1
|
Xia J, Zhang T, Sun Y, Huang Z, Shi D, Qin D, Yang X, Liu H, Yao G, Wei L, Chang X, Gao J, Guo Y, Hou XY. Suppression of neuronal CDK9/p53/VDAC signaling provides bioenergetic support and improves post-stroke neuropsychiatric outcomes. Cell Mol Life Sci 2024; 81:384. [PMID: 39235466 PMCID: PMC11377386 DOI: 10.1007/s00018-024-05428-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Revised: 07/30/2024] [Accepted: 08/27/2024] [Indexed: 09/06/2024]
Abstract
Bioenergy decline occurs with reperfusion following acute ischemic stroke. However, the molecular mechanisms that limit energy metabolism and their impact on post-stroke cognitive and emotional complications are still unclear. In the present study, we demonstrate that the p53 transcriptional response is responsible for neuronal adenosine triphosphate (ATP) deficiency and progressively neuropsychiatric disturbances, involving the downregulation of mitochondrial voltage-dependent anion channels (VDACs). Neuronal p53 transactivated the promoter of microRNA-183 (miR-183) cluster, thereby upregulating biogenesis of miR-183-5p (miR-183), miR-96-5p (miR-96), and miR-182-5p. Both miR-183 and miR-96 directly targeted and post-transcriptionally suppressed VDACs. Neuronal ablation of p53 protected against ATP deficiency and neurological deficits, whereas post-stroke rescue of miR-183/VDAC signaling reversed these benefits. Interestingly, cyclin-dependent kinase 9 (CDK9) was found to be enriched in cortical neurons and upregulated the p53-induced transcription of the miR-183 cluster in neurons after ischemia. Post-treatment with the CDK9 inhibitor oroxylin A promoted neuronal ATP production mainly through suppressing the miR-183 cluster/VDAC axis, further improved long-term sensorimotor abilities and spatial memory, and alleviated depressive-like behaviors in mice following stroke. Our findings reveal an intrinsic CDK9/p53/VDAC pathway that drives neuronal bioenergy decline and underlies post-stroke cognitive impairment and depression, thus highlighting the therapeutic potential of oroxylin A for better outcomes.
Collapse
Affiliation(s)
- Jing Xia
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Tingting Zhang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
- Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Ying Sun
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Zhu Huang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Dingfang Shi
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Dongshen Qin
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Xuejun Yang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Hao Liu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
- Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Guiying Yao
- Research Center for Biochemistry and Molecular Biology, Xuzhou Medical University, Xuzhou, Jiangsu, 221004, China
| | - Libin Wei
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China
| | - Xiaoai Chang
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China
| | - Jun Gao
- School of Basic Medical Sciences, Nanjing Medical University, Nanjing, Jiangsu, 211166, China.
| | - Yongjian Guo
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
- School of Biopharmacy, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
| | - Xiao-Yu Hou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
- School of Life Science and Technology, China Pharmaceutical University, Nanjing, Jiangsu, 211198, China.
| |
Collapse
|
2
|
Gupta S, Khan J, Ghosh S. Molecular mechanism of cognitive impairment associated with Parkinson's disease: A stroke perspective. Life Sci 2024; 337:122358. [PMID: 38128756 DOI: 10.1016/j.lfs.2023.122358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 12/03/2023] [Accepted: 12/13/2023] [Indexed: 12/23/2023]
Abstract
Parkinson's disease (PD) is a common neurological illness that causes several motor and non-motor symptoms, most characteristically limb tremors and bradykinesia. PD is a slowly worsening disease that arises due to progressive neurodegeneration of specific areas of the brain, especially the substantia nigra of the midbrain. Even though PD has continuously been linked to a higher mortality risk in numerous epidemiologic studies, there have been significant discoveries regarding the connection between PD and stroke. The incidence of strokes such as cerebral infarction and hemorrhage is substantially associated with the development of PD. Moreover, cognitive impairments, primarily dementia, have been associated with stroke and PD. However, the underlying molecular mechanism of this phenomenon is still obscure. This concise review focuses on the relationship between stroke and PD, emphasizing the molecular mechanism of cognition deficit and memory loss evident in PD and stroke. Furthermore, we are also highlighting some potential drug molecules that can target both PD and stroke.
Collapse
Affiliation(s)
- Sanju Gupta
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur (IIT-Jodhpur), Rajasthan 342037, India
| | - Juhee Khan
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur (IIT-Jodhpur), Rajasthan 342037, India
| | - Surajit Ghosh
- Department of Bioscience & Bioengineering, Indian Institute of Technology Jodhpur (IIT-Jodhpur), Rajasthan 342037, India.
| |
Collapse
|
3
|
Chen B, Jin W. A comprehensive review of stroke-related signaling pathways and treatment in western medicine and traditional Chinese medicine. Front Neurosci 2023; 17:1200061. [PMID: 37351420 PMCID: PMC10282194 DOI: 10.3389/fnins.2023.1200061] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/19/2023] [Indexed: 06/24/2023] Open
Abstract
This review provides insight into the complex network of signaling pathways and mechanisms involved in stroke pathophysiology. It summarizes the historical progress of stroke-related signaling pathways, identifying potential interactions between them and emphasizing that stroke is a complex network disease. Of particular interest are the Hippo signaling pathway and ferroptosis signaling pathway, which remain understudied areas of research, and are therefore a focus of the review. The involvement of multiple signaling pathways, including Sonic Hedgehog (SHH), nuclear factor erythroid 2-related factor 2 (Nrf2)/antioxidant response element (ARE), hypoxia-inducible factor-1α (HIF-1α), PI3K/AKT, JAK/STAT, and AMPK in pathophysiological mechanisms such as oxidative stress and apoptosis, highlights the complexity of stroke. The review also delves into the details of traditional Chinese medicine (TCM) therapies such as Rehmanniae and Astragalus, providing an analysis of the recent status of western medicine in the treatment of stroke and the advantages and disadvantages of TCM and western medicine in stroke treatment. The review proposes that since stroke is a network disease, TCM has the potential and advantages of a multi-target and multi-pathway mechanism of action in the treatment of stroke. Therefore, it is suggested that future research should explore more treasures of TCM and develop new therapies from the perspective of stroke as a network disease.
Collapse
Affiliation(s)
- Binhao Chen
- The First School of Clinical Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Weifeng Jin
- College of Pharmaceutical Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
4
|
Tian H, Chen X, Liao J, Yang T, Cheng S, Mei Z, Ge J. Mitochondrial quality control in stroke: From the mechanisms to therapeutic potentials. J Cell Mol Med 2022; 26:1000-1012. [PMID: 35040556 PMCID: PMC8831937 DOI: 10.1111/jcmm.17189] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 12/17/2021] [Accepted: 01/03/2022] [Indexed: 12/14/2022] Open
Abstract
Mitochondrial damage is a critical contributor to stroke‐induced injury, and mitochondrial quality control (MQC) is the cornerstone of restoring mitochondrial homeostasis and plays an indispensable role in alleviating pathological process of stroke. Mitochondria quality control promotes neuronal survival via various adaptive responses for preserving mitochondria structure, morphology, quantity and function. The processes of mitochondrial fission and fusion allow for damaged mitochondria to be segregated and facilitate the equilibration of mitochondrial components such as DNA, proteins and metabolites. The process of mitophagy is responsible for the degradation and recycling of damaged mitochondria. This review aims to offer a synopsis of the molecular mechanisms involved in MQC for recapitulating our current understanding of the complex role that MQC plays in the progression of stroke. Speculating on the prospect that targeted manipulation of MQC mechanisms may be exploited for the rationale design of novel therapeutic interventions in the ischaemic stroke and haemorrhagic stroke. In the review, we highlight the potential of MQC as therapeutic targets for stroke treatment and provide valuable insights for clinical strategies.
Collapse
Affiliation(s)
- Heyan Tian
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-cerebral Disease, Hunan University of Chinese Medicine, Changsha, China
| | - Xiangyu Chen
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-cerebral Disease, Hunan University of Chinese Medicine, Changsha, China
| | - Jun Liao
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-cerebral Disease, Hunan University of Chinese Medicine, Changsha, China
| | - Tong Yang
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-cerebral Disease, Hunan University of Chinese Medicine, Changsha, China
| | - Shaowu Cheng
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-cerebral Disease, Hunan University of Chinese Medicine, Changsha, China
| | - Zhigang Mei
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-cerebral Disease, Hunan University of Chinese Medicine, Changsha, China
| | - Jinwen Ge
- Key Laboratory of Hunan Province for Integrated Traditional Chinese and Western Medicine on Prevention and Treatment of Cardio-cerebral Disease, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
5
|
Abstract
Mitochondria play a central role in the pathophysiological processes of acute ischemic stroke. Disruption of the cerebral blood flow during acute ischemic stroke interrupts oxygen and glucose delivery, leading to the dysfunction of mitochondrial oxidative phosphorylation and cellular bioenergetic stress. Cells can respond to such stress by activating mitochondrial quality control mechanisms, including the mitochondrial unfolded protein response, mitochondrial fission and fusion, mitophagy, mitochondrial biogenesis, and intercellular mitochondrial transfer. Collectively, these adaptive response strategies contribute to retaining the integrity and function of the mitochondrial network, thereby helping to recover the homeostasis of the neurovascular unit. In this review, we focus on mitochondrial quality control mechanisms occurring in acute ischemic stroke. A better understanding of how these regulatory pathways work in maintaining mitochondrial homeostasis will provide a rationale for developing innovative neuroprotectants when these mechanisms fail in acute ischemic stroke.
Collapse
Affiliation(s)
- Hong An
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China
| | - Bing Zhou
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, China.,Interdisciplinary Innovation Institute of Medicine and Engineering Interdisciplinary, Beihang University, Beijing, China
| | - Xunming Ji
- Department of Neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Capital Medical University, Beijing, China.,Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, China.,Interdisciplinary Innovation Institute of Medicine and Engineering Interdisciplinary, Beihang University, Beijing, China.,Department of Neurosurgery, 71044Xuanwu Hospital, Xuanwu Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
6
|
Narasimhan M, Schwartz R, Halliday G. Parkinsonism and cerebrovascular disease. J Neurol Sci 2021; 433:120011. [PMID: 34686356 DOI: 10.1016/j.jns.2021.120011] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/01/2021] [Accepted: 09/29/2021] [Indexed: 11/27/2022]
Abstract
The relationship between cerebrovascular disease and parkinsonism is commonly seen in everyday clinical practice but remains ill-defined and under-recognised with little guidance for the practising neurologist. We attempt to define this association and to illustrate key clinical, radiological and pathological features of the syndrome of Vascular Parkinsonism (VaP). VaP is a major cause of morbidity in the elderly associated with falls, hip fractures and cognitive impairment. Although acute parkinsonism is reported in the context of an acute cerebrovascular event, the vast majority of VaP presents as an insidious syndrome usually in the context of vascular risk factors and radiological evidence of small vessel disease. There may be an anatomic impact on basal ganglia neuronal networks, however the effect of small vessel disease (SVD) on these pathways is not clear. There are now established reporting standards for radiological features of SVD on MRI. White matter hyperintensities and lacunes have been thought to be the representative radiological features of SVD but other features such as the perivascular space are gaining more importance, especially in context of the glymphatic system. It is important to consider VaP in the differential diagnosis of Parkinson disease (PD) and in these situations, neuroimaging may offer diagnostic benefit especially in those patients with atypical presentations or refractoriness to levodopa. Proactive management of vascular risk factors, monitoring of bone density and an exercise program may offer easily attainable therapeutic targets in PD and VaP. Levodopa therapy should be considered in patients with VaP, however the dose and effect may be different from use in PD. This article is part of the Special Issue "Parkinsonism across the spectrum of movement disorders and beyond" edited by Joseph Jankovic, Daniel D. Truong and Matteo Bologna.
