1
|
Zhou Q, Meng Y, Le J, Sun Y, Dian Y, Yao L, Xiong Y, Zeng F, Chen X, Deng G. Ferroptosis: mechanisms and therapeutic targets. MedComm (Beijing) 2024; 5:e70010. [PMID: 39568772 PMCID: PMC11577302 DOI: 10.1002/mco2.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 11/22/2024] Open
Abstract
Ferroptosis is a nonapoptotic form of cell death characterized by iron-dependent lipid peroxidation in membrane phospholipids. Since its identification in 2012, extensive research has unveiled its involvement in the pathophysiology of numerous diseases, including cancers, neurodegenerative disorders, organ injuries, infectious diseases, autoimmune conditions, metabolic disorders, and skin diseases. Oxidizable lipids, overload iron, and compromised antioxidant systems are known as critical prerequisites for driving overwhelming lipid peroxidation, ultimately leading to plasma membrane rupture and ferroptotic cell death. However, the precise regulatory networks governing ferroptosis and ferroptosis-targeted therapy in these diseases remain largely undefined, hindering the development of pharmacological agonists and antagonists. In this review, we first elucidate core mechanisms of ferroptosis and summarize its epigenetic modifications (e.g., histone modifications, DNA methylation, noncoding RNAs, and N6-methyladenosine modification) and nonepigenetic modifications (e.g., genetic mutations, transcriptional regulation, and posttranslational modifications). We then discuss the association between ferroptosis and disease pathogenesis and explore therapeutic approaches for targeting ferroptosis. We also introduce potential clinical monitoring strategies for ferroptosis. Finally, we put forward several unresolved issues in which progress is needed to better understand ferroptosis. We hope this review will offer promise for the clinical application of ferroptosis-targeted therapies in the context of human health and disease.
Collapse
Affiliation(s)
- Qian Zhou
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Yu Meng
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Jiayuan Le
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Yuming Sun
- Department of Plastic and Cosmetic Surgery Xiangya Hospital Central South University Changsha Hunan Province China
| | - Yating Dian
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Lei Yao
- Department of General Surgery Xiangya Hospital Central South University Changsha Hunan Province China
| | - Yixiao Xiong
- Department of Dermatology Tongji Hospital Huazhong University of Science and Technology Wuhan Hubei China
| | - Furong Zeng
- Department of Oncology Xiangya Hospital Central South University Changsha Hunan Province China
| | - Xiang Chen
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Guangtong Deng
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| |
Collapse
|
2
|
Kaur M, Aran KR, Paswan R. A potential role of gut microbiota in stroke: mechanisms, therapeutic strategies and future prospective. Psychopharmacology (Berl) 2024; 241:2409-2430. [PMID: 39463207 DOI: 10.1007/s00213-024-06708-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Accepted: 10/18/2024] [Indexed: 10/29/2024]
Abstract
RATIONALE Neurological conditions like Stroke and Alzheimer's disease (AD) often include inflammatory responses in the nervous system. Stroke, linked to high disability and mortality rates, poses challenges related to organ-related complications. Recent focus on understanding the pathophysiology of ischemic stroke includes aspects like cellular excitotoxicity, oxidative stress, cell death mechanisms, and neuroinflammation. OBJECTIVE The objective of this paper is to summarize and explore the pathophysiology of ischemic stroke, elucidates the gut-brain axis mechanism, and discusses recent clinical trials, shedding light on novel treatments and future possibilities. RESULTS Changes in gut architecture and microbiota contribute to dementia by enhancing intestinal permeability, activating the immune system, elevating proinflammatory mediators, altering blood-brain barrier (BBB) permeability, and ultimately leading to neurodegenerative diseases (NDDs). The gut-brain axis's potential role in disease pathophysiology offers new avenues for cell-based regenerative medicine in treating neurological conditions. CONCLUSION In conclusion, the gut microbiome significantly impacts stroke prognosis by highlighting the role of the gut-brain axis in ischemic stroke mechanisms. This insight suggests potential therapeutic strategies for improving outcomes.
Collapse
Affiliation(s)
- Manpreet Kaur
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India
| | - Khadga Raj Aran
- Neuropharmacology Division, Department of Pharmacology, ISF College of Pharmacy, Moga, Punjab, 142001, India.
| | - Raju Paswan
- Department of Pharmacy Practice, ISF College of Pharmacy, Moga, Punjab, 142001, India
| |
Collapse
|
3
|
Yaqubi S, Karimian M. Stem cell therapy as a promising approach for ischemic stroke treatment. CURRENT RESEARCH IN PHARMACOLOGY AND DRUG DISCOVERY 2024; 6:100183. [PMID: 38831867 PMCID: PMC11144755 DOI: 10.1016/j.crphar.2024.100183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 04/23/2024] [Accepted: 05/14/2024] [Indexed: 06/05/2024] Open
Abstract
Ischemia as the most common type of stroke is the main cause of death and disability in the world. However, there are few therapeutic approaches to treat ischemic stroke. The common approach to the treatment of ischemia includes surgery-cum-chemical drugs. Surgery and chemical drugs are used to remove blood clots to prevent the deterioration of the nervous system. Given the surgical hazards and the challenges associated with chemical drugs, these cannot be considered safe approaches to the treatment of brain ischemia. Besides surgery-cum-chemical drugs, different types of stem cells including mesenchymal stem cells and neurological stem cells have been considered to treat ischemic stroke. Therapeutic approaches utilizing stem cells to treat strokes are promising because of their neuroprotective and regenerative benefits. However, the mechanisms by which the transplanted stem cells perform their precisely actions are unknown. The purpose of this study is to critically review stem cell-based therapeutic approaches for ischemia along with related challenges.
Collapse
Affiliation(s)
- Sahar Yaqubi
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, Iran
| | - Mohammad Karimian
- Department of Molecular and Cell Biology, Faculty of Basic Sciences, University of Mazandaran, Babolsar, Iran
| |
Collapse
|
4
|
Zhang Y, Zou Z, Liu S, Chen F, Li M, Zou H, Liu H, Ding J. Edaravone-loaded poly(amino acid) nanogel inhibits ferroptosis for neuroprotection in cerebral ischemia injury. Asian J Pharm Sci 2024; 19:100886. [PMID: 38590795 PMCID: PMC10999513 DOI: 10.1016/j.ajps.2024.100886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Revised: 11/29/2023] [Accepted: 01/11/2024] [Indexed: 04/10/2024] Open
Abstract
Neurological injury caused by ischemic stroke is a major cause of permanent disability and death. The currently available neuroprotective drugs fail to achieve desired therapeutic efficacy mainly due to short circulation half-life and poor blood-brain barrier (BBB) permeability. For that, an edaravone-loaded pH/glutathione (pH/GSH) dual-responsive poly(amino acid) nanogel (NG/EDA) was developed to improve the neuroprotection of EDA. The nanogel was triggered by acidic and EDA-induced high-level GSH microenvironments, which enabled the selective and sustained release of EDA at the site of ischemic injury. NG/EDA exhibited a uniform sub-spherical morphology with a mean hydrodynamic diameter of 112.3 ± 8.2 nm. NG/EDA efficiently accumulated at the cerebral ischemic injury site of permanent middle cerebral artery occlusion (pMCAO) mice, showing an efficient BBB crossing feature. Notably, NG/EDA with 50 µM EDA significantly increased neuron survival (29.3%) following oxygen and glucose deprivation by inhibiting ferroptosis. In addition, administering NG/EDA for 7 d significantly reduced infarct volume to 22.2% ± 7.2% and decreased neurobehavioral scores from 9.0 ± 0.6 to 2.0 ± 0.8. Such a pH/GSH dual-responsive nanoplatform might provide a unique and promising modality for neuroprotection in ischemic stroke and other central nervous system diseases.
Collapse
Affiliation(s)
- Yunhan Zhang
- Key Laboratory of Pathobiology Ministry of Education, Department of Anatomy, College of Basic Medical Sciences, Jilin University, Changchun 130061, China
| | - Zhulin Zou
- Key Laboratory of Pathobiology Ministry of Education, Department of Anatomy, College of Basic Medical Sciences, Jilin University, Changchun 130061, China
| | - Shuang Liu
- Key Laboratory of Pathobiology Ministry of Education, Department of Anatomy, College of Basic Medical Sciences, Jilin University, Changchun 130061, China
| | - Fangfang Chen
- Department of Gastrointestinal, Colorectal, and Anal Surgery, China-Japan Union Hospital of Jilin University, Changchun 130033, China
| | - Minglu Li
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Haoyang Zou
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| | - Haiyan Liu
- Key Laboratory of Pathobiology Ministry of Education, Department of Anatomy, College of Basic Medical Sciences, Jilin University, Changchun 130061, China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun 130022, China
| |
Collapse
|
5
|
Passarelli JP, Nimjee SM, Townsend KL. Stroke and Neurogenesis: Bridging Clinical Observations to New Mechanistic Insights from Animal Models. Transl Stroke Res 2024; 15:53-68. [PMID: 36462099 DOI: 10.1007/s12975-022-01109-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 11/15/2022] [Accepted: 11/16/2022] [Indexed: 12/04/2022]
Abstract
Stroke was the 2nd leading cause of death and a major cause of morbidity. Unfortunately, there are limited means to promote neurological recovery post-stroke, but research has unearthed potential targets for therapies to encourage post-stroke neurogenesis and neuroplasticity. The occurrence of neurogenesis in adult mammalian brains, including humans, was not widely accepted until the 1990s. Now, adult neurogenesis has been extensively studied in human and mouse neurogenic brain niches, of which the subventricular zone of the lateral ventricles and subgranular zone of the dentate gyrus are best studied. Numerous other niches are under investigation for neurogenic potential. This review offers a basic overview to stroke in the clinical setting, a focused summary of recent and foundational research literature on cortical neurogenesis and post-stroke brain plasticity, and insights regarding how the meninges and choroid plexus have emerged as key players in neurogenesis and neuroplasticity in the context of focal cerebral ischemia disrupting the anterior circulation. The choroid plexus and meninges are vital as they are integral sites for neuroimmune interactions, glymphatic perfusion, and niche signaling pertinent to neural stem cells and neurogenesis. Modulating neuroimmune interactions with a focus on astrocyte activity, potentially through manipulation of the choroid plexus and meningeal niches, may reduce the exacerbation of stroke by inflammatory mediators and create an environment conducive to neurorecovery. Furthermore, addressing impaired glymphatic perfusion after ischemic stroke likely supports a neurogenic environment by clearing out inflammatory mediators, neurotoxic metabolites, and other accumulated waste. The meninges and choroid plexus also contribute more directly to promoting neurogenesis: the meninges are thought to harbor neural stem cells and are a niche amenable to neural stem/progenitor cell migration. Additionally, the choroid plexus has secretory functions that directly influences stem cells through signaling mechanisms and growth factor actions. More research to better understand the functions of the meninges and choroid plexus may lead to novel approaches for stimulating neuronal recovery after ischemic stroke.
Collapse
Affiliation(s)
| | - Shahid M Nimjee
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Biomedical Research Tower, 460 W 12th Avenue, Columbus, OH, 43210, USA
| | - Kristy L Townsend
- Department of Neurological Surgery, The Ohio State University Wexner Medical Center, Biomedical Research Tower, 460 W 12th Avenue, Columbus, OH, 43210, USA.
| |
Collapse
|
6
|
Xiong S, Xiao H, Sun M, Liu Y, Gao L, Xu K, Liang H, Jiang N, Lin Y, Chang L, Wu H, Zhu D, Luo C. Glutamate-releasing BEST1 channel is a new target for neuroprotection against ischemic stroke with wide time window. Acta Pharm Sin B 2023; 13:3008-3026. [PMID: 37521872 PMCID: PMC10372917 DOI: 10.1016/j.apsb.2023.05.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 01/13/2023] [Accepted: 04/03/2023] [Indexed: 08/01/2023] Open
Abstract
Many efforts have been made to understand excitotoxicity and develop neuroprotectants for the therapy of ischemic stroke. The narrow treatment time window is still to be solved. Given that the ischemic core expanded over days, treatment with an extended time window is anticipated. Bestrophin 1 (BEST1) belongs to a bestrophin family of calcium-activated chloride channels. We revealed an increase in neuronal BEST1 expression and function within the peri-infarct from 8 to 48 h after ischemic stroke in mice. Interfering the protein expression or inhibiting the channel function of BEST1 by genetic manipulation displayed neuroprotective effects and improved motor functional deficits. Using electrophysiological recordings, we demonstrated that extrasynaptic glutamate release through BEST1 channel resulted in delayed excitotoxicity. Finally, we confirmed the therapeutic efficacy of pharmacological inhibition of BEST1 during 6-72 h post-ischemia in rodents. This delayed treatment prevented the expansion of infarct volume and the exacerbation of neurological functions. Our study identifies the glutamate-releasing BEST1 channel as a potential therapeutic target against ischemic stroke with a wide time window.
Collapse
Affiliation(s)
- Shuai Xiong
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Hui Xiao
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Meng Sun
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yunjie Liu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Ling Gao
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Ke Xu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Haiying Liang
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Nan Jiang
- Department of Medicinal Chemistry, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Yuhui Lin
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Lei Chang
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
| | - Haiyin Wu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
- Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Dongya Zhu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
- Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China
| | - Chunxia Luo
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing 211166, China
- Collaborative Innovation Center for Cardiovascular Disease Translational Medicine, Nanjing Medical University, Nanjing 211166, China
| |
Collapse
|
7
|
Dergunova LV, Filippenkov IB, Limborska SA, Myasoedov NF. Neuroprotective Peptides and New Strategies for Ischemic Stroke Drug Discoveries. Genes (Basel) 2023; 14:genes14050953. [PMID: 37239313 DOI: 10.3390/genes14050953] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 04/15/2023] [Accepted: 04/20/2023] [Indexed: 05/28/2023] Open
Abstract
Ischemic stroke continues to be one of the leading causes of death and disability in the adult population worldwide. The currently used pharmacological methods for the treatment of ischemic stroke are not effective enough and require the search for new tools and approaches to identify therapeutic targets and potential neuroprotectors. Today, in the development of neuroprotective drugs for the treatment of stroke, special attention is paid to peptides. Namely, peptide action is aimed at blocking the cascade of pathological processes caused by a decrease in blood flow to the brain tissues. Different groups of peptides have therapeutic potential in ischemia. Among them are small interfering peptides that block protein-protein interactions, cationic arginine-rich peptides with a combination of various neuroprotective properties, shuttle peptides that ensure the permeability of neuroprotectors through the blood-brain barrier, and synthetic peptides that mimic natural regulatory peptides and hormones. In this review, we consider the latest achievements and trends in the development of new biologically active peptides, as well as the role of transcriptomic analysis in identifying the molecular mechanisms of action of potential drugs aimed at the treatment of ischemic stroke.
