1
|
Tabassum S, Wu S, Lee CH, Yang BSK, Gusdon AM, Choi HA, Ren XS. Mitochondrial-targeted therapies in traumatic brain injury: From bench to bedside. Neurotherapeutics 2025; 22:e00515. [PMID: 39721917 PMCID: PMC11840356 DOI: 10.1016/j.neurot.2024.e00515] [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: 07/30/2024] [Revised: 12/03/2024] [Accepted: 12/10/2024] [Indexed: 12/28/2024] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of morbidity and mortality worldwide, with limited effective therapeutic options currently available. Recent research has highlighted the pivotal role of mitochondrial dysfunction in the pathophysiology of TBI, making mitochondria an attractive target for therapeutic intervention. This review comprehensively examines advancements in mitochondrial-targeted therapies for TBI, bridging the gap from basic research to clinical applications. We discuss the underlying mechanisms of mitochondrial damage in TBI, including oxidative stress, impaired bioenergetics, mitochondrial dynamics, and apoptotic pathways. Furthermore, we highlight the complex interplay between mitochondrial dysfunction, inflammation, and blood-brain barrier (BBB) integrity, elucidating how these interactions exacerbate injury and impede recovery. We also evaluate various preclinical studies exploring pharmacological agents, gene therapy, and novel drug delivery systems designed to protect and restore mitochondrial function. Clinical trials and their outcomes are assessed to evaluate the translational potential of mitochondrial-targeted therapies in TBI. By integrating findings from bench to bedside, this review emphasizes promising therapeutic avenues and addresses remaining challenges. It also provides guidance for future research to pave the way for innovative treatments that improve patient outcomes in TBI.
Collapse
Affiliation(s)
- Sidra Tabassum
- Novel Treatments for Acute Brain Injury Institute, Texas Medical Center, TX, USA; Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Silin Wu
- Novel Treatments for Acute Brain Injury Institute, Texas Medical Center, TX, USA; Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Chang-Hun Lee
- Department of New Biology, Daegu Gyeongbuk Institute of Science and Technology, Daegu, Republic of Korea
| | - Bosco Seong Kyu Yang
- Novel Treatments for Acute Brain Injury Institute, Texas Medical Center, TX, USA; Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Aaron M Gusdon
- Novel Treatments for Acute Brain Injury Institute, Texas Medical Center, TX, USA; Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Huimahn A Choi
- Novel Treatments for Acute Brain Injury Institute, Texas Medical Center, TX, USA; Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA
| | - Xuefang S Ren
- Novel Treatments for Acute Brain Injury Institute, Texas Medical Center, TX, USA; Department of Neurosurgery, McGovern Medical School, University of Texas Health Science Center, Houston, TX, USA.
| |
Collapse
|
2
|
Luo Y, Liu R, Yuan G, Pan Y. Polyphenols for stroke therapy: the role of oxidative stress regulation. Food Funct 2024; 15:11383-11399. [PMID: 39497601 DOI: 10.1039/d4fo01900h] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2024]
Abstract
Stroke is associated with a high incidence and disability rate, which seriously endangers human health. Oxidative stress (OS) plays a crucial role in the underlying pathologic progression of cerebral damage in stroke. Emerging experimental studies suggest that polyphenols have antioxidant potential and express protective effects after different types of strokes, but no breakthrough has been achieved in clinical studies. Nanomaterials, due to small characteristic sizes, can be used to deliver drugs, and have shown excellent performance in the treatment of various diseases. The drug delivery capability of nanomaterials has significant implications for the clinical translation and application of polyphenols. This comprehensive review introduces the mechanism of oxidative stress in stroke, and also summarizes the antioxidant effects of polyphenols on reactive oxygen species generation and oxidative stress after stroke. Also, the application characteristics and research progress of nanomaterials in the treatment of stroke with antioxidants are presented.
Collapse
Affiliation(s)
- Yusong Luo
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China.
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Ruolan Liu
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China.
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Guoqiang Yuan
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China.
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Yawen Pan
- Department of Neurosurgery, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China.
- Gansu Provincial Clinical Research Center for Neurological Diseases, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
- Academician Workstation, the Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| |
Collapse
|
3
|
Lee J, Geum D, Park DH, Kim JH. Molecular Targeting of Ischemic Stroke: The Promise of Naïve and Engineered Extracellular Vesicles. Pharmaceutics 2024; 16:1492. [PMID: 39771472 PMCID: PMC11678501 DOI: 10.3390/pharmaceutics16121492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 11/18/2024] [Accepted: 11/19/2024] [Indexed: 01/04/2025] Open
Abstract
Ischemic stroke (IS) remains a leading cause of mortality and long-term disability worldwide, with limited therapeutic options available. Despite the success of early interventions, such as tissue-type plasminogen activator administration and mechanical thrombectomy, many patients continue to experience persistent neurological deficits. The pathophysiology of IS is multifaceted, encompassing excitotoxicity, oxidative and nitrosative stress, inflammation, and blood-brain barrier disruption, all of which contribute to neural cell death, further complicating the treatment of IS. Recently, extracellular vesicles (EVs) secreted naturally by various cell types have emerged as promising therapeutic agents because of their ability to facilitate selective cell-to-cell communication, neuroprotection, and tissue regeneration. Furthermore, engineered EVs, designed to enhance targeted delivery and therapeutic cargo, hold the potential to improve their therapeutic benefits by mitigating neuronal damage and promoting neurogenesis and angiogenesis. This review summarizes the characteristics of EVs, the molecular mechanisms underlying IS pathophysiology, and the emerging role of EVs in IS treatment at the molecular level. This review also explores the recent advancements in EV engineering, including the incorporation of specific proteins, RNAs, or pharmacological agents into EVs to enhance their therapeutic efficacy.
Collapse
Affiliation(s)
- Jihun Lee
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea;
| | - Dongho Geum
- Department of Medical Science, College of Medicine, Korea University, Seoul 02841, Republic of Korea;
| | - Dong-Hyuk Park
- Department of Neurosurgery, Anam Hospital, College of Medicine, Korea University, Seoul 02841, Republic of Korea;
| | - Jong-Hoon Kim
- Laboratory of Stem Cells and Tissue Regeneration, Department of Biotechnology, College of Life Sciences and Biotechnology, Korea University, Seoul 02841, Republic of Korea;
| |
Collapse
|
4
|
Zhang H, Zhang H, Wang J, Fan L, Mu W, Jin Y, Wang Z. Small-molecular cyclic peptide exerts viability suppression effects on HepG2 cells via triggering p53 apoptotic pathways. Chem Biol Interact 2023; 382:110633. [PMID: 37451662 DOI: 10.1016/j.cbi.2023.110633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Revised: 07/06/2023] [Accepted: 07/11/2023] [Indexed: 07/18/2023]
Abstract
Cyclic peptides have become an attractive modality for drug development due to their high specificity, metabolic stability and higher cell permeability. In an effort to explore novel antitumor compounds based on natural cyclopeptide from the phakellistatin family, we found an isoindolinone-containing analog (S-PK6) of phakellistatin 6 capable of suppressing the viability and proliferation of HepG2 cells. The aim of the present study is to shed light on the mechanism of action of this novel compound. We have detected differences in gene expression before and after treatment with S-PK6 in human hepatocellular carcinoma HepG2 cell line by transcriptome sequencing. To further investigate biological effects, we have also extensively investigated the tumor cell cycle, mitochondrial membrane potential, and intracellular Ca2+ concentration after S-PK6 treatment. Based on the finding that the apoptosis was associated with the p53 signaling pathway and MAPK signaling pathway, western blotting tests were used to assess the expression level of p53 protein and its degenerative regulator MDM2 protein, which showed that S-PK6 could increase p53 levels efficiently. In summary, our results demonstrate the mechanism of action of a small-molecule cyclopeptide, which could be very useful for examining of the possible mechanisms of natural cyclopeptides.
Collapse
Affiliation(s)
- Hui Zhang
- Key Laboratory of Molecular Cytogenetics and Genetic Breeding of Heilongjiang Province, College of Life Science and Technology, Harbin Normal University, Harbin, 150025, China
| | - Huanli Zhang
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, 150025, China
| | - Jingchun Wang
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar, China
| | - Li Fan
- Institute of Medicine and Drug Research, Qiqihar Medical University, Qiqihar, China
| | - Weijie Mu
- Key Laboratory of Molecular Cytogenetics and Genetic Breeding of Heilongjiang Province, College of Life Science and Technology, Harbin Normal University, Harbin, 150025, China.
| | - Yingxue Jin
- Key Laboratory of Molecular Cytogenetics and Genetic Breeding of Heilongjiang Province, College of Life Science and Technology, Harbin Normal University, Harbin, 150025, China; Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, 150025, China.
| | - Zhiqiang Wang
- Key Laboratory for Photonic and Electronic Bandgap Materials, Ministry of Education, College of Chemistry & Chemical Engineering, Harbin Normal University, Harbin, 150025, China.
| |
Collapse
|
5
|
Hou JY, Zhou XL, Wang XY, Liang J, Xue Q. Peroxiredoxin-6 Released by Astrocytes Contributes to Neuroapoptosis During Ischemia. Neuroscience 2023; 512:59-69. [PMID: 36642396 DOI: 10.1016/j.neuroscience.2023.01.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2022] [Revised: 12/26/2022] [Accepted: 01/03/2023] [Indexed: 01/15/2023]
Abstract
Peroxiredoxin-6 (PRDX6), a member of the peroxiredoxin family, has progressively emerged as a possible therapeutic target for a variety of brain diseases, particularly Alzheimer's disease and ischemic stroke. However, the role of PRDX6 in neurons under ischemic conditions has remained elusive. Here, we found that astrocytes could release PRDX6 extracellularly after OGD/R, and that PRDX6 release actually worsened neuroapoptosis under OGD/R. We discovered a unique PRDX6/RAGE/JNK signaling pathway that contributes to the effect of neuroapoptosis. We applied a specific inhibitor of the RAGE signaling pathway in a mouse MCAO model and observed significant alterations in animal behavior. Considered together, our findings show the crucial role of the astrocyte-released PRDX6 in the process of neuroapoptosis caused by OGD/R, and could provide novel insights for investigating the molecular mechanism of protecting brain function from ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Jin-Yi Hou
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China; Department of Acupuncture and Moxibustion, Xuzhou Hospital of Traditional Chinese Medicine, Xuzhou, Jiangsu 221002, China
| | - Xiao-Ling Zhou
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Xiao-Yuan Wang
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China
| | - Jia Liang
- Department of Neurobiology, Xuzhou Medical University, Xuzhou 221002, China
| | - Qun Xue
- Department of Neurology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China; Institute of Clinical Immunology, Jiangsu Key Laboratory of Clinical Immunology, First Affiliated Hospital of Soochow University, Suzhou, Jiangsu 215006, China.
| |
Collapse
|
6
|
Lei L, Lu Q, Ma G, Li T, Deng J, Li W. P53 protein and the diseases in central nervous system. Front Genet 2023; 13:1051395. [PMID: 36712862 PMCID: PMC9880595 DOI: 10.3389/fgene.2022.1051395] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 12/08/2022] [Indexed: 01/11/2023] Open
Abstract
P53 protein is the product of P53 gene, which is a well acknowledged tumor suppressor gene. The function of P53 and the relevant mechanisms of anti-neoplasm have raised the interest of researchers since many years ago. It is demonstrated that P53 is a basic cell cycle regulator and a strong inhibitor for versatile cancers in humans. However, most research focuses on other organs and systems instead of the central nervous system (CNS). In fact, in recent years, more and more studies have been suggesting that P53 plays a significant role in multiple CNS tumors and other diseases and disorders such as cerebral stroke and neurodegenerative diseases. In this work, we mainly reviewed the P53's relationship with CNS tumors, cerebral stroke and neurodegenerative diseases, together with the relevant mechanisms, aiming to summarize the research achievements and providing new insight to the future study on diseases in CNS.
Collapse
Affiliation(s)
- Li Lei
- The Affiliated Hospital of Kunming University of Science and Technology, The Department of Neurosurgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Qixiong Lu
- The Affiliated Hospital of Kunming University of Science and Technology, The Department of Neurosurgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Guifang Ma
- Department of Ear, Nose and Throat (ENT) and Head and Neck (HN) Surgery, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China
| | - Tao Li
- The Affiliated Hospital of Kunming University of Science and Technology, The Department of Neurosurgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China
| | - Jiahong Deng
- Department of Ear, Nose and Throat (ENT) and Head and Neck (HN) Surgery, The First People’s Hospital of Yunnan Province, The Affiliated Hospital of Kunming University of Science and Technology, Kunming, China,*Correspondence: Jiahong Deng, ; Weijia Li,
| | - Weijia Li
- The Affiliated Hospital of Kunming University of Science and Technology, The Department of Neurosurgery, The First People's Hospital of Yunnan Province, Kunming, Yunnan, China,*Correspondence: Jiahong Deng, ; Weijia Li,
| |
Collapse
|
7
|
Khan MM, Paez HG, Pitzer CR, Alway SE. The Therapeutic Potential of Mitochondria Transplantation Therapy in Neurodegenerative and Neurovascular Disorders. Curr Neuropharmacol 2023; 21:1100-1116. [PMID: 36089791 PMCID: PMC10286589 DOI: 10.2174/1570159x05666220908100545] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 07/27/2022] [Accepted: 08/01/2022] [Indexed: 11/22/2022] Open
Abstract
Neurodegenerative and neurovascular disorders affect millions of people worldwide and account for a large and increasing health burden on the general population. Thus, there is a critical need to identify potential disease-modifying treatments that can prevent or slow the disease progression. Mitochondria are highly dynamic organelles and play an important role in energy metabolism and redox homeostasis, and mitochondrial dysfunction threatens cell homeostasis, perturbs energy production, and ultimately leads to cell death and diseases. Impaired mitochondrial function has been linked to the pathogenesis of several human neurological disorders. Given the significant contribution of mitochondrial dysfunction in neurological disorders, there has been considerable interest in developing therapies that can attenuate mitochondrial abnormalities and proffer neuroprotective effects. Unfortunately, therapies that target specific components of mitochondria or oxidative stress pathways have exhibited limited translatability. To this end, mitochondrial transplantation therapy (MTT) presents a new paradigm of therapeutic intervention, which involves the supplementation of healthy mitochondria to replace the damaged mitochondria for the treatment of neurological disorders. Prior studies demonstrated that the supplementation of healthy donor mitochondria to damaged neurons promotes neuronal viability, activity, and neurite growth and has been shown to provide benefits for neural and extra-neural diseases. In this review, we discuss the significance of mitochondria and summarize an overview of the recent advances and development of MTT in neurodegenerative and neurovascular disorders, particularly Parkinson's disease, Alzheimer's disease, and stroke. The significance of MTT is emerging as they meet a critical need to develop a diseasemodifying intervention for neurodegenerative and neurovascular disorders.