Collapse
Affiliation(s)
- Manisha Narasimhan
- Brain and Mind Centre and Faculty of Health and Medical Sciences, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia.
| | - Raymond Schwartz
- Brain and Mind Centre and Faculty of Health and Medical Sciences, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
| | - Glenda Halliday
- Brain and Mind Centre and Faculty of Health and Medical Sciences, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
| |
Collapse
|
7
|
Wen H, Li L, Zhan L, Zuo Y, Li K, Qiu M, Li H, Sun W, Xu E. Hypoxic postconditioning promotes mitophagy against transient global cerebral ischemia via PINK1/Parkin-induced mitochondrial ubiquitination in adult rats. Cell Death Dis 2021; 12:630. [PMID: 34145219 PMCID: PMC8213752 DOI: 10.1038/s41419-021-03900-8] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2020] [Revised: 06/01/2021] [Accepted: 06/03/2021] [Indexed: 11/08/2022]
Abstract
Mitophagy alleviates neuronal damage after cerebral ischemia by selectively removing dysfunctional mitochondria. Phosphatase and tensin homolog (PTEN) induced putative kinase 1 (PINK1)/Parkin-mediated mitophagy is the most well-known type of mitophagy. However, little is known about the role of PINK1/Parkin-mediated mitophagy in ischemic tolerance induced by hypoxic postconditioning (HPC) with 8% O2 against transient global cerebral ischemia (tGCI). Hence, we aimed to test the hypothesis that HPC-mediated PINK1/Parkin-induced mitochondrial ubiquitination and promotes mitophagy, thus exerting neuroprotection in the hippocampal CA1 subregion against tGCI. We found that mitochondrial clearance was disturbed at the late phase of reperfusion after tGCI, which was reversed by HPC, as evidenced by the reduction of the translocase of outer mitochondrial membrane 20 homologs (TOMM20), translocase of inner mitochondrial membrane 23 (TIMM23) and heat shock protein 60 (HSP60) in CA1 after HPC. In addition, HPC further increased the ratio of LC3II/I in mitochondrial fraction and promoted the formation of mitophagosomes in CA1 neurons after tGCI. The administration of lysosome inhibitor chloroquine (CQ) intraperitoneally or mitophagy inhibitor (Mdivi-1) intracerebroventricularly abrogated HPC-induced mitochondrial turnover and neuroprotection in CA1 after tGCI. We also found that HPC activated PINK1/Parkin pathway after tGCI, as shown by the augment of mitochondrial PINK1 and Parkin and the promotion of mitochondrial ubiquitination in CA1. In addition, PINK1 or Parkin knockdown with small-interfering RNA (siRNA) suppressed the activation of PINK1/Parkin pathway and hampered mitochondrial clearance and attenuated neuroprotection induced by HPC, whereas PINK1 overexpression promoted PINK1/Parkin-mediated mitophagy and ameliorated neuronal damage in CA1 after tGCI. Taken together, the new finding in this study is that HPC-induced neuroprotection against tGCI through promoting mitophagy mediated by PINK1/Parkin-dependent pathway.
Collapse
Affiliation(s)
- Haixia Wen
- Institute of Neuroscience and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, P. R. China
- Department of Neurology, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Luxi Li
- Institute of Neuroscience and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, P. R. China
- Department of Neurology, Affiliated Brain Hospital of Guangzhou Medical University, Guangzhou Huiai Hospital, Guangzhou, China
| | - Lixuan Zhan
- Institute of Neuroscience and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, P. R. China
| | - Yunyan Zuo
- Institute of Neuroscience and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, P. R. China
| | - Kongping Li
- Institute of Neuroscience and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, P. R. China
| | - Meiqian Qiu
- Institute of Neuroscience and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, P. R. China
| | - Heying Li
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, 510530, China
| | - Weiwen Sun
- Institute of Neuroscience and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, P. R. China
| | - En Xu
- Institute of Neuroscience and Department of Neurology of the Second Affiliated Hospital of Guangzhou Medical University and Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou, 510260, P. R. China.
| |
Collapse
|
8
|
Murillo-González FE, García-Aguilar R, Vega L, Elizondo G. Regulation of Parkin expression as the key balance between neural survival and cancer cell death. Biochem Pharmacol 2021; 190:114650. [PMID: 34111426 DOI: 10.1016/j.bcp.2021.114650] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 06/02/2021] [Accepted: 06/04/2021] [Indexed: 11/25/2022]
Abstract
Parkin is a cytosolic E3 ubiquitin ligase that plays an important role in neuroprotection by targeting several proteins to be degraded by the 26S proteasome. Its dysfunction has been associated not only with Parkinson's disease (PD) but also with other neurodegenerative pathologies, such as Alzheimer's disease and Huntington's disease. More recently, Parkin has been identified as a tumor suppressor gene implicated in cancer development. Due to the important roles that this E3 ubiquitin ligase plays in cellular homeostasis, its expression, activity, and turnover are tightly regulated. Several reviews have addressed Parkin regulation; however, genetic and epigenetic regulation have been excluded. In addition to posttranslational modifications (PTMs), this review examines the regulatory mechanisms that control Parkin function through gene expression, epigenetic regulation, and degradation. Furthermore, the consequences of disrupting these regulatory processes on human health are discussed.
Collapse
Affiliation(s)
| | | | - Libia Vega
- Department of Toxicology, CINVESTAV-IPN, Av. IPN 2508, C.P. 07360 Mexico City, Mexico
| | - Guillermo Elizondo
- Department of Cellular Biology, CINVESTAV-IPN, Av. IPN 2508, C.P. 07360 Mexico City, Mexico.
| |
Collapse
|
9
|
Mohsin M, Tabassum G, Ahmad S, Ali S, Ali Syed M. The role of mitophagy in pulmonary sepsis. Mitochondrion 2021; 59:63-75. [PMID: 33894359 DOI: 10.1016/j.mito.2021.04.009] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 04/13/2021] [Accepted: 04/19/2021] [Indexed: 12/16/2022]
Abstract
Sepsis is a systemic inflammatory disease with an unacceptably high mortality rate caused by an infection or trauma that involves both innate and adaptive immune systems. Inflammatory events activate different downstream pathways leading to tissue damage and ultimately multi-organ failure. Mitochondria are responsible for cellular energy, thermoregulation, metabolite biosynthesis, intracellular calcium regulation, and cell death. Damaged mitochondria induce the high Ca2+ influx through mitochondrial calcium uniporter (MCU). It also generates excessive Reactive oxygen species (ROS) and releases mtDNA into the cytoplasm, which causes induction of NLRP3 inflammasome and apoptosis. Mitophagy (Autophagy of damaged mitochondria) controls mitochondrial dynamics and function. It also maintains cellular homeostasis. This review is about how pulmonary sepsis affects the body. What is the aftermath of sepsis, and how mitophagy affects Acute Lung Injury and macrophage polarisation to overcome the damages.
Collapse
Affiliation(s)
- Mohd Mohsin
- Translational Research Lab, Department of Biotechnology, Jamia Millia Islamia, New Delhi 110025, India
| | - Gulnaz Tabassum
- Translational Research Lab, Department of Biotechnology, Jamia Millia Islamia, New Delhi 110025, India
| | - Shaniya Ahmad
- Translational Research Lab, Department of Biotechnology, Jamia Millia Islamia, New Delhi 110025, India
| | - Shakir Ali
- Department of Biochemistry, Jamia Hamdard, New Delhi 110019, India
| | - Mansoor Ali Syed
- Translational Research Lab, Department of Biotechnology, Jamia Millia Islamia, New Delhi 110025, India.
| |
Collapse
|
10
|
Perwez A, Wahabi K, Rizvi MA. Parkin: A targetable linchpin in human malignancies. Biochim Biophys Acta Rev Cancer 2021; 1876:188533. [PMID: 33785381 DOI: 10.1016/j.bbcan.2021.188533] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/21/2021] [Accepted: 03/21/2021] [Indexed: 12/16/2022]
Abstract
Parkin, an E3 ubiquitin ligase has been found to be deregulated in a variety of human cancers. Our current understanding is endowed with strong evidences that Parkin plays crucial role in the pathogenesis of cancer by controlling/interfering with major hallmarks of cancer delineated till today. Consistent with the idea of mitophagy, the existing studies imitates the tumor suppressive potential of Parkin, resolved by its capacity to regulate cell proliferation, cell migration, angiogenesis, apoptosis and overall cellular survival. Dysfunction of Parkin has resulted in the loss of ubiquitination of cell cycle components followed by their accumulation leading to genomic instability, perturbed cell cycle and eventually tumor progression. In this review, we provide an overview of current knowledge about the critical role of Parkin in cancer development and progression and have focussed on its therapeutic implications highlighting the diagnostic and prognostic value of Parkin as a biomarker. We earnestly hope that an in-depth knowledge of Parkin will provide a linchpin to target in various cancers that will open a new door of clinical applications and therapeutics.
Collapse
Affiliation(s)
- Ahmad Perwez
- Genome Biology Laboratory, Department of Biosciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Khushnuma Wahabi
- Genome Biology Laboratory, Department of Biosciences, Jamia Millia Islamia, New Delhi 110025, India
| | - Moshahid A Rizvi
- Genome Biology Laboratory, Department of Biosciences, Jamia Millia Islamia, New Delhi 110025, India.
| |
Collapse
|
11
|
White matter hyperintensities and the progression from mild parkinsonian signs to parkinsonism and Parkinson's disease. Neurobiol Aging 2020; 96:267-276. [PMID: 33130544 DOI: 10.1016/j.neurobiolaging.2020.08.005] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Revised: 08/01/2020] [Accepted: 08/03/2020] [Indexed: 12/11/2022]
Abstract
This study investigated the impact of white matter hyperintensities (WMHs) on the progression from mild parkinsonian signs (MPS) to parkinsonism and Parkinson's disease (PD). Participants with MPS completed 5 years of follow-up. WMHs were divided into periventricular WMHs and deep WMHs according to magnetic resonance imaging scans. The diagnosis of MPS, parkinsonism, and PD was based on the motor portion of the Unified Parkinson's Disease Rating Scale. Cox proportional hazard models were used to identify the association between WMHs and MPS progression. Of the 636 participants, 166 (26.1%) with MPS developed parkinsonism and PD after follow-up. After adjusting for potential factors, severe WMHs were associated with an increased risk of MPS progression, moderate and severe periventricular WMHs and severe deep WMHs were associated with the risk of MPS progression, and severe WMHs were associated with the progression of gait/balance impairment, bradykinesia, and rigidity. Additionally, participants treated for vascular risk factors such as hypertension, diabetes mellitus, and hypercholesterolemia had a lower risk of MPS progression.
Collapse
|
12
|
Wang H, Cheung F, Stoll AC, Rockwell P, Figueiredo-Pereira ME. Mitochondrial and calcium perturbations in rat CNS neurons induce calpain-cleavage of Parkin: Phosphatase inhibition stabilizes pSer 65Parkin reducing its calpain-cleavage. Biochim Biophys Acta Mol Basis Dis 2019; 1865:1436-1450. [PMID: 30796971 DOI: 10.1016/j.bbadis.2019.02.016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2018] [Revised: 02/04/2019] [Accepted: 02/18/2019] [Indexed: 02/07/2023]
Abstract
Mitochondrial impairment and calcium (Ca++) dyshomeostasis are associated with Parkinson's disease (PD). When intracellular ATP levels are lowered, Ca++-ATPase pumps are impaired causing cytoplasmic Ca++ to be elevated and calpain activation. Little is known about the effect of calpain activation on Parkin integrity. To address this gap, we examined the effects of mitochondrial inhibitors [oligomycin (Oligo), antimycin and rotenone] on endogenous Parkin integrity in rat midbrain and cerebral cortical cultures. All drugs induced calpain-cleavage of Parkin to ~36.9/43.6 kDa fragments. In contrast, treatment with the proinflammatory prostaglandin J2 (PGJ2) and the proteasome inhibitor epoxomicin induced caspase-cleavage of Parkin to fragments of a different size, previously shown by others to be triggered by apoptosis. Calpain-cleaved Parkin was enriched in neuronal mitochondrial fractions. Pre-treatment with the phosphatase inhibitor okadaic acid prior to Oligo-treatment, stabilized full-length Parkin phosphorylated at Ser65, and reduced calpain-cleavage of Parkin. Treatment with the Ca++ ionophore A23187, which facilitates Ca++ transport across the plasma membrane, mimicked the effect of Oligo by inducing calpain-cleavage of Parkin. Removing extracellular Ca++ from the media prevented oligomycin- and ionophore-induced calpain-cleavage of Parkin. Computational analysis predicted that calpain-cleavage of Parkin liberates its UbL domain. The phosphagen cyclocreatine moderately mitigated Parkin cleavage by calpain. Moreover, the pituitary adenylate cyclase activating peptide (PACAP27), which stimulates cAMP production, prevented caspase but not calpain-cleavage of Parkin. Overall, our data support a link between Parkin phosphorylation and its cleavage by calpain. This mechanism reflects the impact of mitochondrial impairment and Ca++-dyshomeostasis on Parkin integrity and could influence PD pathogenesis.
Collapse
Affiliation(s)
- Hu Wang
- Department of Biological Sciences, Hunter College and Graduate Center, City University of New York, NY 10065, USA
| | - Fanny Cheung
- Department of Biological Sciences, Hunter College and Graduate Center, City University of New York, NY 10065, USA
| | - Anna C Stoll
- Department of Biological Sciences, Hunter College and Graduate Center, City University of New York, NY 10065, USA
| | - Patricia Rockwell
- Department of Biological Sciences, Hunter College and Graduate Center, City University of New York, NY 10065, USA
| | - Maria E Figueiredo-Pereira
- Department of Biological Sciences, Hunter College and Graduate Center, City University of New York, NY 10065, USA.
| |
Collapse
|
13
|
Degradation of TRPML1 in Neurons Reduces Neuron Survival in Transient Global Cerebral Ischemia. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:4612727. [PMID: 30662583 PMCID: PMC6312622 DOI: 10.1155/2018/4612727] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/10/2018] [Revised: 09/28/2018] [Accepted: 10/15/2018] [Indexed: 11/17/2022]
Abstract
Postcardiac arrest syndrome yields poor neurological outcomes, but the mechanisms underlying this condition remain poorly understood. Autophagy plays an important role in neuronal apoptosis induced by ischemia. However, whether autophagy is involved in neuron apoptosis induced by cardiac arrest has been less studied. This study found that TRPML1 participates in cerebral ischemic reperfusion injury. Primary neurons were isolated and treated with mucolipin synthetic agonist 1 (ML-SA1), as well as infected with the recombinant lentivirus TRPML1 overexpression vector in vitro. ML-SA1 was delivered intracerebroventricularly in transient global ischemia model. Protein expression levels were determined by western blot. Neurological deficit score and the infarct volume were analyzed for the detection of neuronal damage. We found that TRPML1 was significantly downregulated in vivo and in vitro ischemic reperfusion model. We also observed that TRPML1 overexpression or treatment with the ML-SA1 attenuated neuronal death in primary neurons and ameliorated neurological dysfunction in vivo. Our findings suggested that autophagy and apoptosis were activated after transient global ischemia. Administration of ML-SA1 before transient global ischemia ameliorated neurological dysfunction possibly through the promotion of autophagy and the inhibition of apoptosis.