Collapse
Affiliation(s)
- Lyudmila V Dergunova
- Institute of Molecular Genetics, National Research Center "Kurchatov Institute", Kurchatov Sq. 2, 123182 Moscow, Russia
| | - Ivan B Filippenkov
- Institute of Molecular Genetics, National Research Center "Kurchatov Institute", Kurchatov Sq. 2, 123182 Moscow, Russia
| | - Svetlana A Limborska
- Institute of Molecular Genetics, National Research Center "Kurchatov Institute", Kurchatov Sq. 2, 123182 Moscow, Russia
| | - Nikolay F Myasoedov
- Institute of Molecular Genetics, National Research Center "Kurchatov Institute", Kurchatov Sq. 2, 123182 Moscow, Russia
| |
Collapse
|
8
|
Liang Z, Lou Y, Hao Y, Li H, Feng J, Liu S. The Relationship of Astrocytes and Microglia with Different Stages of Ischemic Stroke. Curr Neuropharmacol 2023; 21:2465-2480. [PMID: 37464832 PMCID: PMC10616922 DOI: 10.2174/1570159x21666230718104634] [Citation(s) in RCA: 24] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2022] [Revised: 01/31/2023] [Accepted: 02/04/2023] [Indexed: 07/20/2023] Open
Abstract
Ischemic stroke is the predominant cause of severe morbidity and mortality worldwide. Post-stroke neuroinflammation has recently received increasing attention with the aim of providing a new effective treatment strategy for ischemic stroke. Microglia and astrocytes are major components of the innate immune system of the central nervous system. They can be involved in all phases of ischemic stroke, from the early stage, contributing to the first wave of neuronal cell death, to the late stage involving phagocytosis and repair. In the early stage of ischemic stroke, a vicious cycle exists between the activation of microglia and astrocytes (through astrocytic connexin 43 hemichannels), aggravating neuroinflammatory injury post-stroke. However, in the late stage of ischemic stroke, repeatedly activated microglia can induce the formation of glial scars by triggering reactive astrogliosis in the peri-infarct regions, which may limit the movement of activated microglia in reverse and restrict the diffusion of inflammation to healthy brain tissues, alleviating the neuroinflammatory injury poststroke. In this review, we elucidated the various roles of astrocytes and microglia and summarized their relationship with neuroinflammation. We also examined how astrocytes and microglia influence each other at different stages of ischemic stroke. Several potential therapeutic approaches targeting astrocytes and microglia in ischemic stroke have been reviewed. Understanding the details of astrocytemicroglia interaction processes will contribute to a better understanding of the mechanisms underlying ischemic stroke, contributing to the identification of new therapeutic interventions.
Collapse
Affiliation(s)
- Zhen Liang
- Department of Neurology, China-Japan Union Hospital, Jilin University, Changchun, China
| | - Yingyue Lou
- Department of Rehabilitation, The Second Hospital of Jilin University, Changchun, China
| | - Yulei Hao
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Hui Li
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Jiachun Feng
- Department of Neurology and Neuroscience Center, The First Hospital of Jilin University, Changchun, China
| | - Songyan Liu
- Department of Neurology, China-Japan Union Hospital, Jilin University, Changchun, China
| |
Collapse
|
9
|
He Y, Ying J, Tang J, Zhou R, Qu H, Qu Y, Mu D. Neonatal Arterial Ischaemic Stroke: Advances in Pathologic Neural Death, Diagnosis, Treatment, and Prognosis. Curr Neuropharmacol 2022; 20:2248-2266. [PMID: 35193484 PMCID: PMC9890291 DOI: 10.2174/1570159x20666220222144744] [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: 11/29/2021] [Revised: 02/04/2022] [Accepted: 02/18/2022] [Indexed: 12/29/2022] Open
Abstract
Neonatal arterial ischaemic stroke (NAIS) is caused by focal arterial occlusion and often leads to severe neurological sequelae. Neural deaths after NAIS mainly include necrosis, apoptosis, necroptosis, autophagy, ferroptosis, and pyroptosis. These neural deaths are mainly caused by upstream stimulations, including excitotoxicity, oxidative stress, inflammation, and death receptor pathways. The current clinical approaches to managing NAIS mainly focus on supportive treatments, including seizure control and anticoagulation. In recent years, research on the pathology, early diagnosis, and potential therapeutic targets of NAIS has progressed. In this review, we summarise the latest progress of research on the pathology, diagnosis, treatment, and prognosis of NAIS and highlight newly potential diagnostic and treatment approaches.
Collapse
Affiliation(s)
- Yang He
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Junjie Ying
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Jun Tang
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Ruixi Zhou
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Haibo Qu
- Department of Radiology, West China Second University Hospital, Sichuan University, Chengdu, China
| | - Yi Qu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| | - Dezhi Mu
- Department of Pediatrics, West China Second University Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Sichuan University, Ministry of Education, Chengdu, China
| |
Collapse
|
10
|
Zhang T, Han H, Zhou Y, Liu Z, Ma T, Cao X. MicroRNA-454 modulates the oxidative stress and neuronal apoptosis after cerebral ischemia/reperfusion injury via targeting NADPH oxidase 4 (NOX4). J Biochem Mol Toxicol 2022; 36:e23153. [PMID: 36043333 PMCID: PMC9786265 DOI: 10.1002/jbt.23153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 04/06/2022] [Accepted: 05/29/2022] [Indexed: 12/30/2022]
Abstract
To investigate the function of miR-454 in ischemic stroke, this study was carried out. Cerebral ischemia/reperfusion (I/R) injury animal model and a SHSY5Y cell culture model of oxygen-glucose deprivation/reoxygenation (OGD/R) were constructed. The effects of miR-454 were detected by evaluating the levels of biochemical markers, gene expression, and pathophysiological markers. The results showed that NOX4 level was elevated, while miR-454 expression was reduced in I/R brain samples and in OGD/R-treated cells. The miR-454 agomir declined NOX4 level and reactive oxygen species (ROS) production in rats suffering from I/R. Furthermore, microRNA-145 (miR-454) overexpression inhibited NOX4 level and ROS production in cells treated by OGD/R and decreased luciferase activity in cells transfected with NOX4-wild type (WT) reporter plasmid. Meanwhile, our results proved that the protected effects of miR-454 on SH-SY5Y cells treated by OGD/R were reversed by pcDNA-NOX4 transfection. MiR-454 protected animals from brain injury induced by cerebral I/R via directly regulating its target gene NOX4, illustrating a curatively potential target for treating ischemic stroke.
Collapse
Affiliation(s)
- Tao Zhang
- Department of NeurologyBaoan District Central HospitalShenzhen CityGuangdong ProvinceP.R. China
| | - Haiping Han
- Department of NeurologyNingxia Hui Autonomous Region People's HospitalYinchuan CityNingxia Hui Autonomous RegionP.R. China
| | - Yan Zhou
- Department of Thoracic SurgeryNingxia Hui Autonomous Region People's HospitalYinchuan CityNingxia Hui Autonomous RegionP.R. China
| | - Zhimei Liu
- Department of NeurologyNingxia Hui Autonomous Region People's HospitalYinchuan CityNingxia Hui Autonomous RegionP.R. China
| | - Tingjie Ma
- Department of NeurologyNingxia Hui Autonomous Region People's HospitalYinchuan CityNingxia Hui Autonomous RegionP.R. China
| | - Xuqing Cao
- Department of NeurologyNingxia Hui Autonomous Region People's HospitalYinchuan CityNingxia Hui Autonomous RegionP.R. China
| |
Collapse
|
11
|
Understanding Acquired Brain Injury: A Review. Biomedicines 2022; 10:biomedicines10092167. [PMID: 36140268 PMCID: PMC9496189 DOI: 10.3390/biomedicines10092167] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 08/02/2022] [Accepted: 08/26/2022] [Indexed: 01/19/2023] Open
Abstract
Any type of brain injury that transpires post-birth is referred to as Acquired Brain Injury (ABI). In general, ABI does not result from congenital disorders, degenerative diseases, or by brain trauma at birth. Although the human brain is protected from the external world by layers of tissues and bone, floating in nutrient-rich cerebrospinal fluid (CSF); it remains susceptible to harm and impairment. Brain damage resulting from ABI leads to changes in the normal neuronal tissue activity and/or structure in one or multiple areas of the brain, which can often affect normal brain functions. Impairment sustained from an ABI can last anywhere from days to a lifetime depending on the severity of the injury; however, many patients face trouble integrating themselves back into the community due to possible psychological and physiological outcomes. In this review, we discuss ABI pathologies, their types, and cellular mechanisms and summarize the therapeutic approaches for a better understanding of the subject and to create awareness among the public.
Collapse
|
12
|
Kwon B, Song Y, Kim JG, Lee D, Lee SH, Cho YK, Kim JS, Suh DC. Preventive effects of ginseng against atherosclerosis and subsequent ischemic stroke: A randomized controlled trial (PEGASUS trial). J Ginseng Res 2022; 46:585-591. [PMID: 35818424 PMCID: PMC9270648 DOI: 10.1016/j.jgr.2021.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 10/30/2021] [Accepted: 11/04/2021] [Indexed: 11/30/2022] Open
Abstract
Background Korean Red Ginseng (KRG) extract has been shown to have beneficial effects in patients with atherosclerosis, suggesting that KRG extract may be effective in preventing subsequent ischemic stroke in patients with severe atherosclerosis. Methods This double-blind, placebo-controlled trial randomized patients with severe atherosclerosis in major intracranial arteries or extracranial carotid artery, to ginseng group and placebo group. They were given two 500-mg KRG tablets or identical placebo tablets twice daily for 12 months according to randomization. The primary endpoint was the composite of cerebral ischemic stroke and transient ischemic attack during 12 months after randomization. The secondary endpoints were change in volumetric blood flow of the intracranial vessels and the incidence of newly developed asymptomatic ischemic lesions. Any adverse events were monitored. Results Fifty-eight patients were randomized from June 2016 to June 2017, 29 to ginseng and 29 to placebo, and 52 (28 and 24, respectively) completed the study. One patient in the placebo group, but none in the ginseng group, experienced ischemic symptoms (p = 0.46). Changes in volumetric blood flow and the presence of ischemic brain lesions did not differ significantly in the two groups, and none of these patients experienced adverse drug reactions. Conclusion Ginseng was well tolerated by patients with severe atherosclerosis, with these patients showing good compliance with ginseng dosing. Ginseng did not show significant effects compared with placebo, although none of the ginseng-treated patients experienced ischemic events. Long-term studies in larger patient populations are required to test the effect of ginseng.
Collapse
Affiliation(s)
- Boseong Kwon
- Neurointervention Clinic, Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Republic of Korea
| | - Yunsun Song
- Neurointervention Clinic, Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Republic of Korea
| | - Joong-Goo Kim
- Department of Neurology, Jeju National University Hospital, Republic of Korea
| | - Dongwhane Lee
- Department of Neurology, Uijeongbu Eulji Medical Center, Eulji University School of Medicine, Republic of Korea
| | - Sang-hun Lee
- Department of Neurology, Korea University Ansan Hospital, Korea University College of Medicine, Republic of Korea
| | - Young-Keol Cho
- Department of Microbiology, University of Ulsan College of Medicine, Republic of Korea
| | - Jong S. Kim
- Department of Neurology, Asan Medical Center, University of Ulsan College of Medicine, Republic of Korea
| | - Dae Chul Suh
- Neurointervention Clinic, Department of Radiology, Asan Medical Center, University of Ulsan College of Medicine, Republic of Korea
| |
Collapse
|
13
|
Wang P, Ren Q, Shi M, Liu Y, Bai H, Chang YZ. Overexpression of Mitochondrial Ferritin Enhances Blood–Brain Barrier Integrity Following Ischemic Stroke in Mice by Maintaining Iron Homeostasis in Endothelial Cells. Antioxidants (Basel) 2022; 11:antiox11071257. [PMID: 35883748 PMCID: PMC9312053 DOI: 10.3390/antiox11071257] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 06/23/2022] [Accepted: 06/24/2022] [Indexed: 12/04/2022] Open
Abstract
Blood–brain barrier (BBB) breakdown, a characteristic feature of ischemic stroke, contributes to poor patient outcomes. Brain microvascular endothelial cells (BMVECs) are a key component of the BBB and dysfunction or death of these cells following cerebral ischemia reperfusion (I/R) injury can disrupt the BBB, leading to leukocyte infiltration, brain edema and intracerebral hemorrhage. We previously demonstrated that mitochondrial ferritin (FtMt) can alleviate I/R-induced neuronal ferroptosis by inhibiting inflammation-regulated iron deposition. However, whether FtMt is involved in BBB disruption during cerebral I/R is still unknown. In the present study, we found that FtMt expression in BMVECs is upregulated after I/R and overexpression of FtMt attenuates I/R-induced BBB disruption. Mechanistically, we found that FtMt prevents tight junction loss and apoptosis by inhibiting iron dysregulation and reactive oxygen species (ROS) accumulation in I/R-treated BMVECs. Chelating excess iron with deferoxamine alleviates apoptosis in the brain endothelial cell line bEnd.3 under oxygen glucose deprivation followed by reoxygenation (OGD/R) insult. In summary, our data identify a previously unexplored effect for FtMt in the BBB and provide evidence that iron-mediated oxidative stress in BMVECs is an early cause of BMVECs damage and BBB breakdown in ischemic stroke.
Collapse
Affiliation(s)
- Peina Wang
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China; (P.W.); (Q.R.); (M.S.); (Y.L.); (H.B.)
- Department of Histology and Embryology, College of Basic Medical Sciences, Hebei Medical University, Shijiazhuang 050017, China
| | - Qianqian Ren
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China; (P.W.); (Q.R.); (M.S.); (Y.L.); (H.B.)
| | - Mengtong Shi
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China; (P.W.); (Q.R.); (M.S.); (Y.L.); (H.B.)
| | - Yuanyuan Liu
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China; (P.W.); (Q.R.); (M.S.); (Y.L.); (H.B.)
| | - Huiyuan Bai
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China; (P.W.); (Q.R.); (M.S.); (Y.L.); (H.B.)
| | - Yan-Zhong Chang
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Science, Hebei Normal University, Shijiazhuang 050024, China; (P.W.); (Q.R.); (M.S.); (Y.L.); (H.B.)
