathi Sepramaniam
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, 8 Medical Drive, 117597, Singapore, Singapore
| | - Arunmozhiarasi Armugam
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, 8 Medical Drive, 117597, Singapore, Singapore
| | - Peter T. H. Wong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, 10 Medical Drive, 117597, Singapore, Singapore
| | - Kandiah Jeyaseelan
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University Health System, National University of Singapore, 8 Medical Drive, 117597, Singapore, Singapore
- Department of Anatomy and Developmental Biology, School of Biomedical Sciences, Faculty of Medicine, Nursing and Health Sciences, Monash University, Clayton, Victoria, 3800, Australia
- * E-mail:
| |
Collapse
|
140
|
Electroacupuncture Pretreatment Attenuates Cerebral Ischemic Injury via Notch Pathway-Mediated Up-Regulation of Hypoxia Inducible Factor-1α in Rats. Cell Mol Neurobiol 2015; 35:1093-103. [PMID: 25976178 PMCID: PMC4602051 DOI: 10.1007/s10571-015-0203-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2015] [Accepted: 04/29/2015] [Indexed: 12/16/2022]
Abstract
We have reported electroacupuncture (EA) pretreatment induced
the tolerance against focal cerebral ischemia through activation of canonical Notch pathway. However, the underlying mechanisms have not been fully understood. Evidences suggest that up-regulation of hypoxia inducible factor-1α (HIF-1α) contributes to neuroprotection against ischemia which could interact with Notch signaling pathway in this process. Therefore, the current study is to test that up-regulation of HIF-1α associated with Notch pathway contributes to the neuroprotection of EA pretreatment. Sprague–Dawley rats were treated with EA at the acupoint “Baihui (GV 20)” 30 min per day for successive 5 days before MCAO. HIF-1α levels were measured before and after reperfusion. Then, HIF-1α antagonist 2ME2 and γ-secretase inhibitor MW167 were used. Neurologic deficit scores, infarction volumes, neuronal apoptosis, and Bcl2/Bax were evaluated. HIF-1α and Notch1 intracellular domain (NICD) were assessed. The results showed EA pretreatment enhanced the neuronal expression of HIF-1α, reduced infarct volume, improved neurological outcome, inhibited neuronal apoptosis, up-regulated expression of Bcl-2, and down-regulated expression of Bax after reperfusion in the penumbra, while the beneficial effects were attenuated by 2ME2. Furthermore, intraventricular injection with MW167 efficiently suppressed both up-regulation of NICD and HIF-1α after reperfusion. However, administration with 2ME2 could only decrease the expression of HIF-1α in the penumbra. In conclusion, EA pretreatment exerts neuroprotection against ischemic injury through Notch pathway-mediated up-regulation of HIF-1α.
Collapse
|
141
|
Li R, Wang Y, Yang Z, He Y, Zhao T, Fan M, Wang X, Zhu L, Wang X. Hypoxia-inducible factor-1α regulates the expression of L-type voltage-dependent Ca(2+) channels in PC12 cells under hypoxia. Cell Stress Chaperones 2015; 20:507-16. [PMID: 25648081 PMCID: PMC4406929 DOI: 10.1007/s12192-015-0575-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2014] [Revised: 01/16/2015] [Accepted: 01/23/2015] [Indexed: 10/24/2022] Open
Abstract
Hypoxia is an important factor in regulation of cell behavior both under physiological and pathological conditions. The mechanisms of hypoxia-induced cell death have not been completely elucidated yet. It is well known that Ca(2+) is critically related to cell survival. Hypoxia-inducible factor-1α (HIF-1α) is a core regulatory factor during hypoxia, and L-type voltage-dependent Ca(2+) channels (L-VDCCs) have been reported to play a critical role in cell survival. This study was conducted to explore the relationship between L-VDCC expression and HIF-1α regulation in PC12 cells under hypoxia. PC12 cells were treated at 20 or 3 % O2 to observe its proliferation and the intracellular calcium concentration. Then, we detected the protein expression of HIF-1α and L-VDCCs subtypes, Cav1.2 and Cav1.3. At last, to verify the relationship between HIF-1α and Cav1.2 and Cav1.3, we