Collapse
|
14
|
Pecanka J, Jonker MA, Bochdanovits Z, Van Der Vaart AW. A powerful and efficient two-stage method for detecting gene-to-gene interactions in GWAS. Biostatistics 2018; 18:477-494. [PMID: 28334077 DOI: 10.1093/biostatistics/kxw060] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2016] [Accepted: 11/05/2016] [Indexed: 11/13/2022] Open
Abstract
For over a decade functional gene-to-gene interaction (epistasis) has been suspected to be a determinant in the "missing heritability" of complex traits. However, searching for epistasis on the genome-wide scale has been challenging due to the prohibitively large number of tests which result in a serious loss of statistical power as well as computational challenges. In this article, we propose a two-stage method applicable to existing case-control data sets, which aims to lessen both of these problems by pre-assessing whether a candidate pair of genetic loci is involved in epistasis before it is actually tested for interaction with respect to a complex phenotype. The pre-assessment is based on a two-locus genotype independence test performed in the sample of cases. Only the pairs of loci that exhibit non-equilibrium frequencies are analyzed via a logistic regression score test, thereby reducing the multiple testing burden. Since only the computationally simple independence tests are performed for all pairs of loci while the more demanding score tests are restricted to the most promising pairs, genome-wide association study (GWAS) for epistasis becomes feasible. By design our method provides strong control of the type I error. Its favourable power properties especially under the practically relevant misspecification of the interaction model are illustrated. Ready-to-use software is available. Using the method we analyzed Parkinson's disease in four cohorts and identified possible interactions within several SNP pairs in multiple cohorts.
Collapse
Affiliation(s)
- Jakub Pecanka
- Leiden University Medical Center, Department of Medical Statistics and Bioinformatics, Leiden, The Netherlands and VU University, Department of Mathematics, Amsterdam, the Netherlands
| | - Marianne A Jonker
- VU University Medical Center, Department of Epidemiology and Biostatistics, Amsterdam, The Netherlands and Radboud University medical center, Radboud Institute for Health Sciences, Nijmegen, The Netherlands
| | | | - Zoltan Bochdanovits
- VU University Medical Center, Department of Clinical Genetics, Amsterdam, The Netherlands
| | | |
Collapse
|
15
|
Hochrainer K. Protein Modifications with Ubiquitin as Response to Cerebral Ischemia-Reperfusion Injury. Transl Stroke Res 2017; 9:157-173. [DOI: 10.1007/s12975-017-0567-x] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 08/11/2017] [Accepted: 08/17/2017] [Indexed: 12/12/2022]
|
16
|
Yuan Y, Zheng Y, Zhang X, Chen Y, Wu X, Wu J, Shen Z, Jiang L, Wang L, Yang W, Luo J, Qin Z, Hu W, Chen Z. BNIP3L/NIX-mediated mitophagy protects against ischemic brain injury independent of PARK2. Autophagy 2017; 13:1754-1766. [PMID: 28820284 DOI: 10.1080/15548627.2017.1357792] [Citation(s) in RCA: 211] [Impact Index Per Article: 26.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022] Open
Abstract
Cerebral ischemia induces massive mitochondrial damage. These damaged mitochondria are cleared, thus attenuating brain injury, by mitophagy. Here, we identified the involvement of BNIP3L/NIX in cerebral ischemia-reperfusion (I-R)-induced mitophagy. Bnip3l knockout (bnip3l-/-) impaired mitophagy and aggravated cerebral I-R injury in mice, which can be rescued by BNIP3L overexpression. The rescuing effects of BNIP3L overexpression can be observed in park2-/- mice, which showed mitophagy deficiency after I-R. Interestingly, bnip3l and park2 double-knockout mice showed a synergistic mitophagy deficiency with I-R treatment, which further highlighted the roles of BNIP3L-mediated mitophagy as being independent from PARK2. Further experiments indicated that phosphorylation of BNIP3L serine 81 is critical for BNIP3L-mediated mitophagy. Nonphosphorylatable mutant BNIP3LS81A failed to counteract both mitophagy impairment and neuroprotective effects in bnip3l-/- mice. Our findings offer insights into mitochondrial quality control in ischemic stroke and bring forth the concept that BNIP3L could be a potential therapeutic target for ischemic stroke, beyond its accepted role in reticulocyte maturation.
Collapse
Affiliation(s)
- Yang Yuan
- a Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Department of Pharmacology, Key Laboratory of Medical Neurobiology of The Ministry of Health of China , Zhejiang University , Hangzhou , China
| | - Yanrong Zheng
- a Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Department of Pharmacology, Key Laboratory of Medical Neurobiology of The Ministry of Health of China , Zhejiang University , Hangzhou , China
| | - Xiangnan Zhang
- a Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Department of Pharmacology, Key Laboratory of Medical Neurobiology of The Ministry of Health of China , Zhejiang University , Hangzhou , China.,b Collaborative Innovation Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine , Zhejiang University , Hangzhou , China
| | - Ying Chen
- a Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Department of Pharmacology, Key Laboratory of Medical Neurobiology of The Ministry of Health of China , Zhejiang University , Hangzhou , China
| | - Xiaoli Wu
- a Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Department of Pharmacology, Key Laboratory of Medical Neurobiology of The Ministry of Health of China , Zhejiang University , Hangzhou , China
| | - Jiaying Wu
- a Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Department of Pharmacology, Key Laboratory of Medical Neurobiology of The Ministry of Health of China , Zhejiang University , Hangzhou , China
| | - Zhe Shen
- a Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Department of Pharmacology, Key Laboratory of Medical Neurobiology of The Ministry of Health of China , Zhejiang University , Hangzhou , China
| | - Lei Jiang
- a Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Department of Pharmacology, Key Laboratory of Medical Neurobiology of The Ministry of Health of China , Zhejiang University , Hangzhou , China
| | - Lu Wang
- a Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Department of Pharmacology, Key Laboratory of Medical Neurobiology of The Ministry of Health of China , Zhejiang University , Hangzhou , China
| | - Wei Yang
- a Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Department of Pharmacology, Key Laboratory of Medical Neurobiology of The Ministry of Health of China , Zhejiang University , Hangzhou , China
| | - Jianhong Luo
- a Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Department of Pharmacology, Key Laboratory of Medical Neurobiology of The Ministry of Health of China , Zhejiang University , Hangzhou , China
| | - Zhenghong Qin
- c Department of Pharmacology and Laboratory of Aging and Nervous Diseases, Jiangsu Key Laboratory of Translational Research and Therapy for Neuro-Psycho-Diseases , Soochow University School of Pharmaceutical Science , Suzhou , China
| | - Weiwei Hu
- a Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Department of Pharmacology, Key Laboratory of Medical Neurobiology of The Ministry of Health of China , Zhejiang University , Hangzhou , China.,b Collaborative Innovation Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine , Zhejiang University , Hangzhou , China
| | - Zhong Chen
- a Institute of Pharmacology & Toxicology, College of Pharmaceutical Sciences, Department of Pharmacology, Key Laboratory of Medical Neurobiology of The Ministry of Health of China , Zhejiang University , Hangzhou , China.,b Collaborative Innovation Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine , Zhejiang University , Hangzhou , China
| |
Collapse
|
17
|
Poststroke Induction of α-Synuclein Mediates Ischemic Brain Damage. J Neurosci 2017; 36:7055-65. [PMID: 27358461 DOI: 10.1523/jneurosci.1241-16.2016] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2016] [Accepted: 05/23/2016] [Indexed: 01/24/2023] Open
Abstract
UNLABELLED α-Synuclein (α-Syn), one of the most abundant proteins in the CNS, is known to be a major player in the neurodegeneration observed in Parkinson's disease. We currently report that transient focal ischemia upregulates α-Syn protein expression and nuclear translocation in neurons of the adult rodent brain. We further show that knockdown or knock-out of α-Syn significantly decreases the infarction and promotes better neurological recovery in rodents subjected to focal ischemia. Furthermore, α-Syn knockdown significantly reduced postischemic induction of phospho-Drp1, 3-nitrotyrosine, cleaved caspase-3, and LC-3 II/I, indicating its role in modulating mitochondrial fragmentation, oxidative stress, apoptosis, and autophagy, which are known to mediate poststroke neuronal death. Transient focal ischemia also significantly upregulated serine-129 (S129) phosphorylation (pα-Syn) of α-Syn and nuclear translocation of pα-Syn. Furthermore, knock-out mice that lack PLK2 (the predominant kinase that mediates S129 phosphorylation) showed better functional recovery and smaller infarcts when subjected to transient focal ischemia, indicating a detrimental role of S129 phosphorylation of α-Syn. In conclusion, our studies indicate that α-Syn is a potential therapeutic target to minimize poststroke brain damage. SIGNIFICANCE STATEMENT Abnormal aggregation of α-synuclein (α-Syn) has been known to cause Parkinson's disease and other chronic synucleinopathies. However, even though α-Syn is linked to pathophysiological mechanisms similar to those that produce acute neurodenegerative disorders, such as stroke, the role of α-Syn in such disorder is not clear. We presently studied whether α-Syn mediates poststroke brain damage and more importantly whether preventing α-Syn expression is neuroprotective and leads to better physiological and functional outcome after stroke. Our study indicates that α-Syn is a potential therapeutic target for stroke therapy.
Collapse
|
18
|
Kim T, Vemuganti R. Mechanisms of Parkinson's disease-related proteins in mediating secondary brain damage after cerebral ischemia. J Cereb Blood Flow Metab 2017; 37:1910-1926. [PMID: 28273718 PMCID: PMC5444552 DOI: 10.1177/0271678x17694186] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Both Parkinson's disease (PD) and stroke are debilitating conditions that result in neuronal death and loss of neurological functions. These two conditions predominantly affect aging populations with the deterioration of the quality of life for the patients themselves and a tremendous burden to families. While the neurodegeneration and symptomology of PD develop chronically over the years, post-stroke neuronal death and dysfunction develop rapidly in days. Despite the discrepancy in the pathophysiological time frame and severity, both conditions share common molecular mechanisms that include oxidative stress, mitochondrial dysfunction, inflammation, endoplasmic reticulum stress, and activation of various cell death pathways (apoptosis/necrosis/autophagy) that synergistically modulate the neuronal death. Emerging evidence indicates that several proteins associated with early-onset familial PD play critical roles in mediating the neuronal death. Importantly, mutations in the genes encoding Parkin, PTEN-induced putative kinase 1 and DJ-1 mediate autosomal recessive forms of PD, whereas mutations in the genes encoding leucine-rich repeat kinase 2 and α-synuclein are responsible for autosomal dominant PD. This review discusses the significance of these proteins with the emphasis on the role of α-synuclein in mediating post-ischemic brain damage.
Collapse
Affiliation(s)
- TaeHee Kim
- 1 Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA.,2 Neuroscience Training Program, Madison, WI, USA
| | - Raghu Vemuganti
- 1 Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA.,2 Neuroscience Training Program, Madison, WI, USA.,3 Cellular & Molecular Pathology Graduate Program, University of Wisconsin, Madison, WI, USA.,4 William S. Middleton Memorial Veterans Administration Hospital, Madison, WI, USA
| |
Collapse
|
19
|
Li H, Wu J, Shen H, Yao X, Liu C, Pianta S, Han J, Borlongan CV, Chen G. Autophagy in hemorrhagic stroke: Mechanisms and clinical implications. Prog Neurobiol 2017; 163-164:79-97. [PMID: 28414101 DOI: 10.1016/j.pneurobio.2017.04.002] [Citation(s) in RCA: 50] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Revised: 02/13/2017] [Accepted: 04/08/2017] [Indexed: 02/07/2023]
Abstract
Accumulating evidence advances the critical role of autophagy in brain pathology after stroke. Investigations employing autophagy induction or inhibition using pharmacological tools or autophagy-related gene knockout mice have recently revealed the biological significance of intact and functional autophagy in stroke. Most of the reported cases attest to a pro-survival role for autophagy in stroke, by facilitating removal of damaged proteins and organelles, which can be recycled for energy generation and cellular defenses. However, these observations are difficult to reconcile with equally compelling evidence demonstrating stroke-induced upregulation of brain cell death index that parallels enhanced autophagy. This begs the question of whether drug-induced autophagy during stroke culminates in improved or worsened pathological outcomes. A corollary fascinating hypothesis, but presents as a tricky conundrum, involves the effects of autophagy on cell death and inflammation, which are two main culprits in the disease progression of stroke-induced brain injury. Evidence has extended the roles of autophagy in inflammation via cytokine regulation in an unconventional secretion manner or by targeting inflammasomes for degradation. Moreover, in the recently concluded Vancouver Autophagy Symposium (VAS) held in 2014, the potential of selective autophagy for clinical treatment has been recognized. The role of autophagy in ischemic stroke has been reviewed previously in detail. Here, we evaluate the strength of laboratory and clinical evidence by providing a comprehensive summary of the literature on autophagy, and thereafter we offer our perspectives on exploiting autophagy as a drug target for cerebral ischemia, especially in hemorrhagic stroke.