- Correspondence: ; Tel./Fax: +86-311-80787539
| |
Collapse
|
14
|
Qiu M, Xu E, Zhan L. Epigenetic Regulations of Microglia/Macrophage Polarization in Ischemic Stroke. Front Mol Neurosci 2021; 14:697416. [PMID: 34707480 PMCID: PMC8542724 DOI: 10.3389/fnmol.2021.697416] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Accepted: 08/26/2021] [Indexed: 01/04/2023] Open
Abstract
Ischemic stroke is one of the leading causes of death and disability worldwide. Microglia/macrophages (MMs)-mediated neuroinflammation contributes significantly to the pathological process of ischemic brain injury. Microglia, serving as resident innate immune cells in the central nervous system, undergo pro-inflammatory phenotype or anti-inflammatory phenotype in response to the microenvironmental changes after cerebral ischemia. Emerging evidence suggests that epigenetics modifications, reversible modifications of the phenotype without changing the DNA sequence, could play a pivotal role in regulation of MM polarization. However, the knowledge of the mechanism of epigenetic regulations of MM polarization after cerebral ischemia is still limited. In this review, we present the recent advances in the mechanisms of epigenetics involved in regulating MM polarization, including histone modification, non-coding RNA, and DNA methylation. In addition, we discuss the potential of epigenetic-mediated MM polarization as diagnostic and therapeutic targets for ischemic stroke. It is valuable to identify the underlying mechanisms between epigenetics and MM polarization, which may provide a promising treatment strategy for neuronal damage after cerebral ischemia.
Collapse
Affiliation(s)
- Meiqian Qiu
- Institute of Neurosciences 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, China
| | - En Xu
- Institute of Neurosciences 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, China
| | - Lixuan Zhan
- Institute of Neurosciences 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, China
| |
Collapse
|
15
|
Mulder IA, van Bavel ET, de Vries HE, Coutinho JM. Adjunctive cytoprotective therapies in acute ischemic stroke: a systematic review. Fluids Barriers CNS 2021; 18:46. [PMID: 34666786 PMCID: PMC8524879 DOI: 10.1186/s12987-021-00280-1] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Accepted: 09/29/2021] [Indexed: 01/08/2023] Open
Abstract
With the introduction of endovascular thrombectomy (EVT), a new era for treatment of acute ischemic stroke (AIS) has arrived. However, despite the much larger recanalization rate as compared to thrombolysis alone, final outcome remains far from ideal. This raises the question if some of the previously tested neuroprotective drugs warrant re-evaluation, since these compounds were all tested in studies where large-vessel recanalization was rarely achieved in the acute phase. This review provides an overview of compounds tested in clinical AIS trials and gives insight into which of these drugs warrant a re-evaluation as an add-on therapy for AIS in the era of EVT. A literature search was performed using the search terms "ischemic stroke brain" in title/abstract, and additional filters. After exclusion of papers using pre-defined selection criteria, a total of 89 trials were eligible for review which reported on 56 unique compounds. Trial compounds were divided into 6 categories based on their perceived mode of action: systemic haemodynamics, excitotoxicity, neuro-inflammation, blood-brain barrier and vasogenic edema, oxidative and nitrosative stress, neurogenesis/-regeneration and -recovery. Main trial outcomes and safety issues are summarized and promising compounds for re-evaluation are highlighted. Looking at group effect, drugs intervening with oxidative and nitrosative stress and neurogenesis/-regeneration and -recovery appear to have a favourable safety profile and show the most promising results regarding efficacy. Finally, possible theories behind individual and group effects are discussed and recommendation for promising treatment strategies are described.
Collapse
Affiliation(s)
- I A Mulder
- Department of Biomedical Engineering and Physics, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands.
| | - E T van Bavel
- Department of Biomedical Engineering and Physics, Amsterdam Cardiovascular Sciences, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| | - H E de Vries
- Department of Molecular Cell Biology and Immunology, Amsterdam Neuroscience, Amsterdam UMC, Vrije Universiteit Amsterdam, Amsterdam, The Netherlands
| | - J M Coutinho
- Department of Neurology, Amsterdam Neuroscience, Amsterdam UMC, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
16
|
Fang Z, Wu D, Deng J, Yang Q, Zhang X, Chen J, Wang S, Hu S, Hou W, Ning S, Ding Y, Fan Z, Jiang Z, Kang J, Liu Y, Miao J, Ji X, Dong H, Xiong L. An MD2-perturbing peptide has therapeutic effects in rodent and rhesus monkey models of stroke. Sci Transl Med 2021; 13:13/597/eabb6716. [PMID: 34108252 DOI: 10.1126/scitranslmed.abb6716] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2020] [Revised: 12/08/2020] [Accepted: 03/26/2021] [Indexed: 12/21/2022]
Abstract
Studies have failed to translate more than 1000 experimental treatments from bench to bedside, leaving stroke as the second leading cause of death in the world. Thrombolysis within 4.5 hours is the recommended therapy for stroke and cannot be performed until neuroimaging is used to distinguish ischemic stroke from hemorrhagic stroke. Therefore, finding a common and critical therapeutic target for both ischemic and hemorrhagic stroke is appealing. Here, we report that the expression of myeloid differentiation protein 2 (MD2), which is traditionally regarded to be expressed only in microglia in the normal brain, was markedly increased in cortical neurons after stroke. We synthesized a small peptide, Trans-trans-activating (Tat)-cold-inducible RNA binding protein (Tat-CIRP), which perturbed the function of MD2 and strongly protected neurons against excitotoxic injury in vitro. In addition, systemic administration of Tat-CIRP or genetic deletion of MD2 induced robust neuroprotection against ischemic and hemorrhagic stroke in mice. Tat-CIRP reduced the brain infarct volume and preserved neurological function in rhesus monkeys 30 days after ischemic stroke. Tat-CIRP efficiently crossed the blood-brain barrier and showed a wide therapeutic index for stroke because no toxicity was detected when high doses were administered to the mice. Furthermore, we demonstrated that MD2 elicited neuronal apoptosis and necroptosis via a TLR4-independent, Sam68-related cascade. In summary, Tat-CIRP provides robust neuroprotection against stroke in rodents and gyrencephalic nonhuman primates. Further efforts should be made to translate these findings to treat both ischemic and hemorrhagic stroke in patients.
Collapse
Affiliation(s)
- Zongping Fang
- Department of Anesthesiology and Perioperative Medicine and Department of Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Di Wu
- Department of neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 10053, China
| | - Jiao Deng
- Department of Anesthesiology and Perioperative Medicine and Department of Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Qianzi Yang
- Department of Anesthesiology and Perioperative Medicine and Department of Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xijing Zhang
- Department of Anesthesiology and Perioperative Medicine and Department of Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jian Chen
- Department of neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 10053, China
| | - Shiquan Wang
- Department of Anesthesiology and Perioperative Medicine and Department of Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Sijun Hu
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Wugang Hou
- Department of Anesthesiology and Perioperative Medicine and Department of Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Siming Ning
- State Key Laboratory of Cancer Biology, Department of Biochemistry and Molecular Biology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Yi Ding
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Zhongmin Fan
- Department of Anesthesiology and Perioperative Medicine and Department of Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Zhenhua Jiang
- Department of Anesthesiology and Perioperative Medicine and Department of Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Junjun Kang
- Department of Neurobiology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Yingying Liu
- Department of Neurobiology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Jinlin Miao
- National Translational Science Center for Molecular Medicine and Department of Cell Biology, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| | - Xunming Ji
- Department of neurology and China-America Institute of Neuroscience, Xuanwu Hospital, Beijing Institute of Brain Disorders, Capital Medical University, Beijing 10053, China.
| | - Hailong Dong
- Department of Anesthesiology and Perioperative Medicine and Department of Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China.
| | - Lize Xiong
- Translational Research Institute of Brain and Brain-Like Intelligence and Department of Anesthesiology and Perioperative Medicine, Shanghai Fourth People's Hospital, Tongji University School of Medicine, Shanghai 200434, China. .,Department of Anesthesiology and Perioperative Medicine and Department of Critical Care Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi 710032, China
| |
Collapse
|
17
|
Hernández IH, Villa-González M, Martín G, Soto M, Pérez-Álvarez MJ. Glial Cells as Therapeutic Approaches in Brain Ischemia-Reperfusion Injury. Cells 2021; 10:1639. [PMID: 34208834 PMCID: PMC8305833 DOI: 10.3390/cells10071639] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/24/2021] [Accepted: 06/26/2021] [Indexed: 02/07/2023] Open
Abstract
Ischemic stroke is the second cause of mortality and the first cause of long-term disability constituting a serious socioeconomic burden worldwide. Approved treatments include thrombectomy and rtPA intravenous administration, which, despite their efficacy in some cases, are not suitable for a great proportion of patients. Glial cell-related therapies are progressively overcoming inefficient neuron-centered approaches in the preclinical phase. Exploiting the ability of microglia to naturally switch between detrimental and protective phenotypes represents a promising therapeutic treatment, in a similar way to what happens with astrocytes. However, the duality present in many of the roles of these cells upon ischemia poses a notorious difficulty in disentangling the precise pathways to target. Still, promoting M2/A2 microglia/astrocyte protective phenotypes and inhibiting M1/A1 neurotoxic profiles is globally rendering promising results in different in vivo models of stroke. On the other hand, described oligodendrogenesis after brain ischemia seems to be strictly beneficial, although these cells are the less studied players in the stroke paradigm and negative effects could be described for oligodendrocytes in the next years. Here, we review recent advances in understanding the precise role of mentioned glial cell types in the main pathological events of ischemic stroke, including inflammation, blood brain barrier integrity, excitotoxicity, reactive oxygen species management, metabolic support, and neurogenesis, among others, with a special attention to tested therapeutic approaches.
Collapse
Affiliation(s)
- Ivó H Hernández
- Genomic Instability Group, Spanish National Cancer Research Centre (CNIO), 28029 Madrid, Spain
- Center for Molecular Biology "Severo Ochoa" (CBMSO) UAM/CSIC, 28049 Madrid, Spain
- Networking Research Center on Neurodegenerative Diseases (CIBERNED), Instituto de Salud Carlos III, 28031 Madrid, Spain
| | - Mario Villa-González
- Center for Molecular Biology "Severo Ochoa" (CBMSO) UAM/CSIC, 28049 Madrid, Spain
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Gerardo Martín
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - Manuel Soto
- Center for Molecular Biology "Severo Ochoa" (CBMSO) UAM/CSIC, 28049 Madrid, Spain
- Departamento de Biología Molecular, Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| | - María José Pérez-Álvarez
- Center for Molecular Biology "Severo Ochoa" (CBMSO) UAM/CSIC, 28049 Madrid, Spain
- Departamento de Biología (Fisiología Animal), Facultad de Ciencias, Universidad Autónoma de Madrid, 28049 Madrid, Spain
| |
Collapse
|
18
|
Wang P, Cui Y, Ren Q, Yan B, Zhao Y, Yu P, Gao G, Shi H, Chang S, Chang YZ. Mitochondrial ferritin attenuates cerebral ischaemia/reperfusion injury by inhibiting ferroptosis. Cell Death Dis 2021; 12:447. [PMID: 33953171 PMCID: PMC8099895 DOI: 10.1038/s41419-021-03725-5] [Citation(s) in RCA: 130] [Impact Index Per Article: 32.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2020] [Revised: 04/15/2021] [Accepted: 04/16/2021] [Indexed: 02/08/2023]
Abstract
Ischaemic stroke is becoming the most common cerebral disease in aging populations, but the underlying molecular mechanism of the disease has not yet been fully elucidated. Increasing evidence has indicated that an excess of iron contributes to brain damage in cerebral ischaemia/reperfusion (I/R) injury. Although mitochondrial ferritin (FtMt) plays a critical role in iron homeostasis, the molecular function of FtMt in I/R remains unknown. We herein report that FtMt levels are upregulated in the ischaemic brains of mice. Mice lacking FtMt experience more severe brain damage and neurological deficits, accompanied by typical molecular features of ferroptosis, including increased lipid peroxidation and disturbed glutathione (GSH) after cerebral I/R. Conversely, FtMt overexpression reverses these changes. Further investigation shows that Ftmt ablation promotes I/R-induced inflammation and hepcidin-mediated decreases in ferroportin1, thus markedly increasing total and chelatable iron. The elevated iron consequently facilitates ferroptosis in the brain of I/R. In brief, our results provide evidence that FtMt plays a critical role in protecting against cerebral I/R-induced ferroptosis and subsequent brain damage, thus providing a new potential target for the treatment/prevention of ischaemic stroke.
Collapse
Affiliation(s)
- Peina Wang
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Science, Hebei Normal University, 050024, Shijiazhuang, Hebei Province, China
| | - Yanmei Cui
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Science, Hebei Normal University, 050024, Shijiazhuang, Hebei Province, China
| | - Qianqian Ren
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Science, Hebei Normal University, 050024, Shijiazhuang, Hebei Province, China
| | - Bingqi Yan
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Science, Hebei Normal University, 050024, Shijiazhuang, Hebei Province, China
| | - Yashuo Zhao
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Science, Hebei Normal University, 050024, Shijiazhuang, Hebei Province, China
- Scientific Research Center, Hebei University of Chinese Medicine, 050200, Shijiazhuang, Hebei Province, China
| | - Peng Yu
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Science, Hebei Normal University, 050024, Shijiazhuang, Hebei Province, China
| | - Guofen Gao
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Science, Hebei Normal University, 050024, Shijiazhuang, Hebei Province, China
| | - Honglian Shi
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Kansas, 1251 Wescoe Hall Drive, Malott Hall 5044, Lawrence, KS, 66045, USA
| | - Shiyang Chang
- College of basic medicine, Hebei Medical University, 050017, Shijiazhuang, Hebei Province, China.
| | - Yan-Zhong Chang
- Laboratory of Molecular Iron Metabolism, Key Laboratory of Animal Physiology, Biochemistry and Molecular Biology of Hebei Province, Ministry of Education Key Laboratory of Molecular and Cellular Biology, College of Life Science, Hebei Normal University, 050024, Shijiazhuang, Hebei Province, China.
| |
Collapse
|
19
|
Higher uric acid is associated with better discharge recovery and short-term outcome in stroke patients treated with thrombolysis. Neurol Sci 2020; 42:3225-3231. [PMID: 33241534 DOI: 10.1007/s10072-020-04919-z] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Accepted: 11/16/2020] [Indexed: 12/17/2022]
Abstract
BACKGROUND Uric acid (UA) possesses antioxidant features and potential neuroprotective effects. However, conflicting results regarding the association between serum uric acid (SUA) levels and the prognosis of stroke have been obtained. We aimed to assess whether SUA is related to discharge recovery and short-term outcomes in patients who underwent thrombolysis therapy. METHODS We recruited 393 consecutive patients who were diagnosed with acute ischaemic stroke (AIS) and treated with thrombolysis. The demographic information, including sex and age, was collected. Haematology tests, including SUA, fasting plasma glucose (FPG), and blood lipid parameters, were performed under fasting conditions the morning after admission. The modified Rankin Scale (mRS) was used to assess the functional outcome of patients at discharge and 3 months after onset. RESULTS A negative correlation was observed between the levels of SUA and the National Institute of Health Stroke Scale (NIHSS) score at discharge (r = - 0.171, P = 0.003). Additionally, a positive correlation was observed between the levels of SUA and the difference between the baseline NIHSS and discharge NIHSS (r = 0.118, P = 0.032). The levels of SUA in the patients with good outcomes (353.76 ± 93.05) were higher than those in the patients with poor outcomes (301.99 ± 92.24; P = 0.015) at 3 months. The multivariate logistic regression analysis demonstrated that a higher SUA level (odds ratio 0.988, 95% confidence interval 0.985-0.991, P = 0.002) was an independent predictor of a good outcome at 3 months. CONCLUSION Higher SUA levels were associated with better discharge recovery and 3-month outcomes in patients with ischaemic stroke who received thrombolysis.