Collapse
Affiliation(s)
- Mohammad Moshahid Khan
- Department of Neurology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
- Neuroscience Institute, University of Tennessee Health Science Center, Memphis, TN, USA
- Center for Muscle, Metabolism and Neuropathology, Division of Regenerative and Rehabilitation Sciences and Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Hector G. Paez
- Center for Muscle, Metabolism and Neuropathology, Division of Regenerative and Rehabilitation Sciences and Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, USA
- Laboratory of Muscle Biology and Sarcopenia, Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
- Integrated Biomedical Sciences Graduate Program, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Christopher R. Pitzer
- Center for Muscle, Metabolism and Neuropathology, Division of Regenerative and Rehabilitation Sciences and Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, USA
- Laboratory of Muscle Biology and Sarcopenia, Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
- Integrated Biomedical Sciences Graduate Program, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, TN, 38163, USA
| | - Stephen E. Alway
- Center for Muscle, Metabolism and Neuropathology, Division of Regenerative and Rehabilitation Sciences and Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, USA
- Laboratory of Muscle Biology and Sarcopenia, Department of Physical Therapy, College of Health Professions, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Physiology, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
- The Tennessee Institute of Regenerative Medicine, 910 Madison Avenue, Memphis, TN, 38163, USA
| |
Collapse
|
8
|
Han X, Du J, Shi D, Li L, Li D, Zhang K, Lin S, Zhu J, Huang Z, Zhou Y, Fang Z. Improving Reporter Gene Assay Methodology for Evaluating the Ability of Compounds to Restore P53 Activity. Int J Mol Sci 2022; 23:ijms232213867. [PMID: 36430341 PMCID: PMC9694221 DOI: 10.3390/ijms232213867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/30/2022] [Accepted: 11/04/2022] [Indexed: 11/12/2022] Open
Abstract
Tumor suppressor protein P53 induces cycle arrest and apoptosis by mediating the transcriptional expression of its target genes. Mutations causing conformational abnormalities and post-translational modifications that promote degradation are the main reasons for the loss of P53 function in tumor cells. Reporter gene assays that can scientifically reflect the biological function can help discover the mechanism and therapeutic strategies that restore P53 function. In the reporter gene system of this work, tetracycline-inducible expression of wild-type P53 was used to provide a fully activated state as a 100% activity reference for the objective measurement of biological function. It was confirmed by RT-qPCR, cell viability assay, immunofluorescence, and Western blot analysis that the above-mentioned reporter gene system could correctly reflect the differences in biological activity between the wild-type and mutants. After that, the system was tentatively used for related mechanism research and compound activity evaluation. Through the tetracycline-induced co-expression of wild-type P53 and mutant P53 in exact proportion, it was observed that the response modes of typical transcriptional response elements (TREs) to dominant negative P53 mutation effect were not exactly the same. Compared to the relative multiple-to-solvent control, the activity percentage relative to the 100% activity reference of wild-type P53 can better reflect the actual influence of the so-called P53 mutant reactivator. Similarly, relative to the 100% activity reference, it can objectively reflect the biological effects caused by the inhibitor of P53 negative factors, such as MDM2. In conclusion, this study provides a 100% activity reference and a reliable calculation model for relevant basic research and drug development.
Collapse
Affiliation(s)
- Xinle Han
- Biomedical Research Institute, Shenzhen Peking University—The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
- Department of Pathology, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200011, China
| | - Jun Du
- Department of Hematology, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200127, China
| | - Dandan Shi
- Shenzhen Chipscreen Biosciences Co., Ltd., Shenzhen Hi-Tech Industrial Park, Shenzhen 518057, China
| | - Lingjie Li
- Shenzhen Chipscreen Biosciences Co., Ltd., Shenzhen Hi-Tech Industrial Park, Shenzhen 518057, China
| | - Dandan Li
- Shenzhen Chipscreen Biosciences Co., Ltd., Shenzhen Hi-Tech Industrial Park, Shenzhen 518057, China
| | - Kun Zhang
- Shenzhen Chipscreen Biosciences Co., Ltd., Shenzhen Hi-Tech Industrial Park, Shenzhen 518057, China
| | - Suwen Lin
- Biomedical Research Institute, Shenzhen Peking University—The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
| | - Jingzhong Zhu
- Shenzhen Chipscreen Biosciences Co., Ltd., Shenzhen Hi-Tech Industrial Park, Shenzhen 518057, China
| | - Zoufang Huang
- Ganzhou Key Laboratory of Hematology, Department of Hematology, The First Affiliated Hospital of Gannan Medical University, Ganzhou 341000, China
| | - You Zhou
- Shenzhen Chipscreen Biosciences Co., Ltd., Shenzhen Hi-Tech Industrial Park, Shenzhen 518057, China
- Correspondence: (Y.Z.); (Z.F.)
| | - Zhengyu Fang
- Biomedical Research Institute, Shenzhen Peking University—The Hong Kong University of Science and Technology Medical Center, Shenzhen 518036, China
- Department of Pathology, Peking University Shenzhen Hospital, Shenzhen 518036, China
- Correspondence: (Y.Z.); (Z.F.)
| |
Collapse
|
9
|
Cameron SJ, Sheng J, Hosseinian F, Willmore WG. Nanoparticle Effects on Stress Response Pathways and Nanoparticle-Protein Interactions. Int J Mol Sci 2022; 23:7962. [PMID: 35887304 PMCID: PMC9323783 DOI: 10.3390/ijms23147962] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/01/2022] [Accepted: 07/11/2022] [Indexed: 12/12/2022] Open
Abstract
Nanoparticles (NPs) are increasingly used in a wide variety of applications and products; however, NPs may affect stress response pathways and interact with proteins in biological systems. This review article will provide an overview of the beneficial and detrimental effects of NPs on stress response pathways with a focus on NP-protein interactions. Depending upon the particular NP, experimental model system, and dose and exposure conditions, the introduction of NPs may have either positive or negative effects. Cellular processes such as the development of oxidative stress, the initiation of the inflammatory response, mitochondrial function, detoxification, and alterations to signaling pathways are all affected by the introduction of NPs. In terms of tissue-specific effects, the local microenvironment can have a profound effect on whether an NP is beneficial or harmful to cells. Interactions of NPs with metal-binding proteins (zinc, copper, iron and calcium) affect both their structure and function. This review will provide insights into the current knowledge of protein-based nanotoxicology and closely examines the targets of specific NPs.
Collapse
Affiliation(s)
- Shana J. Cameron
- Department of Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada; (S.J.C.); (F.H.)
| | - Jessica Sheng
- Department of Biology, Carleton University, Ottawa, ON K1S 5B6, Canada;
| | - Farah Hosseinian
- Department of Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada; (S.J.C.); (F.H.)
| | - William G. Willmore
- Department of Chemistry, Carleton University, Ottawa, ON K1S 5B6, Canada; (S.J.C.); (F.H.)
- Department of Biology, Carleton University, Ottawa, ON K1S 5B6, Canada;
- Institute of Biochemistry, Carleton University, Ottawa, ON K1S 5B6, Canada
| |
Collapse
|
10
|
Roshan-Milani S, Sattari P, Ghaderi-Pakdel F, Naderi R. miR-23b/TAB3/NF-κB/p53 axis is involved in hippocampus injury induced by cerebral ischemia-reperfusion in rats: The protective effect of chlorogenic acid. Biofactors 2022; 48:908-917. [PMID: 35201648 DOI: 10.1002/biof.1830] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 02/01/2022] [Indexed: 12/18/2022]
Abstract
Apoptosis is the main pathological aspect of neuronal injury after cerebral ischemia-reperfusion (I/R) injury. However the detailed molecular mediators are still under debate. The aim of this study is to explore the effect of cerebral I/R on miR-23a/TGF-β-activated kinase 1 binding protein 3 (TAB3)/nuclear factor kappa B (NF-κB)/p53 axis in rat hippocampus alone and in combination with chlorogenic acid (CGA). Common carotid artery occlusion (CCAO) was performed by nylon monofilament for 20 min to establish a model of ischemic brain injury. CGA (30 mg/kg) was administered intraperitoneally (ip), 10 min prior to ischemia and 10 min before reperfusion. Examination of hippocampus neurons by terminal deoxynucleotidyl transferase-mediated dUTP nick-end labeling staining showed that the number of apoptotic neurons was elevated at 24 h after reperfusion. At the molecular levels, I/R injury resulted in an increased protein expression of p53 with a concomitant upregulation of cleaved-caspase3/phosphorelated-caspase3 ratio and cytochrome c level. Further miR-23b gene expression was significantly downregulated after 24 h of reperfusion. Also, we observed increased TAB3 and NF-κB protein expressions after 24 h following CCAO. Treatment with CGA significantly reduced the apoptotic damage and also reversed miR-23b gene expression, TAB3 and NF-κB protein expressions in hippocampus neurons in I/R rats. In conclusion our data suggest that miR-23b/TAB3/NF-κB/p53 axis could play a regulatory role in hippocampus cell death, which provide a new target for novel therapeutic interventions during transit ischemic stroke. It also demonstrated that CGA could reverse these molecular alterations indicating an effective component against hippocampus apoptotic insult following acute I/R injury.
Collapse
Affiliation(s)
- Shiva Roshan-Milani
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Parisa Sattari
- Student Research Committee, Urmia University of Medical Sciences, Urmia, Iran
| | - Firouz Ghaderi-Pakdel
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Roya Naderi
- Neurophysiology Research Center, Cellular and Molecular Medicine Institute, Urmia University of Medical Sciences, Urmia, Iran
- Department of Physiology, Faculty of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| |
Collapse
|
11
|
Mesenchymal Stem Cell Therapy: A Potential Treatment Targeting Pathological Manifestations of Traumatic Brain Injury. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:4645021. [PMID: 35757508 PMCID: PMC9217616 DOI: 10.1155/2022/4645021] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 05/30/2022] [Indexed: 01/02/2023]
Abstract
Traumatic brain injury (TBI) makes up a large proportion of acute brain injuries and is a major cause of disability globally. Its complicated etiology and pathogenesis mainly include primary injury and secondary injury over time, which can cause cognitive deficits, physical disabilities, mood changes, and impaired verbal communication. Recently, mesenchymal stromal cell- (MSC-) based therapy has shown significant therapeutic potential to target TBI-induced pathological processes, such as oxidative stress, neuroinflammation, apoptosis, and mitochondrial dysfunction. In this review, we discuss the main pathological processes of TBI and summarize the underlying mechanisms of MSC-based TBI treatment. We also discuss research progress in the field of MSC therapy in TBI as well as major shortcomings and the great potential shown.
Collapse
|
12
|
Cao S, Chen S, Qiao X, Guo Y, Liu F, Ding Z, Jin B. Protocatechualdehyde Rescues Oxygen-Glucose Deprivation/Reoxygenation-Induced Endothelial Cells Injury by Inducing Autophagy and Inhibiting Apoptosis via Regulation of SIRT1. Front Pharmacol 2022; 13:846513. [PMID: 35431914 PMCID: PMC9008765 DOI: 10.3389/fphar.2022.846513] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2021] [Accepted: 03/14/2022] [Indexed: 01/30/2023] Open
Abstract
Background: Oxidative stress-induced endothelial cell death, such as apoptosis and autophagy, plays a critical role in ischemia-reperfusion injury. Protocatechualdehyde (PCA) is a major bioactive component of the traditional Chinese medicine Salvia miltiorrhiza Bunge (Lamiaceae), and it has been proved to be effective in the prevention and treatment of ischemic cardiovascular and cerebrovascular diseases. However, its role in oxidative stress-induced endothelial cell death and its underlying mechanisms remains unclear. This study aims to investigate the effects and mechanisms of PCA on endothelial cell apoptosis and autophagy induced by oxygen-glucose deprivation/reoxygenation (OGD/R) injury. Methods: After OGD/R induction, human umbilical vein endothelial cells (HUVECs) were treated with different concentrations of PCA. Cell viability, apoptosis, and autophagy were detected by Cell Counting Kit-8 assay, flow cytometry, and monodansylcadaverine assay, respectively. Western blot was applied to explore the effects of PCA on the expression levels of relevant protein factors. Results: The results show that PCA significantly promoted cell survival rate and cell proliferation and enhanced the antioxidant activity in OGD/R-induced HUVECs. PCA inhibited HUVECs apoptosis, as evidenced by decreased expression of cleaved-caspase-3, Bcl2-associated X (BAX), and increased expression of Bcl-2. PCA induced autophagy by reducing the expression of P62 while increasing the expression of Beclin-1 and LC3 II/I. Meanwhile, PCA enhanced the expression of Sirtuin 1 (SIRT1) and suppressed the expression of P53. When SIRT1 was inhibited by selisistat or SIRT1 small-interfering RNA, the anti-apoptotic and pro-autophagy abilities of PCA were attenuated. Conclusion: These results demonstrated that PCA rescued HUVECs from OGD/R-induced injury by promoting autophagy and inhibiting apoptosis through SIRT1 and could be developed as a potential therapeutic agent against ischemic diseases.