got the expression of Cav1.2 and Cav1.3 with Western blot and luciferase report gene assays after PC12 cells were treated by echinomycin, which is an HIF-1α inhibitor. Compared with 20 % O2 (normoxia), 3 % O2 (hypoxia) inhibited cell proliferation, increased the intracellular calcium concentration, and induced protein expression of HIF-1α. The protein expression of two L-VDCCs subtypes expressed in the nervous system, Cav1.2 and Cav1.3, was upregulated by hypoxia and reduced dose dependently by treatment with echinomycin, a HIF-1α inhibitor. Luciferase report gene assays showed that the expression of Cav1.2 and Cav1.3 genes was augmented under 3 % O2. However, echinomycin only slightly and dose dependently decreased expression of the Cav1.2 gene, but not that of the Cav1.3 gene. These data indicated that Cav1.2 might be regulated by HIF-1α as one of its downstream target genes and involved in regulation of PC12 cells death under hypoxia.
Collapse
Affiliation(s)
- Ran Li
- />Department of Physiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, 10# You An Men, Beijing, 100069 People’s Republic of China
- />Department of Rehabilitation Medicine, Xuan Xu Hospital, Capital Medical University, 45# Changchun Street, Beijing, 100053 People’s Republic of China
| | - Yong Wang
- />Department of Physiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, 10# You An Men, Beijing, 100069 People’s Republic of China
| | - Zhaofei Yang
- />Department of Physiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, 10# You An Men, Beijing, 100069 People’s Republic of China
| | - Yunling He
- />Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, 27# Taiping Road, Beijing, 100850 People’s Republic of China
| | - Tong Zhao
- />Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, 27# Taiping Road, Beijing, 100850 People’s Republic of China
| | - Ming Fan
- />Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, 27# Taiping Road, Beijing, 100850 People’s Republic of China
| | - Xuan Wang
- />Department of Physiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, 10# You An Men, Beijing, 100069 People’s Republic of China
| | - Lingling Zhu
- />Department of Brain Protection and Plasticity, Institute of Basic Medical Sciences, 27# Taiping Road, Beijing, 100850 People’s Republic of China
| | - Xiaomin Wang
- />Department of Physiology, Key Laboratory for Neurodegenerative Disorders of the Ministry of Education, Capital Medical University, 10# You An Men, Beijing, 100069 People’s Republic of China
- />Beijing Institute for Brain Disorder, 10# You An Men, Beijing, 100069 People’s Republic of China
| |
Collapse
|
142
|
Abstract
Preconditioning (PC) using a preceding sublethal ischemic insult is an attractive strategy for protecting neurons by inducing ischemic tolerance in the brain. Although the underlying molecular mechanisms have been extensively studied, almost all studies have focused on neurons. Here, using a middle cerebral artery occlusion model in mice, we show that astrocytes play an essential role in the induction of brain ischemic tolerance. PC caused activation of glial cells without producing any noticeable brain damage. The spatiotemporal pattern of astrocytic, but not microglial, activation correlated well with that of ischemic tolerance. Interestingly, such activation in astrocytes lasted at least 8 weeks. Importantly, inhibiting astrocytes with fluorocitrate abolished the induction of ischemic tolerance. To investigate the underlying mechanisms, we focused on the P2X7 receptor as a key molecule in astrocyte-mediated ischemic tolerance. P2X7 receptors were dramatically upregulated in activated astrocytes. PC-induced ischemic tolerance was abolished in P2X7 receptor knock-out mice. Moreover, our results suggest that hypoxia-inducible factor-1α, a well known mediator of ischemic tolerance, is involved in P2X7 receptor-mediated ischemic tolerance. Unlike previous reports focusing on neuron-based mechanisms, our results show that astrocytes play indispensable roles in inducing ischemic tolerance, and that upregulation of P2X7 receptors in astrocytes is essential.