Collapse
Affiliation(s)
- Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University,188 Shizi Street, Suzhou 215006, China
| | - Jiang Wu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University,188 Shizi Street, Suzhou 215006, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University,188 Shizi Street, Suzhou 215006, China
| | - Xiyang Yao
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University,188 Shizi Street, Suzhou 215006, China
| | - Chenglin Liu
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University,188 Shizi Street, Suzhou 215006, China
| | - S Pianta
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery & Brain Repair, University of South Florida Morsani College of Medicine,12901 Bruce B Downs Blvd Tampa, FL 33612 USA
| | - J Han
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery & Brain Repair, University of South Florida Morsani College of Medicine,12901 Bruce B Downs Blvd Tampa, FL 33612 USA
| | - C V Borlongan
- Center of Excellence for Aging and Brain Repair, Department of Neurosurgery & Brain Repair, University of South Florida Morsani College of Medicine,12901 Bruce B Downs Blvd Tampa, FL 33612 USA
| | - Gang Chen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University,188 Shizi Street, Suzhou 215006, China.
| |
Collapse
|
20
|
Lu Y, Chen W, Lin C, Wang J, Zhu M, Chen J, Miao C. The protective effects of propofol against CoCl 2-induced HT22 cell hypoxia injury via PP2A/CAMKIIα/nNOS pathway. BMC Anesthesiol 2017; 17:32. [PMID: 28241801 PMCID: PMC5329915 DOI: 10.1186/s12871-017-0327-1] [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: 10/11/2016] [Accepted: 02/20/2017] [Indexed: 02/07/2023] Open
Abstract
Background Perioperative cerebral ischemia/hypoxia could induce hippocampal injury and has been reported to induce cognitive impairment. In this study, we used cobalt chloride (CoCl2) to build a hypoxia model in mouse hippocampal cell lines. Propofol, a widely used intravenous anesthetic agent, has been demonstrated to have neuroprotective effect. Here, we explored whether and how propofol attenuated CoCl2-induced mouse hippocampal HT22 cell injury. Methods Mouse hippocampal HT22 cells were pretreated with propofol, and then stimulated with CoCl2. Cell viability was measured by cell counting kit 8 (CCK8). The effect of propofol on CoCl2-modulated expressions of B-cell lymphoma 2 (Bcl-2), BAX, cleaved caspase 3, phosphatase A2 (PP2A), and the phosphorylation of Ca2+/Calmodulin (CaM)-dependent protein kinase II (pCAMKIIα), neuron nitric oxide synthase at Ser1412 (pnNOS-Ser1412), neuron nitric oxide synthase at Ser847 (pnNOS-Ser847) were detected by Western blot analysis. Results Compared with control, CoCl2 treatment could significantly decrease cell viability, which could be reversed by propofol. Further, we found CoCl2 treatment could up-regulate the expression of PP2A, BAX, cleaved caspase three and cause the phosphorylation of nNOS-Ser1412, but it down-regulated the expression of Bcl-2 and the phosphorylation of CAMKIIα and nNOS-Ser847. More importantly, these CoCl2-mediated effects were attentuated by propofol. In addition, we demonstrated that propofol could exert similar effect to that of the PP2A inhibitor (okadaic acid). Further, the PP2A activator (FTY720) and the CAMKIIα inhibitor (KN93) could reverse the neuroprotective effect of propofol. Conclusion Our data indicated that propofol could attenuate CoCl2-induced HT22 cells hypoxia injury via PP2A/CAMKIIα/nNOS pathway.
Collapse
Affiliation(s)
- Yan Lu
- Department of Anesthesiology, Fudan University Shanghai Cancer Centre, No. 270 DongAn Road, Shanghai, 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Wei Chen
- Department of Anesthesiology, Fudan University Shanghai Cancer Centre, No. 270 DongAn Road, Shanghai, 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Chen Lin
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China.,Department of Medical Oncology, Fudan University Shanghai Cancer Centre, No. 270 DongAn Road, Shanghai, 200032, People's Republic of China
| | - Jiaqiang Wang
- Department of Anesthesiology, Fudan University Shanghai Cancer Centre, No. 270 DongAn Road, Shanghai, 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Minmin Zhu
- Department of Anesthesiology, Fudan University Shanghai Cancer Centre, No. 270 DongAn Road, Shanghai, 200032, People's Republic of China.,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China
| | - Jiawei Chen
- Department of Anesthesiology, Fudan University Shanghai Cancer Centre, No. 270 DongAn Road, Shanghai, 200032, People's Republic of China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China.
| | - Changhong Miao
- Department of Anesthesiology, Fudan University Shanghai Cancer Centre, No. 270 DongAn Road, Shanghai, 200032, People's Republic of China. .,Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, 200032, People's Republic of China.
| |
Collapse
|
21
|
Abstract
Ischemic disorders, such as myocardial infarction, stroke, and peripheral vascular disease, are the most common causes of debilitating disease and death in westernized cultures. The extent of tissue injury relates directly to the extent of blood flow reduction and to the length of the ischemic period, which influence the levels to which cellular ATP and intracellular pH are reduced. By impairing ATPase-dependent ion transport, ischemia causes intracellular and mitochondrial calcium levels to increase (calcium overload). Cell volume regulatory mechanisms are also disrupted by the lack of ATP, which can induce lysis of organelle and plasma membranes. Reperfusion, although required to salvage oxygen-starved tissues, produces paradoxical tissue responses that fuel the production of reactive oxygen species (oxygen paradox), sequestration of proinflammatory immunocytes in ischemic tissues, endoplasmic reticulum stress, and development of postischemic capillary no-reflow, which amplify tissue injury. These pathologic events culminate in opening of mitochondrial permeability transition pores as a common end-effector of ischemia/reperfusion (I/R)-induced cell lysis and death. Emerging concepts include the influence of the intestinal microbiome, fetal programming, epigenetic changes, and microparticles in the pathogenesis of I/R. The overall goal of this review is to describe these and other mechanisms that contribute to I/R injury. Because so many different deleterious events participate in I/R, it is clear that therapeutic approaches will be effective only when multiple pathologic processes are targeted. In addition, the translational significance of I/R research will be enhanced by much wider use of animal models that incorporate the complicating effects of risk factors for cardiovascular disease. © 2017 American Physiological Society. Compr Physiol 7:113-170, 2017.
Collapse
Affiliation(s)
- Theodore Kalogeris
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
| | - Christopher P. Baines
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
- Department of Biomedical Sciences, University of Missouri College of Veterinary Medicine, Columbia, Missouri, USA
| | - Maike Krenz
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| | - Ronald J. Korthuis
- Department of Medical Pharmacology and Physiology, University of Missouri School of Medicine, Columbia, Missouri, USA
- Dalton Cardiovascular Research Center, University of Missouri, Columbia, Missouri, USA
| |
Collapse
|
22
|
Abstract
Mitochondria play a key role in various cell processes including ATP production, Ca2+ homeostasis, reactive oxygen species (ROS) generation, and apoptosis. The selective removal of impaired mitochondria by autophagosome is known as mitophagy. Cerebral ischemia is a common form of stroke caused by insufficient blood supply to the brain. Emerging evidence suggests that mitophagy plays important roles in the pathophysiological process of cerebral ischemia. This review focuses on the relationship between ischemic brain injury and mitophagy. Based on the latest research, it describes how the signaling pathways of mitophagy appear to be involved in cerebral ischemia.
Collapse
|
23
|
Parkin Protects against Oxygen-Glucose Deprivation/Reperfusion Insult by Promoting Drp1 Degradation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:8474303. [PMID: 27597885 PMCID: PMC5002297 DOI: 10.1155/2016/8474303] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/27/2016] [Revised: 06/13/2016] [Accepted: 07/11/2016] [Indexed: 11/17/2022]
Abstract
Ischemic stroke results in severe brain damage and remains one of the leading causes of death and disability worldwide. Effective neuroprotective therapies are needed to reduce brain damage resulting from ischemic stroke. Mitochondria are crucial for cellular energy production and homeostasis. Modulation of mitochondrial function mediates neuroprotection against ischemic brain damage. Dynamin-related protein 1 (Drp1) and parkin play a key role in regulating mitochondrial dynamics. They are potential therapeutic targets for neuroprotection in ischemic stroke. Protective effects of parkin-Drp1 pathway on mitochondria were assessed in a cellular ischemia-reperfusion injury model. Mouse neuroblastoma Neuro2a (N2a) cells were subjected to oxygen-glucose deprivation/reperfusion (OGDR) insult. OGDR induces mitochondrial fragmentation. The expression of Drp1 protein is increased after OGDR insult, while the parkin protein level is decreased. The altered protein level of Drp1 after OGDR injury is mediated by parkin through ubiquitin proteasome system (UPS). Drp1 depletion protects against OGDR induced mitochondrial damage and apoptosis. Meanwhile, parkin overexpression protects against OGDR induced apoptosis and mitochondrial dysfunction, which is attenuated by increased expression of Drp1. Our data demonstrate that parkin protects against OGDR insult through promoting degradation of Drp1. This neuroprotective potential of parkin-Drp1 pathway against OGDR insult will pave the way for developing novel neuroprotective agents for cerebral ischemia-reperfusion related disorders.
Collapse
|
24
|
Gong B, Radulovic M, Figueiredo-Pereira ME, Cardozo C. The Ubiquitin-Proteasome System: Potential Therapeutic Targets for Alzheimer's Disease and Spinal Cord Injury. Front Mol Neurosci 2016; 9:4. [PMID: 26858599 PMCID: PMC4727241 DOI: 10.3389/fnmol.2016.00004] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2015] [Accepted: 01/07/2016] [Indexed: 01/20/2023] Open
Abstract
The ubiquitin-proteasome system (UPS) is a crucial protein degradation system in eukaryotes. Herein, we will review advances in the understanding of the role of several proteins of the UPS in Alzheimer’s disease (AD) and functional recovery after spinal cord injury (SCI). The UPS consists of many factors that include E3 ubiquitin ligases, ubiquitin hydrolases, ubiquitin and ubiquitin-like molecules, and the proteasome itself. An extensive body of work links UPS dysfunction with AD pathogenesis and progression. More recently, the UPS has been shown to have vital roles in recovery of function after SCI. The ubiquitin hydrolase (Uch-L1) has been proposed to increase cellular levels of mono-ubiquitin and hence to increase rates of protein turnover by the UPS. A low Uch-L1 level has been linked with Aβ accumulation in AD and reduced neuroregeneration after SCI. One likely mechanism for these beneficial effects of Uch-L1 is reduced turnover of the PKA regulatory subunit and consequently, reduced signaling via CREB. The neuron-specific F-box protein Fbx2 ubiquitinates β-secretase thus targeting it for proteasomal degradation and reducing generation of Aβ. Both Uch-L1 and Fbx2 improve synaptic plasticity and cognitive function in mouse AD models. The role of Fbx2 after SCI has not been examined, but abolishing ß-secretase reduces neuronal recovery after SCI, associated with reduced myelination. UBB+1, which arises through a frame-shift mutation in the ubiquitin gene that adds 19 amino acids to the C-terminus of ubiquitin, inhibits proteasomal function and is associated with increased neurofibrillary tangles in patients with AD, Pick’s disease and Down’s syndrome. These advances in understanding of the roles of the UPS in AD and SCI raise new questions but, also, identify attractive and exciting targets for potential, future therapeutic interventions.