Collapse
Affiliation(s)
- Shidong Cao
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Senmiao Chen
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Xilin Qiao
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Yan Guo
- School of Basic Medicine, Zhejiang Chinese Medical University, Hangzhou, China
| | - Fang Liu
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| | - Zhishan Ding
- School of Medical Technology and Information Engineering, Zhejiang Chinese Medical University, Hangzhou, China
| | - Bo Jin
- School of Life Science, Zhejiang Chinese Medical University, Hangzhou, China
| |
Collapse
|
13
|
Taheri F, Sattari E, Hormozi M, Ahmadvand H, Bigdeli MR, Kordestani-Moghadam P, Anbari K, Milanizadeh S, Moghaddasi M. Dose-Dependent Effects of Astaxanthin on Ischemia/Reperfusion Induced Brain Injury in MCAO Model Rat. Neurochem Res 2022; 47:1736-1750. [DOI: 10.1007/s11064-022-03565-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 02/22/2022] [Accepted: 02/28/2022] [Indexed: 12/20/2022]
|
14
|
Chavda V, Chaurasia B, Garg K, Deora H, Umana GE, Palmisciano P, Scalia G, Lu B. Molecular mechanisms of oxidative stress in stroke and cancer. BRAIN DISORDERS 2022. [DOI: 10.1016/j.dscb.2021.100029] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
|
15
|
Pluta R, Kiś J, Januszewski S, Jabłoński M, Czuczwar SJ. Cross-Talk between Amyloid, Tau Protein and Free Radicals in Post-Ischemic Brain Neurodegeneration in the Form of Alzheimer’s Disease Proteinopathy. Antioxidants (Basel) 2022; 11:antiox11010146. [PMID: 35052650 PMCID: PMC8772936 DOI: 10.3390/antiox11010146] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/05/2022] [Accepted: 01/09/2022] [Indexed: 02/04/2023] Open
Abstract
Recent years have seen remarkable progress in research into free radicals oxidative stress, particularly in the context of post-ischemic recirculation brain injury. Oxidative stress in post-ischemic tissues violates the integrity of the genome, causing DNA damage, death of neuronal, glial and vascular cells, and impaired neurological outcome after brain ischemia. Indeed, it is now known that DNA damage and repair play a key role in post-stroke white and gray matter remodeling, and restoring the integrity of the blood-brain barrier. This review will present one of the newly characterized mechanisms that emerged with genomic and proteomic development that led to brain ischemia to a new level of post-ischemic neuropathological mechanisms, such as the presence of amyloid plaques and the development of neurofibrillary tangles, which further exacerbate oxidative stress. Finally, we hypothesize that modified amyloid and the tau protein, along with the oxidative stress generated, are new key elements in the vicious circle important in the development of post-ischemic neurodegeneration in a type of Alzheimer’s disease proteinopathy.
Collapse
Affiliation(s)
- Ryszard Pluta
- Laboratory of Ischemic and Neurodegenerative Brain Research, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 Str., 02-106 Warsaw, Poland;
- Correspondence: ; Tel.: +48-22-608-6540
| | - Jacek Kiś
- Department of Urology, 1st Military Clinical Hospital with the Outpatient Clinic, Al. Racławickie 23, 20-049 Lublin, Poland;
| | - Sławomir Januszewski
- Laboratory of Ischemic and Neurodegenerative Brain Research, Mossakowski Medical Research Institute, Polish Academy of Sciences, Pawińskiego 5 Str., 02-106 Warsaw, Poland;
| | - Mirosław Jabłoński
- Department of Rehabilitation and Orthopedics, Medical University of Lublin, Jaczewskiego 8 Str., 20-090 Lublin, Poland;
| | - Stanisław J. Czuczwar
- Department of Pathophysiology, Medical University of Lublin, Jaczewskiego 8b Str., 20-090 Lublin, Poland;
| |
Collapse
|
16
|
Tsai YT, Huang HC, Kao ST, Chang TT, Cheng CY. Neuroprotective Effects of Alpinia oxyphylla Miq against Mitochondria-Related Apoptosis by the Interactions between Upregulated p38 MAPK Signaling and Downregulated JNK Signaling in the Subacute Phase of Cerebral Ischemia-Reperfusion in Rats. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2022; 50:2057-2083. [DOI: 10.1142/s0192415x22500884] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Apoptosis in the penumbra region is the major cell death mechanism occurring during ischemia–reperfusion injury’s early phase. Here, we evaluated how the Alpinia oxyphylla Miq (AOM) affects mitochondria-related apoptosis 3 days after transient middle cerebral artery occlusion (MCAo) and examined the mechanisms underlying the regulation of MAPK-mediated mitochondria-related apoptotic signaling in the peri-infarct cortex in rats. The rats were administered the AOM extract intraperitoneally at doses of 0.2[Formula: see text]g/kg (AOM-0.2[Formula: see text]g), 0.4[Formula: see text]g/kg (AOM-0.4[Formula: see text]g), or 0.8[Formula: see text]g/kg (AOM-0.8[Formula: see text]g) at MCAo initiation. The AOM-0.4[Formula: see text]g and AOM-0.8[Formula: see text]g significantly ameliorated apoptotic cell death and considerably downregulated cytochrome c (cyto c) and cleaved caspase-3 immunoreactivity 3 days after reperfusion. Simultaneously, they significantly downregulated cytosolic p-JNK/JNK, cathepsin B/actin, cyto c/actin, Smac/DIABLO/actin, cleaved caspase-3/actin, and AIF/actin and mitochondrial p53/HSP60 and Bax/HSP60 fractions but upregulated cytosolic p-p38 MAPK/p38 MAPK, p-p90RSK/actin, p-Bad/Bad, p-CREB/actin, and XIAP/actin and cytosolic and mitochondrial Bcl-2/Bax and Bcl-xL/Bax fractions in the peri-infarct cortex. Pretreatment with SB203580 — a p38 MAPK inhibitor — completely abrogated the effects of AOM-0.8[Formula: see text]g on the aforementioned protein expression, whereas treatment with SP600125 — a JNK inhibitor — exerted protective effects similar to those of AOM-0.8[Formula: see text]g. Treatment with 0.4 or 0.8[Formula: see text]g/kg AOM has neuroprotective effects against mitochondria-related apoptosis by suppressing cyto c, Smac/DIABLO, and AIF release from the mitochondria to cytosol. The anti-mitochondria related apoptotic effects of the AOM extract are attributable to the interactions between upregulated p38 MAPK/p90RSK-mediated p-Bad and CREB signaling and downregulated JNK/cathepsin B-mediated Bax and p53 signaling in the peri-infarct cortex 3 days after transient MCAo.
Collapse
Affiliation(s)
- Yueh-Ting Tsai
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University Taichung 40402, Taiwan
- Department of Traditional Chinese Medicine, Kuang Tien General Hospital, Taichung, Taiwan
| | - Hui-Chi Huang
- Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University Taichung 40402, Taiwan
| | - Shung-Te Kao
- School of Chinese Medicine, College of Chinese Medicine, China Medical University Taichung 40402, Taiwan
| | - Tung-Ti Chang
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University Taichung 40402, Taiwan
| | - Chin-Yi Cheng
- School of Post-Baccalaureate Chinese Medicine, College of Chinese Medicine, China Medical University Taichung 40402, Taiwan
- Department of Chinese Medicine, Hui-Sheng Hospital, Taichung 42056, Taiwan
- Department of Chinese Medicine, China Medical University Hospital, Taichung 42056, Taiwan
| |
Collapse
|
17
|
Yang T, Guo R, Ofengeim D, Hwang JY, Zukin RS, Chen J, Zhang F. Molecular and Cellular Mechanisms of Ischemia-Induced Neuronal Death. Stroke 2022. [DOI: 10.1016/b978-0-323-69424-7.00005-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
18
|
Chi OZ, Theis T, Kumar S, Chiricolo A, Liu X, Farooq S, Trivedi N, Young W, Schachner M, Weiss HR. Adhesion molecule L1 inhibition increases infarct size in cerebral ischemia-reperfusion without change in blood-brain barrier disruption. Neurol Res 2021; 43:751-759. [PMID: 34057049 DOI: 10.1080/01616412.2021.1934311] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 05/20/2021] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Neural cell adhesion molecule L1CAM (L1) is involved in neuroprotection. To investigate a possible neuroprotective effect of L1 during ischemia, we determined whether blocking L1 with an antagonistic antibody would worsen the outcome of focal cerebral ischemia-reperfusion and increase blood-brain barrier (BBB) disruption. METHODS Transient middle cerebral artery occlusion (MCAO) was performed in anesthetized rats. Five µg of antagonistic mouse IgG monoclonal L1 antibody 324 or non-immune control mouse IgG was applied on the ischemic-reperfused cortex during one hour of MCAO and two hours of reperfusion. At two hours of reperfusion, BBB permeability, size of infarct using tetrazolium staining, number of TUNEL-labeled apoptotic cells, and immunohistochemistry for expression of PTEN and p53 were studied. RESULTS The antagonistic L1 antibody 324 increased the percentage of cortical infarct area (+36%), but did not affect BBB permeability in the ischemic-reperfused cortex. The antagonistic L1 antibody increased number of apoptotic neurons and p53 expression, but decreased PTEN expression. CONCLUSION Functional antagonism of L1 increases infarct size by increasing numbers of apoptotic neurons without affecting BBB permeability during the early stage of cerebral ischemia-reperfusion. Our data suggest that L1 affects primarily the brain parenchyma rather than BBB during early stages of cerebral ischemia-reperfusion and that endogenous brain L1 may be neuroprotective.
Collapse
Affiliation(s)
- Oak Z Chi
- Department of Anesthesiology and Perioperative Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Thomas Theis
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Suneel Kumar
- Department of Biomedical Engineering, Rutgers University, Piscataway, NJ, USA
| | - Antonio Chiricolo
- Department of Anesthesiology and Perioperative Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Xia Liu
- Department of Anesthesiology and Perioperative Medicine, Rutgers Robert Wood Johnson Medical School, New Brunswick, NJ, USA
| | - Saad Farooq
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Nishta Trivedi
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Wise Young
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Melitta Schachner
- Keck Center for Collaborative Neuroscience and Department of Cell Biology and Neuroscience, Rutgers, The State University of New Jersey, Piscataway, NJ, USA
| | - Harvey R Weiss
- Department of Neuroscience and Cell Biology, Rutgers Robert Wood Johnson Medical School, Piscataway, NJ, USA
| |
Collapse
|
19
|
Liu Y, Wu X, Du D, Liu J, Zhang W, Gao Y, Zhang H. p53 Inhibition Provides a Pivotal Protective Effect against Cerebral Ischemia-Reperfusion Injury via the Wnt Signaling Pathway. Cerebrovasc Dis 2021; 50:682-690. [PMID: 34340236 DOI: 10.1159/000516889] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2021] [Accepted: 04/26/2021] [Indexed: 11/19/2022] Open
Abstract
INTRODUCTION Cerebral ischemia-reperfusion injury enhances brain injury and increases its morbidity and mortality. The purpose of our study was to further explore the specific pathogenesis of cerebral ischemia disease by studying the role of p53 in cerebral ischemia-reperfusion injury and its mechanism to provide a new target for the treatment of cerebral ischemia. METHODS Middle cerebral artery occlusion (MCAo) was established in rats. The changes in p53 and apoptotic proteins in the rat model were detected by quantitative real-time polymerase chain reaction (qRT-PCR) and Western blot. The effects of p53 inhibitors on cerebral is-chemia-reperfusion injury in rats were evaluated by modified neurological severity score (mNSS) and infarct area. Subsequently, neural stem cells (NSCs) were isolated and cultured in vitro, and oxygen and glucose deprivation (OGD) was induced to establish an in vitro ischemia-reperfusion injury model. Cell viability and migration were detected by CCK-8 and transwell assays. Apoptosis of NSCs was detected by flow cytometry. Finally, protein expression in the Wnt pathway activated by p53 was detected by Western blotting. RESULTS Compared with the sham group, p53 levels, mNSS, cerebral infarction area, and apoptosis were significantly increased in the MCAo group (p < 0.05). When the p53 inhibitor PFT-α was injected, the increase in these levels was reversed. Also, the viability and migration of cells decreased and apo-ptosis increased in the in vitro OGD model, whereas the viability, migration, and apoptosis were significantly reversed after the addition of p53 inhibitors (p < 0.05). Finally, p53 induced Wnt signaling pathway proteins β-catenin and cyclin D1 decrease in the MCAo group, while p53 inhibitors reversed their inhibitory effect on the Wnt signaling pathway. CONCLUSION We confirmed in vivo and in vitro that inhibition of p53 has a protective effect on the cerebral ischemia-reperfusion injury, which may be related to the activation of the Wnt signaling pathway.