Collapse
|
143
|
Fumagalli S, Perego C, Pischiutta F, Zanier ER, De Simoni MG. The ischemic environment drives microglia and macrophage function. Front Neurol 2015; 6:81. [PMID: 25904895 PMCID: PMC4389404 DOI: 10.3389/fneur.2015.00081] [Citation(s) in RCA: 190] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2015] [Accepted: 03/25/2015] [Indexed: 12/16/2022] Open
Abstract
Cells of myeloid origin, such as microglia and macrophages, act at the crossroads of several inflammatory mechanisms during pathophysiology. Besides pro-inflammatory activity (M1 polarization), myeloid cells acquire protective functions (M2) and participate in the neuroprotective innate mechanisms after brain injury. Experimental research is making considerable efforts to understand the rules that regulate the balance between toxic and protective brain innate immunity. Environmental changes affect microglia/macrophage functions. Hypoxia can affect myeloid cell distribution, activity, and phenotype. With their intrinsic differences, microglia and macrophages respond differently to hypoxia, the former depending on ATP to activate and the latter switching to anaerobic metabolism and adapting to hypoxia. Myeloid cell functions include homeostasis control, damage-sensing activity, chemotaxis, and phagocytosis, all distinctive features of these cells. Specific markers and morphologies enable to recognize each functional state. To ensure homeostasis and activate when needed, microglia/macrophage physiology is finely tuned. Microglia are controlled by several neuron-derived components, including contact-dependent inhibitory signals and soluble molecules. Changes in this control can cause chronic activation or priming with specific functional consequences. Strategies, such as stem cell treatment, may enhance microglia protective polarization. This review presents data from the literature that has greatly advanced our understanding of myeloid cell action in brain injury. We discuss the selective responses of microglia and macrophages to hypoxia after stroke and review relevant markers with the aim of defining the different subpopulations of myeloid cells that are recruited to the injured site. We also cover the functional consequences of chronically active microglia and review pivotal works on microglia regulation that offer new therapeutic possibilities for acute brain injury.
Collapse
Affiliation(s)
- Stefano Fumagalli
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri , Milan , Italy ; Department of Pathophysiology and Transplantation, Fondazione IRCCS Ca' Granda-Ospedale Maggiore Policlinico , Milan , Italy
| | - Carlo Perego
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri , Milan , Italy
| | - Francesca Pischiutta
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri , Milan , Italy
| | - Elisa R Zanier
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri , Milan , Italy
| | - Maria-Grazia De Simoni
- Department of Neuroscience, IRCCS-Istituto di Ricerche Farmacologiche Mario Negri , Milan , Italy
| |
Collapse
|
144
|
Hypoxia Inducible Factor-1 α in Astrocytes and/or Myeloid Cells Is Not Required for the Development of Autoimmune Demyelinating Disease. eNeuro 2015. [PMID: 26213713 PMCID: PMC4511492 DOI: 10.1523/eneuro.0050-14.2015] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Despite numerous reports indicating HIF-1α expression in glia, neurons, and inflammatory cells in the CNS of MS patients, the cell-specific contribution of HIF-1α to disease pathogenesis remains unclear. Here we show that although HIF-1α is dramatically upregulated in astrocytes and myeloid cells in EAE, cell-specific depletion of HIF-1α in these two cell types surprisingly does not affect the development of neuroinflammatory disease. Hypoxia-like tissue alterations, characterized by the upregulation of hypoxia-inducible factor-1α (HIF-1α), have been described in the normal appearing white matter and pre-demyelinating lesions of multiple sclerosis (MS) patients. As HIF-1α regulates