Collapse
Affiliation(s)
- Bing Gong
- Department of Medicine, Mount Sinai School of MedicineNew York, NY, USA; Medicine, James J. Peters Veteran Affairs Medical CenterBronx, NY, USA
| | - Miroslav Radulovic
- Department of Medicine, Mount Sinai School of MedicineNew York, NY, USA; Medicine, James J. Peters Veteran Affairs Medical CenterBronx, NY, USA; National Center of Excellence for the Medical Consequences of Spinal Cord Injury (SCI)Bronx, NY, USA
| | - Maria E Figueiredo-Pereira
- Department of Biological Sciences, Hunter College, and the Graduate School and University Center, The City University of New York New York, NY, USA
| | - Christopher Cardozo
- Department of Medicine, Mount Sinai School of MedicineNew York, NY, USA; Medicine, James J. Peters Veteran Affairs Medical CenterBronx, NY, USA; National Center of Excellence for the Medical Consequences of Spinal Cord Injury (SCI)Bronx, NY, USA
| |
Collapse
|
25
|
Li H, Qiu S, Li X, Li M, Peng Y. Autophagy biomarkers in CSF correlates with infarct size, clinical severity and neurological outcome in AIS patients. J Transl Med 2015; 13:359. [PMID: 26576535 PMCID: PMC4650838 DOI: 10.1186/s12967-015-0726-3] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 11/03/2015] [Indexed: 12/12/2022] Open
Abstract
BACKGROUND Autophagy is demonstrated to be involved in acute ischemic stroke(AIS), which, however, is confined to cells and/or animals levels. The aim of this study was to determine two autophagy biomarkers, Beclin1 and LC3B, in cerebrospinal fluid (CSF) and serum of patients with AIS, and to evaluate a possible correlation between levels of Beclin1 and LC3B and severity of neurological deficit and clinical outcome of stroke patients. METHODS Levels of Beclin1 and LC3B were quantified by ELISA in CSF and serum collected from 37 AIS patients and 21 controls. The clinical severity at stroke onset was determined by the National Institute of Health Stroke Scale (NIHSS) and the neurological outcome was determined by the Modified Rankin Scale (mRs) and the improvement in NIHSS between stroke onset and 3 months later. Associations between autophagy biomarkers and infarct volume, NIHSS and mRs were assessed using Pearson analysis. RESULTS The levels of Beclin1 and LC3B were increased both in CSF and serum of AIS patients relative to controls. In CSF, they were positively correlated with infarct volume and NIHSS scores, and negatively correlated with mRs scores, but no significant association was observed in serum. Moreover, AIS patients with higher levels of Beclin1 and LC3B in CSF had significantly higher improvement in NIHSS. CONCLUSION CSF and serum levels of autophagy biomarkers are altered in AIS patients. CSF levels of autophagy biomarkers are associated with infarct volume, clinical severity of and neurological outcome.
Collapse
Affiliation(s)
- Honghong Li
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 West Yanjiang Road, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| | - Shuwei Qiu
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 West Yanjiang Road, Guangzhou, 510120, China.
- Department of Neurology, the First Affiliated Hospital of Soochow University, Suzhou, China.
| | - Xiangpen Li
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 West Yanjiang Road, Guangzhou, 510120, China.
| | - Mei Li
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 West Yanjiang Road, Guangzhou, 510120, China.
| | - Ying Peng
- Department of Neurology, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, No. 107 West Yanjiang Road, Guangzhou, 510120, China.
- Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou, China.
| |
Collapse
|
26
|
Palubinsky AM, Stankowski JN, Kale AC, Codreanu SG, Singer RJ, Liebler DC, Stanwood GD, McLaughlin B. CHIP Is an Essential Determinant of Neuronal Mitochondrial Stress Signaling. Antioxid Redox Signal 2015; 23:535-49. [PMID: 25602369 PMCID: PMC4544748 DOI: 10.1089/ars.2014.6102] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
AIMS Determine the mechanism by which C-terminus of HSC70-interacting protein (CHIP) induction alters neuronal survival under conditions of mitochondrial stress induced by oxygen glucose deprivation. RESULTS We report that animals deficient in the E3 ubiquitin ligase, CHIP, have high baseline levels of central nervous system protein oxidation and lipid peroxidation, reduced antioxidant defenses, and decreased energetic status. Stress-associated molecules typically linked to Parkinson's disease such as the mitochondrial kinase, PTEN-inducible putative kinase 1 (PINK1), and another E3 ligase, Parkin, are upregulated in brains from CHIP knockout (KO) animals. Utilizing a novel biotin-avidin capture technique, we found that the oxidation status of Parkin and the mitochondrial fission protein, dynamin-related protein 1 (Drp1), are altered in a CHIP-dependent manner. We also found that following oxygen-glucose deprivation (OGD), the expression of CHIP, PINK1, and the autophagic marker, LC3, increase and there is activation of the redox-sensitive kinase p66(shc). Under conditions of OGD, CHIP relocalizes from the cytosol to mitochondria. Mitochondria from CHIP KO mice have profound impairments in stress response induced by calcium overload, resulting in accelerated permeability transition activity. While CHIP-deficient neurons are morphologically intact, they are more susceptible to OGD consistent with a previously unknown neuroprotective role for CHIP in maintaining mitochondrial homeostasis. INNOVATION CHIP relocalization to the mitochondria is essential for the regulation of mitochondrial integrity and neuronal survival following OGD. CONCLUSIONS CHIP is an essential regulator of neuronal bioenergetics and redox tone. Altering the expression of this protein has profound effects on neuronal survival when cells are exposed to OGD.
Collapse
Affiliation(s)
- Amy M Palubinsky
- 1 Neuroscience Graduate Program, Vanderbilt Brain Institute, Vanderbilt University , Nashville, Tennessee.,2 Clinical Neuroscience Scholars Program, Vanderbilt University , Nashville, Tennessee.,3 J.B. Marshall Laboratory for Neurovascular Therapeutics, Vanderbilt University , Nashville, Tennessee.,4 Kennedy Center for Research on Human Development, Vanderbilt University , Nashville, Tennessee
| | - Jeannette N Stankowski
- 1 Neuroscience Graduate Program, Vanderbilt Brain Institute, Vanderbilt University , Nashville, Tennessee.,4 Kennedy Center for Research on Human Development, Vanderbilt University , Nashville, Tennessee.,5 Department of Neuroscience, Mayo Clinic , Jacksonville, Florida
| | - Alixandra C Kale
- 3 J.B. Marshall Laboratory for Neurovascular Therapeutics, Vanderbilt University , Nashville, Tennessee.,4 Kennedy Center for Research on Human Development, Vanderbilt University , Nashville, Tennessee.,6 Department of Neurology, Vanderbilt University , Nashville, Tennessee
| | - Simona G Codreanu
- 7 Department of Biochemistry, Vanderbilt University , Nashville, Tennessee.,8 Department of Biomedical Informatics, Vanderbilt University , Nashville, Tennessee
| | - Robert J Singer
- 3 J.B. Marshall Laboratory for Neurovascular Therapeutics, Vanderbilt University , Nashville, Tennessee.,9 Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University , Nashville, Tennessee.,10 Department of Neurosurgery, Dartmouth Hitchcock Hospital , Lebanon , New Hampshire
| | - Daniel C Liebler
- 7 Department of Biochemistry, Vanderbilt University , Nashville, Tennessee.,8 Department of Biomedical Informatics, Vanderbilt University , Nashville, Tennessee
| | - Gregg D Stanwood
- 4 Kennedy Center for Research on Human Development, Vanderbilt University , Nashville, Tennessee.,11 Department of Pharmacology, Vanderbilt University , Nashville, Tennessee
| | - BethAnn McLaughlin
- 3 J.B. Marshall Laboratory for Neurovascular Therapeutics, Vanderbilt University , Nashville, Tennessee.,4 Kennedy Center for Research on Human Development, Vanderbilt University , Nashville, Tennessee.,6 Department of Neurology, Vanderbilt University , Nashville, Tennessee.,9 Vanderbilt Institute for Integrative Biosystems Research and Education, Vanderbilt University , Nashville, Tennessee.,11 Department of Pharmacology, Vanderbilt University , Nashville, Tennessee
| |
Collapse
|
27
|
Abstract
The selective degradation of damaged or excessive mitochondria by autophagy is termed mitophagy. Mitophagy is crucial for mitochondrial quality control and has been implicated in several neurodegenerative disorders as well as in ischemic brain injury. Emerging evidence suggested that the role of mitophagy in cerebral ischemia may depend on different pathological processes. In particular, a neuroprotective role of mitophagy has been proposed, and the regulation of mitophagy seems to be important in cell survival. For these reasons, extensive investigations aimed to profile the mitophagy process and its underlying molecular mechanisms have been executed in recent years. In this review, we summarize the current knowledge regarding the mitophagy process and its role in cerebral ischemia, and focus on the pathological events and molecules that regulate mitophagy in ischemic brain injury.
Collapse
Affiliation(s)
- Yang Yuan
- Department of Pharmacology, Key Laboratory of Medical Neurobiology (Ministry of Health of China), College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, 310058, China
| | | | | | | |
Collapse
|
28
|
Li F, Gao B, Dong H, Shi J, Fang D. Icariin induces synoviolin expression through NFE2L1 to protect neurons from ER stress-induced apoptosis. PLoS One 2015; 10:e0119955. [PMID: 25806530 PMCID: PMC4373914 DOI: 10.1371/journal.pone.0119955] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2014] [Accepted: 12/26/2014] [Indexed: 11/19/2022] Open
Abstract
By suppressing neuronal apoptosis, Icariin is a potential therapeutic drug for neuronal degenerative diseases. The molecular mechanisms of Icariin anti-apoptotic functions are still largely unclear. In this report, we found that Icariin induces the expression of Synoviolin, an endoplasmic reticulum (ER)-anchoring E3 ubiquitin ligase that functions as a suppressor of ER stress-induced apoptosis. The nuclear factor erythroid 2-related factor 1 (NFE2L1) is responsible for Icariin-mediated Synoviolin gene expression. Mutation of the NFE2L1-binding sites in a distal region of the Synoviolin promoter abolished Icariin-induced Synoviolin promoter activity, and knockdown of NFE2L1 expression prevented Icariin-stimulated Synoviolin expression. More importantly, Icariin protected ER stress-induced apoptosis of PC12 cells in a Synoviolin-dependent manner. Therefore, our study reveals Icariin-induced Synoviolin expression through NFE2L1 as a previously unappreciated molecular mechanism underlying the neuronal protective function of Icariin.
Collapse
Affiliation(s)
- Fei Li
- Department of Pharmacology and the Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical College, Zunyi, China
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave, Chicago, IL, 60611, United States of America
- * E-mail: (FL); (DF)
| | - Beixue Gao
- Department of Pathology, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave, Chicago, IL, 60611, United States of America
| | - Hongxin Dong
- Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave, Chicago, IL, 60611, United States of America
| | - Jingshan Shi
- Department of Pharmacology and the Key Laboratory of Basic Pharmacology of Guizhou Province, Zunyi Medical College, Zunyi, China
| | - Deyu Fang
- Department of Pathology, Northwestern University Feinberg School of Medicine, 303 E. Chicago Ave, Chicago, IL, 60611, United States of America
| |
Collapse
|
29
|
Romero JI, Hanschmann EM, Gellert M, Eitner S, Holubiec MI, Blanco-Calvo E, Lillig CH, Capani F. Thioredoxin 1 and glutaredoxin 2 contribute to maintain the phenotype and integrity of neurons following perinatal asphyxia. Biochim Biophys Acta Gen Subj 2015; 1850:1274-85. [PMID: 25735211 DOI: 10.1016/j.bbagen.2015.02.015] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2014] [Revised: 02/14/2015] [Accepted: 02/24/2015] [Indexed: 01/23/2023]
Abstract
BACKGROUND Thioredoxin (Trx) family proteins are crucial mediators of cell functions via regulation of the thiol redox state of various key proteins and the levels of the intracellular second messenger hydrogen peroxide. Their expression, localization and functions are altered in various pathologies. Here, we have analyzed the impact of Trx family proteins in neuronal development and recovery, following hypoxia/ischemia and reperfusion. METHODS We have analyzed the regulation and potential functions of Trx family proteins during hypoxia/ischemia and reoxygenation of the developing brain in both an animal and a cellular model of perinatal asphyxia. We have analyzed the distribution of 14 Trx family and related proteins in the cerebellum, striatum, and hippocampus, three areas of the rat brain that are especially susceptible to hypoxia. Using SH-SY5Y cells subjected to hypoxia and reoxygenation, we have analyzed the functions of some redoxins suggested by the animal experiment. RESULTS AND CONCLUSIONS We have described/discovered a complex, cell-type and tissue-specific expression pattern following the hypoxia/ischemia and reoxygenation. Particularly, Grx2 and Trx1 showed distinct changes during tissue recovery following hypoxia/ischemia and reoxygenation. Silencing of these proteins in SH-SY5Y cells subjected to hypoxia-reoxygenation confirmed that these proteins are required to maintain the normal neuronal phenotype. GENERAL SIGNIFICANCE These findings demonstrate the significance of redox signaling in cellular pathways. Grx2 and Trx1 contribute significantly to neuronal integrity and could be clinically relevant in neuronal damage following perinatal asphyxia and other neuronal disorders.