Collapse
|
20
|
Voelz C, Habib P, Köberlein S, Beyer C, Slowik A. Alteration of miRNA Biogenesis Regulating Proteins in the Human Microglial Cell Line HMC-3 After Ischemic Stress. Mol Neurobiol 2020; 58:1535-1549. [PMID: 33210205 DOI: 10.1007/s12035-020-02210-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 11/12/2020] [Indexed: 12/21/2022]
Abstract
MicroRNAs (miRNA) are small noncoding sequences that control apoptosis, proliferation, and neuroinflammatory pathways in microglia cells. The expression of distinct miRNAs is altered after ischemia in the brain. Only minor information is available about the biogenesis and maturation of miRNAs after ischemia. We aimed at examining the impact of oxygen-glucose deprivation (OGD) and hydrogen peroxide (H2O2)-induced stress on the expression of miRNA regulating proteins such as DROSHA, DGCR8, XPO5, DICER, TARBP2, and AGO2 in the cultured human microglial cell line HMC-3 (human microglial cell line clone 3). OGD duration of 2.5 h or H2O2 stimulation at a concentration of 100 μM for 24 h resulted in a marked increase of the hypoxia sensor hypoxia-inducible factor1-α in HMC-3 cells. These treatments also led to an upregulation of DROSHA, DICER1, and AGO2 detected by semiquantitative real-time PCR (qrtPCR). XPO5 and TARBP2 were only upregulated after stimulation with H2O2, while DGCR8 responded only to OGD. We found elevated DICER1, DROSHA, and AGO2 protein levels by western blot and immunohistochemistry staining. Interestingly, the latter also exposed a colocalization of AGO2 with stress granules (G3BP1) after OGD. Our data indicate that DICER, DROSHA, and AGO2 are induced in microglial cells under hypoxia-like conditions. It might be speculated that their inductions might increase the miRNA synthesis rate. Future studies should investigate this correlation to determine which miRNAs are preferably expressed by microglia cells after ischemia and which functions they could exert.
Collapse
Affiliation(s)
- Clara Voelz
- Institute of Neuroanatomy, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany
| | - Pardes Habib
- Department of Neurology, Medical Faculty, RWTH Aachen University, Aachen, Germany
| | - Stefan Köberlein
- Institute of Neuroanatomy, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany
| | - Cordian Beyer
- Institute of Neuroanatomy, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany
- JARA-Brain, RWTH Aachen University, Aachen, Germany
| | - Alexander Slowik
- Institute of Neuroanatomy, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany.
- Department of Anatomy and Cell Biology, Medical Faculty, RWTH Aachen University, 52074, Aachen, Germany.
| |
Collapse
|
21
|
Mallah K, Couch C, Borucki DM, Toutonji A, Alshareef M, Tomlinson S. Anti-inflammatory and Neuroprotective Agents in Clinical Trials for CNS Disease and Injury: Where Do We Go From Here? Front Immunol 2020; 11:2021. [PMID: 33013859 PMCID: PMC7513624 DOI: 10.3389/fimmu.2020.02021] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 07/27/2020] [Indexed: 02/06/2023] Open
Abstract
Neurological disorders are major contributors to death and disability worldwide. The pathology of injuries and disease processes includes a cascade of events that often involve molecular and cellular components of the immune system and their interaction with cells and structures within the central nervous system. Because of this, there has been great interest in developing neuroprotective therapeutic approaches that target neuroinflammatory pathways. Several neuroprotective anti-inflammatory agents have been investigated in clinical trials for a variety of neurological diseases and injuries, but to date the results from the great majority of these trials has been disappointing. There nevertheless remains great interest in the development of neuroprotective strategies in this arena. With this in mind, the complement system is being increasingly discussed as an attractive therapeutic target for treating brain injury and neurodegenerative conditions, due to emerging data supporting a pivotal role for complement in promoting multiple downstream activities that promote neuroinflammation and degeneration. As we move forward in testing additional neuroprotective and immune-modulating agents, we believe it will be useful to review past trials and discuss potential factors that may have contributed to failure, which will assist with future agent selection and trial design, including for complement inhibitors. In this context, we also discuss inhibition of the complement system as a potential neuroprotective strategy for neuropathologies of the central nervous system.
Collapse
Affiliation(s)
- Khalil Mallah
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
| | - Christine Couch
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Health Sciences and Research, College of Health Professions, Medical University of South Carolina, Charleston, SC, United States
| | - Davis M. Borucki
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, United States
- Medical Scientist Training Program, Medical University of South Carolina, Charleston, SC, United States
| | - Amer Toutonji
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Neurosciences, Medical University of South Carolina, Charleston, SC, United States
- Medical Scientist Training Program, Medical University of South Carolina, Charleston, SC, United States
| | - Mohammed Alshareef
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Department of Neurological Surgery, Medical University of South Carolina, Charleston, SC, United States
| | - Stephen Tomlinson
- Department of Microbiology and Immunology, Medical University of South Carolina, Charleston, SC, United States
- Ralph Johnson VA Medical Center, Charleston, SC, United States
| |
Collapse
|
22
|
Chen J, Wang Y, Wu J, Yang J, Li M, Chen Q. The Potential Value of Targeting Ferroptosis in Early Brain Injury After Acute CNS Disease. Front Mol Neurosci 2020; 13:110. [PMID: 32625062 PMCID: PMC7314952 DOI: 10.3389/fnmol.2020.00110] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 05/26/2020] [Indexed: 02/06/2023] Open
Abstract
Acute central nervous system (CNS) disease is very common and with high mortality. Many basic studies have confirmed the molecular mechanism of early brain injury (EBI) after acute CNS disease. Neuron death and dysfunction are important reasons for the neurological dysfunction in patients with acute CNS disease. Ferroptosis is a nonapoptotic form of cell death, the classical characteristic of which is based on the iron-dependent accumulation of toxic lipid reactive oxygen species. Previous studies have indicated that this mechanism is critical in the cell death events observed in many diseases, including cancer, tumor resistance, Alzheimer’s disease, Parkinson’s disease, stroke, and intracerebral hemorrhage (ICH). Ferroptosis may also play a very important role in EBI after acute CNS disease. Unresolved issues include the relationship between ferroptosis and other forms of cell death after acute CNS disease, the specific molecular mechanisms of EBI, the strategies to activate or inhibit ferroptosis to achieve desirable attenuation of EBI, and the need to find new molecular markers of ferroptosis that can be used to detect and study this process in vivo after acute CNS disease.
Collapse
Affiliation(s)
- Junhui Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China.,Department of Neurosurgery, 904th Hospital of Joint Logistic Support Force of PLA, Wuxi Clinical College of Anhui Medical University, Wuxi, China
| | - Yuhai Wang
- Department of Neurosurgery, 904th Hospital of Joint Logistic Support Force of PLA, Wuxi Clinical College of Anhui Medical University, Wuxi, China
| | - Jiyun Wu
- Department of Orthopedic, 904th Hospital of Joint Logistic Support Force of PLA, Wuxi Clinical College of Anhui Medical University, Wuxi, China
| | - Jiaji Yang
- Department of Orthopedic, 904th Hospital of Joint Logistic Support Force of PLA, Wuxi Clinical College of Anhui Medical University, Wuxi, China
| | - Mingchang Li
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| | - Qianxue Chen
- Department of Neurosurgery, Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
23
|
Brain-Derived Neurotrophic Factor and Its Potential Therapeutic Role in Stroke Comorbidities. Neural Plast 2020; 2020:1969482. [PMID: 32399020 PMCID: PMC7204205 DOI: 10.1155/2020/1969482] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2019] [Revised: 10/14/2019] [Accepted: 11/08/2019] [Indexed: 12/17/2022] Open
Abstract
With the rise in the aging global population, stroke comorbidities have become a serious health threat and a tremendous economic burden on human society. Current therapeutic strategies mainly focus on protecting neurons from cytotoxic damage at the acute phase upon stroke onset, which not only is a difficult way to ameliorate stroke symptoms but also presents a challenge for the patients to receive effective treatment in time. The brain-derived neurotrophic factor (BDNF) is the most abundant neurotrophin in the adult brain, which possesses a remarkable capability to repair brain damage. Recent promising preclinical outcomes have made BDNF a popular late-stage target in the development of novel stroke treatments. In this review, we aim to summarize the latest progress in the understanding of the cellular/molecular mechanisms underlying stroke pathogenesis, current strategies and difficulties in drug development, the mechanism of BDNF action in poststroke neurorehabilitation and neuroplasticity, and recent updates in novel therapeutic methods.
Collapse
|
24
|
Dhir N, Medhi B, Prakash A, Goyal MK, Modi M, Mohindra S. Pre-clinical to Clinical Translational Failures and Current Status of Clinical Trials in Stroke Therapy: A Brief Review. Curr Neuropharmacol 2020; 18:596-612. [PMID: 31934841 PMCID: PMC7457423 DOI: 10.2174/1570159x18666200114160844] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Revised: 10/31/2019] [Accepted: 12/28/2019] [Indexed: 12/16/2022] Open
Abstract
In stroke (cerebral ischemia), despite continuous efforts both at the experimental and clinical level, the only approved pharmacological treatment has been restricted to tissue plasminogen activator (tPA). Stroke is the leading cause of functional disability and mortality throughout worldwide. Its pathophysiology starts with energy pump failure, followed by complex signaling cascade that ultimately ends in neuronal cell death. Ischemic cascade involves excessive glutamate release followed by raised intracellular sodium and calcium influx along with free radicals' generation, activation of inflammatory cytokines, NO synthases, lipases, endonucleases and other apoptotic pathways leading to cell edema and death. At the pre-clinical stage, several agents have been tried and proven as an effective neuroprotectant in animal models of ischemia. However, these agents failed to show convincing results in terms of efficacy and safety when the trials were conducted in humans following stroke. This article highlights the various agents which have been tried in the past but failed to translate into stroke therapy along with key points that are responsible for the lagging of experimental success to translational failure in stroke treatment.
Collapse
Affiliation(s)
| | - Bikash Medhi
- Address correspondence to this author at the Department of Pharmacology, Research Block B, 4th Floor, Room no 4043, Postgraduate Institute of Medical Education & Research (PGIMER), Chandigarh, 160012, India; E-mail:
| | | | | | | | | |
Collapse
|
25
|
Kobayashi S, Fukuma S, Ikenoue T, Fukuhara S, Kobayashi S. Effect of Edaravone on Neurological Symptoms in Real-World Patients With Acute Ischemic Stroke. Stroke 2019; 50:1805-1811. [PMID: 31164072 DOI: 10.1161/strokeaha.118.024351] [Citation(s) in RCA: 57] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Background and Purpose- In Japan, nearly half of ischemic stroke patients receive edaravone for acute treatment. The purpose of this study was to assess the effect of edaravone on neurological symptoms in patients with ischemic stroke stratified by stroke subtype. Methods- Study subjects were 61 048 patients aged 18 years or older who were hospitalized ≤14 days after onset of an acute ischemic stroke and were registered in the Japan Stroke Data Bank, a hospital-based multicenter stroke registration database, between June 2001 and July 2013. Patients were stratified according to ischemic stroke subtype (large-artery atherosclerosis, cardioembolism, small-vessel occlusion, and cryptogenic/undetermined) and then divided into 2 groups (edaravone-treated and no edaravone). Neurological symptoms were evaluated using the National Institutes of Health Stroke Scale (NIHSS). The primary outcome was changed in neurological symptoms during the hospital stay (ΔNIHSS=NIHSS score at discharge-NIHSS score at admission). Data were analyzed using multivariate linear regression with inverse probability of treatment weighting after adjusting for the following confounding factors: age, gender, and systolic and diastolic blood pressure at the start of treatment, NIHSS score at admission, time from stroke onset to hospital admission, infarct size, comorbidities, concomitant medication, clinical department, history of smoking, alcohol consumption, and history of stroke. Results- After adjusting for potential confounders, the improvement in NIHSS score from admission to discharge was greater in the edaravone-treated group than in the no edaravone group for all ischemic stroke subtypes (mean [95% CI] difference in ΔNIHSS: -0.46 [-0.75 to -0.16] for large-artery atherosclerosis, -0.64 [-1.09 to -0.2] for cardioembolism, and -0.25 [-0.4 to -0.09] for small-vessel occlusion). Conclusions- For any ischemic stroke subtype, edaravone use (compared with no use) was associated with a greater improvement in neurological symptoms, although the difference was small (<1 point NIHSS) and of limited clinical significance.