the transcription of a wide set of genes involved in neuroprotection and neuroinflammation, HIF-1α expression may contribute to the pathogenesis of inflammatory demyelination. To test this hypothesis, we analyzed the effect of cell-specific genetic ablation or overexpression of HIF-1α on the onset and progression of experimental autoimmune encephalomyelitis (EAE), a mouse model for MS. HIF-1α was mainly expressed in astrocytes and microglia/macrophages in the mouse spinal cord at the peak of EAE. However, genetic ablation of HIF-1α in astrocytes and/or myeloid cells did not ameliorate clinical symptoms. Furthermore, conditional knock-out of Von Hippel Lindau, a negative regulator of HIF-1α stabilization, failed to exacerbate the clinical course of EAE. In accordance with clinical symptoms, genetic ablation or overexpression of HIF-1α did not change the extent of spinal cord inflammation and demyelination. Overall, our data indicate that despite dramatic upregulation of HIF-1α in astrocytes and myeloid cells in EAE, HIF-1α expression in these two cell types is not required for the development of inflammatory demyelination. Despite numerous reports indicating HIF-1α expression in glia, neurons, and inflammatory cells in the CNS of MS patients, the cell-specific contribution of HIF-1α to disease pathogenesis remains unclear. Here we show that although HIF-1α is dramatically upregulated in astrocytes and myeloid cells in EAE, cell-specific depletion of HIF-1α in these two cell types surprisingly does not affect the development of neuroinflammatory disease. Together with two recently published studies showing a role for oligodendrocyte-specific HIF-1α in myelination and T-cell-specific HIF-1α in EAE, our results demonstrate a tightly regulated cellular specificity for HIF-1α contribution in nervous system pathogenesis.
Collapse
|
145
|
López-Hernández B, Ceña V, Posadas I. The endoplasmic reticulum stress and the HIF-1 signalling pathways are involved in the neuronal damage caused by chemical hypoxia. Br J Pharmacol 2015; 172:2838-51. [PMID: 25625917 DOI: 10.1111/bph.13095] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 12/23/2014] [Accepted: 01/15/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND AND PURPOSE Hypoxia inducible factor-1 (HIF-1) promotes transitory neuronal survival suggesting that additional mechanisms such as the endoplasmic reticulum (ER) stress might be involved in determining neuronal survival or death. Here, we examined the involvement of ER stress in hypoxia-induced neuronal death and analysed the relationship between ER stress and the HIF-1 pathways. EXPERIMENTAL APPROACH Cultures of rat cortical neurons were exposed to chemical hypoxia induced by 200 μM CoCl2 , and its effect on neuronal viability was assessed by 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assay and counting apoptotic nuclei. Protein levels were determined by Western blot analysis. RT-PCR was performed to analyse the content and the t1/2 of HIF-1α mRNA. KEY RESULTS Chemical hypoxia induced neuronal apoptosis in a time-dependent manner and activated the ER stress PRK-like endoplasmic reticulum kinase (PERK)-dependent pathway. At later stages, chemical hypoxia increased the expression of the C/EBP homologous protein (CHOP) and caspase 12 activity. CoCl2 reduced HIF-1α mRNA t1/2 leading to a decrease in HIF-1α mRNA and protein content, simultaneously activating the ER stress PERK-dependent pathway. Salubrinal, a selective inhibitor of phospho-eIF2α phosphatase, protected neurons from chemical hypoxia by reducing CHOP levels and caspase 12 activity, and increasing the t1/2 of HIF-1α mRNA and the levels of HIF-1α protein. Knocking down HIF-1α blocked the neuroprotective effects of salubrinal. CONCLUSIONS AND IMPLICATIONS Neuronal apoptosis induced by chemical hypoxia is a process regulated by HIF-1α stabilization early on and by ER stress activation at later stages. Our data also suggested that HIF-1α levels were regulated by ER stress.