Collapse
Affiliation(s)
- Juan Ignacio Romero
- Instituto de Investigaciones Cardiológicas "Prof. Dr. Alberto C. Taquini" (ININCA), Facultad de Medicina, UBA-CONICET, Marcelo T. de Alvear 2270, C1122AAJ, Ciudad de Buenos Aires, Argentina
| | - Eva-Maria Hanschmann
- Institute for Medical Biochemistry and Molecular Biology, Universitätsmedizin Greifswald, Ernst-Moritz-Arndt-Universität Greifswald, 17475 Greifswald, Germany
| | - Manuela Gellert
- Institute for Medical Biochemistry and Molecular Biology, Universitätsmedizin Greifswald, Ernst-Moritz-Arndt-Universität Greifswald, 17475 Greifswald, Germany
| | - Susanne Eitner
- Institute for Medical Biochemistry and Molecular Biology, Universitätsmedizin Greifswald, Ernst-Moritz-Arndt-Universität Greifswald, 17475 Greifswald, Germany
| | - Mariana Inés Holubiec
- Instituto de Investigaciones Cardiológicas "Prof. Dr. Alberto C. Taquini" (ININCA), Facultad de Medicina, UBA-CONICET, Marcelo T. de Alvear 2270, C1122AAJ, Ciudad de Buenos Aires, Argentina
| | - Eduardo Blanco-Calvo
- Instituto de Investigaciones Cardiológicas "Prof. Dr. Alberto C. Taquini" (ININCA), Facultad de Medicina, UBA-CONICET, Marcelo T. de Alvear 2270, C1122AAJ, Ciudad de Buenos Aires, Argentina; Facultat d'Educació, Psicologia i Treball Social Universitat de Lleida Av. de l'Estudi General, 4, 25001 Lleida, Spain
| | - Christopher Horst Lillig
- Institute for Medical Biochemistry and Molecular Biology, Universitätsmedizin Greifswald, Ernst-Moritz-Arndt-Universität Greifswald, 17475 Greifswald, Germany
| | - Francisco Capani
- Instituto de Investigaciones Cardiológicas "Prof. Dr. Alberto C. Taquini" (ININCA), Facultad de Medicina, UBA-CONICET, Marcelo T. de Alvear 2270, C1122AAJ, Ciudad de Buenos Aires, Argentina; Departamento de Biología, UAJFK, C1197AAR, Ciudad de Buenos Aires, Argentina.
| |
Collapse
|
30
|
Zhang X, Yuan Y, Jiang L, Zhang J, Gao J, Shen Z, Zheng Y, Deng T, Yan H, Li W, Hou WW, Lu J, Shen Y, Dai H, Hu WW, Zhang Z, Chen Z. Endoplasmic reticulum stress induced by tunicamycin and thapsigargin protects against transient ischemic brain injury: Involvement of PARK2-dependent mitophagy. Autophagy 2014; 10:1801-13. [PMID: 25126734 PMCID: PMC4198364 DOI: 10.4161/auto.32136] [Citation(s) in RCA: 218] [Impact Index Per Article: 19.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
Transient cerebral ischemia leads to endoplasmic reticulum (ER) stress. However, the contributions of ER stress to cerebral ischemia are not clear. To address this issue, the ER stress activators tunicamycin (TM) and thapsigargin (TG) were administered to transient middle cerebral artery occluded (tMCAO) mice and oxygen-glucose deprivation-reperfusion (OGD-Rep.)-treated neurons. Both TM and TG showed significant protection against ischemia-induced brain injury, as revealed by reduced brain infarct volume and increased glucose uptake rate in ischemic tissue. In OGD-Rep.-treated neurons, 4-PBA, the ER stress releasing mechanism, counteracted the neuronal protection of TM and TG, which also supports a protective role of ER stress in transient brain ischemia. Knocking down the ER stress sensor Eif2s1, which is further activated by TM and TG, reduced the OGD-Rep.-induced neuronal cell death. In addition, both TM and TG prevented PARK2 loss, promoted its recruitment to mitochondria, and activated mitophagy during reperfusion after ischemia. The neuroprotection of TM and TG was reversed by autophagy inhibition (3-methyladenine and Atg7 knockdown) as well as Park2 silencing. The neuroprotection was also diminished in Park2(+/-) mice. Moreover, Eif2s1 and downstream Atf4 silencing reduced PARK2 expression, impaired mitophagy induction, and counteracted the neuroprotection. Taken together, the present investigation demonstrates that the ER stress induced by TM and TG protects against the transient ischemic brain injury. The PARK2-mediated mitophagy may be underlying the protection of ER stress. These findings may provide a new strategy to rescue ischemic brains by inducing mitophagy through ER stress activation.
Collapse
Affiliation(s)
- Xiangnan Zhang
- Department of Pharmacology; Key Laboratory of Medical Neurobiology of the Ministry of Health of China; Zhejiang Province Key Laboratory of Neurobiology; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou, China
- Collaborative Innovation Center for Infectious Diseases; Zhejiang University; Hangzhou, China
| | - Yang Yuan
- Department of Pharmacology; Key Laboratory of Medical Neurobiology of the Ministry of Health of China; Zhejiang Province Key Laboratory of Neurobiology; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou, China
| | - Lei Jiang
- Department of Pharmacology; Key Laboratory of Medical Neurobiology of the Ministry of Health of China; Zhejiang Province Key Laboratory of Neurobiology; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou, China
| | - Jingying Zhang
- Department of Pharmacology; Key Laboratory of Medical Neurobiology of the Ministry of Health of China; Zhejiang Province Key Laboratory of Neurobiology; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou, China
| | - Jieqiong Gao
- Zhejiang Provincial Key Laboratory of Medical Genetics; School of Life Sciences; Wenzhou Medical College; Wenzhou, China
| | - Zhe Shen
- Department of Pharmacology; Key Laboratory of Medical Neurobiology of the Ministry of Health of China; Zhejiang Province Key Laboratory of Neurobiology; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou, China
| | - Yanrong Zheng
- Department of Pharmacology; Key Laboratory of Medical Neurobiology of the Ministry of Health of China; Zhejiang Province Key Laboratory of Neurobiology; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou, China
| | - Tian Deng
- Department of Pharmacology; Key Laboratory of Medical Neurobiology of the Ministry of Health of China; Zhejiang Province Key Laboratory of Neurobiology; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou, China
| | - Haijing Yan
- Department of Pharmacology; Key Laboratory of Medical Neurobiology of the Ministry of Health of China; Zhejiang Province Key Laboratory of Neurobiology; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou, China
| | - Wenlu Li
- Department of Pharmacology; Key Laboratory of Medical Neurobiology of the Ministry of Health of China; Zhejiang Province Key Laboratory of Neurobiology; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou, China
- Department of Pharmacy; the Second Affiliated Hospital; School of Medicine; Zhejiang University; Hangzhou, China
| | - Wei-Wei Hou
- Department of Pharmacology; Key Laboratory of Medical Neurobiology of the Ministry of Health of China; Zhejiang Province Key Laboratory of Neurobiology; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou, China
| | - Jianxin Lu
- Zhejiang Provincial Key Laboratory of Medical Genetics; School of Life Sciences; Wenzhou Medical College; Wenzhou, China
| | - Yao Shen
- Zhejiang Provincial Key Laboratory of Medical Genetics; School of Life Sciences; Wenzhou Medical College; Wenzhou, China
| | - Haibing Dai
- Department of Pharmacy; the Second Affiliated Hospital; School of Medicine; Zhejiang University; Hangzhou, China
| | - Wei-Wei Hu
- Department of Pharmacology; Key Laboratory of Medical Neurobiology of the Ministry of Health of China; Zhejiang Province Key Laboratory of Neurobiology; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou, China
- Collaborative Innovation Center for Infectious Diseases; Zhejiang University; Hangzhou, China
| | - Zhuohua Zhang
- State Key Laboratory of Medical Genetics; Central South University; Changsha, China
| | - Zhong Chen
- Department of Pharmacology; Key Laboratory of Medical Neurobiology of the Ministry of Health of China; Zhejiang Province Key Laboratory of Neurobiology; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou, China
- Collaborative Innovation Center for Infectious Diseases; Zhejiang University; Hangzhou, China
- Correspondence to: Zhong Chen,
| |
Collapse
|
31
|
Zhang X, Yan H, Yuan Y, Gao J, Shen Z, Cheng Y, Shen Y, Wang RR, Wang X, Hu WW, Wang G, Chen Z. Cerebral ischemia-reperfusion-induced autophagy protects against neuronal injury by mitochondrial clearance. Autophagy 2013; 9:1321-33. [PMID: 23800795 DOI: 10.4161/auto.25132] [Citation(s) in RCA: 413] [Impact Index Per Article: 34.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
Cerebral ischemia-reperfusion (I-R) is a complex pathological process. Although autophagy can be evoked by ischemia, its involvement in the reperfusion phase after ischemia and its contribution to the fate of neurons remains largely unknown. In the present investigation, we found that autophagy was activated in the reperfusion phase, as revealed in both mice with middle cerebral artery occlusion and oxygen-glucose deprived cortical neurons in culture. Interestingly, in contrast to that in permanent ischemia, inhibition of autophagy (by 3-methyladenine, bafilomycin A 1, Atg7 knockdown or in atg5(-/-) MEF cells) in the reperfusion phase reinforced, rather than reduced, the brain and cell injury induced by I-R. Inhibition of autophagy either with 3-methyladenine or Atg7 knockdown enhanced the I-R-induced release of cytochrome c and the downstream activation of apoptosis. Moreover, MitoTracker Red-labeled neuronal mitochondria increasingly overlapped with GFP-LC3-labeled autophagosomes during reperfusion, suggesting the presence of mitophagy. The mitochondrial clearance in I-R was reversed by 3-methyladenine and Atg7 silencing, further suggesting that mitophagy underlies the neuroprotection by autophagy. In support, administration of the mitophagy inhibitor mdivi-1 in the reperfusion phase aggravated the ischemia-induced neuronal injury both in vivo and in vitro. PARK2 translocated to mitochondria during reperfusion and Park2 knockdown aggravated ischemia-induced neuronal cell death. In conclusion, the results indicated that autophagy plays different roles in cerebral ischemia and subsequent reperfusion. The protective role of autophagy during reperfusion may be attributable to mitophagy-related mitochondrial clearance and inhibition of downstream apoptosis. PARK2 may be involved in the mitophagy process.
Collapse
Affiliation(s)
- Xiangnan Zhang
- Department of Pharmacology; Key Laboratory of Medical Neurobiology of The Ministry of Health of China; Zhejiang Province Key Laboratory of Neurobiology; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Mitochondria play a central role in cell fate after stressors such as ischemic brain injury. The convergence of intracellular signaling pathways on mitochondria and their release of critical factors are now recognized as a default conduit to cell death or survival. Besides the individual processes that converge on or emanate from mitochondria, a mitochondrial organellar response to changes in the cellular environment has recently been described. Whereas mitochondria have previously been perceived as a major center for cellular signaling, one can postulate that the organelle's dynamics themselves affect cell survival. This brief perspective review puts forward the concept that disruptions in mitochondrial dynamics--biogenesis, clearance, and fission/fusion events--may underlie neural diseases and thus could be targeted as neuroprotective strategies in the context of ischemic injury. To do so, we present a general overview of the current understanding of mitochondrial dynamics and regulation. We then review emerging studies that correlate mitochondrial biogenesis, mitophagy, and fission/fusion events with neurologic disease and recovery. An overview of the system as it is currently understood is presented, and current assessment strategies and their limitations are discussed.
Collapse
|
33
|
Grimaldi M, Romer I, de Apodaca MTG, Iturbe L, Catania ID, González J, Kolliker-Fres R, Barreto G, Capani F. Early changes in the synapses of the neostriatum induced by perinatal asphyxia. Nutr Neurosci 2012; 15:103-10. [PMID: 22732353 DOI: 10.1179/1476830511y.0000000026] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
Perinatal asphyxia (PA) is a medical condition associated with a high short-term morbimortality and different long-term neurological diseases. In previous work we have observed at 6 months post-synaptic densities (PSDs) alterations compatible with neurodegeneration highly correlated with the increment in the ubiquitination. Although alterations in the synaptic organization and function have been related with neuronal death after hypoxia, little is known about the synaptic changes in young animals exposed to PA. The main aim of this work is to study the PSDs changes in striatum of 30-day-old rats subjected to PA. Using two-dimensional electron microscopic analyses of synapses staining with ethanolic phosphotungstic acid we observed an increment of PSD thickness in severe hypoxic rats. These data are consistent with the western blot analysis that showed an increment in ubiquitination levels in the synapses of severe hypoxic rat. We did observe any alterations neither in synaptic structure nor in ubiquitinization in mild asphyctic rats. These data suggest that hypoxia might cause early misfolding and aggregation of synaptic proteins in severe anoxic animas that could induce long-term neurodegeneration.