Collapse
Affiliation(s)
- Susumu Kobayashi
- From the Department of General Internal Medicine, Fukushima Medical University, Japan (Susumu Kobayashi)
| | - Shingo Fukuma
- Human Health Sciences, Kyoto University Graduate School of Medicine, Japan (S.F., T.I.)
| | - Tatsuyoshi Ikenoue
- Human Health Sciences, Kyoto University Graduate School of Medicine, Japan (S.F., T.I.)
| | - Shunichi Fukuhara
- Department of Healthcare Epidemiology, Kyoto University Graduate School of Medicine and Public Health, Japan (S.F.)
| | - Shotai Kobayashi
- Shimane University, School of Medicine, Japan (Shotai Kobayashi)
| |
Collapse
|
26
|
Modern Concepts in Regenerative Therapy for Ischemic Stroke: From Stem Cells for Promoting Angiogenesis to 3D-Bioprinted Scaffolds Customized via Carotid Shear Stress Analysis. Int J Mol Sci 2019; 20:ijms20102574. [PMID: 31130624 PMCID: PMC6566983 DOI: 10.3390/ijms20102574] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2019] [Revised: 05/20/2019] [Accepted: 05/22/2019] [Indexed: 02/06/2023] Open
Abstract
Ischemic stroke is associated with a tremendous economic and societal burden, and only a few therapies are currently available for the treatment of this devastating disease. The main therapeutic approaches used nowadays for the treatment of ischemic brain injury aim to achieve reperfusion, neuroprotection and neurorecovery. Therapeutic angiogenesis also seems to represent a promising tool to improve the prognosis of cerebral ischemia. This review aims to present the modern concepts and the current status of regenerative therapy for ischemic stroke and discuss the main results of major clinical trials addressing the effectiveness of stem cell therapy for achieving neuroregeneration in ischemic stroke. At the same time, as a glimpse into the future, this article describes modern concepts for stroke prevention, such as the implantation of bioprinted scaffolds seeded with stem cells, whose 3D geometry is customized according to carotid shear stress.
Collapse
|
27
|
Gu B, Piebalgs A, Huang Y, Longstaff C, Hughes AD, Chen R, Thom SA, Xu XY. Mathematical Modelling of Intravenous Thrombolysis in Acute Ischaemic stroke: Effects of Dose Regimens on Levels of Fibrinolytic Proteins and Clot Lysis Time. Pharmaceutics 2019; 11:E111. [PMID: 30866489 PMCID: PMC6471481 DOI: 10.3390/pharmaceutics11030111] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2019] [Revised: 02/28/2019] [Accepted: 03/03/2019] [Indexed: 12/19/2022] Open
Abstract
Thrombolytic therapy is one of the medical procedures in the treatment of acute ischaemic stroke (AIS), whereby the tissue plasminogen activator (tPA) is intravenously administered to dissolve the obstructive blood clot. The treatment of AIS by thrombolysis can sometimes be ineffective and it can cause serious complications, such as intracranial haemorrhage (ICH). In this study, we propose an efficient mathematical modelling approach that can be used to evaluate the therapeutic efficacy and safety of thrombolysis in various clinically relevant scenarios. Our model combines the pharmacokinetics and pharmacodynamics of tPA with local clot lysis dynamics. By varying the drug dose, bolus-infusion delay time, and bolus-infusion ratio, with the FDA approved dosing protocol serving as a reference, we have used the model to simulate 13 dose regimens. Simulation results are compared for temporal concentrations of fibrinolytic proteins in plasma and the time that is taken to achieve recanalisation. Our results show that high infusion rates can cause the rapid degradation of plasma fibrinogen, indicative of increased risk for ICH, but they do not necessarily lead to fast recanalisation. In addition, a bolus-infusion delay results in an immediate drop in plasma tPA concentration, which prolongs the time to achieve recanalisation. Therefore, an optimal administration regimen should be sought by keeping the tPA level sufficiently high throughout the treatment and maximising the lysis rate while also limiting the degradation of fibrinogen in systemic plasma. This can be achieved through model-based optimisation in the future.
Collapse
Affiliation(s)
- Boram Gu
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, UK.
| | - Andris Piebalgs
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, UK.
| | - Yu Huang
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, UK.
| | - Colin Longstaff
- Biotherapeutics Section, National Institute for Biological Standards and Control, South Mimms, Hertfordshire EN6 3QG, UK.
| | - Alun D Hughes
- Institute of Cardiovascular Science, University College London, London WC1E 6DD, UK.
- MRC Unit for Lifelong Health and Ageing at University College London, London WC1B 5JU, UK.
| | - Rongjun Chen
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, UK.
| | - Simon A Thom
- National Heart & Lung Institute, Imperial College London, Hammersmith Campus, London W12 0NN, UK.
| | - Xiao Yun Xu
- Department of Chemical Engineering, Imperial College London, South Kensington Campus, London SW7 2AZ, UK.
| |
Collapse
|
28
|
Meythaler J, Fath J, Fuerst D, Zokary H, Freese K, Martin HB, Reineke J, Peduzzi-Nelson J, Roskos PT. Safety and feasibility of minocycline in treatment of acute traumatic brain injury. Brain Inj 2019; 33:679-689. [PMID: 30744442 DOI: 10.1080/02699052.2019.1566968] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
BACKGROUND Minocycline is a pleomorphic neuroprotective agent well studied in animal models of traumatic brain injury (TBI) and brain ischemia. METHODS To test the hypothesis that administration of minocycline in moderate to severe TBI (Glasgow Coma Score 3-12). Fifteen patients were enrolled in a two-dose escalation study of minocycline to evaluate the safety of twice the recommended antibiotic dosage; tier 1 n = 7 at a loading dose of 800 mg followed by 200 mg twice a day (BID) for 7 days; tier 2 n = 8 at a loading dose of 800 mg followed by 400 mg BID for 7 days. RESULTS The mean initial GCS was 5.6 for Tier 1 patients and 5.4 for Tier 2. The Disability Rating Scale (DRS) had a trend towards improvement with the higher dose 12.5 SD ± 7.7 (N = 5) for Tier 1 at 4 weeks and 8.5 SD ± 9.9 at week 12 (N = 5), whereas for Tier 2 it was 9.7 ± 6.9 (N = 6) for week 4 and 6.0 SD ± 6.1 (N = 7) for week 12 (p = .251 repeated measures ANOVA). Liver function tests increased but resolved after the first week and there were no infections. CONCLUSIONS Minocycline was safe for moderate to severe TBI at a dose twice that as recommended for treatment of infection. The higher dose did trend towards an improved outcome.
Collapse
Affiliation(s)
- Jay Meythaler
- a Department of Physical Medicine and Rehabilitation-Oakwood, School of Medicine , Wayne State University , Taylor , MI , USA
| | - John Fath
- b Department of Surgery - Trauma Division , Oakwood Dearborn Hospital , Dearborn , Michigan
| | - Darren Fuerst
- a Department of Physical Medicine and Rehabilitation-Oakwood, School of Medicine , Wayne State University , Taylor , MI , USA
| | - Hashem Zokary
- a Department of Physical Medicine and Rehabilitation-Oakwood, School of Medicine , Wayne State University , Taylor , MI , USA
| | - Kristina Freese
- a Department of Physical Medicine and Rehabilitation-Oakwood, School of Medicine , Wayne State University , Taylor , MI , USA
| | - Heidi Baird Martin
- a Department of Physical Medicine and Rehabilitation-Oakwood, School of Medicine , Wayne State University , Taylor , MI , USA
| | - Joshua Reineke
- c School of Pharmacy , South Dakota State University , Dearborn , Michigan
| | - Jean Peduzzi-Nelson
- a Department of Physical Medicine and Rehabilitation-Oakwood, School of Medicine , Wayne State University , Taylor , MI , USA
| | - P Tyler Roskos
- a Department of Physical Medicine and Rehabilitation-Oakwood, School of Medicine , Wayne State University , Taylor , MI , USA
| |
Collapse
|
29
|
Kikuchi K, Setoyama K, Tanaka E, Otsuka S, Terashi T, Nakanishi K, Takada S, Sakakima H, Ampawong S, Kawahara KI, Nagasato T, Hosokawa K, Harada Y, Yamamoto M, Kamikokuryo C, Kiyama R, Morioka M, Ito T, Maruyama I, Tancharoen S. Uric acid enhances alteplase-mediated thrombolysis as an antioxidant. Sci Rep 2018; 8:15844. [PMID: 30367108 PMCID: PMC6203847 DOI: 10.1038/s41598-018-34220-1] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2018] [Accepted: 10/15/2018] [Indexed: 01/01/2023] Open
Abstract
Uric acid (UA) therapy may prevent early ischemic worsening after acute stroke in thrombolysis patients. The aim of this study was to examine the influence of UA on the thrombolytic efficacy of alteplase in human blood samples by measuring thrombolysis under flow conditions using a newly developed microchip-based flow-chamber assay. Human blood samples from healthy volunteers were exposed to UA, alteplase, or a combination of UA and alteplase. Whole blood and platelet-rich plasma were perfused over a collagen- and thromboplastin-coated microchip, and capillary occlusion was monitored with a video microscope and flow-pressure sensor. The area under the curve (extent of thrombogenesis or thrombolysis) at 30 minutes was 92% lower in the UA-alteplase-treated group compared with the alteplase-treated group. D-dimers were measured to evaluate these effects in human platelet-poor plasma samples. Although hydrogen peroxide significantly decreased the elevation of D-dimers by alteplase, UA significantly inhibited the effect of hydrogen peroxide. Meanwhile, rat models of thromboembolic cerebral ischemia were treated with either alteplase or UA-alteplase combination therapy. Compared with alteplase alone, the combination therapy reduced the infarct volume and inhibited haemorrhagic transformation. UA enhances alteplase-mediated thrombolysis, potentially by preventing oxidative stress, which inhibits fibrinolysis by alteplase in thrombi.
Collapse
Affiliation(s)
- Kiyoshi Kikuchi
- Division of Brain Science, Department of Physiology, Kurume University School of Medicine, Kurume, Japan.,Department of Neurosurgery, Kurume University School of Medicine, Kurume, Japan.,Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, Japan.,Department of Pharmacology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand
| | - Kentaro Setoyama
- Natural Science Center for Research and Education, Division of Laboratory Animal Science, Kagoshima University, Kagoshima, Japan
| | - Eiichiro Tanaka
- Division of Brain Science, Department of Physiology, Kurume University School of Medicine, Kurume, Japan
| | - Shotaro Otsuka
- Course of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, Kagoshima, Japan
| | - Takuto Terashi
- Course of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, Kagoshima, Japan
| | - Kazuki Nakanishi
- Course of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, Kagoshima, Japan
| | - Seiya Takada
- Course of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, Kagoshima, Japan
| | - Harutoshi Sakakima
- Course of Physical Therapy, School of Health Sciences, Faculty of Medicine, Kagoshima University, Kagoshima, Japan
| | - Sumate Ampawong
- Department of Tropical Pathology, Faculty of Tropical Medicine, (S.A.), Mahidol University, Bangkok, Thailand
| | - Ko-Ichi Kawahara
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, Japan.,Laboratory of Functional Foods, Department of Biomedical Engineering, Osaka Institute of Technology, Osaka, Japan
| | - Tomoka Nagasato
- Research Institute, Fujimori Kogyo Co., Yokohama, Kanagawa, Japan
| | - Kazuya Hosokawa
- Research Institute, Fujimori Kogyo Co., Yokohama, Kanagawa, Japan
| | - Yoichiro Harada
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, Japan
| | - Mika Yamamoto
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, Japan
| | - Chinatsu Kamikokuryo
- Department of Emergency and Critical Care Medicine, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, Japan
| | - Ryoji Kiyama
- School of Health Sciences, Faculty of Medicine, Kagoshima University, Kagoshima, Japan
| | - Motohiro Morioka
- Department of Neurosurgery, Kurume University School of Medicine, Kurume, Japan
| | - Takashi Ito
- Department of Emergency and Critical Care Medicine, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, Japan
| | - Ikuro Maruyama
- Department of Systems Biology in Thromboregulation, Kagoshima University Graduate School of Medical and Dental Science, Kagoshima, Japan
| | - Salunya Tancharoen
- Department of Pharmacology, Faculty of Dentistry, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
30
|
Khoury N, Xu J, Stegelmann SD, Jackson CW, Koronowski KB, Dave KR, Young JI, Perez-Pinzon MA. Resveratrol Preconditioning Induces Genomic and Metabolic Adaptations within the Long-Term Window of Cerebral Ischemic Tolerance Leading to Bioenergetic Efficiency. Mol Neurobiol 2018; 56:4549-4565. [PMID: 30343466 DOI: 10.1007/s12035-018-1380-6] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2018] [Accepted: 10/04/2018] [Indexed: 01/23/2023]
Abstract
Neuroprotective agents administered post-cerebral ischemia have failed so far in the clinic to promote significant recovery. Thus, numerous efforts were redirected toward prophylactic approaches such as preconditioning as an alternative therapeutic strategy. Our laboratory has revealed a novel long-term window of cerebral ischemic tolerance mediated by resveratrol preconditioning (RPC) that lasts for 2 weeks in mice. To identify its mediators, we conducted an RNA-seq experiment on the cortex of mice 2 weeks post-RPC, which revealed 136 differentially expressed genes. The majority of genes (116/136) were downregulated upon RPC and clustered into biological processes involved in transcription, synaptic signaling, and neurotransmission. The downregulation in these processes was reminiscent of metabolic depression, an adaptation used by hibernating animals to survive severe ischemic states by downregulating energy-consuming pathways. Thus, to assess metabolism, we used a neuronal-astrocytic co-culture model and measured the cellular respiration rate at the long-term window post-RPC. Remarkably, we observed an increase in glycolysis and mitochondrial respiration efficiency upon RPC. We also observed an increase in the expression of genes involved in pyruvate uptake, TCA cycle, and oxidative phosphorylation, all of which indicated an increased reliance on energy-producing pathways. We then revealed that these nuclear and mitochondrial adaptations, which reduce the reliance on energy-consuming pathways and increase the reliance on energy-producing pathways, are epigenetically coupled through acetyl-CoA metabolism and ultimately increase baseline ATP levels. This increase in ATP would then allow the brain, a highly metabolic organ, to endure prolonged durations of energy deprivation encountered during cerebral ischemia.