Collapse
Affiliation(s)
- Beatriz López-Hernández
- Departamento de Ciencias Médicas, Unidad Asociada Neurodeath CSIC-Universidad de Castilla-La Mancha, Albacete, Spain
| | - Valentin Ceña
- Departamento de Ciencias Médicas, Unidad Asociada Neurodeath CSIC-Universidad de Castilla-La Mancha, Albacete, Spain.,CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| | - Inmaculada Posadas
- Departamento de Ciencias Médicas, Unidad Asociada Neurodeath CSIC-Universidad de Castilla-La Mancha, Albacete, Spain.,CIBERNED, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
146
|
Smeyne M, Sladen P, Jiao Y, Dragatsis I, Smeyne RJ. HIF1α is necessary for exercise-induced neuroprotection while HIF2α is needed for dopaminergic neuron survival in the substantia nigra pars compacta. Neuroscience 2015; 295:23-38. [PMID: 25796140 DOI: 10.1016/j.neuroscience.2015.03.015] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2014] [Revised: 03/05/2015] [Accepted: 03/07/2015] [Indexed: 02/05/2023]
Abstract
Exercise reduces the risk of developing a number of neurological disorders and increases the efficiency of cellular energy production. However, overly strenuous exercise produces oxidative stress. Proper oxygenation is crucial for the health of all tissues, and tight regulation of cellular oxygen is critical to balance O2 levels and redox homeostasis in the brain. Hypoxia Inducible Factor (HIF)1α and HIF2α are transcription factors regulated by cellular oxygen concentration that initiate gene regulation of vascular development, redox homeostasis, and cell cycle control. HIF1α and HIF2α contribute to important adaptive mechanisms that occur when oxygen and ROS homeostasis become unbalanced. It has been shown that preconditioning by exposure to a stressor prior to a hypoxic event reduces damage that would otherwise occur. Previously we reported that 3 months of exercise protects SNpc dopaminergic (DA) neurons from toxicity caused by Complex I inhibition. Here, we identify the cells in the SNpc that express HIF1α and HIF2α and show that running exercise produces hypoxia in SNpc DA neurons, and alters the expression of HIF1α and HIF2α. In mice carrying a conditional knockout of Hif1α in postnatal neurons we observe that exercise alone produces SNpc TH+ DA neuron loss. Loss of HIF1α also abolishes exercise-induced neuroprotection. In mice lacking Hif2α in postnatal neurons, the number of TH+ DA neurons in the adult SNpc is diminished, but 3months of exercise rescues this loss. We conclude that HIF1α is necessary for exercise-induced neuroprotection and both HIF1α and HIF2α are necessary for the survival and function of adult SNpc DA neurons.
Collapse
Affiliation(s)
- M Smeyne
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, United States
| | - P Sladen
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, United States
| | - Y Jiao
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, United States
| | - I Dragatsis
- Department of Physiology, The University of Tennessee Health Science Center, 894 Union Avenue, Memphis, TN 38163, United States
| | - R J Smeyne
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, 262 Danny Thomas Place, Memphis, TN 38105, United States.
| |
Collapse
|
147
|
Chen PY, Ho YR, Wu MJ, Huang SP, Chen PK, Tai MH, Ho CT, Yen JH. Cytoprotective effects of fisetin against hypoxia-induced cell death in PC12 cells. Food Funct 2015; 6:287-96. [DOI: 10.1039/c4fo00948g] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Fisetin protects cells under hypoxia through ROS scavenging and the HIF1α-, MAPK/ERK-, p38 MAPK- and PI3 K/Akt-dependent pathways in PC12 cells.