Collapse
Affiliation(s)
- Yanwei Liu
- Department of Emergency Internal Medicine, the Affiliated Hospital of Qingdao University, Qingdao, China
| | - Xinning Wu
- Department of Cardiovascular Medicine, People's Hospital of Rizhao, Rizhao, China
| | - Deyong Du
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou, China
| | - Jing Liu
- Bincheng Municipal Hospital, Binzhou, China
| | - Wensheng Zhang
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou, China
| | - Yang Gao
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou, China
| | - Haitao Zhang
- Department of Neurosurgery, Binzhou Medical University Hospital, Binzhou, China
| |
Collapse
|
20
|
Shin EJ, Jeong JH, Hwang Y, Sharma N, Dang DK, Nguyen BT, Nah SY, Jang CG, Bing G, Nabeshima T, Kim HC. Methamphetamine-induced dopaminergic neurotoxicity as a model of Parkinson's disease. Arch Pharm Res 2021; 44:668-688. [PMID: 34286473 DOI: 10.1007/s12272-021-01341-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 07/06/2021] [Indexed: 12/01/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease with a high prevalence, approximately 1 % in the elderly population. Numerous studies have demonstrated that methamphetamine (MA) intoxication caused the neurological deficits and nigrostriatal damage seen in Parkinsonian conditions, and subsequent rodent studies have found that neurotoxic binge administration of MA reproduced PD-like features, in terms of its symptomatology and pathology. Several anti-Parkinsonian medications have been shown to attenuate the motor impairments and dopaminergic damage induced by MA. In addition, it has been recognized that mitochondrial dysfunction, oxidative stress, pro-apoptosis, proteasomal/autophagic impairment, and neuroinflammation play important roles in inducing MA neurotoxicity. Importantly, MA neurotoxicity has been shown to share a common mechanism of dopaminergic toxicity with that of PD pathogenesis. This review describes the major findings on the neuropathological features and underlying neurotoxic mechanisms induced by MA and compares them with Parkinsonian pathogenesis. Taken together, it is suggested that neurotoxic binge-type administration of MA in rodents is a valid animal model for PD that may provide knowledge on the neuropathogenesis of PD.
Collapse
Affiliation(s)
- Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea
| | - Ji Hoon Jeong
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, 06974, Seoul, Republic of Korea
| | - Yeonggwang Hwang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea
| | - Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea.,Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, 06974, Seoul, Republic of Korea
| | - Duy-Khanh Dang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea.,Pharmacy Faculty, Can Tho University of Medicine and Pharmacy, 900000, Can Tho City, Vietnam
| | - Bao-Trong Nguyen
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory, Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, 05029, Seoul, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, 16419, Suwon, Republic of Korea
| | - Guoying Bing
- Department of Neuroscience, College of Medicine, University of Kentucky, KY, 40536, Lexington, USA
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Graduate School of Health Science, Fujita Health University, 470-1192, Toyoake, Japan
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea. .,Neuropsychopharmacology & Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea.
| |
Collapse
|
21
|
Padmavathi G, Ramkumar KM. MicroRNA mediated regulation of the major redox homeostasis switch, Nrf2, and its impact on oxidative stress-induced ischemic/reperfusion injury. Arch Biochem Biophys 2021; 698:108725. [PMID: 33326800 DOI: 10.1016/j.abb.2020.108725] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 11/21/2020] [Accepted: 12/11/2020] [Indexed: 12/13/2022]
Abstract
Ischemia/reperfusion injury (IRI) initiates from oxidative stress caused by lack of blood supply and subsequent reperfusion. It is often associated with sterile inflammation, cell death and microvascular dysfunction, which ultimately results in myocardial, cerebral and hepatic IRIs. Reportedly, deregulation of Nrf2 pathway plays a significant role in the oxidative stress-induced IRIs. Further, microRNAs (miRNAs/miRs) are proved to regulate the expression and activation of Nrf2 by targeting either the 3'-UTR or the upstream regulators of Nrf2. Additionally, compounds (crocin, ZnSO4 and ginsenoside Rg1) that modulate the levels of the Nrf2-regulating miRNAs were found to exhibit a protective effect against IRIs of different organs. Therefore, the current review briefs the impact of ischemia reperfusion (I/R) pathogenesis in various organs, role of miRNAs in the regulation of Nrf2 and the I/R protective effect of compounds that alter their expression.
Collapse
Affiliation(s)
- Ganesan Padmavathi
- SRM Research Institute, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India
| | - Kunka Mohanram Ramkumar
- SRM Research Institute, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India; Department of Biotechnology, School of Bioengineering, SRM Institute of Science and Technology, Kattankulathur, 603 203, Tamil Nadu, India.
| |
Collapse
|
22
|
Ludhiadch A, Sharma R, Muriki A, Munshi A. Role of Calcium Homeostasis in Ischemic Stroke: A Review. CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2021; 21:52-61. [DOI: 10.2174/1871527320666210212141232] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/10/2020] [Revised: 07/10/2020] [Accepted: 12/01/2020] [Indexed: 11/22/2022]
Abstract
:
Stroke is the second most common cause of death worldwide. It occurs due to the insufficient supply of oxygen-rich blood to the brain. It is a complex disease with multiple associated risk factors including smoking, alcoholism, age, sex, ethnicity, etc. Calcium ions are known to play a vital role in cell death pathways, which is a ubiquitous intracellular messenger during and immediately after an ischemic period. Disruption in normal calcium hemostasis is known to be a major initiator and activator of the ischemic cell death pathway. Under Ischemic stroke conditions, glutamate is released from the neurons and glia which further activates the N-methyl-D-aspartate (NMDA) receptor and triggers the rapid translocation of Ca2+ from extracellular to intracellular spaces in cerebral tissues and vice versa. Various studies indicated that Ca2+ could have harmful effects on neurons under acute ischemic conditions. Mitochondrial dysfunction also contributes to delayed neuronal death, and it was established decades ago that massive calcium accumulation triggers mitochondrial damage. Elevated Ca2+ levels cause mitochondria to swell and release their contents. As a result oxidative stress and mitochondrial calcium accumulation activate mitochondrial permeability transition and lead to depolarization-coupled production of reactive oxygen species. This association between calcium levels and mitochondrial death suggests that elevated calcium levels might have a role in the neurological outcome in ischemic stroke. Previous studies have also reported that elevated Ca2+ levels play a role in the determination of infarct size, outcome, and recurrence of ischemic stroke. The current review has been compiled to understand the multidimensional role of altered Ca2+ levels in the initiation and alteration of neuronal death after ischemic attack. The underlying mechanisms understood to date have also been discussed.
Collapse
Affiliation(s)
- Abhilash Ludhiadch
- Department of Human Genetics and Molecular Medicine Central University of Punjab, Bathinda-151001, India
| | - Rashmi Sharma
- Department of Human Genetics and Molecular Medicine Central University of Punjab, Bathinda-151001, India
| | - Aishwarya Muriki
- Department of Human Genetics and Molecular Medicine Central University of Punjab, Bathinda-151001, India
| | - Anjana Munshi
- Department of Human Genetics and Molecular Medicine Central University of Punjab, Bathinda-151001, India
| |
Collapse
|
23
|
Chen Y, Yu T. Involvement of p53 in the Responses of Cardiac Muscle Cells to Heat Shock Exposure and Heat Acclimation. J Cardiovasc Transl Res 2020; 13:928-937. [PMID: 32314164 DOI: 10.1007/s12265-020-10003-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Accepted: 04/07/2020] [Indexed: 11/28/2022]
Abstract
Intense heat stress induces damage to the heart, whereas mild to moderate heat stress protects the heart against subsequent ischemic injury. The mechanisms underlying the detrimental and beneficial effects of heat stress remain unclear. In this study, we investigated the role of p53 in the responses of cardiac muscle cells to acute heat exposure and heat acclimation (HA). Heat exposure increased the levels of caspase and annexin, and levels of cytosolic, nuclear, and mitochondrial p53 protein in H9c2 cells. Pifithrin-α or pifithrin-μ reduced heat-induced apoptotic response in these cells. HA reduced localization of p53 in the mitochondria and improved cell viability during heat exposure. The effects of heat exposure and HA on p53 were further verified in vivo in mouse heart tissue. These results suggest that p53 plays a role in heat-induced apoptosis in cardiac muscle cells. The protective effect of HA against heat injury likely involves a p53-dependent mechanism.
Collapse
Affiliation(s)
- Yifan Chen
- Department of Military and Emergency Medicine, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA.
| | - Tianzheng Yu
- Department of Military and Emergency Medicine, Uniformed Services University, 4301 Jones Bridge Road, Bethesda, MD, 20814, USA
- Henry M. Jackson Foundation for the Advancement of Military Medicine, Bethesda, MD, USA
| |
Collapse
|
24
|
Yang Y, Gao L, Niu Y, Li X, Liu W, Jiang X, Liu Y, Zhao Q. Kukoamine A Protects against NMDA-Induced Neurotoxicity Accompanied with Down-Regulation of GluN2B-Containing NMDA Receptors and Phosphorylation of PI3K/Akt/GSK-3β Signaling Pathway in Cultured Primary Cortical Neurons. Neurochem Res 2020; 45:2703-2711. [PMID: 32892226 DOI: 10.1007/s11064-020-03114-y] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Revised: 08/07/2020] [Accepted: 08/14/2020] [Indexed: 12/19/2022]
Abstract
Kukoamine (KuA) is a spermine alkaloid present in traditional Chinese medicine Cortex Lycii radices, which possesses various pharmacological properties. Our previous studies have demonstrated that KuA exerts neuroprotective effects against H2O2-induced oxidative stress, radiation-induced neuroinflammation, oxidative stress and neuronal apoptosis, as well as neurotoxin-induced Parkinson's disease through apoptosis inhibition and autophagy enhancement. The present study aimed to investigate the neuroprotective effects of KuA against NMDA-induced neuronal injury in cultured primary cortical neurons and explore the underlying mechanism. Incubation with 200 μM NMDA for 30 min induced excitotoxicity in primary cultured cortical neurons. The results demonstrated that pretreatment with KuA attenuated NMDA induced cell injury, LDH leakage and neuronal apoptosis. KuA also regulated apoptosis-related proteins. Thus, incubation with the alkaloid decreased the ratio of Bax/Bcl-2, and inhibited the release of cytochrome C, the expression of p53 and the cleavage of caspase-3. Moreover, KuA prevented the upregulation of GluN2B-containing NMDA receptors (NMDAR). Additionally, pretreatment with KuA reversed NMDA-induced dephosphorylation of Akt and GSK-3β and the protective effect of KuA on NMDA-induced cytotoxicity was abolished by wortmannin, a PI3K inhibitor. Taken together, these results indicated that KuA exerted neuroprotective effects against NMDA-induced neurotoxicity in cultural primary cortical neurons and caused the down-regulation of GluN2B-containing NMDARs as well as the phosphorylation of proteins belonging to the PI3K/Akt/GSK-3β signaling pathway.
Collapse
Affiliation(s)
- Yue Yang
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China.,Department of Pharmacy, General Hospital of Northern Military Area, 83 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China
| | - Lingyue Gao
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China
| | - Yixuan Niu
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China
| | - Xiang Li
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China.,Department of Pharmacy, General Hospital of Northern Military Area, 83 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China
| | - Wenwu Liu
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China.,Department of Pharmacy, General Hospital of Northern Military Area, 83 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China
| | - Xiaowen Jiang
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China.,Department of Pharmacy, General Hospital of Northern Military Area, 83 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China
| | - Yaqian Liu
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China.,Department of Pharmacy, General Hospital of Northern Military Area, 83 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China
| | - Qingchun Zhao
- School of Life Sciences and Biopharmaceutics, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China. .,Department of Pharmacy, General Hospital of Northern Military Area, 83 Wenhua Road, Shenyang, Liaoning, 110016, People's Republic of China.
| |
Collapse
|
25
|
Dapagliflozin improves behavioral dysfunction of Huntington's disease in rats via inhibiting apoptosis-related glycolysis. Life Sci 2020; 257:118076. [PMID: 32659371 DOI: 10.1016/j.lfs.2020.118076] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2020] [Revised: 06/24/2020] [Accepted: 07/07/2020] [Indexed: 12/31/2022]
Abstract
AIMS Huntington's disease is a rare neurodegenerative disorder which is associated with defected glucose metabolism with consequent behavioral disturbance including memory and locomotion. 3-nitropropionic acid (3-NP) can cause, in high single dose, an acute striatal injury/Huntington's disease. Dapagliflozin, which is one of the longest duration of action of SGLTIs family, may be able to diminish that injury and its resultant behavioral disturbances. MATERIAL AND METHODS Forty rats were divided into four groups (n = 10 in each group): normal control group (CTRL), dapagliflozin (CTRL + DAPA) group, 3-nitropropionic acid (3-NP) group, and dapagliflozin plus 3-nitropropionic acid (DAPA + 3-NP) group. Behavioral tests (beam walking test, hanging wire test, limb withdrawal test, Y-maze spontaneous alteration, elevated plus maze) were performed with evaluating neurological scoring. In striatum, neurotransmitters (glutamate, aspartate, GABA, ACh and AChE activity) were measured. In addition, apoptosis and glycolysis markers (NF-κB, Cyt-c, lactate, HK-II activity, P53, calpain, PEA15 and TIGAR) were determined. Inflammation (IL-1β, IL-6, IL-8 and TNF-α) and autophagy (beclin-1, LC3 and DRAM) indicators were measured. Additionally, histopathological screening was conducted. KEY FINDINGS 3-Nitropropionic acid had the ability to perturb the neurotransmission which was reflected in impaired behavioral outcome. All of glycolysis, apoptosis and inflammation markers were elevated after 3-NP acute intoxication but autophagy parameters, except DRAM, were reduced. However, DAPA markedly reversed the abovementioned parameters. SIGNIFICANCE Dapagliflozin demonstrated anti-glycolytic, anti-apoptotic, anti-inflammatory and autophagic effects on 3-NP-damaged striatal cells and promoted the behavioral outcome.