Collapse
Affiliation(s)
- M Grimaldi
- Universidad Argentina John F Kennedy, Buenos Aires, Argentina
| | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Sadleir KR, Vassar R. Cdk5 protein inhibition and Aβ42 increase BACE1 protein level in primary neurons by a post-transcriptional mechanism: implications of CDK5 as a therapeutic target for Alzheimer disease. J Biol Chem 2012; 287:7224-35. [PMID: 22223639 DOI: 10.1074/jbc.m111.333914] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The β-secretase enzyme BACE1 initiates production of the amyloid-β (Aβ) peptide that comprises plaques in Alzheimer disease (AD) brain. BACE1 levels are increased in AD, potentially accelerating Aβ generation, but the mechanisms of BACE1 elevation are not fully understood. Cdk5/p25 has been implicated in neurodegeneration and BACE1 regulation, suggesting therapeutic Cdk5 inhibition for AD. In addition, caspase 3 has been implicated in BACE1 elevation. Here, we show that the Cdk5 level and p25:p35 ratio were elevated and correlated with BACE1 level in brains of AD patients and 5XFAD transgenic mice. Mouse primary cortical neurons treated with Aβ42 oligomers had increased BACE1 level and p25:p35 ratio. Surprisingly, the Aβ42-induced BACE1 elevation was not blocked by Cdk5 inhibitors CP68130 and roscovitine, and instead the BACE1 level was increased greater than with Aβ42 treatment alone. Moreover, Cdk5 inhibitors alone elevated BACE1 in a time- and dose-dependent manner that coincided with increased caspase 3 cleavage and decreased Cdk5 level. Caspase 3 inhibitor benzyloxycarbonyl-VAD failed to prevent the Aβ42-induced BACE1 increase. Further experiments suggested that the Aβ42-induced BACE1 elevation was the result of a post-transcriptional mechanism. We conclude that Aβ42 may increase the BACE1 level independently of either Cdk5 or caspase 3 and that Cdk5 inhibition for AD may cause BACE1 elevation, a potentially negative therapeutic outcome.
Collapse
Affiliation(s)
- Katherine R Sadleir
- Department of Cell and Molecular Biology, Feinberg School of Medicine, Northwestern University, Chicago, Illinois 60611, USA
| | | |
Collapse
|
35
|
Liang J, Yao J, Wang G, Wang Y, Wang B, Ge P. Ischemic postconditioning protects neuronal death caused by cerebral ischemia and reperfusion via attenuating protein aggregation. Int J Med Sci 2012; 9:923-32. [PMID: 23236262 PMCID: PMC3520018 DOI: 10.7150/ijms.4878] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2012] [Accepted: 11/08/2012] [Indexed: 11/05/2022] Open
Abstract
OBJECTIVE To investigate the effect of ischemic postconditioning on protein aggregation caused by transient ischemia and reperfusion and to clarify its underlying mechanism. METHODS Two-vessel-occluded transient global ischemia rat model was used. The rats in ischemic postconditioning group were subjected to three cycles of 30-s/30-s reperfusion/clamping after 15 min of ischemia. Neuronal death in the CA1 region was observed by hematoxylin-eosin staining, and number of live neurons was assessed by cell counting under a light microscope. Succinyl-LLVY-AMC was used as substrate to assay proteasome activity in vitro. Protein carbonyl content was spectrophotometrically measured to analyze protein oxidization. Immunochemistry and laser scanning confocal microscopy were used to observe the distribution of ubiquitin in the CA1 neurons. Western blotting was used to analyze the quantitative alterations of protein aggregates, proteasome, hsp70 and hsp40 in cellular fractions under different ischemic conditions. RESULTS Histological examination showed that the percentage of live neurons in the CA1 region was elevated from 5.21% ± 1.21% to 55.32% ± 5.34% after administration of ischemic postconditioning (P = 0.0087). Western blotting analysis showed that the protein aggregates in the ischemia group was 32.12 ± 4.87, 41.86 ± 4.71 and 34.51 ± 5.18 times higher than that in the sham group at reperfusion 12h, 24h and 48h, respectively. However, protein aggregates were alleviated significantly by ischemic postconditioning to 2.84 ± 0.97, 13.72 ± 2.13 and 14.37 ± 2.42 times at each indicated time point (P = 0.000032, 0.0000051 and 0.0000082). Laser scanning confocal images showed ubiquitin labeled protein aggregates could not be discerned in the ischemic postconditioning group. Further study showed that ischemic postconditioning suppressed the production of carbonyl derivatives, elevated proteasome activity that was damaged by ischemia and reperfusion, increased the expression of chaperone hsp70, and maintained the quantity of chaperone hsp40. CONCLUSION Ischemic postconditioning could rescue significantly neuronal death in the CA1 region caused by transient ischemia and reperfusion, which is closely associated with suppressing the formation of protein aggregation.
Collapse
Affiliation(s)
- Jianmin Liang
- Department of Pediatrics, First Bethune Hospital of Jilin University, Changchun 130021, Jilin Province, PR China
| | | | | | | | | | | |
Collapse
|
36
|
Bosco DA, LaVoie MJ, Petsko GA, Ringe D. Proteostasis and movement disorders: Parkinson's disease and amyotrophic lateral sclerosis. Cold Spring Harb Perspect Biol 2011; 3:a007500. [PMID: 21844169 DOI: 10.1101/cshperspect.a007500] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Parkinson's disease (PD) is a movement disorder that afflicts over one million in the U.S.; amyotrophic lateral sclerosis (ALS or Lou Gehrig's disease) is less prevalent but also has a high incidence. The two disorders sometimes present together, making a comparative study of interest. Both ALS and PD are neurodegenerative diseases, and are characterized by the presence of intraneuronal inclusions; however, different classes of neurons are affected and the primary protein in the inclusions differs between the diseases, and in some cases is different in distinct forms of the same disease. These observations might suggest that the more general approach of proteostasis pathway alteration would be a powerful one in treating these disorders. Examining results from human genetics and studies in model organisms, as well as from biochemical and biophysical characterization of the proteins involved in both diseases, we find that most instances of PD can be considered as arising from the misfolding, and self-association to a toxic species, of the small neuronal protein α-synuclein, and that proteostasis strategies are likely to be of value for this disorder. For ALS, the situation is much more complex and less clear-cut; the available data are most consistent with a view that ALS may actually be a family of disorders, presenting similarly but arising from distinct and nonoverlapping causes, including mislocalization of some properly folded proteins and derangement of RNA quality control pathways. Applying proteostasis approaches to this disease may require rethinking or broadening the concept of what proteostasis means.
Collapse
Affiliation(s)
- Daryl A Bosco
- Department of Neurology, University of Massachusetts Medical Center, Worcester, Massachusetts 01655, USA
| | | | | | | |
Collapse
|
37
|
Kumar A, Singh HP. Information homeostasis as a fundamental principle governing the cell division and death. Med Hypotheses 2011; 77:318-22. [PMID: 21616604 DOI: 10.1016/j.mehy.2011.05.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2010] [Revised: 04/04/2011] [Accepted: 05/02/2011] [Indexed: 11/29/2022]
Abstract
To express the genetic information with minimal error is one of the key functions of a cell. Here we propose an information theory based, phenomenological model for the expression of genetic information. Based on the model we propose the concept of 'information homeostasis' which ensures that genetic information is expressed with minimal error. We suggest that together with energy homeostasis, information homeostasis is a fundamental working principle of a biological cell. This model proposes a novel explanation of why a cell divides and why it stops to divide and, thus, provides novel insights into oncogenesis and various neuro-degenerative diseases. Moreover, the model suggests a theoretical framework to understand cell division and death, beyond specific biochemical pathways.
Collapse
Affiliation(s)
- Arvind Kumar
- Bernstein Centre Freiburg, and Neurobiology and Biophysics, Faculty of Biology, University of Freiburg, Freiburg, Germany.
| | | |
Collapse
|
38
|
Tay SP, Yeo CWS, Chai C, Chua PJ, Tan HM, Ang AXY, Yip DLH, Sung JX, Tan PH, Bay BH, Wong SH, Tang C, Tan JMM, Lim KL. Parkin enhances the expression of cyclin-dependent kinase 6 and negatively regulates the proliferation of breast cancer cells. J Biol Chem 2010; 285:29231-8. [PMID: 20630868 DOI: 10.1074/jbc.m110.108241] [Citation(s) in RCA: 90] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Although mutations in the parkin gene are frequently associated with familial Parkinsonism, emerging evidence suggests that parkin also plays a role in cancers as a putative tumor suppressor. Supporting this, we show here that parkin expression is dramatically reduced in several breast cancer-derived cell lines as well as in primary breast cancer tissues. Importantly, we found that ectopic parkin expression in parkin-deficient breast cancer cells mitigates their proliferation rate both in vitro and in vivo, as well as reduces the capacity of these cells to migrate. Cell cycle analysis revealed the arrestment of a significant percentage of parkin-expressing breast cancer cells at the G1-phase. However, we did not observe significant changes in the levels of the G1-associated cyclin D1 and E. On the other hand, the level of cyclin-dependent kinase 6 (CDK6) is dramatically and selectively elevated in parkin-expressing breast cancer cells, the extent of which correlates well with the expression of parkin. Interestingly, a recent study demonstrated that CDK6 restrains the proliferation of breast cancer cells. Taken together, our results support a negative role for parkin in tumorigenesis and provide a potential mechanism by which parkin exerts its suppressing effects on breast cancer cell proliferation.
Collapse
Affiliation(s)
- Shiam-Peng Tay
- Neurodegeneration Research Laboratory, National Neuroscience Institute, Singapore 308433, Singapore
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
The endoplasmic reticulum and neurological diseases. Exp Neurol 2009; 219:376-81. [DOI: 10.1016/j.expneurol.2009.07.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2009] [Revised: 06/29/2009] [Accepted: 07/09/2009] [Indexed: 12/21/2022]
|
40
|
Protein ubiquitination in postsynaptic densities after hypoxia in rat neostriatum is blocked by hypothermia. Exp Neurol 2009; 219:404-13. [PMID: 19555686 DOI: 10.1016/j.expneurol.2009.06.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2009] [Revised: 05/24/2009] [Accepted: 06/14/2009] [Indexed: 11/23/2022]
Abstract
Synaptic dysfunction has been associated with neuronal cell death following hypoxia. The lack of knowledge on the mechanisms underlying this dysfunction prompted us to investigate the morphological changes in the postsynaptic densities (PSDs) induced by hypoxia. The results presented here demonstrate that PSDs of the rat neostriatum are highly modified and ubiquitinated 6 months after induction of hypoxia in a model of perinatal asphyxia. Using both two dimensional (2D) and three dimensional (3D) electron microscopic analyses of synapses stained with ethanolic phosphotungstic acid (E-PTA), we observed an increment of PSD thickness dependent on the duration and severity of the hypoxic insult. The PSDs showed clear signs of damage and intense staining for ubiquitin. These morphological and molecular changes were effectively blocked by hypothermia treatment, one of the most effective strategies for hypoxia-induced brain injury available today. Our data suggest that synaptic dysfunction following hypoxia may be caused by long-term misfolding and aggregation of proteins in the PSD.
Collapse
|
41
|
Ischemic preconditioning induces chaperone hsp70 expression and inhibits protein aggregation in the CA1 neurons of rats. Neurosci Bull 2009; 24:288-96. [PMID: 18839022 DOI: 10.1007/s12264-008-0623-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
OBJECTIVE To investigate the effect of ischemic preconditioning on chaperone hsp70 expression and protein aggregation in the CA1 neurons of rats, and to further explore its potential neuroprotective mechanism. METHODS Two-vesseloccluded transient global ischemia rat model was used. The rats were divided into sublethal 3-min ischemia group, lethal 10-min ischemia group and ischemic preconditioning group. Neuronal death in the CA1 region was observed by hematoxylineosin staining, and number of live neurons was assessed by cell counting under a light microscope. Immunochemistry and laser scanning confocal microscopy were used to observe the distribution of chaperone hsp70 in the CA1 neurons. Differential centrifuge was used to isolate cytosol, nucleus and protein aggregates fractions. Western blot was used to analyze the quantitative alterations of protein aggregates and inducible chaperone hsp70 in cellular fractions and in protein aggregates under different ischemic conditions. RESULTS Histological examination showed that ischemic preconditioning significantly reduced delayed neuronal death in the hippocampus CA1 region (P < 0.01 vs 10-min ischemia group). Sublethal ischemic preconditioning induced chaperone hsp70 expression in the CA1 neurons after 24 h reperfusion following 10-min ischemia. Induced-hsp70 combined with the abnormal proteins produced during the secondary lethal 10-min ischemia and inhibited the formation of cytotoxic protein aggregates (P < 0.01 vs 10-min ischemia group). CONCLUSION Ischemic preconditioning induced chaperone hsp70 expression and inhibited protein aggregates formation in the CA1 neurons when suffered secondary lethal ischemia, which may protect neurons from death.