Collapse
Affiliation(s)
- Pei-Yi Chen
- Center of Medical Genetics
- Buddhist Tzu Chi General Hospital
- Hualien 970
- Taiwan
| | - Yi-Ru Ho
- Department of Molecular Biology and Human Genetics
- Tzu Chi University
- Hualien 970
- Taiwan
| | - Ming-Jiuan Wu
- Department of Biotechnology
- Chia Nan University of Pharmacy and Science
- Tainan 717
- Taiwan
| | - Shun-Ping Huang
- Department of Molecular Biology and Human Genetics
- Tzu Chi University
- Hualien 970
- Taiwan
| | - Po-Kong Chen
- Department of Molecular Biology and Human Genetics
- Tzu Chi University
- Hualien 970
- Taiwan
| | - Mi-Hsueh Tai
- Department of Molecular Biology and Human Genetics
- Tzu Chi University
- Hualien 970
- Taiwan
| | - Chi-Tang Ho
- Department of Food Science
- Rutgers University
- New Brunswick
- USA
| | - Jui-Hung Yen
- Department of Molecular Biology and Human Genetics
- Tzu Chi University
- Hualien 970
- Taiwan
| |
Collapse
|
148
|
Moisan A, Favre IM, Rome C, Grillon E, Naegele B, Barbieux M, De Fraipont F, Richard MJ, Barbier EL, Rémy C, Detante O. Microvascular plasticity after experimental stroke: a molecular and MRI study. Cerebrovasc Dis 2014; 38:344-53. [PMID: 25427570 DOI: 10.1159/000368597] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2014] [Accepted: 09/23/2014] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Microvasculature plays a key role in stroke pathophysiology both during initial damage and extended neural repair. Moreover, angiogenesis processes seem to be a promising target for future neurorestorative therapies. However, dynamic changes of microvessels after stroke still remain unclear, and MRI follow-up could be interesting as an in vivo biomarker of these. METHODS The aim of this study is to characterize the microvascular plasticity 25 days after ischemic stroke using both in vivo microvascular 7T-MRI (vascular permeability, cerebral blood volume (CBV), vessel size index (VSI), vascular density) and quantification of angiogenic factor expressions by RT-qPCR in a transient middle cerebral artery occlusion rat model. CBV and VSI (perfused vessel caliber) imaging was performed using a steady-state approach with a multi gradient-echo spin-echo sequence before and 2 min after intravenous (IV) injection of ultrasmall superparamagnetic iron particles. Vascular density (per mm2) was derived from the ratio [ΔR₂/(ΔR₂*)²/³]. Blood brain barrier leakage was assessed using T₁W images before and after IV injection of Gd-DOTA. Additionally, microvessel immunohistology was done. RESULTS 3 successive stages were observed: 1) 'Acute stage' from day 1 to day 3 post-stroke (D1-D3) characterized by high levels of angiopoietin-2 (Ang2), vascular endothelial growth factor receptor-2 (VEGFR-2) and endothelial NO synthase (eNOS) that may be associated with deleterious vascular permeability and vasodilation; 2) 'Transition stage' (D3-D7) that involves transforming the growth factors β1 (TGFβ1), Ang1, and tyrosine kinase with immunoglobulin-like and endothelial growth factor-like domains 1 (Tie1), stromal-derived factor-1 (SDF-1), chemokine receptor type 4 (CXCR-4); and 3) 'Subacute stage' (D7-D25) with high levels of Ang1, Ang2, VEGF, VEGFR-1 and TGFβ1 leading to favorable stabilization and maturation of microvessels. In vivo MRI appeared in line with the angiogenic factors changes with a delay of at least 1 day. All MRI parameters varied over time, revealing the different aspects of the post-stroke microvascular plasticity. At D25, despite a normal CBV, MRI revealed a limited microvessel density, which is insufficient to support a good neural repair. CONCLUSIONS Microvasculature MRI can provide imaging of different states of functional (perfused) microvessels after stroke. These results highlight that multiparametric MRI is useful to assess post-stroke angiogenesis, and could be used as a biomarker notably for neurorestorative therapy studies. Additionally, we identified that endogenous vessel maturation and stabilization occur during the 'subacute stage'. Thus, pro-angiogenic treatments, such as cell-based therapy, would be relevant during this subacute phase of stroke.