Collapse
|
26
|
Remote Ischemic Postconditioning Inhibits Hippocampal Neuronal Apoptosis and Mitophagy After Cardiopulmonary Resuscitation in Rats. Shock 2020; 55:74-82. [PMID: 32590695 DOI: 10.1097/shk.0000000000001596] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
BACKGROUND Studies have shown that remote ischemic post-conditioning can improve brain damage caused by ischemia and hypoxia. However, the specific mechanism underlying this phenomenon is still unclear. The purpose of this study was to investigate the effects of remote ischemic post-conditioning on neuronal apoptosis and mitophagy after cardiopulmonary resuscitation (CPR) in rats. METHODS Male Sprague-Dawley rats were used to establish an asphyxia cardiac arrest model by clamping the tracheal duct. First, the expression levels of P53, Cytochrome c (Cytc), and Parkin in the cytoplasm and mitochondria were observed at 3, 6, 24, and 72 h after the restoration of spontaneous circulation (ROSC). Then neurological deficit scores, hippocampal neuron apoptosis, mitochondrial P53 and Parkin, cytoplasmic Cytc, and neuron ultrastructure were evaluated 24 h after ROSC. RESULTS P53 and Parkin can translocate from the cytoplasm to the mitochondria, promoting the translocation of cytoplasmic Cytc to mitochondria after CPR, reaching a peak at 24 h after the ROSC. The P53 inhibitor Pifithrin-μ reduced apoptosis induced by P53 mitochondrial translocation. Apoptosis was induced after cardiac arrest and attenuated by remote ischemic postconditioning via inhibiting P53 mitochondrial translocation and the release of Cytc to the cytoplasm. In addition, remote ischemic postconditioning could inhibit Parkin-mediated mitophagy. CONCLUSION Taken together, our results show that remote ischemic post-conditioning improves neural function after CPR by inhibiting P53 mitochondrial translocation-induced apoptosis and Parkin-mediated mitophagy.
Collapse
|
27
|
Li Y, Ding H, Liu L, Song Y, Du X, Feng S, Wang X, Li X, Wang Z, Li X, Li J, Wu J, Liu G. Non-esterified Fatty Acid Induce Dairy Cow Hepatocytes Apoptosis via the Mitochondria-Mediated ROS-JNK/ERK Signaling Pathway. Front Cell Dev Biol 2020; 8:245. [PMID: 32411699 PMCID: PMC7198733 DOI: 10.3389/fcell.2020.00245] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 03/24/2020] [Indexed: 12/30/2022] Open
Abstract
Elevated plasma non-esterified fatty acid (NEFA) levels and hepatocytes damage are characteristics of ketosis in dairy cows. Oxidative stress is associated with the pathogenesis of NEFA-induced liver damage. However, the exact mechanism by which oxidative stress mediates NEFA-induced hepatocytes apoptosis and liver injury remains poorly understood. The results of the present study demonstrated that NEFA contribute to reactive oxygen species (ROS) generation, resulting in an imbalance between oxidative and antioxidant species, transcriptional activation of p53, transcriptional inhibition of nuclear factor E2-related factor 2 (Nrf2), loss of mitochondria membrane potential (MMP) and release of apoptosis-inducing factor (AIF) and cytochrome c (cyt c) into the cytosol, leading to hepatocytes apoptosis. Besides, NEFA triggered apoptosis in dairy cow hepatocytes via the regulation of c-Jun N-terminal kinase (JNK), extracellular signal-regulated protein kinases 1 and 2 (ERK1/2), Bcl-2-associated X protein (Bax), B-cell lymphoma gene 2 (Bcl-2), caspase 9 and poly (ADP-ribose) polymerase (PARP). Pretreatment with the inhibitor SP600125 or PD98059 or the antioxidant N-acetylcysteine (NAC) revealed that NEFA-ROS-JNK/ERK-mediated mitochondrial signaling pathway plays a crucial role in NEFA-induced hepatocytes apoptosis. Moreover, the results suggested that the transcription factors p53 and Nrf2 function downstream of this NEFA-ROS-JNK/ERK pathway and are involved in NEFA-induced hepatocytes apoptosis. In conclusion, these findings indicate that the NEFA-ROS-JNK/ERK-mediated mitochondrial pathway plays an important role in NEFA-induced dairy cow hepatocytes apoptosis and strongly suggests that the inhibitors SP600125 and PD98059 and the antioxidant NAC may be developed as therapeutics to prevent hyperlipidemia-induced apoptotic damage in ketotic dairy cows.
Collapse
Affiliation(s)
- Yu Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China.,Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Hongyan Ding
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Leihong Liu
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Yuxiang Song
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xiliang Du
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Shibin Feng
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Xichun Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Xiaobing Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Zhe Wang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xinwei Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jinchun Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Jinjie Wu
- College of Animal Science and Technology, Anhui Agricultural University, Hefei, China
| | - Guowen Liu
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
28
|
Abstract
Traumatic brain injury (TBI) is the leading cause of morbidity and mortality worldwide. Although TBI leads to mechanical damage during initial impact, secondary damage also occurs as results from delayed neurochemical process and intracellular signaling pathways. Accumulated animal and human studies demonstrated that apoptotic mechanism contributes to overall pathology of TBI. Apoptotic cell death has been identified within contusional brain lesion at acute phase of TBI and in region remote from the site directly injured in days to weeks after trauma. TBI is also dynamic conditions that cause neuronal decline overtime and is likely due to neurodegenerative mechanisms years after trauma. Current studies have even suggested association of neuronal damage through apoptotic pathway with mild TBI, which contributes chronic persistent neurological symptoms and cognitive deficits. Thus, a better understanding of the acute and chronic consequences of apoptosis following TBI is required. The purpose of this review is to describe (1) neuronal apoptotic pathway following TBI, (2) contribution of apoptosis to acute and chronic phase of TBI, and (3) current treatment targeting on apoptotic pathway.
Collapse
Affiliation(s)
- Yosuke Akamatsu
- Division of Neurosurgery, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA, USA
- Department of Neurosurgery, Iwate Medical University, Morioka, Japan
| | - Khalid A Hanafy
- Department of Neurology, Harvard Medical School, Boston, MA, USA.
- Division of Neurointensive Care, Beth Israel Deaconess Medical Center, Harvard Medical School, 3 Blackfan Circle Rm 639, Boston, MA, 02115, USA.
| |
Collapse
|
29
|
He Z, Ning N, Zhou Q, Khoshnam SE, Farzaneh M. Mitochondria as a therapeutic target for ischemic stroke. Free Radic Biol Med 2020; 146:45-58. [PMID: 31704373 DOI: 10.1016/j.freeradbiomed.2019.11.005] [Citation(s) in RCA: 163] [Impact Index Per Article: 32.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 10/07/2019] [Accepted: 11/03/2019] [Indexed: 12/24/2022]
Abstract
Stroke is the leading cause of death and physical disability worldwide. Mitochondrial dysfunction has been considered as one of the hallmarks of ischemic stroke and contributes to the pathology of ischemia and reperfusion. Mitochondria is essential in promoting neural survival and neurological improvement following ischemic stroke. Therefore, mitochondria represent an important drug target for stroke treatment. This review discusses the mitochondrial molecular mechanisms underlying cerebral ischemia and involved in reactive oxygen species generation, mitochondrial electron transport dysfunction, mitochondria-mediated regulation of inflammasome activation, mitochondrial dynamics and biogenesis, and apoptotic cell death. We highlight the potential of mitochondrial transfer by stem cells as a therapeutic target for stroke treatment and provide valuable insights for clinical strategies. A better understanding of the roles of mitochondria in ischemia-induced cell death and protection may provide a rationale design of novel therapeutic interventions in the ischemic stroke.
Collapse
Affiliation(s)
- Zhi He
- Department of Pharmacy, Luohe Medical College, Luohe, 462000, China
| | - Niya Ning
- Department of Obstetrics and Gynecology, Shaoling District People's Hospital of Luohe City, Luohe, 462300, China
| | - Qiongxiu Zhou
- Institute of Blood Transfusion, Chinese Academy of Medical Sciences, Chengdu, 610052, China.
| | - Seyed Esmaeil Khoshnam
- Physiology Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran.
| | - Maryam Farzaneh
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| |
Collapse
|
30
|
Ginkgetin attenuates cerebral ischemia-reperfusion induced autophagy and cell death via modulation of the NF-κB/p53 signaling pathway. Biosci Rep 2019; 39:BSR20191452. [PMID: 31420372 PMCID: PMC6732367 DOI: 10.1042/bsr20191452] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2019] [Revised: 07/27/2019] [Accepted: 08/01/2019] [Indexed: 12/12/2022] Open
Abstract
Background: Cerebral ischemia–reperfusion (I/R) injury is the key to fatality in cerebrovascular accident, hence further endeavor is warranted to delineate the mechanism underlying its lethal aggravation procedure. In the present study, we aimed to elucidate the anti-autophagy and anti-apoptosis effects of ginkgetin via nuclear factor κB (NF-κB)/p53 pathway in cerebral I/R rats. Methods: Rats were administrated 2-h occlusion of right middle cerebral artery before the 24-h reperfusion followed. There were three doses of ginkgetin (25, 50, 100 mg/kg) given intraperitoneally (i.p.) after the 2-h ischemia, and Pifithrin-α (PFT-α, p53 inhibitor), SN50 (NF-κB inhibitor) and 3-methyladenine (3-MA, autophagy inhibitor) was administered 20 min before the ischemia, respectively. Results: The neurological deficits decreased significantly with the administration of ginkgetin. The concentrations of microtubule-associated protein 1 light chain 3-II and p53 were significantly decreased by PFT-α, 3-MA and ginkgetin. The concentrations of Beclin 1, damage-regulated autophagy modulator, cathepsin B and cathepsin D were significantly decreased due to the administration of PFT-α, ginkgetin and SN50. Furthermore, the concentrations of Bax and p53-upregulated modulator of apoptosis were significantly decreased with that of Bcl-2 being significantly increased by administration of SN50, PFT-α and ginkgetin. Conclusion: Ginkgetin can alleviate cerebral ischemia/reperfusion induced autophagy and apoptosis by inhibiting the NF-κB/p53 signaling pathway.
Collapse
|
31
|
Pifithrin-Alpha Reduces Methamphetamine Neurotoxicity in Cultured Dopaminergic Neurons. Neurotox Res 2019; 36:347-356. [DOI: 10.1007/s12640-019-00050-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/26/2019] [Accepted: 04/16/2019] [Indexed: 12/28/2022]
|
32
|
Genetic depletion of p53 attenuates cocaine-induced hepatotoxicity in mice. Biochimie 2018; 158:53-61. [PMID: 30576773 DOI: 10.1016/j.biochi.2018.12.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2018] [Accepted: 12/14/2018] [Indexed: 12/13/2022]
Abstract
Cocaine, an addictive drug, is known to induce hepatotoxicity via oxidative damage and proapoptosis. Since p53, a tumor suppressor gene, plays a major role in inducing oxidative stress and apoptosis, we examined the role of p53 inhibition against cocaine-induced hepatotoxicity. Cocaine treatment significantly increased oxidative parameters (i.e., reactive oxygen species, 4-hydroxylnonenal, and protein carbonyl) in the liver of wild type (WT) mice. We found that the pharmacological (i.e. pifithrin-α) and genetic (i.e. p53 knockout) inhibition of p53 significantly attenuates cocaine-induced hepatotoxicity. Cocaine treatment increased alanine aminotransferase (ALT) and aspartate aminotransferase (AST) levels in the serum of mice, signifying hepatic damage. Consistently, these increases were attenuated by inhibition of p53, implying protection against cocaine-induced hepatic damage. In addition, cocaine treatment significantly increased PKCδ, cleaved PKCδ and p53 levels in the liver of WT mice. These increases were followed by the interaction between p53 and PKCδ, and pro-apoptotic consequences (i.e., cytosolic release of cytochrome c, activation of caspase-3, increase in Bax level and decreases in Bcl-2 and Bcl-xL levels). These changes were attenuated by p53 depletion, reflecting that the critical role of PKCδ in p53-mediated apoptotic potentials. Combined, our results suggest that the inhibition of p53 is important for protection against oxidative burdens, pro-apoptotic events, and hepatic degeneration induced by cocaine.
Collapse
|
33
|
Dehghani A, Karatas H, Can A, Erdemli E, Yemisci M, Eren-Kocak E, Dalkara T. Nuclear expansion and pore opening are instant signs of neuronal hypoxia and can identify poorly fixed brains. Sci Rep 2018; 8:14770. [PMID: 30282977 PMCID: PMC6170374 DOI: 10.1038/s41598-018-32878-1] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Accepted: 09/11/2018] [Indexed: 11/09/2022] Open
Abstract
The initial phase of neuronal death is not well characterized. Here, we show that expansion of the nuclear membrane without losing its integrity along with peripheralization of chromatin are immediate signs of neuronal injury. Importantly, these changes can be identified with commonly used nuclear stains and used as markers of poor perfusion-fixation. Although frozen sections are widely used, no markers are available to ensure that the observed changes were not confounded by perfusion-induced hypoxia/ischemia. Moreover, HMGB1 was immediately released and p53 translocated to mitochondria in hypoxic/ischemic neurons, whereas nuclear pore complex inhibitors prevented the nuclear changes, identifying novel neuroprotection targets.