Collapse
|
42
|
Gupta S, Kühnisch J, Mustafa A, Lhotak S, Schlachterman A, Slifker MJ, Klein-Szanto A, High KA, Austin RC, Kruger WD. Mouse models of cystathionine beta-synthase deficiency reveal significant threshold effects of hyperhomocysteinemia. FASEB J 2008; 23:883-93. [PMID: 18987302 DOI: 10.1096/fj.08-120584] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Untreated cystathionine beta-synthase (CBS) deficiency in humans is characterized by extremely elevated plasma total homocysteine (tHcy>200 microM), with thrombosis as the major cause of morbidity. Treatment with vitamins and diet leads to a dramatic reduction in thrombotic events, even though patients often still have severe elevations in tHcy (>80 microM). To understand the difference between extreme and severe hyperhomocysteinemia, we have examined two mouse models of CBS deficiency: Tg-hCBS Cbs(-/-) mice, with a mean serum tHcy of 169 microM, and Tg-I278T Cbs(-/-) mice, with a mean tHcy of 296 microM. Only Tg-I278T Cbs(-/-) animals exhibited strong biological phenotypes, including facial alopecia, osteoporosis, endoplasmic reticulum (ER) stress in the liver and kidney, and a 20% reduction in mean survival time. Metabolic profiling of serum and liver reveals that Tg-I278T Cbs(-/-) mice have significantly elevated levels of free oxidized homocysteine but not protein-bound homocysteine in serum and elevation of all forms of homocysteine and S-adenosylhomocysteine in the liver compared to Tg-hCBS Cbs(-/-) mice. RNA profiling of livers indicate that Tg-I278T Cbs(-/-) and Tg-hCBS Cbs(-/-) mice have unique gene signatures, with minimal overlap. Our results indicate that there is a clear pathogenic threshold effect for tHcy and bring into question the idea that mild elevations in tHcy are directly pathogenic.
Collapse
Affiliation(s)
- Sapna Gupta
- Division of Population Science, Fox Chase Cancer Center, 333 Cottman Ave., Philadelphia, PA 19111, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
|
44
|
Wang HQ, Imai Y, Kataoka A, Takahashi R. Cell type-specific upregulation of Parkin in response to ER stress. Antioxid Redox Signal 2007; 9:533-42. [PMID: 17465879 DOI: 10.1089/ars.2006.1522] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Parkin is the gene responsible for a familial form of Parkinson's disease (PD) termed autosomal recessive juvenile parkinsonism (AR-JP)/PARK2. Parkin has been shown to protect cells from endoplasmic reticulum (ER) stress and oxidative stress, presumably due to its ubiquitin ligase (E3) activity that targets proteins for proteasomal degradation. Although the authors showed that parkin is upregulated in response to ER stress, subsequent reports suggest that it does not represent a universal unfolded protein response (UPR). Here the authors report different regulation of parkin in response to ER stress in different cell lines, demonstrating upregulation of parkin as a cell type-specific response to ER stress. 2-Mercaptoethanol (2-ME) and tunicamycin increased the expression of parkin in SH-SY5Y (H) cells, Neuro2a cells, Goto-P3 cells, but not in SH-SY5Y (J) cells and IMR32 cells. In parallel with these studies, similar upregulation of the parkin coregulated gene (PACRG)/gene adjacent to parkin (Glup) was also observed by ER stress. Luciferase assays failed to detect the transcriptional activation of 500 bp parkin/Glup promoter in response to ER stress. These results indicate that induction of parkin by ER stress represents a cell type-specific response.
Collapse
Affiliation(s)
- Hua-Qin Wang
- Department of Neurology, Kyoto University Graduate School of Medicine, Kyoto, Japan
| | | | | | | |
Collapse
|
45
|
Abstract
Neuronal cell death is a major feature of various diseases, including brain ischemia, neuronal degenerative diseases, and traumatic injury, suggesting the importance of investigating the mechanisms that mediate neuronal cell death. Although the various factors that contribute to brain ischemia have been defined and the mechanism through which each factor causes neuronal cell death has been investigated, definite strategies have not been established. In this brief review, we focus on two important mechanisms that contribute to the pathogenesis of brain ischemia. First, we discuss the glutamate theory, a proposed mechanism for the understanding of ischemia-induced neuronal cell death. Second, an accumulation of recent molecular neurobiology evidence regarding the dysfunction of a cellular organelle, the endoplasmic reticulum (ER), suggests that it plays a major role in the pathogenesis of neuronal cell death. Whereas the former theory reflects the role of neuron-specific factors in the induction of cell death, the stress response of the ER for maintenance of its function is regarded as a defense mechanism. Because hypoxia, another major factor in ischemia, results in further dysfunction of the ER, studies on the malfunction of this cellular organelle may facilitate the development of novel strategies to block ischemia-induced cell death.
Collapse
Affiliation(s)
- Satoshi Ogawa
- Department of Neuroanatomy, Kanazawa University Medical School, Takara-machi, Kanazawa City, Ishikawa, Japan.
| | | | | |
Collapse
|
46
|
O'Duffy AE, Bordelon YM, McLaughlin B. Killer proteases and little strokes--how the things that do not kill you make you stronger. J Cereb Blood Flow Metab 2007; 27:655-68. [PMID: 16896349 PMCID: PMC2881558 DOI: 10.1038/sj.jcbfm.9600380] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The phenomenon of ischemic preconditioning was initially observed over 20 years ago. The basic tenant is that if stimuli are applied at a subtoxic level, cells upregulate endogenous protective mechanisms to block injury induced by subsequent stress. Since this discovery, many conserved signaling mechanisms that contribute to activation of this potent protective program have been identified in the brain. A clinical correlate of this basic research finding can be found in patients with a history of transient ischemic attack (TIA), who have a decreased morbidity after stroke. In spite of multidisciplinary efforts to design safer, more effective stroke therapies, we have thus far failed to translate our understanding of endogenous protective pathways to treatments for neurodegeneration. This review is designed to provide clinicians and basic scientists with an overview of stress biology after TIA and preconditioning, discuss new therapeutic strategies to target the protein dysfunction that follows ischemic injury, and propose enhanced biochemical profiling to identify individuals at risk of stroke after TIA. We pay particular attention to the unanticipated consequences of overly aggressive intervention after TIA in which we have found that traditional cytotoxic agents such as free radicals and apoptosis associated proteases is essential for neuroprotection and communication in the stressed brain. These data emphasize the importance of understanding the complex interplay between chaperones, apoptotic proteases including caspases, and the proteolytic degradation machinery in adaptation to neurological injury.
Collapse
Affiliation(s)
- Anne E O'Duffy
- Department of Neurology, Vanderbilt University, Nashville, Tennessee 37232-8548, USA
| | | | | |
Collapse
|
47
|
Liu JJ, Zhao H, Sung JH, Sun GH, Steinberg GK. Hypothermia blocks ischemic changes in ubiquitin distribution and levels following stroke. Neuroreport 2007; 17:1691-5. [PMID: 17047455 DOI: 10.1097/01.wnr.0000236868.83970.79] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
Dysfunction of the ubiquitin-proteasome system has recently been linked to stroke. Ischemia may cause increased protein misfolding and inhibit the proteasome, shifting the balance from free ubiquitin to conjugated ubiquitin. In this study, we examine the effect of hypothermia on the distribution of total and free ubiquitin, as well as the levels of conjugated ubiquitin after experimental stroke using a focal cerebral ischemia model. We show that hypothermia prevents redistribution of ubiquitin following ischemia, largely through preservation of intracellular cytoplasmic free ubiquitin. We also show that hypothermia blocks the increase in conjugated ubiquitin observed after stroke. Our data indicate that hypothermia's neuroprotection is mediated, in part, through preservation of ubiquitin-proteasome system function.
Collapse
Affiliation(s)
- Jen-Jane Liu
- Department of Neurosurgery and Stanford Stroke Center, Stanford University, Stanford, California 94305-5487, USA
| | | | | | | | | |
Collapse
|
48
|
Abstract
The etiologies of neurodegenerative diseases, such as Alzheimer's disease, Parkinson's disease, polyglutamine diseases, or prion diseases may be diverse; however, aberrations in protein folding, processing, and/or degradation are common features of these entities, implying a role of quality control systems, such as molecular chaperones and the ubiquitin-proteasome pathway. There is substantial evidence for a causal role of protein misfolding in the pathogenic process coming from neuropathology, genetics, animal modeling, and biophysics. The presence of protein aggregates in all neurodegenerative diseases gave rise to the hypothesis that protein aggregates, be it intracellular or extracellular deposits, may perturb the cellular homeostasis and disintegrate neuronal function (Table 1). More recently, however, an increasing number of studies have indicated that protein aggregates are not toxic per se and might even serve a protective role by sequestering misfolded proteins. Specifically, experimental models of polyglutamine diseases, Alzheimer's disease, and Parkinson's disease revealed that the appearance of aggregates can be dissociated from neuronal toxicity, while misfolded monomers or oligomeric intermediates seem to be the toxic species. The unique features of molecular chaperones to assist in the folding of nascent proteins and to prevent stress-induced misfolding was the rationale to exploit their effects in different models of neurodegenerative diseases. This chapter concentrates on two neurodegenerative diseases, Parkinson's disease and prion diseases, with a special focus on protein misfolding and a possible role of molecular chaperones.
Collapse
Affiliation(s)
- K F Winklhofer
- Department of Cellular Biochemistry, Max-Planck-Institute for Biochemistry, Martinsried, Germany.
| | | |
Collapse
|
49
|
Reitz C, Trenkwalder C, Kretzschmar K, Roesler A, V Eckardstein A, Berger K. Relation of cerebral small-vessel disease and brain atrophy to mild parkinsonism in the elderly. Mov Disord 2006; 21:1914-9. [PMID: 16960865 DOI: 10.1002/mds.21085] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022] Open
Abstract
The association between cerebral small-vessel disease, brain atrophy, and the risk and severity of mild parkinsonian signs (MPS) remains unclear. The objective of this study is to examine the effect of lacunar brain infarcts, cerebral white matter lesions (WMLs), and cortical atrophy on the risk and severity of MPS. This study is a cross-sectional community-based cohort study comprising 268 subjects, 65 to 83 years of age, residing in the Augsburg region of southern Germany, and without contraindications for magnetic resonance imaging (MRI) of the brain. Main outcome measures. Subcortical and periventricular WMLs, lacunar brain infarcts, and cortical atrophy determined using a standardized MRI protocol developed for the Rotterdam Scan Study and an established rating scale. MPS, assessed in a standardized neurological examination and based on the Unified Parkinson's Disease Rating Scale motor scale. Lacunar brain infarcts and large subcortical white matter lesions were associated with an elevated risk of resting tremor. More severe cortical atrophy was related to an increased risk of rigidity and bradykinesia. In a linear regression analysis relating each individual MRI measurement with the severity of MPS, the number of lacunar brain infarcts and the degree of brain atrophy were correlated with the severity of resting tremor, whereas the size of subcortical and periventricular WMLs was correlated with the severity of rigidity. A higher degree of brain atrophy was associated with increased severity of either cardinal sign. In our study, presence and volume of lacunar brain infarcts, cerebral WMLs, and cortical atrophy were associated with the risk as well as severity of MPS. Determining the presence of these brain changes using brain imaging might contribute to identify persons at risk for MPS.
Collapse
Affiliation(s)
- Christiane Reitz
- Institute of Epidemiology and Social Medicine, University of Muenster, Muenster, Germany.
| | | | | | | | | | | |
Collapse
|
50
|
Paschen W, Mengesdorf T. Cellular abnormalities linked to endoplasmic reticulum dysfunction in cerebrovascular disease—therapeutic potential. Pharmacol Ther 2005; 108:362-75. [PMID: 16140387 DOI: 10.1016/j.pharmthera.2005.05.008] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2005] [Accepted: 05/26/2005] [Indexed: 01/20/2023]
Abstract
Unfolded proteins accumulate in the lumen of the endoplasmic reticulum (ER) as part of the cellular response to cerebral hypoxia/ischemia and also to the overexpression of the mutant genes responsible for familial forms of degenerative diseases such as Alzheimer's disease, Parkinson's disease, Huntington's disease, amyothrophic lateral sclerosis, and Huntington's disease, as well as other disorders that are caused by an expanded CAG repeat. This accumulation arises from an imbalance between the load of proteins that need to be folded and processed in the ER lumen and the ER folding/processing capacity. To withstand such potentially lethal conditions, stress responses are activated that includes the shutdown of translation to reduce the ER work load and the activation of the expression of genes coding for proteins involved in the folding and processing reactions, to increase folding/processing capacity. In transient cerebral ischemia, ER stress-induced suppression of protein synthesis is believed to be too severe to permit sufficient activation of the genetic arm of the ER stress response. Mutations associated with Alzheimer's disease down-regulate the ER stress response and make cells more vulnerable to conditions associated with ER stress. When the functioning of the ER is severely impaired and affected cells can no longer withstand these stressful conditions, programmed cell death is induced, including a mitochondria-driven apoptotic pathway. Raising the resistance of cells to conditions that interfere with ER functions and activating the degradation and refolding of unfolded proteins accumulated in the ER lumen are possible strategies for blocking the pathological process leading to cell death at an early stage.
Collapse
Affiliation(s)
- Wulf Paschen
- Multidisciplinary Neuroprotection Laboratories, Duke University Medical Center, Department of Anesthesiology, 132 Sands Building, Research Drive, Durham, NC 27710, USA.
| | | |
Collapse
|