Collapse
|
149
|
Hypoxia-inducible factor 1α mediates neuroprotection of hypoxic postconditioning against global cerebral ischemia. J Neuropathol Exp Neurol 2014; 73:975-86. [PMID: 25192050 DOI: 10.1097/nen.0000000000000118] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Hypoxia administered after transient global cerebral ischemia (tGCI) has been shown to induce neuroprotection in adult rats, but the underlying mechanisms for this protection are unclear. Here, we tested the hypothesis that hypoxic postconditioning (HPC) induces neuroprotection through upregulation of hypoxia-inducible factor 1α (HIF-1α) and vascular endothelial growth factor (VEGF), and that this involves phosphatidylinositol-3-kinase (PI3K), p38 mitogen-activated protein kinase (p38 MAPK), and mitogen-activated protein kinase/extracellular signal-regulated kinase kinase (MEK) pathways. The expression of HIF-1α, VEGF, and cleaved caspase-9 were determined by immunohistochemistry and Western blot. As pharmacologic interventions, the HIF-1α inhibitor 2-methoxyestradiol (2ME2), PI3K inhibitor LY294002, p38 MAPK inhibitor SB203580, and MEK inhibitor U0126 were administered before HPC or after tGCI. We found that HPC maintained the higher expression of HIF-1α and VEGF and decreased cleaved caspase-9 levels in CA1 after tGCI. These effects were reversed by 2ME2 administered before HPC, and the neuroprotection of HPC was abolished. LY294002 and SB203580 decreased the expression of HIF-1α and VEGF after HPC, whereas U0126 increased HIF-1α and VEGF after tGCI. These findings suggested that HIF-1α exerts neuroprotection induced by HPC against tGCI through VEGF upregulation and cleaved caspase-9 downregulation, and that the PI3K, p38 MAPK, and MEK pathways are involved in the regulation of HIF-1α and VEGF.
Collapse
|
150
|
Cui H, Han W, Yang L, Chang Y. Expression of hypoxia-inducible factor 1 alpha and oligodendrocyte lineage gene-1 in cultured brain slices after oxygen-glucose deprivation. Neural Regen Res 2014; 8:328-37. [PMID: 25206673 PMCID: PMC4107529 DOI: 10.3969/j.issn.1673-5374.2013.04.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2012] [Accepted: 01/09/2013] [Indexed: 02/03/2023] Open
Abstract
Oligodendrocyte lineage gene-1 expressed in oligodendrocytes may trigger the repair of neuronal myelin impairment, and play a crucial role in myelin repair. Hypoxia-inducible factor 1α, a transcription factor, is of great significance in premature infants with hypoxic-ischemic brain damage. There is little evidence of direct regulatory effects of hypoxia-inducible factor 1α on oligodendrocyte lineage gene-1. In this study, brain slices of Sprague-Dawley rats were cultured and subjected to oxygen-glucose deprivation. Then, slices were transfected with hypoxia-inducible factor 1α or oligodendrocyte lineage gene-1. The expression levels of hypoxia-inducible factor 1α and oligodendrocyte lineage gene-1 were significantly up-regulated in rat brains prior to transfection, as detected by immunohistochemical staining. Eight hours after transfection of slices with hypoxia-inducible factor 1α, oligodendrocyte lineage gene-1 expression was upregulated, and reached a peak 24 hours after transfection. Oligodendrocyte lineage gene-1 transfection induced no significant differences in hypoxia-inducible factor 1α levels in rat brain tissues with oxygen-glucose deprivation. These experimental findings indicate that hypoxia-inducible factor 1α can regulate oligodendrocyte lineage gene-1 expression in hypoxic brain tissue, thus repairing the neural impairment.
Collapse
Affiliation(s)
- Hong Cui
- College of Life Science, Hebei Normal University, Shijiazhuang 050016, Hebei Province, China ; Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Weijuan Han
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Lijun Yang
- Department of Pediatrics, Beijing Friendship Hospital, Capital Medical University, Beijing 100050, China
| | - Yanzhong Chang
- College of Life Science, Hebei Normal University, Shijiazhuang 050016, Hebei Province, China
| |
Collapse
|