Collapse
Affiliation(s)
- Anisa Dehghani
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, Ankara, 06100, Turkey
| | - Hulya Karatas
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, Ankara, 06100, Turkey
| | - Alp Can
- Ankara University, School of Medicine, Department of Histology and Embryology, Ankara, 06100, Turkey
| | - Esra Erdemli
- Ankara University, School of Medicine, Department of Histology and Embryology, Ankara, 06100, Turkey
| | - Muge Yemisci
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, Ankara, 06100, Turkey.,Hacettepe University, Faculty of Medicine, Department of Neurology, Ankara, 06100, Turkey
| | - Emine Eren-Kocak
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, Ankara, 06100, Turkey.,Hacettepe University, Faculty of Medicine, Department of Psychiatry, Ankara, 06100, Turkey
| | - Turgay Dalkara
- Hacettepe University, Institute of Neurological Sciences and Psychiatry, Ankara, 06100, Turkey. .,Hacettepe University, Faculty of Medicine, Department of Neurology, Ankara, 06100, Turkey.
| |
Collapse
|
34
|
Sun M, Izumi H, Shinoda Y, Fukunaga K. Neuroprotective effects of protein tyrosine phosphatase 1B inhibitor on cerebral ischemia/reperfusion in mice. Brain Res 2018; 1694:1-12. [PMID: 29705606 DOI: 10.1016/j.brainres.2018.04.029] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2018] [Revised: 04/10/2018] [Accepted: 04/24/2018] [Indexed: 12/11/2022]
Abstract
Akt (Protein kinase B, PKB), a serine/threonine kinase, plays a critical role in cell development, growth, and survival. Akt phosphorylation mediates a neuroprotective effect against ischemic injury. Recently, a protein-tyrosine phosphatase-1B (PTP1B) inhibitor (KY-226) was developed to elicit anti-diabetic and anti-obesity effects via enhancement of insulin signaling. Previously, we reported that the nonselective PTP1B inhibitor, sodium orthovanadate, rescued neurons from delayed neuronal death during brain ischemia. In this study, we confirmed the ameliorative effects of KY-226 on ischemia/reperfusion (I/R) injury using a murine model of middle cerebral artery occlusion (MCAO). ICR mice were subjected to MCAO for 2 h followed by reperfusion. Although KY-226 permeability was poor through the blood-brain barrier (BBB) of normal mice, it could penetrate through the BBB of mice after I/R insult. Intraperitoneal KY-226 administration elicited dose-dependent reductions in infarcted brain areas and improved neurological deficits. The neuroprotective effects of KY-266 were obtained when administered within 0.5 h after reperfusion. KY-226 (10 mg/kg) also restored reduced Akt phosphorylation and eNOS phosphorylation (Ser-1177) levels following I/R insult. Moreover, 10 mg/kg of KY-226 improved I/R-induced decreased extracellular signal-regulated kinase (ERK) phosphorylation. Furthermore, KY-226 attenuated the generation of reactive oxygen species (ROS) in mouse cortex. These results suggest that KY-226 may act as a novel therapeutic candidate for ischemic stroke. Activation of Akt and ERK possibly underlie the neuroprotective mechanism of KY-226.
Collapse
Affiliation(s)
- Meiling Sun
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai Japan
| | - Hisanao Izumi
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai Japan
| | - Yasuharu Shinoda
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai Japan
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai Japan.
| |
Collapse
|
35
|
Yang JL, Mukda S, Chen SD. Diverse roles of mitochondria in ischemic stroke. Redox Biol 2018; 16:263-275. [PMID: 29549824 PMCID: PMC5854930 DOI: 10.1016/j.redox.2018.03.002] [Citation(s) in RCA: 320] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/01/2018] [Accepted: 03/06/2018] [Indexed: 12/15/2022] Open
Abstract
Stroke is the leading cause of adult disability and mortality in most developing and developed countries. The current best practices for patients with acute ischemic stroke include intravenous tissue plasminogen activator and endovascular thrombectomy for large-vessel occlusion to improve clinical outcomes. However, only a limited portion of patients receive thrombolytic therapy or endovascular treatment because the therapeutic time window after ischemic stroke is narrow. To address the current shortage of stroke management approaches, it is critical to identify new potential therapeutic targets. The mitochondrion is an often overlooked target for the clinical treatment of stroke. Early studies of mitochondria focused on their bioenergetic role; however, these organelles are now known to be important in a wide range of cellular functions and signaling events. This review aims to summarize the current knowledge on the mitochondrial molecular mechanisms underlying cerebral ischemia and involved in reactive oxygen species generation and scavenging, electron transport chain dysfunction, apoptosis, mitochondrial dynamics and biogenesis, and inflammation. A better understanding of the roles of mitochondria in ischemia-related neuronal death and protection may provide a rationale for the development of innovative therapeutic regimens for ischemic stroke and other stroke syndromes. Review of current treatment of ischemic stroke indicates deficiency in the contemporary methods. Discuss the mitochondrial ROS-related signaling that affect neuronal fate after ischemic stroke. Mechanisms of mitochondrial dynamics and mitophagy could be pivotal for ischemic stroke. Inhibiting mitochondrion-induced inflammatory response is a potential treatment for ischemic stroke.
Collapse
Affiliation(s)
- Jenq-Lin Yang
- Institute for Translation Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, 123 Dapi Road, Kaohsiung 83301, Taiwan, ROC
| | - Sujira Mukda
- Institute for Translation Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, 123 Dapi Road, Kaohsiung 83301, Taiwan, ROC; Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, 25/25 Phuttamonthon 4 Road, Salaya, Nakhon Pathom 73170, Thailand
| | - Shang-Der Chen
- Institute for Translation Research in Biomedicine, Kaohsiung Chang Gung Memorial Hospital, 123 Dapi Road, Kaohsiung 83301, Taiwan, ROC; Department of Neurology, Kaohsiung Chang Gung Memorial Hospital, 123 Dapi Road, Kaohsiung 83301, Taiwan, ROC; College of Medicine, Chang Gung University, 259 Wenhua 1st Road, Taoyuan 33302, Taiwan, ROC.
| |
Collapse
|
36
|
bFGF plays a neuroprotective role by suppressing excessive autophagy and apoptosis after transient global cerebral ischemia in rats. Cell Death Dis 2018; 9:172. [PMID: 29416039 PMCID: PMC5833346 DOI: 10.1038/s41419-017-0229-7] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2017] [Revised: 11/20/2017] [Accepted: 12/14/2017] [Indexed: 12/31/2022]
Abstract
Transient global cerebral ischemia (tGCI) is a cerebrovascular disorder that can cause apoptotic neuronal damage and functional deficits. Basic fibroblast growth factor (bFGF) was reported to be highly expressed in the central nervous system (CNS) and to exert neuroprotective effects against different CNS diseases. However, the effects of bFGF on tGCI have not been studied intensively. This study was conducted to investigate the effect of bFGF and its underlying mechanism in an animal model of tGCI. After intracerebroventricular (i.c.v.) injection of bFGF, functional improvement was observed, and the number of viable neurons increased in the ischemia-vulnerable hippocampal CA1 region. Apoptosis was induced after tGCI and could be attenuated by bFGF treatment via inhibition of p53 mitochondrial translocation. In addition, autophagy was activated during this process, and bFGF could inhibit activation of autophagy through the mTOR pathway. Rapamycin, an activator of autophagy, was utilized to explore the relationship among bFGF, apoptosis, and autophagy. Apoptosis deteriorated after rapamycin treatment, which indicated that excessive autophagy could contribute to the apoptosis process. In conclusion, these results demonstrate that bFGF could exert neuroprotective effects in the hippocampal CA1 region by suppressing excessive autophagy via the mTOR pathway and inhibiting apoptosis by preventing p53 mitochondrial translocation. Furthermore, our results suggest that bFGF may be a promising therapeutic agent to for treating tGCI in response to major adverse events, including cardiac arrest, shock, extracorporeal circulation, traumatic hemorrhage, and asphyxiation.
Collapse
|
37
|
Free Radical Damage in Ischemia-Reperfusion Injury: An Obstacle in Acute Ischemic Stroke after Revascularization Therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:3804979. [PMID: 29770166 PMCID: PMC5892600 DOI: 10.1155/2018/3804979] [Citation(s) in RCA: 320] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2017] [Accepted: 12/07/2017] [Indexed: 12/16/2022]
Abstract
Acute ischemic stroke is a common cause of morbidity and mortality worldwide. Thrombolysis with recombinant tissue plasminogen activator and endovascular thrombectomy are the main revascularization therapies for acute ischemic stroke. However, ischemia-reperfusion injury after revascularization therapy can result in worsening outcomes. Among all possible pathological mechanisms of ischemia-reperfusion injury, free radical damage (mainly oxidative/nitrosative stress injury) has been found to play a key role in the process. Free radicals lead to protein dysfunction, DNA damage, and lipid peroxidation, resulting in cell death. Additionally, free radical damage has a strong connection with inducing hemorrhagic transformation and cerebral edema, which are the major complications of revascularization therapy, and mainly influencing neurological outcomes due to the disruption of the blood-brain barrier. In order to get a better clinical prognosis, more and more studies focus on the pharmaceutical and nonpharmaceutical neuroprotective therapies against free radical damage. This review discusses the pathological mechanisms of free radicals in ischemia-reperfusion injury and adjunctive neuroprotective therapies combined with revascularization therapy against free radical damage.
Collapse
|
38
|
Cao L, Zhang Y, Zhang S, Jiang TP, Chen L, Liu J, Zhou S. MicroRNA-29b alleviates oxygen and glucose deprivation/reperfusion-induced injury via inhibition of the p53-dependent apoptosis pathway in N2a neuroblastoma cells. Exp Ther Med 2017; 15:67-74. [PMID: 29399057 PMCID: PMC5766061 DOI: 10.3892/etm.2017.5410] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2016] [Accepted: 07/20/2017] [Indexed: 12/14/2022] Open
Abstract
Cerebral ischemic injury causes severe brain damage and remains one of the leading causes of morbidity and mortality worldwide. Members of the microRNA-29 (miR-29) family are involved in regulating the process of ischemia and may be developed as biomarkers to diagnose and treat cerebral ischemia. The role of miR-29b in cerebral ischemia injury remains poorly understood. The purpose of the present study was to investigate whether miR-29b overexpression suppressed cerebral ischemic injury and to explore its underlying mechanism of action. The results demonstrated that levels of miR-29b in N2a neuroblastoma cells decreased following oxygen and glucose deprivation/reperfusion (OGD/R) treatment. Transfection with miR-29b mimics significantly increased cell viability, decreased lactate dehydrogenase (LDH) leakage, inhibited apoptosis by decreasing morphological changes occurring in the nuclei and reduced caspase-3 activity in OGD/R-treated N2a cells. Conversely, miR-29b inhibitors enhanced OGD/R-induced cytotoxicity and apoptosis. In addition, the miR-29b mimics blocked the increase in Bax and p53 expression and decreased Bcl-2 expression in OGD/R-treated N2a cells, whereas miR-29b inhibitors exacerbated the changes in the expression of these apoptosis-associated proteins caused by OGD/R. p53 knockdown using p53 small interfering RNA decreased cell viability and increased LDH leakage, reversing the improvements that the miR-29b mimics induced in damaged cells. Taken together, the results of the present study demonstrated that miR-29b attenuates ischemic injury by negatively regulating the p53-dependent apoptosis pathway and may therefore be a novel potential therapeutic target for treating ischemic stroke.
Collapse
Affiliation(s)
- Lei Cao
- Department of Interventional Radiology, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Yu Zhang
- Department of Interventional Radiology, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Shuai Zhang
- Department of Interventional Radiology, Tumor Hospital Affiliated with Guizhou Medical University, Guiyang, Guizhou 550000, P.R. China
| | - Tian-Peng Jiang
- Department of Interventional Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Li Chen
- Department of Interventional Radiology, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Jing Liu
- Department of Interventional Radiology, Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| | - Shi Zhou
- Department of Interventional Radiology, The Affiliated Hospital of Guizhou Medical University, Guiyang, Guizhou 550004, P.R. China
| |
Collapse
|
39
|
Abd El-Aal SA, Abd El-Fattah MA, El-Abhar HS. CoQ10 Augments Rosuvastatin Neuroprotective Effect in a Model of Global Ischemia via Inhibition of NF-κB/JNK3/Bax and Activation of Akt/FOXO3A/Bim Cues. Front Pharmacol 2017; 8:735. [PMID: 29081748 PMCID: PMC5645536 DOI: 10.3389/fphar.2017.00735] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Accepted: 09/29/2017] [Indexed: 12/12/2022] Open
Abstract
Statins were reported to lower the Coenzyme Q10 (CoQ10) content upon their inhibition of HMG-CoA reductase enzyme and both are known to possess neuroprotective potentials; therefore, the aim is to assess the possible use of CoQ10 as an adds-on therapy to rosuvastatin to improve its effect using global I/R model. Rats were allocated into sham, I/R, rosuvastatin (10 mg/kg), CoQ10 (10 mg/kg) and their combination. Drugs were administered orally for 7 days before I/R. Pretreatment with rosuvastatin and/or CoQ10 inhibited the hippocampal content of malondialdehyde, nitric oxide, and boosted glutathione and superoxide dismutase. They also opposed the upregulation of gp91phox, and p47phox subunits of NADPH oxidase. Meanwhile, both agents reduced content/expression of TNF-α, iNOS, NF-κBp65, ICAM-1, and MPO. Besides, all regimens abated cytochrome c, caspase-3 and Bax, but increased Bcl-2 in favor of cell survival. On the molecular level, they increased p-Akt and its downstream target p-FOXO3A, with the inhibition of the nuclear content of FOXO3A to downregulate the expression of Bim, a pro-apoptotic gene. Additionally, both treatments downregulate the JNK3/c-Jun signaling pathway. The effect of the combination regimen overrides that of either treatment alone. These effects were reflected on the alleviation of the hippocampal damage in CA1 region inflicted by I/R. Together, these findings accentuate the neuroprotective potentials of both treatments against global I/R by virtue of their rigorous multi-pronged actions, including suppression of hippocampal oxidative stress, inflammation, and apoptosis with the involvement of the Akt/FOXO3A/Bim and JNK3/c-Jun/Bax signaling pathways. The study also nominates CoQ10 as an adds-on therapy with statins.