Collapse
Affiliation(s)
- Nathalie Khoury
- Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA.,Department of Neurology, University of Miami, Miller School of Medicine, P.O. Box 016960, Miami, FL, 33101, USA.,Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
| | - Jing Xu
- Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA.,Department of Neurology, University of Miami, Miller School of Medicine, P.O. Box 016960, Miami, FL, 33101, USA.,Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
| | - Samuel D Stegelmann
- Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA.,Department of Neurology, University of Miami, Miller School of Medicine, P.O. Box 016960, Miami, FL, 33101, USA
| | - Charles W Jackson
- Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA.,Department of Neurology, University of Miami, Miller School of Medicine, P.O. Box 016960, Miami, FL, 33101, USA.,Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
| | - Kevin B Koronowski
- Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA.,Department of Neurology, University of Miami, Miller School of Medicine, P.O. Box 016960, Miami, FL, 33101, USA.,Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
| | - Kunjan R Dave
- Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA.,Department of Neurology, University of Miami, Miller School of Medicine, P.O. Box 016960, Miami, FL, 33101, USA.,Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
| | - Juan I Young
- Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA.,John P. Hussman Institute for Human Genomics, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA.,Department of Human Genetics, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA
| | - Miguel A Perez-Pinzon
- Cerebral Vascular Disease Research Laboratories, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA. .,Department of Neurology, University of Miami, Miller School of Medicine, P.O. Box 016960, Miami, FL, 33101, USA. .,Neuroscience Program, University of Miami Leonard M. Miller School of Medicine, Miami, FL, 33136, USA.
| |
Collapse
|
31
|
Suvanish Kumar VS, Pretorius E, Rajanikant GK. The Synergistic Combination of Everolimus and Paroxetine Exerts Post-ischemic Neuroprotection In Vitro. Cell Mol Neurobiol 2018; 38:1383-1397. [PMID: 30062636 PMCID: PMC11481845 DOI: 10.1007/s10571-018-0605-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Accepted: 07/24/2018] [Indexed: 01/08/2023]
Abstract
Ischemic stroke is a debilitating multi-factorial cerebrovascular disorder, representing an area of tremendous unmet medical need. Combination treatment has been proposed as a promising therapeutic approach towards combating ischemic stroke. The present study employs in vitro oxygen glucose deprivation (OGD) model to evaluate the post-ischemic neuroprotective efficacy of Everolimus and Paroxetine, alone and in combination. Post-OGD treatment with Everolimus and Paroxetine, alone or in combination, significantly improved the cell survival (~ 80%) when compared to the cells subjected to ischemic injury alone. The individual neuroprotective doses of Everolimus and Paroxetine were found to be at 6.25 and 25 nM, respectively. Whereas, the synergistic neuroprotective dose for Everolimus:Paroxetine was 2:10 nM, calculated using the Chou-Talalay combination index and other four mathematical models. The synergistic combination dose downregulated neuroinflammatory genes (Tnf-α, Il1b, Nf-κB, and iNos) and upregulated the neuroprotective genes (Bcl-2, Bcl-xl, Hif-1, and Epo). The mitochondrial functioning and ROS neutralizing ability increased with combination treatment. Further, the active role of nitric oxide synthase and calmodulin were revealed while exploring the bio-activity of Everolimus and Paroxetine through network pharmacology. The present study for the first time demonstrates the synergistic post-ischemic neuroprotective efficacy of combination treatment with Everolimus and Paroxetine in vitro. Taken together, these findings clearly suggest that Everolimus in combination with Paroxetine may represent a promising therapeutic strategy for the treatment of ischemic stroke, further supporting the combination treatment strategy for this debilitating disorder.
Collapse
Affiliation(s)
- V S Suvanish Kumar
- School of Biotechnology, National Institute of Technology Calicut, Calicut, 673601, India
| | - Etheresia Pretorius
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch Private Bag X1, Matieland, 7602, South Africa
| | - G K Rajanikant
- School of Biotechnology, National Institute of Technology Calicut, Calicut, 673601, India.
| |
Collapse
|
32
|
Chen Z, Bai S, Hu Q, Shen P, Wang T, Liang Z, Wang W, Qi X, Xie P. Ginkgo biloba extract and its diterpene ginkgolide constituents ameliorate the metabolic disturbances caused by recombinant tissue plasminogen activator in rat prefrontal cortex. Neuropsychiatr Dis Treat 2018; 14:1755-1772. [PMID: 30013348 PMCID: PMC6037272 DOI: 10.2147/ndt.s167448] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
INTRODUCTION Although recombinant tissue plasminogen activator (rtPA) is a widely used therapy in patients with acute ischemic stroke, rtPA-induced toxicity or its adverse effects have been reported in our previous studies. However, Ginkgo biloba extract (GBE) may provide neuroprotective effects against rtPA-induced toxicity. Thus, in the present study, we investigated whether a single administration of rtPA caused neurotoxicity in the prefrontal cortex (PFC) of rats and determined whether GBE or its diterpene ginkgolide (DG) constituents were neuroprotective against any rtPA-induced toxicity. MATERIALS AND METHODS We randomly divided adult Sprague-Dawley rats into four groups that were intravenously administered saline, rtPA, rtPA+DG, or rtPA+GBE. The rats were sacrificed 24 hours later and the whole brain removed. A gas chromatography-mass spectrometry metabolomic approach was used to detect molecular changes in the PFC among the groups. Multivariate statistical and pathway analyses were used to determine the relevant metabolites as well as their functions and pathways. RESULTS We found 32 metabolites differentially altered in the four groups that were primarily involved in neurotransmitter, amino acid, energy, lipid, and nucleotide metabolism. Our results indicated that a single rtPA administration caused metabolic disturbances in the PFC. Both GBE and DG effectively ameliorated these rtPA-induced disturbances, although DG better controlled the rtPA-induced glutamate and aspartate excitotoxicity and the activation of NMDA receptor. CONCLUSION Our results provide important novel mechanistic insights into the adverse effects of rtPA and offer directions for future exploration on the thrombolytic effects of rtPA combined with the administration of DG or GBE for the treatment of acute ischemic stroke in humans.
Collapse
Affiliation(s)
- Zhi Chen
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China,
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China,
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
| | - Shunjie Bai
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China,
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China,
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
- Key Laboratory of Laboratory Medical Diagnostics of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China,
| | - Qingchuan Hu
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China,
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China,
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
- Key Laboratory of Laboratory Medical Diagnostics of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China,
| | - Peng Shen
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China,
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China,
| | - Ting Wang
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China,
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
- Key Laboratory of Laboratory Medical Diagnostics of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China,
| | - Zihong Liang
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China,
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China,
- Department of Neurology, The Inner Mongolia Autonomous Region People's Hospital, Hohhot, Inner Mongolia, China,
| | - Wei Wang
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China,
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China,
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
| | - Xunzhong Qi
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China,
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China,
| | - Peng Xie
- Department of Neurology, Yongchuan Hospital, Chongqing Medical University, Chongqing, China,
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China,
- Key Laboratory of Laboratory Medical Diagnostics of Education, Department of Laboratory Medicine, Chongqing Medical University, Chongqing, China,
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China,
- Department of Neurology, The Inner Mongolia Autonomous Region People's Hospital, Hohhot, Inner Mongolia, China,
| |
Collapse
|
33
|
Martin LJ, Chang Q. DNA Damage Response and Repair, DNA Methylation, and Cell Death in Human Neurons and Experimental Animal Neurons Are Different. J Neuropathol Exp Neurol 2018; 77:636-655. [PMID: 29788379 PMCID: PMC6005106 DOI: 10.1093/jnen/nly040] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Neurological disorders affecting individuals in infancy to old age elude interventions for meaningful protection against neurodegeneration, and preclinical work has not translated to humans. We studied human neuron responses to injury and death stimuli compared to those of animal neurons in culture under similar settings of insult (excitotoxicity, oxidative stress, and DNA damage). Human neurons were differentiated from a cortical neuron cell line and the embryonic stem cell-derived H9 line. Mouse neurons were differentiated from forebrain neural stem cells and embryonic cerebral cortex; pig neurons were derived from forebrain neural stem cells. Mitochondrial morphology was different in human and mouse neurons. Human and mouse neurons challenged with DNA-damaging agent camptothecin showed different chromatin condensation, cell death, and DNA damage sensor activation. DNA damage accumulation and repair kinetics differed among human, mouse, and pig neurons. Promoter CpG island methylation microarrays showed significant differential DNA methylation in human and mouse neurons after injury. Therefore, DNA damage response, DNA repair, DNA methylation, and autonomous cell death mechanisms in human neurons and experimental animal neurons are different.
Collapse
Affiliation(s)
- Lee J Martin
- Department of Pathology, Division of Neuropathology
- Pathobiology Graduate Training Program
- Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Qing Chang
- Department of Pathology, Division of Neuropathology
| |
Collapse
|
34
|
Li G, Morris-Blanco KC, Lopez MS, Yang T, Zhao H, Vemuganti R, Luo Y. Impact of microRNAs on ischemic stroke: From pre- to post-disease. Prog Neurobiol 2018; 163-164:59-78. [PMID: 28842356 PMCID: PMC11884751 DOI: 10.1016/j.pneurobio.2017.08.002] [Citation(s) in RCA: 128] [Impact Index Per Article: 18.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2017] [Revised: 06/12/2017] [Accepted: 08/16/2017] [Indexed: 12/21/2022]
Abstract
Stroke is the number one cause of neurological dysfunction in adults and has a heavy socioeconomic burden worldwide. The etiological origins of ischemic stroke and resulting pathological processes are mediated by a multifaceted cascade of molecular mechanisms that are in part modulated by posttranscriptional activity. Accumulating evidence has revealed a role for microRNAs (miRNAs) as essential mediators of posttranscriptional gene silencing in both the physiology of brain development and pathology of ischemic stroke. In this review, we compile miRNAs that have been reported to regulate various stroke risk factors and pre-disease mechanisms, including hypertension, atherosclerosis, and diabetes, followed by an in-depth analysis of miRNAs in ischemic stroke pathogenesis, such as excitotoxicity, oxidative stress, inflammation, apoptosis, angiogenesis and neurogenesis. Since promoting or suppressing expression of miRNAs by specific pharmaceutical and non-pharmaceutical therapies may be beneficial to post-stroke recovery, we also highlight the potential therapeutic value of miRNAs in clinical settings.
Collapse
Affiliation(s)
- Guangwen Li
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 10053, China
| | | | - Mary S Lopez
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA; Cellular and Molecular Pathology Graduate Program, University of Wisconsin, Madison, WI, USA
| | - Tuo Yang
- Department of Neurology, University of Pittsburgh School of Medicine, PA, USA
| | - Haiping Zhao
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 10053, China
| | - Raghu Vemuganti
- Department of Neurological Surgery, University of Wisconsin, Madison, WI, USA; Cellular and Molecular Pathology Graduate Program, University of Wisconsin, Madison, WI, USA; William S. Middleton VA Hospital, Madison, WI, USA.
| | - Yumin Luo
- Cerebrovascular Diseases Research Institute and Department of Neurology, Xuanwu Hospital of Capital Medical University, Beijing 10053, China; Beijing Institute for Brain Disorders and Beijing Key Laboratory of Translational Medicine for Cerebrovascular Diseases, Beijing 10053, China.
| |
Collapse
|
35
|
Khoury N, Koronowski KB, Young JI, Perez-Pinzon MA. The NAD +-Dependent Family of Sirtuins in Cerebral Ischemia and Preconditioning. Antioxid Redox Signal 2018; 28:691-710. [PMID: 28683567 PMCID: PMC5824497 DOI: 10.1089/ars.2017.7258] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/25/2017] [Accepted: 07/04/2017] [Indexed: 12/11/2022]
Abstract
SIGNIFICANCE Sirtuins are an evolutionarily conserved family of NAD+-dependent lysine deacylases and ADP ribosylases. Their requirement for NAD+ as a cosubstrate allows them to act as metabolic sensors that couple changes in the energy status of the cell to changes in cellular physiological processes. NAD+ levels are affected by several NAD+-producing and NAD+-consuming pathways as well as by cellular respiration. Thus their intracellular levels are highly dynamic and are misregulated in a spectrum of metabolic disorders including cerebral ischemia. This, in turn, compromises several NAD+-dependent processes that may ultimately lead to cell death. Recent Advances: A number of efforts have been made to replenish NAD+ in cerebral ischemic injuries as well as to understand the functions of one its important mediators, the sirtuin family of proteins through the use of pharmacological modulators or genetic manipulation approaches either before or after the insult. Critical Issues and Future Directions: The results of these studies have regarded the sirtuins as promising therapeutic targets for cerebral ischemia. Yet, additional efforts are needed to understand the role of some of the less characterized members and to address the sex-specific effects observed with some members. Sirtuins also exhibit cell-type-specific expression in the brain as well as distinct subcellular and regional localizations. As such, they are involved in diverse and sometimes opposing cellular processes that can either promote neuroprotection or further contribute to the injury; which also stresses the need for the development and use of sirtuin-specific pharmacological modulators. Antioxid. Redox Signal. 28, 691-710.
Collapse
Affiliation(s)
- Nathalie Khoury
- Department of Neurology; Cerebral Vascular Research Laboratories; and Neuroscience Program, Miller School of Medicine, University of Miami, Miami, Florida
| | - Kevin B. Koronowski
- Department of Neurology; Cerebral Vascular Research Laboratories; and Neuroscience Program, Miller School of Medicine, University of Miami, Miami, Florida
| | - Juan I. Young
- Dr. John T. Macdonald Foundation Department of Human Genetics; Hussman Institute for Human Genomics, and Neuroscience Program, Miller School of Medicine, University of Miami, Miami, Florida
| | - Miguel A. Perez-Pinzon
- Department of Neurology; Cerebral Vascular Research Laboratories; and Neuroscience Program, Miller School of Medicine, University of Miami, Miami, Florida
| |
Collapse
|
36
|
5d, a novel analogue of 3-n-butylphthalide, decreases NADPH oxidase activity through the positive regulation of CK2 after ischemia/reperfusion injury. Oncotarget 2018; 7:39444-39457. [PMID: 27276705 PMCID: PMC5129944 DOI: 10.18632/oncotarget.8548] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 05/05/2016] [Indexed: 12/19/2022] Open
Abstract
5d, a novel analogue of the racemic 3-n-butylphthalide (NBP), has been reported for its free radical scavenging activity in vitro and preventive neuroprotection in vivo. Nevertheless, the mechanism by which 5d attenuated ischemia/reperfusion (I/R) injury is still unknown. Our results showed that 5d significantly increased CK2 activity as well as CK2α and 2α' protein levels after I/R injury. Besides, 5d suppressed the translocation of cytosolic p47phox and Rac1 to the membrane, decreased NOX4 expression and ROS generation. Furthermore, 5d blocked the dissociation between CK2α and Rac1 so as to decrease NADPH oxidase activity. Based on these findings, we propose that the neuroprotective effect of 5d is due to an increase of CK2 activity, which blocks I/R-induced dissociation between CK2α and Rac1, decreases NADPH oxidase activity, inhibits ROS production and finally realizes the neuroprotection of I/R. These findings point to that 5d might be considered an attractive candidate for further studies in ischemic stroke.