Collapse
Affiliation(s)
- Sarah A Abd El-Aal
- Department of Pharmacology and Toxicology, October 6 University, Cairo, Egypt
| | - Mai A Abd El-Fattah
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Hanan S El-Abhar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| |
Collapse
|
40
|
Chen J, Wang Z, Zheng Z, Chen Y, Khor S, Shi K, He Z, Wang Q, Zhao Y, Zhang H, Li X, Li J, Yin J, Wang X, Xiao J. Neuron and microglia/macrophage-derived FGF10 activate neuronal FGFR2/PI3K/Akt signaling and inhibit microglia/macrophages TLR4/NF-κB-dependent neuroinflammation to improve functional recovery after spinal cord injury. Cell Death Dis 2017; 8:e3090. [PMID: 28981091 PMCID: PMC5682656 DOI: 10.1038/cddis.2017.490] [Citation(s) in RCA: 120] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2017] [Revised: 08/07/2017] [Accepted: 08/28/2017] [Indexed: 01/09/2023]
Abstract
Therapeutics used to treat central nervous system (CNS) injury were designed to repair neurites and inhibit cell apoptosis. Previous studies have shown that neuron-derived FGF10 exerts potential neuroprotective effects after cerebral ischemia injury. However, little is known about the role of endogenous FGF10 in the recovery process after spinal cord injury (SCI). In this study, we found that FGF10 is mainly produced by neuron and microglia/macrophages, and its expression is increased after SCI. Exogenous treatment of FGF10 improved functional recovery after injury by reducing apoptosis, as well as repairing neurites via FGFR2/PI3K/Akt pathway. On another hand, inhibiting the PI3K/Akt pathway with LY294002 partially reversed the therapeutic effects of FGF10. In addition, small interfering RNA knockdown of FGFR2 suppressed PI3K/Akt pathway activation by FGF10 and abolished its anti-apoptotic and neurite repair effects in vitro. Furthermore, FGF10 treatment inhibited the activation and proliferation of microglia/macrophages through regulation of TLR4/NF-κB pathway, and attenuated the release of pro-inflammatory cytokines after SCI. Thus, the increased expression of FGF10 after acute SCI is an endogenous self-protective response, suggesting that FGF10 could be a potential treatment for CNS injury.
Collapse
Affiliation(s)
- Jian Chen
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zhouguang Wang
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - ZengMing Zheng
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Yu Chen
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Sinan Khor
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, USA
| | - KeSi Shi
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - ZiLi He
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Qingqing Wang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Yingzheng Zhao
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Hongyu Zhang
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiaokun Li
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Jiawei Li
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiayu Yin
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| | - Xiangyang Wang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jian Xiao
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
41
|
Ye Z, Li Q, Guo Q, Xiong Y, Guo D, Yang H, Shu Y. Ketamine induces hippocampal apoptosis through a mechanism associated with the caspase-1 dependent pyroptosis. Neuropharmacology 2017; 128:63-75. [PMID: 28963039 DOI: 10.1016/j.neuropharm.2017.09.035] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2017] [Revised: 08/24/2017] [Accepted: 09/25/2017] [Indexed: 12/11/2022]
Abstract
Ketamine, a pediatric anesthetic, is widely used in clinical practice. There was growing evidence showing that ketamine can promote neuronal death in developing brains of both humans and animals. In this study, we used in vivo neonatal and juvenile mouse models to induce ketamine-related neurotoxicity in the hippocampus. Active caspase-3 and -9 proteins, which are responsible for the release of cytochrome C, and the mitochondrial translocation of p53, which is associated with mitochondrial apoptosis, were found to be significantly up-regulated in the ketamine-induced hippocampal neurotoxicity. Furthermore, we demonstrated that the levels of pyroptosis-related proteins, including caspase-1 and -11, NOD-like receptor family, pyrin domain containing 3 (NLRP3), and IL-1β and IL-18, significantly increased after multiple doses of ketamine administration. We speculated that ketamine triggered the formation of NLRP3 and caspase-1 complex and its translocation to the mitochondria. In consistent with this, ketamine treatment was found to induce pyroptosis in mouse primary hippocampal neurons, which was characterized by increased pore formation and elevated lactate dehydrogenase release in mitochondria. Silencing caspase-1 with lentivirus-mediated short hairpin RNA (shRNA) significantly decreased the levels of not only pyroptosis-related proteins but also mitochondrial apoptosis-associated proteins in mouse primary hippocampal neurons. We conclude that caspase-1-dependent pyroptosis is an important event which may be an essential pathway involved in the mitochondria-associated apoptosis in ketamine-induced hippocampal neurotoxicity.
Collapse
Affiliation(s)
- Zhi Ye
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, MD, USA; Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Qing Li
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, MD, USA; Institute of Clinical Pharmacology, Central South University, Hunan 410078, China
| | - Qulian Guo
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Yunchuan Xiong
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Dong Guo
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, MD, USA
| | - Hong Yang
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, MD, USA
| | - Yan Shu
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Maryland at Baltimore, MD, USA.
| |
Collapse
|
42
|
Liu T, Wang F, LePochat P, Woo JAA, Bukhari MZ, Hong KW, Trotter C, Kang DE. Cofilin-mediated Neuronal Apoptosis via p53 Translocation and PLD1 Regulation. Sci Rep 2017; 7:11532. [PMID: 28912445 PMCID: PMC5599510 DOI: 10.1038/s41598-017-09996-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2017] [Accepted: 08/01/2017] [Indexed: 01/15/2023] Open
Abstract
Amyloid β (Aβ) accumulation is an early event in the pathogenesis of Alzheimer’s disease (AD), leading to mitochondrial and synaptic dysfunction, tau accumulation, and eventual neuronal death. While the p53 apoptotic pathway has clearly been associated with Aβ deposits and neuronal apoptosis, the critical upstream factors contributing to p53 activation in AD are not well understood. We have previously shown that cofilin activation plays a pivotal role in Aβ-induced mitochondrial and synaptic dysfunction. In this study, we show that activated cofilin (S3A) preferentially forms a complex with p53 and promotes its mitochondrial and nuclear localization, resulting in transcription of p53-responsive genes and promotion of apoptosis. Conversely, reduction of endogenous cofilin by knockdown or genetic deficiency inhibits mitochondrial and nuclear translocation of p53 in cultured cells and in APP/PS1 mice. This cofilin-p53 pro-apoptotic pathway is subject to negative regulation by PLD1 thorough cofilin inactivation and inhibition of cofilin/p53 complex formation. Finally, activated cofilin is unable to induce apoptosis in cells genetically lacking p53. These findings taken together indicate that cofilin coopts and requires the nuclear and mitochondrial pro-apoptotic p53 program to induce and execute apoptosis, while PLD1 functions in a regulatory multi-brake capacity in this pathway.
Collapse
Affiliation(s)
- Tian Liu
- USF Health Byrd Alzheimer's Institute, Department of Molecular of Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL, 33613, USA
| | - Fang Wang
- USF Health Byrd Alzheimer's Institute, Department of Molecular of Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL, 33613, USA
| | - Patrick LePochat
- USF Health Byrd Alzheimer's Institute, Department of Molecular of Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL, 33613, USA
| | - Jung-A A Woo
- USF Health Byrd Alzheimer's Institute, Department of Molecular of Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL, 33613, USA
| | - Mohammed Zaheen Bukhari
- USF Health Byrd Alzheimer's Institute, Department of Molecular of Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL, 33613, USA
| | - Kyung Woo Hong
- USF Health Byrd Alzheimer's Institute, Department of Molecular of Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL, 33613, USA
| | - Courtney Trotter
- USF Health Byrd Alzheimer's Institute, Department of Molecular of Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL, 33613, USA
| | - David E Kang
- USF Health Byrd Alzheimer's Institute, Department of Molecular of Medicine, University of South Florida, Morsani College of Medicine, Tampa, FL, 33613, USA. .,James A. Haley Veteran's Administration Hospital, Tampa, FL, 33612, USA.
| |
Collapse
|
43
|
Chen J, Wang Z, Mao Y, Zheng Z, Chen Y, Khor S, Shi K, He Z, Li J, Gong F, Liu Y, Hu A, Xiao J, Wang X. Liraglutide activates autophagy via GLP-1R to improve functional recovery after spinal cord injury. Oncotarget 2017; 8:85949-85968. [PMID: 29156769 PMCID: PMC5689659 DOI: 10.18632/oncotarget.20791] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 07/13/2017] [Indexed: 12/18/2022] Open
Abstract
Therapeutics used to treat central nervous system (CNS) injury are designed to promote axonal regeneration and inhibit cell death. Previous studies have shown that liraglutide exerts potent neuroprotective effects after brain injury. However, little is known if liraglutide treatment has neuroprotective effects after spinal cord injury (SCI). This study explores the neuroprotective effects of liraglutide and associated underlying mechanisms. Our results showed that liraglutide could improve recovery after injury by decreasing apoptosis as well as increasing microtubulin acetylation, and autophagy. Autophagy inhibition with 3-methyladenine (3-MA) partially reversed the preservation of spinal cord tissue and decreased microtubule acetylation and polymerization. Additionally, siRNA knockdown of GLP-1R suppressed autophagy and reversed mTOR inhibition induced by liraglutide in vitro, indicating that GLP-1R regulates autophagic flux. GLP-1R knockdown ameliorated the mTOR inhibition and autophagy induction seen with liraglutide treatment in PC12 cells under H2O2 stimulation. Taken together, our study demonstrated that liraglutide could reduce apoptosis, improve functional recovery, and increase microtubule acetylation via autophagy stimulation after SCI. GLP-1R was associated with both the induction of autophagy and suppression of apoptosis in neuronal cultures.
Collapse
Affiliation(s)
- Jian Chen
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zhouguang Wang
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yuqin Mao
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Zengming Zheng
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yu Chen
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Sinan Khor
- Department of Molecular Pharmacology, Albert Einstein College of Medicine, Bronx, NY, U.S.A
| | - Kesi Shi
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Zili He
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Jiawei Li
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| | - Fanghua Gong
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Yanlong Liu
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Aiping Hu
- Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Jian Xiao
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China.,Molecular Pharmacology Research Center, School of Pharmaceutical Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Xiangyang Wang
- Department of Orthopaedic Surgery, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
44
|
Pilchova I, Klacanova K, Dibdiakova K, Saksonova S, Stefanikova A, Vidomanova E, Lichardusova L, Hatok J, Racay P. Proteasome Stress Triggers Death of SH-SY5Y and T98G Cells via Different Cellular Mechanisms. Neurochem Res 2017; 42:3170-3185. [PMID: 28725954 DOI: 10.1007/s11064-017-2355-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2017] [Revised: 07/07/2017] [Accepted: 07/12/2017] [Indexed: 01/24/2023]
Abstract
Overload or dysfunction of ubiquitin-proteasome system (UPS) is implicated in mechanisms of neurodegeneration associated with neurodegenerative diseases, e.g. Parkinson and Alzheimer disease, and ischemia-reperfusion injury. The aim of this study was to investigate the possible association between viability of neuroblastoma SH-SY5Y and glioblastoma T98G cells treated with bortezomib, inhibitor of 26S proteasome, and accumulation of ubiquitin-conjugated proteins with respect to direct cytotoxicity of aggregates of ubiquitin-conjugated proteins. Bortezomib-induced death of SH-SY5Y cells was documented after 24 h of treatment while death of T98G cells was delayed up to 48 h. Already after 4 h of treatment of both SH-SY5Y and T98G cells with bortezomib, increased levels of both ubiquitin-conjugated proteins with molecular mass more than 150 kDa and Hsp70 were observed whereas Hsp90 was elevated in T98G cells and decreased in SH-SY5Y cells. With respect to the cell death mechanism, we have documented bortezomib-induced activation of caspase 3 in SH-SY5Y cells that was probably a result of increased expression of pro-apoptotic proteins, PUMA and Noxa. In T98G cells, bortezomib-induced expression of caspase 4, documented after 24 h of treatment, with further activation of caspase 3, observed after 48 h of treatment. The delay in activation of caspase 3 correlated well with the delay of death of T98G cells. Our results do not support the possibility about direct cytotoxicity of aggregates of ubiquitin-conjugated proteins. They are more consistent with a view that proteasome inhibition is associated with both transcription-dependent and -independent changes in expression of pro-apoptotic proteins and consequent cell death initiation associated with caspase 3 activation.