Collapse
|
37
|
Miyazaki T, Miyazaki A. Defective Protein Catabolism in Atherosclerotic Vascular Inflammation. Front Cardiovasc Med 2017; 4:79. [PMID: 29270409 PMCID: PMC5725411 DOI: 10.3389/fcvm.2017.00079] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 11/23/2017] [Indexed: 01/08/2023] Open
Abstract
Vascular inflammation in atheroprone vessels propagates throughout the arterial tree in dyslipidemic patients, thereby accelerating atherosclerotic progression. To elucidate the mechanism of vascular inflammation, most previous studies have focused on inflammation-related signals that are sent in response to vasoactive stimuli. However, it is also important to understand how normal blood vessels become defective and start degenerating. Growing evidence suggests that major protein catabolism pathways, including the ubiquitin-proteasome, autophagy, and calpain systems, are disturbed in atheroprone vessels and contribute to the pathogenesis of atherosclerosis. Indeed, dysregulation of ubiquitin-proteasome pathways results in the accumulation of defective proteins in blood vessels, leading to vascular endothelial dysfunction and apoptosis in affected cells. Impaired autophagy-lysosomal degradation affects smooth muscle cell transformation and proliferation, as well as endothelial integrity and phagocytic clearance of cellular corpses. Dysregulation of the calpain system confers proatherogenic properties to endothelial cells, smooth muscle cells, and macrophages. In this review article, we will discuss the current information available on defective protein catabolism in atheroprone vessels and its potential interrelation with inflammation-related signals.
Collapse
Affiliation(s)
- Takuro Miyazaki
- Department of Biochemistry, School of Medicine, Showa University, Tokyo, Japan
| | - Akira Miyazaki
- Department of Biochemistry, School of Medicine, Showa University, Tokyo, Japan
| |
Collapse
|
38
|
Edaravone, a Synthetic Free Radical Scavenger, Enhances Alteplase-Mediated Thrombolysis. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:6873281. [PMID: 29259732 PMCID: PMC5702421 DOI: 10.1155/2017/6873281] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/07/2017] [Accepted: 09/07/2017] [Indexed: 12/12/2022]
Abstract
The combination of alteplase, a recombinant tissue plasminogen activator, and edaravone, an antioxidant, reportedly enhances recanalization after acute ischemic stroke. We examined the influence of edaravone on the thrombolytic efficacy of alteplase by measuring thrombolysis using a newly developed microchip-based flow-chamber assay. Rat models of embolic cerebral ischemia were treated with either alteplase or alteplase-edaravone combination therapy. The combination therapy significantly reduced the infarct volume and improved neurological deficits. Human blood samples from healthy volunteers were exposed to edaravone, alteplase, or a combination of alteplase and edaravone or hydrogen peroxide. Whole blood was perfused over a collagen- and thromboplastin-coated microchip; capillary occlusion was monitored with a video microscope and flow-pressure sensor. The area under the curve (extent of thrombogenesis or thrombolysis) at 30 minutes was 69.9% lower in the edaravone-alteplase- than alteplase-treated group. The thrombolytic effect of alteplase was significantly attenuated in the presence of hydrogen peroxide, suggesting that oxidative stress might hinder thrombolysis. D-dimers were measured to evaluate these effects in human platelet-poor plasma samples. Although hydrogen peroxide significantly decreased the elevation of D-dimers by alteplase, edaravone significantly inhibited the decrease. Edaravone enhances alteplase-mediated thrombolysis, likely by preventing oxidative stress, which inhibits fibrinolysis by alteplase in thrombi.
Collapse
|
39
|
Yu C, Shen S, Jiang L, Li J, Lu Y, Li T, Yao C. NHC-catalyzed regiodivergent syntheses of difunctionalized 3-pyrazolidinones from α-bromoenal and monosubstituted hydrazine. Org Biomol Chem 2017; 15:9149-9155. [PMID: 29058746 DOI: 10.1039/c7ob02041d] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
A formal [3 + 2] annulation of α-bromoenal with monosubstituted hydrazine could give 1,5 or 2,5-difunctionalized 3-pyrazolidinone regiodivergently by tuning the structure of the N-Heterocyclic Carbene (NHC) catalyst. Moderate to high yields, mild reaction conditions, good regioselectivity and potential biological significance of the final product have made this protocol attractive for the assembly of 3-pyrazolidinone.
Collapse
Affiliation(s)
- Chenxia Yu
- Jiangsu Key Laboratory of Green Synthetic Chemistry for Functional Materials, School of Chemistry and Materials Science, Jiangsu Normal University, Xuzhou 221116, P. R. China.
| | | | | | | | | | | | | |
Collapse
|
40
|
Tuo QZ, Lei P, Jackman KA, Li XL, Xiong H, Li XL, Liuyang ZY, Roisman L, Zhang ST, Ayton S, Wang Q, Crouch PJ, Ganio K, Wang XC, Pei L, Adlard PA, Lu YM, Cappai R, Wang JZ, Liu R, Bush AI. Tau-mediated iron export prevents ferroptotic damage after ischemic stroke. Mol Psychiatry 2017; 22:1520-1530. [PMID: 28886009 DOI: 10.1038/mp.2017.171] [Citation(s) in RCA: 512] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/20/2017] [Accepted: 07/06/2017] [Indexed: 02/05/2023]
Abstract
Functional failure of tau contributes to age-dependent, iron-mediated neurotoxicity, and as iron accumulates in ischemic stroke tissue, we hypothesized that tau failure may exaggerate ischemia-reperfusion-related toxicity. Indeed, unilateral, transient middle cerebral artery occlusion (MCAO) suppressed hemispheric tau and increased iron levels in young (3-month-old) mice and rats. Wild-type mice were protected by iron-targeted interventions: ceruloplasmin and amyloid precursor protein ectodomain, as well as ferroptosis inhibitors. At this age, tau-knockout mice did not express elevated brain iron and were protected against hemispheric reperfusion injury following MCAO, indicating that tau suppression may prevent ferroptosis. However, the accelerated age-dependent brain iron accumulation that occurs in tau-knockout mice at 12 months of age negated the protective benefit of tau suppression against MCAO-induced focal cerebral ischemia-reperfusion injury. The protective benefit of tau knockout was revived in older mice by iron-targeting interventions. These findings introduce tau-iron interaction as a pleiotropic modulator of ferroptosis and ischemic stroke outcome.
Collapse
Affiliation(s)
- Q-Z Tuo
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China.,Oxidation Biology Unit, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia.,Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, China
| | - P Lei
- Oxidation Biology Unit, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia.,Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, China
| | - K A Jackman
- Oxidation Biology Unit, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - X-L Li
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, China
| | - H Xiong
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, China
| | - X-L Li
- Oxidation Biology Unit, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia.,Department of Neurology, The Fourth Affiliated Hospital, Harbin Medical University, Harbin, China
| | - Z-Y Liuyang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - L Roisman
- Department of Pathology, The University of Melbourne, Melbourne, VIC, Australia
| | - S-T Zhang
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, China
| | - S Ayton
- Oxidation Biology Unit, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Q Wang
- Department of Neurology and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, and Collaborative Innovation Center for Biotherapy, Sichuan, China
| | - P J Crouch
- Department of Pathology, The University of Melbourne, Melbourne, VIC, Australia
| | - K Ganio
- Oxidation Biology Unit, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - X-C Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - L Pei
- Department of Neurobiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Hubei, China
| | - P A Adlard
- Oxidation Biology Unit, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| | - Y-M Lu
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - R Cappai
- Department of Pathology, The University of Melbourne, Melbourne, VIC, Australia
| | - J-Z Wang
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - R Liu
- Department of Pathophysiology, Key Laboratory of Ministry of Education for Neurological Disorders, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - A I Bush
- Oxidation Biology Unit, Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Melbourne, VIC, Australia
| |
Collapse
|
41
|
Miyazaki T, Miyazaki A. Dysregulation of Calpain Proteolytic Systems Underlies Degenerative Vascular Disorders. J Atheroscler Thromb 2017; 25:1-15. [PMID: 28819082 PMCID: PMC5770219 DOI: 10.5551/jat.rv17008] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Chronic vascular diseases such as atherosclerosis, aneurysms, diabetic angiopathy/retinopathy as well as fibrotic and proliferative vascular diseases are generally complicated by the progression of degenerative insults, which are characterized by endothelial dysfunction, apoptotic/necrotic cell death in vascular/immune cells, remodeling of extracellular matrix or breakdown of elastic lamella. Increasing evidence suggests that dysfunctional calpain proteolytic systems and defective calpain protein metabolism in blood vessels contribute to degenerative disorders. In vascular endothelial cells, the overactivation of conventional calpains consisting of calpain-1 and -2 isozymes can lead to the disorganization of cell-cell junctions, dysfunction of nitric oxide synthase, sensitization of Janus kinase/signal transducer and activator of transcription cascades and depletion of prostaglandin I2, which contributes to degenerative disorders. In addition to endothelial cell dysfunctions, calpain overactivation results in inflammatory insults in macrophages and excessive fibrogenic/proliferative signaling in vascular smooth muscle cells. Moreover, calpain-6, a non-proteolytic unconventional calpain, is involved in the conversion of macrophages to a pro-atherogenic phenotype, leading to the pinocytotic deposition of low-density lipoprotein cholesterol in the cells. Here, we discuss the recent progress that has been made in our understanding of how calpain contributes to degenerative vascular disorders.
Collapse
Affiliation(s)
- Takuro Miyazaki
- Department of Biochemistry, Showa University School of Medicine
| | - Akira Miyazaki
- Department of Biochemistry, Showa University School of Medicine
| |
Collapse
|
42
|
Targeting a Potassium Channel/Syntaxin Interaction Ameliorates Cell Death in Ischemic Stroke. J Neurosci 2017; 37:5648-5658. [PMID: 28483976 DOI: 10.1523/jneurosci.3811-16.2017] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2016] [Revised: 04/25/2017] [Accepted: 05/01/2017] [Indexed: 12/12/2022] Open
Abstract
The voltage-gated K+ channel Kv2.1 has been intimately linked with neuronal apoptosis. After ischemic, oxidative, or inflammatory insults, Kv2.1 mediates a pronounced, delayed enhancement of K+ efflux, generating an optimal intracellular environment for caspase and nuclease activity, key components of programmed cell death. This apoptosis-enabling mechanism is initiated via Zn2+-dependent dual phosphorylation of Kv2.1, increasing the interaction between the channel's intracellular C-terminus domain and the SNARE (soluble N-ethylmaleimide-sensitive factor activating protein receptor) protein syntaxin 1A. Subsequently, an upregulation of de novo channel insertion into the plasma membrane leads to the critical enhancement of K+ efflux in damaged neurons. Here, we investigated whether a strategy designed to interfere with the cell death-facilitating properties of Kv2.1, specifically its interaction with syntaxin 1A, could lead to neuroprotection following ischemic injury in vivo The minimal syntaxin 1A-binding sequence of Kv2.1 C terminus (C1aB) was first identified via a far-Western peptide screen and used to create a protherapeutic product by conjugating C1aB to a cell-penetrating domain. The resulting peptide (TAT-C1aB) suppressed enhanced whole-cell K+ currents produced by a mutated form of Kv2.1 mimicking apoptosis in a mammalian expression system, and protected cortical neurons from slow excitotoxic injury in vitro, without influencing NMDA-induced intracellular calcium responses. Importantly, intraperitoneal administration of TAT-C1aB in mice following transient middle cerebral artery occlusion significantly reduced ischemic stroke damage and improved neurological outcome. These results provide strong evidence that targeting the proapoptotic function of Kv2.1 is an effective and highly promising neuroprotective strategy.SIGNIFICANCE STATEMENT Kv2.1 is a critical regulator of apoptosis in central neurons. It has not been determined, however, whether the cell death-enabling function of this K+ channel can be selectively targeted to improve neuronal survival following injury in vivo The experiments presented here demonstrate that the cell death-specific role of Kv2.1 can be uniquely modulated to provide neuroprotection in an animal model of acute ischemic stroke. We thus reveal a novel therapeutic strategy for neurological disorders that are accompanied by Kv2.1-facilitated forms of cell death.
Collapse
|
43
|
Kim Y, Davidson JO, Green CR, Nicholson LFB, O'Carroll SJ, Zhang J. Connexins and Pannexins in cerebral ischemia. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2017; 1860:224-236. [PMID: 28347700 DOI: 10.1016/j.bbamem.2017.03.018] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 12/21/2016] [Revised: 02/24/2017] [Accepted: 03/22/2017] [Indexed: 12/12/2022]
Abstract
A common cause of mortality and long-term adult disability, cerebral ischemia or brain ischemia imposes a significant health and financial burden on communities worldwide. Cerebral ischemia is a condition that arises from a sudden loss of blood flow and consequent failure to meet the high metabolic demands of the brain. The lack of blood flow initiates a sequelae of cell death mechanisms, including the activation of the inflammatory pathway, which can ultimately result in irreversible brain tissue damage. In particular, Connexins and Pannexins are non-selective channels with a large pore that have shown to play time-dependent roles in the perpetuation of ischaemic injury. This review highlights the roles of Connexin and Pannexin channels in cell death mechanisms as a promising therapeutic target in cerebral ischemia, and in particular connexin hemichannels which may contribute most of the ATP release as a result of ischemia as well as during reperfusion. This article is part of a Special Issue entitled: Gap Junction Proteins edited by Jean Claude Herve.
Collapse
Affiliation(s)
- Yeri Kim
- Department of Ophthalmology, Faculty of Medical and Health Sciences, University of Auckland
| | - Joanne O Davidson
- Department of Physiology, Faculty of Medical and Health Sciences, University of Auckland
| | - Colin R Green
- Department of Ophthalmology, Faculty of Medical and Health Sciences, University of Auckland
| | - Louise F B Nicholson
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland
| | - Simon J O'Carroll
- Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland
| | - Jie Zhang
- Department of Ophthalmology, Faculty of Medical and Health Sciences, University of Auckland.
| |
Collapse
|
44
|
Phase I and Phase II Therapies for Acute Ischemic Stroke: An Update on Currently Studied Drugs in Clinical Research. BIOMED RESEARCH INTERNATIONAL 2017; 2017:4863079. [PMID: 28286764 PMCID: PMC5329656 DOI: 10.1155/2017/4863079] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Accepted: 11/23/2016] [Indexed: 02/06/2023]
Abstract
Acute ischemic stroke is a devastating cause of death and disability, consequences of which depend on the time from ischemia onset to treatment, the affected brain region, and its size. The main targets of ischemic stroke therapy aim to restore tissue perfusion in the ischemic penumbra in order to decrease the total infarct area by maintaining blood flow. Advances in research of pathological process and pathways during acute ischemia have resulted in improvement of new treatment strategies apart from restoring perfusion. Additionally, limiting the injury severity by manipulating the molecular mechanisms during ischemia has become a promising approach, especially in animal research. The purpose of this article is to review completed and ongoing phases I and II trials for the treatment of acute ischemic stroke, reviewing studies on antithrombotic, thrombolytic, neuroprotective, and antineuroinflammatory drugs that may translate into more effective treatments.