Collapse
Affiliation(s)
- Ivana Pilchova
- Biomedical Center Martin JFM CU and Department of Medical Biochemistry JFM CU, Jessenius Faculty of Medicine in Martin (JFM CU), Comenius University in Bratislava, Mala Hora 4D, 03601, Martin, Slovak Republic
| | - Katarina Klacanova
- Biomedical Center Martin JFM CU and Department of Medical Biochemistry JFM CU, Jessenius Faculty of Medicine in Martin (JFM CU), Comenius University in Bratislava, Mala Hora 4D, 03601, Martin, Slovak Republic
| | - Katarina Dibdiakova
- Biomedical Center Martin JFM CU and Department of Medical Biochemistry JFM CU, Jessenius Faculty of Medicine in Martin (JFM CU), Comenius University in Bratislava, Mala Hora 4D, 03601, Martin, Slovak Republic
| | - Simona Saksonova
- Biomedical Center Martin JFM CU and Department of Medical Biochemistry JFM CU, Jessenius Faculty of Medicine in Martin (JFM CU), Comenius University in Bratislava, Mala Hora 4D, 03601, Martin, Slovak Republic
| | - Andrea Stefanikova
- Biomedical Center Martin JFM CU and Department of Medical Biochemistry JFM CU, Jessenius Faculty of Medicine in Martin (JFM CU), Comenius University in Bratislava, Mala Hora 4D, 03601, Martin, Slovak Republic
| | - Eva Vidomanova
- Biomedical Center Martin JFM CU and Department of Medical Biochemistry JFM CU, Jessenius Faculty of Medicine in Martin (JFM CU), Comenius University in Bratislava, Mala Hora 4D, 03601, Martin, Slovak Republic
| | - Lucia Lichardusova
- Biomedical Center Martin JFM CU and Department of Medical Biochemistry JFM CU, Jessenius Faculty of Medicine in Martin (JFM CU), Comenius University in Bratislava, Mala Hora 4D, 03601, Martin, Slovak Republic
| | - Jozef Hatok
- Biomedical Center Martin JFM CU and Department of Medical Biochemistry JFM CU, Jessenius Faculty of Medicine in Martin (JFM CU), Comenius University in Bratislava, Mala Hora 4D, 03601, Martin, Slovak Republic
| | - Peter Racay
- Biomedical Center Martin JFM CU and Department of Medical Biochemistry JFM CU, Jessenius Faculty of Medicine in Martin (JFM CU), Comenius University in Bratislava, Mala Hora 4D, 03601, Martin, Slovak Republic.
| |
Collapse
|
45
|
Lv J, Tian J, Zheng G, Zhao J. Sirtuin7 is involved in protecting neurons against oxygen-glucose deprivation and reoxygenation-induced injury through regulation of the p53 signaling pathway. J Biochem Mol Toxicol 2017; 31. [PMID: 28675767 DOI: 10.1002/jbt.21955] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Revised: 05/31/2017] [Accepted: 06/16/2017] [Indexed: 01/04/2023]
Abstract
Sirtuin7 (SIRT7) is known to regulate apoptosis and stress responses. So far, very little is known about the role of SIRT7 in cerebral ischemia/reperfusion injury. In this study, we aimed to investigate the potential role of SIRT7 in regulating oxygen-glucose deprivation and reoxygenation (OGD/R)-induced injury in neurons. We found a significant increase of SIRT7 expression in neurons in response to OGD/R treatment. Knockdown of SIRT7 aggravated OGD/R-induced injury. Knockdown of SIRT7 augmented the levels of total and acetylated p53 protein. Moreover, knockdown of SIRT7 markedly increased the transcriptional activity of p53 toward apoptosis and activated the p53-mediated proapoptotic signaling pathway. By contrast, overexpression of SIRT7 showed the opposite effects. Taken together, the results of our study suggest that SIRT7 is involved in protecting neurons against OGD/R-induced injury, possibly through regulation of the p53-mediated proapoptotic signaling pathway, indicating a potential therapeutic target for cerebral ischemia/reperfusion injury.
Collapse
Affiliation(s)
- Jianrui Lv
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, People's Republic of China
| | - Junbin Tian
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, People's Republic of China
| | - Guoxi Zheng
- Department of Otorhinolaryngology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, People's Republic of China
| | - Jing Zhao
- Department of Anesthesiology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, People's Republic of China
| |
Collapse
|
46
|
Sun Y, Li T, Xie C, Xu Y, Zhou K, Rodriguez J, Han W, Wang X, Kroemer G, Modjtahedi N, Blomgren K, Zhu C. Haploinsufficiency in the mitochondrial protein CHCHD4 reduces brain injury in a mouse model of neonatal hypoxia-ischemia. Cell Death Dis 2017; 8:e2781. [PMID: 28492551 PMCID: PMC5520716 DOI: 10.1038/cddis.2017.196] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/13/2017] [Accepted: 04/03/2017] [Indexed: 12/13/2022]
Abstract
Mitochondria contribute to neonatal hypoxic-ischemic brain injury by releasing potentially toxic proteins into the cytosol. CHCHD4 is a mitochondrial intermembrane space protein that plays a major role in the import of intermembrane proteins and physically interacts with apoptosis-inducing factor (AIF). The purpose of this study was to investigate the impact of CHCHD4 haploinsufficiency on mitochondrial function and brain injury after cerebral hypoxia-ischemia (HI) in neonatal mice. CHCHD4+/- and wild-type littermate mouse pups were subjected to unilateral cerebral HI on postnatal day 9. CHCHD4 haploinsufficiency reduced insult-related AIF and superoxide dismutase 2 release from the mitochondria and reduced neuronal cell death. The total brain injury volume was reduced by 21.5% at 3 days and by 31.3% at 4 weeks after HI in CHCHD4+/- mice. However, CHCHD4 haploinsufficiency had no influence on mitochondrial biogenesis, fusion, or fission; neural stem cell proliferation; or neural progenitor cell differentiation. There were no significant changes in the expression or distribution of p53 protein or p53 pathway-related genes under physiological conditions or after HI. These results suggest that CHCHD4 haploinsufficiency afforded persistent neuroprotection related to reduced release of mitochondrial intermembrane space proteins. The CHCHD4-dependent import pathway might thus be a potential therapeutic target for preventing or treating neonatal brain injury.
Collapse
Affiliation(s)
- Yanyan Sun
- Henan Key Laboratory of Child Brain Injury, Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Tao Li
- Henan Key Laboratory of Child Brain Injury, Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Pediatrics, Zhengzhou Children’s Hospital, Zhengzhou, China
| | - Cuicui Xie
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Yiran Xu
- Henan Key Laboratory of Child Brain Injury, Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kai Zhou
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Juan Rodriguez
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Wei Han
- Henan Key Laboratory of Child Brain Injury, Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
| | - Xiaoyang Wang
- Henan Key Laboratory of Child Brain Injury, Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Perinatal Center, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Guido Kroemer
- Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
- INSERM, U1138, Paris, France
- Equipe 11 labellisée par la Ligue Nationale contre le Cancer, Centre de Recherche des Cordeliers, Paris, France
- Université Paris Descartes/Paris V, Sorbonne Paris Cité, Paris, France
- Metabolomics and Cell Biology Platforms, Gustave Roussy Cancer Campus, Villejuif, France
| | - Nazanine Modjtahedi
- Laboratory of Molecular Radiotherapy, INSERM U1030, Gustave Roussy, Villejuif F-94805, France
- Gustave Roussy, Villejuif F-94805, France
- Department of Medicine, Université Paris-Saclay, Kremlin-Bicêtre, France
| | - Klas Blomgren
- Department of Women’s and Children’s Health, Karolinska Institutet, Stockholm, Sweden
- Department of Pediatric Oncology, Karolinska University Hospital, Stockholm, Sweden
| | - Changlian Zhu
- Henan Key Laboratory of Child Brain Injury, Department of Pediatrics, The Third Affiliated Hospital of Zhengzhou University, Zhengzhou, China
- Center for Brain Repair and Rehabilitation, Institute of Neuroscience and Physiology, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
47
|
Current understanding of methamphetamine-associated dopaminergic neurodegeneration and psychotoxic behaviors. Arch Pharm Res 2017; 40:403-428. [DOI: 10.1007/s12272-017-0897-y] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2016] [Accepted: 12/02/2016] [Indexed: 12/21/2022]
|
48
|
Bakthavachalam P, Shanmugam PST. Mitochondrial dysfunction - Silent killer in cerebral ischemia. J Neurol Sci 2017; 375:417-423. [PMID: 28320180 DOI: 10.1016/j.jns.2017.02.043] [Citation(s) in RCA: 81] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2016] [Revised: 01/27/2017] [Accepted: 02/20/2017] [Indexed: 10/20/2022]
Abstract
Mitochondrial dysfunction aggravates ischemic neuronal injury through activation of various pathophysiological and molecular mechanisms. Ischemic neuronal injury is particularly intensified during reperfusion due to impairment of mitochondrial function. Mitochondrial mutilation instigates alterations in calcium homeostasis in neurons, which plays a pivotal role in the maintenance of normal neuronal function. Increase in intracellular calcium level in mitochondria triggers the opening of mitochondrial transition pore and over production of reactive oxygen species (ROS). Several investigations have concluded that ROS not only contribute to lipids and proteins damage, but also transduce apoptotic signals leading to neuronal death. In addition to the above mentioned reasons, endoplasmic reticulum (ER) stress due to excitotoxicity also leads to neuronal death. Recently, some newer proteins have been claimed to induce "mitophagy" by triggering the receptors on autophagic membranes leading to neurodegeneration. This review summarizes the mechanisms underlying neuronal death involving mitochondrial dysfunction and mitophagy.
Collapse
Affiliation(s)
- Pramila Bakthavachalam
- Sri Ramachandra University, No. 1, Ramachandra Nagar, Porur, Chennai, Tamil Nadu, India.
| | | |
Collapse
|
49
|
SUMO-Modified FADD Recruits Cytosolic Drp1 and Caspase-10 to Mitochondria for Regulated Necrosis. Mol Cell Biol 2017; 37:MCB.00254-16. [PMID: 27799292 DOI: 10.1128/mcb.00254-16] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2016] [Accepted: 10/20/2016] [Indexed: 01/02/2023] Open
Abstract
Fas-associated protein with death domain (FADD) plays a key role in extrinsic apoptosis. Here, we show that FADD is SUMOylated as an essential step during intrinsic necrosis. FADD was modified at multiple lysine residues (K120/125/149) by small ubiquitin-related modifier 2 (SUMO2) during necrosis caused by calcium ionophore A23187 and by ischemic damage. SUMOylated FADD bound to dynamin-related protein 1 (Drp1) in cells both in vitro and in ischemic tissue damage cores, thus promoting Drp1 recruitment by mitochondrial fission factor (Mff) to accomplish mitochondrial fragmentation. Mitochondrial-fragmentation-associated necrosis was blocked by FADD or Drp1 deficiency and SUMO-defective FADD expression. Interestingly, caspase-10, but not caspase-8, formed a ternary protein complex with SUMO-FADD/Drp1 on the mitochondria upon exposure to A23187 and potentiated Drp1 oligomerization for necrosis. Moreover, the caspase-10 L285F and A414V mutants, found in autoimmune lymphoproliferative syndrome and non-Hodgkin lymphoma, respectively, regulated this necrosis. Our study reveals an essential role of SUMOylated FADD in Drp1- and caspase-10-dependent necrosis, providing insights into the mechanism of regulated necrosis by calcium overload and ischemic injury.
Collapse
|
50
|
Lee JC, Park JH, Kim IH, Cho GS, Ahn JH, Tae HJ, Choi SY, Cho JH, Kim DW, Kwon YG, Kang IJ, Won MH, Kim YM. Neuroprotection of ischemic preconditioning is mediated by thioredoxin 2 in the hippocampal CA1 region following a subsequent transient cerebral ischemia. Brain Pathol 2016; 27:276-291. [PMID: 27117068 DOI: 10.1111/bpa.12389] [Citation(s) in RCA: 48] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Accepted: 04/20/2016] [Indexed: 12/26/2022] Open
Abstract
Preconditioning by brief ischemic episode induces tolerance to a subsequent lethal ischemic insult, and it has been suggested that reactive oxygen species are involved in this phenomenon. Thioredoxin 2 (Trx2), a small protein with redox-regulating function, shows cytoprotective roles against oxidative stress. Here, we had focused on the role of Trx2 in ischemic preconditioning (IPC)-mediated neuroprotection against oxidative stress followed by a subsequent lethal transient cerebral ischemia. Animals used in this study were randomly assigned to six groups; sham-operated group, ischemia-operated group, IPC plus (+) sham-operated group, IPC + ischemia-operated group, IPC + auranofin (a TrxR2 inhibitor) + sham-operated group and IPC + auranofin + ischemia-operated group. IPC was subjected to a 2 minutes of sublethal transient ischemia 1 day prior to a 5 minutes of lethal transient ischemia. A significant loss of neurons was found in the stratum pyramidale (SP) of the hippocampal CA1 region (CA1) in the ischemia-operated-group 5 days after ischemia-reperfusion; in the IPC + ischemia-operated-group, pyramidal neurons in the SP were well protected. In the IPC + ischemia-operated-group, Trx2 and TrxR2 immunoreactivities in the SP and its protein level in the CA1 were not significantly changed compared with those in the sham-operated-group after ischemia-reperfusion. In addition, superoxide dismutase 2 (SOD2) expression, superoxide anion radical ( O2-) production, denatured cytochrome c expression and TUNEL-positive cells in the IPC + ischemia-operated-group were similar to those in the sham-operated-group. Conversely, the treatment of auranofin to the IPC + ischemia-operated-group significantly increased cell damage/death and abolished the IPC-induced effect on Trx2 and TrxR2 expressions. Furthermore, the inhibition of Trx2R nearly cancelled the beneficial effects of IPC on SOD2 expression, O2- production, denatured cytochrome c expression and TUNEL-positive cells. In brief, this study shows that IPC conferred neuroprotection against ischemic injury by maintaining Trx2 and suggests that the maintenance or enhancement of Trx2 expression by IPC may be a legitimate strategy for therapeutic intervention of cerebral ischemia.
Collapse
Affiliation(s)
- Jae-Chul Lee
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Joon Ha Park
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - In Hye Kim
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Geum-Sil Cho
- Pharmacology & Toxicology Department, Shinpoong Pharmaceutical Co., Ltd., Ansan, South Korea
| | - Ji Hyeon Ahn
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, South Korea
| | - Hyun-Jin Tae
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, South Korea
| | - Soo Young Choi
- Department of Biomedical Science and Research Institute of Bioscience and Biotechnology, Hallym University, Chuncheon, South Korea
| | - Jun Hwi Cho
- Department of Emergency Medicine, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Dae Won Kim
- Department of Biochemistry and Molecular Biology, Research Institute of Oral Sciences, College of Dentistry, Kangnung-Wonju National University, Gangneung, South Korea
| | - Young-Guen Kwon
- Department of Biochemistry, College of Life Science and Biotechnology, Yonsei University, Seoul, South Korea
| | - Il Jun Kang
- Department of Food Science and Nutrition, Hallym University, Chuncheon, South Korea
| | - Moo-Ho Won
- Department of Neurobiology, School of Medicine, Kangwon National University, Chuncheon, South Korea
| | - Young-Myeong Kim
- Department of Molecular and Cellular Biochemistry, School of Medicine, Kangwon National University, Chuncheon, South Korea
| |
Collapse
|