Collapse
|
45
|
Griffin JH, Mosnier LO, Fernández JA, Zlokovic BV. 2016 Scientific Sessions Sol Sherry Distinguished Lecturer in Thrombosis: Thrombotic Stroke: Neuroprotective Therapy by Recombinant-Activated Protein C. Arterioscler Thromb Vasc Biol 2016; 36:2143-2151. [PMID: 27758767 PMCID: PMC5119536 DOI: 10.1161/atvbaha.116.308038] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2016] [Accepted: 09/21/2016] [Indexed: 01/19/2023]
Abstract
APC (activated protein C), derived from the plasma protease zymogen, is antithrombotic and anti-inflammatory. In preclinical injury models, recombinant APC provides neuroprotection for multiple injuries, including ischemic stroke. APC acts directly on brain endothelial cells and neurons by initiating cell signaling that requires multiple receptors. Two or more major APC receptors mediate APC's neuroprotective cell signaling. When bound to endothelial cell protein C receptor, APC can cleave protease-activated receptor 1, causing biased cytoprotective signaling that reduces ischemia-induced injury. Pharmacological APC alleviates bleeding induced by tissue-type plasminogen activator in murine ischemic stroke studies. Remarkably, APC's signaling promotes neurogenesis. The signaling-selective recombinant variant of APC, 3K3A-APC, was engineered to lack most of the APC's anticoagulant activity but retain APC's cell signaling actions. Recombinant 3K3A-APC is in ongoing National Institutes of Health (NIH)-funded clinical trials for ischemic stroke.
Collapse
Affiliation(s)
- John H Griffin
- From the Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA (J.H.G., L.O.M., J.A.F.); Division of Hematology/Oncology, Department of Medicine, University of California, San Diego (J.H.G.); and Department of Physiology and Biophysics, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles (B.V.Z.).
| | - Laurent O Mosnier
- From the Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA (J.H.G., L.O.M., J.A.F.); Division of Hematology/Oncology, Department of Medicine, University of California, San Diego (J.H.G.); and Department of Physiology and Biophysics, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles (B.V.Z.)
| | - José A Fernández
- From the Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA (J.H.G., L.O.M., J.A.F.); Division of Hematology/Oncology, Department of Medicine, University of California, San Diego (J.H.G.); and Department of Physiology and Biophysics, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles (B.V.Z.)
| | - Berislav V Zlokovic
- From the Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA (J.H.G., L.O.M., J.A.F.); Division of Hematology/Oncology, Department of Medicine, University of California, San Diego (J.H.G.); and Department of Physiology and Biophysics, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles (B.V.Z.)
| |
Collapse
|
46
|
Dong MX, Hu QC, Shen P, Pan JX, Wei YD, Liu YY, Ren YF, Liang ZH, Wang HY, Zhao LB, Xie P. Recombinant Tissue Plasminogen Activator Induces Neurological Side Effects Independent on Thrombolysis in Mechanical Animal Models of Focal Cerebral Infarction: A Systematic Review and Meta-Analysis. PLoS One 2016; 11:e0158848. [PMID: 27387385 PMCID: PMC4936748 DOI: 10.1371/journal.pone.0158848] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2016] [Accepted: 06/22/2016] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND AND PURPOSE Recombinant tissue plasminogen activator (rtPA) is the only effective drug approved by US FDA to treat ischemic stroke, and it contains pleiotropic effects besides thrombolysis. We performed a meta-analysis to clarify effect of tissue plasminogen activator (tPA) on cerebral infarction besides its thrombolysis property in mechanical animal stroke. METHODS Relevant studies were identified by two reviewers after searching online databases, including Pubmed, Embase, and ScienceDirect, from 1979 to 2016. We identified 6, 65, 17, 12, 16, 12 and 13 comparisons reporting effect of endogenous tPA on infarction volume and effects of rtPA on infarction volume, blood-brain barrier, brain edema, intracerebral hemorrhage, neurological function and mortality rate in all 47 included studies. Standardized mean differences for continuous measures and risk ratio for dichotomous measures were calculated to assess the effects of endogenous tPA and rtPA on cerebral infarction in animals. The quality of included studies was assessed using the Stroke Therapy Academic Industry Roundtable score. Subgroup analysis, meta-regression and sensitivity analysis were performed to explore sources of heterogeneity. Funnel plot, Trim and Fill method and Egger's test were obtained to detect publication bias. RESULTS We found that both endogenous tPA and rtPA had not enlarged infarction volume, or deteriorated neurological function. However, rtPA would disrupt blood-brain barrier, aggravate brain edema, induce intracerebral hemorrhage and increase mortality rate. CONCLUSIONS This meta-analysis reveals rtPA can lead to neurological side effects besides thrombolysis in mechanical animal stroke, which may account for clinical exacerbation for stroke patients that do not achieve vascular recanalization with rtPA.
Collapse
Affiliation(s)
- Mei-Xue Dong
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
| | - Qing-Chuan Hu
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
| | - Peng Shen
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Jun-Xi Pan
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
| | - You-Dong Wei
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yi-Yun Liu
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yi-Fei Ren
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Zi-Hong Liang
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Hai-Yang Wang
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
| | - Li-Bo Zhao
- Department of Neurology, Yongchuan Hospital of Chongqing Medical University, Chongqing, China
| | - Peng Xie
- Department of Neurology, the First Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Institute of Neuroscience and the Collaborative Innovation Center for Brain Science, Chongqing Medical University, Chongqing, China
- Chongqing Key Laboratory of Neurobiology, Chongqing, China
| |
Collapse
|
47
|
Khoury N, Koronowski KB, Perez-Pinzon MA. Long-term window of ischemic tolerance: An evolutionarily conserved form of metabolic plasticity regulated by epigenetic modifications? ACTA ACUST UNITED AC 2016; 1:6-12. [PMID: 27796011 DOI: 10.29245/2572.942x/2016/2.1021] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
In the absence of effective neuroprotective agents in the clinic, ischemic and pharmacological preconditioning are gaining increased interest in the field of cerebral ischemia. Our lab recently reported that resveratrol preconditioning affords tolerance against a focal cerebral ischemic insult in mice that can last for at least 14 days in vivo making it the longest window of ischemic tolerance discovered to date by a single administration of a pharmacological agent. The mechanism behind this novel extended window of ischemic tolerance remains elusive. In the below commentary we discuss potential mechanisms that could explain this novel extended window of ischemic tolerance in the context of previously identified windows and the known mechanisms behind them. We also draw parallels from the fields of hibernation and hypoxia-tolerance, which are chronic adaptations to severe conditions of hypoxia and ischemia known to be mediated by a form of metabolic depression. We also briefly discuss the importance of epigenetic modifications in maintaining this depressed state of metabolism.
Collapse
Affiliation(s)
- Nathalie Khoury
- Department of Neurology and Neuroscience Program, Cerebral Vascular Disease Research Laboratories, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Kevin B Koronowski
- Department of Neurology and Neuroscience Program, Cerebral Vascular Disease Research Laboratories, Miller School of Medicine, University of Miami, Miami, Florida, USA
| | - Miguel A Perez-Pinzon
- Department of Neurology and Neuroscience Program, Cerebral Vascular Disease Research Laboratories, Miller School of Medicine, University of Miami, Miami, Florida, USA
| |
Collapse
|
48
|
Yen TL, Chen RJ, Jayakumar T, Lu WJ, Hsieh CY, Hsu MJ, Yang CH, Chang CC, Lin YK, Lin KH, Sheu JR. Andrographolide stimulates p38 mitogen-activated protein kinase-nuclear factor erythroid-2-related factor 2-heme oxygenase 1 signaling in primary cerebral endothelial cells for definite protection against ischemic stroke in rats. Transl Res 2016; 170:57-72. [PMID: 26746802 DOI: 10.1016/j.trsl.2015.12.002] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/25/2015] [Revised: 12/02/2015] [Accepted: 12/09/2015] [Indexed: 12/13/2022]
Abstract
Stroke pathogenesis involves complex oxidative stress-related pathways. The nuclear factor erythroid-2-related factor 2 (Nrf2) and heme oxygenase 1 (HO-1) pathways have been considered molecular targets in pharmacologic intervention for ischemic diseases. Andrographolide, a labdane diterpene, has received increasing attention in recent years because of its various pharmacologic activities. We determined that andrographolide modulates the mitogen-activated protein kinase (MAPK)-Nrf2-HO-1 signaling cascade in primary cerebral endothelial cells (CECs) to provide positive protection against middle cerebral artery occlusion (MCAO)-induced ischemic stroke in rats. In the present study, andrographolide (10 μM) increased HO-1 protein and messenger RNA expressions, Nrf2 phosphorylation, and nuclear translocation in CECs, and these activities were disrupted by a p38 MAPK inhibitor, SB203580, but not by the extracellular signal-regulated kinase inhibitor PD98059 or c-Jun amino-terminal kinase inhibitor SP600125. Similar results were observed in confocal microscopy analysis. Moreover, andrographolide-induced Nrf2 and HO-1 protein expressions were significantly inhibited by Nrf2 small interfering RNA. Moreover, HO-1 knockdown attenuated the protective effect of andrographolide against oxygen-glucose deprivation-induced CEC death. Andrographolide (0.1 mg/kg) significantly suppressed free radical formation, blood-brain barrier disruption, and brain infarction in MCAO-insulted rats, and these effects were reversed by the HO-1 inhibitor zinc protoporphyrin IX. The mechanism is attributable to HO-1 activation, as directly evidenced by andrographolide-induced pronounced HO-1 expression in brain tissues, which was highly localized in the cerebral capillary. In conclusion, andrographolide increased Nrf2-HO-1 expression through p38 MAPK regulation, confirming that it provides protection against MCAO-induced brain injury. These findings provide strong evidence that andrographolide could be a therapeutic agent for treating ischemic stroke or neurodegenerative diseases.
Collapse
Affiliation(s)
- Ting-Lin Yen
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ray-Jade Chen
- School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of General Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei, Taiwan; Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan
| | - Thanasekaran Jayakumar
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Wan-Jung Lu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of General Surgery, Department of Surgery, Taipei Medical University Hospital, Taipei, Taiwan; Department of Medical Research, Taipei Medical University Hospital, Taipei, Taiwan
| | - Cheng-Ying Hsieh
- Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Ming-Jen Hsu
- Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chih-Hao Yang
- Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chao-Chien Chang
- Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Cardiology, Cathay General Hospital, Taipei, Taiwan
| | - Yen-Kuang Lin
- Biostatistics Center, Taipei Medical University, Taipei, Taiwan
| | - Kuan-Hung Lin
- Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei, Taiwan; Central Laboratory, Shin-Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan.
| | - Joen-Rong Sheu
- Graduate Institute of Medical Sciences, College of Medicine, Taipei Medical University, Taipei, Taiwan; Department of Pharmacology, College of Medicine, Taipei Medical University, Taipei, Taiwan.
| |
Collapse
|
49
|
Wang WY, Shen CW, Weng ZJ, Wang TC, Zhang C, Jin XQ, Li JQ. Design, synthesis and biological evaluation of novel dicarbonylalkyl piperazine derivatives as neuroprotective agents. CHINESE CHEM LETT 2016. [DOI: 10.1016/j.cclet.2015.11.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
50
|
Yu H, Liu P, Tang H, Jing J, Lv X, Chen L, Jiang L, Xu J, Li J. Oleuropein, a natural extract from plants, offers neuroprotection in focal cerebral ischemia/reperfusion injury in mice. Eur J Pharmacol 2016; 775:113-9. [PMID: 26872990 DOI: 10.1016/j.ejphar.2016.02.027] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2015] [Revised: 02/03/2016] [Accepted: 02/08/2016] [Indexed: 12/22/2022]
Abstract
Oleuropein (OLE) was found to have anti-inflammatory and anti-oxidant effects. The latest study has shown that it can resist myocardial injury that follows an acute myocardial infarction and can rescue impaired spinal nerve cells. In this study, we investigated the neuroprotective effects of OLE on cerebral ischemia and reperfusion injury in a middle cerebral artery occlusion model in mice.OLE (100 mg/kg) was injected intraperitoneally 1h before ischemia. We found that the volume of cerebral infarction was significantly reduced after 75 min of ischemia and 24 h of reperfusion compared with the I/R (ischemia/reperfusion) group. This protective function occurred in a dose-dependent manner. We also found that treatment with OLE could reduce the cerebral infarct volume. The neuroprotective effect was prolonged from 2 h to 4 h when we injected OLE intracerebroventricularly after reperfusion. We then found that OLE can decrease the level of cleavedcaspase-3, an important marker of apoptosis, in the ischemic mouse brain. Finally, we explored the role of OLE in providing anti-apoptotic effects through the increased expression of Bcl-2 and the decreased expression of Bax, which are important markers in apoptosis. As shown above, the function and safety of OLE in cardiovascular disease may indicate that it is a potential therapeutic for stroke.
Collapse
Affiliation(s)
- Hailong Yu
- Department of Neurology, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou 225001, China
| | - Peipei Liu
- Department of Neurology, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou 225001, China
| | - Hui Tang
- Department of Neurology, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou 225001, China
| | - Jian Jing
- Department of Neurology, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou 225001, China
| | - Xiang Lv
- Department of Neurology, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou 225001, China
| | - Lanlan Chen
- Department of Neurology, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou 225001, China
| | - Li Jiang
- Department of Neurology, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou 225001, China
| | - Jun Xu
- Department of Neurology, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou 225001, China.
| | - Jun Li
- Department of Neurology, Clinical Medical College of Yangzhou University, Subei People's Hospital of Jiangsu Province, Yangzhou 225001, China.
| |
Collapse
|