1
|
Xia S, Li J, Yuan H, Yan W. PIN1‑silencing mitigates keratinocyte proliferation and the inflammatory response in psoriasis by activating mitochondrial autophagy. Exp Ther Med 2024; 28:402. [PMID: 39234585 PMCID: PMC11372252 DOI: 10.3892/etm.2024.12691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Accepted: 04/24/2024] [Indexed: 09/06/2024] Open
Abstract
Peptidyl-prolyl cis/trans isomerase, NIMA-interacting 1 (PIN1) has been suggested to be a critical regulator in skin-related diseases. However, the role and molecular mechanism of PIN1 in psoriasis remain unclear. HaCaT cells were stimulated with five cytokines (M5) to induce psoriatic inflammation-like conditions. Reverse transcription-quantitative PCR and western blotting were performed to examine PIN1 expression in M5-induced HaCaT cells. A Cell Counting Kit-8 assay and 5-ethynyl-2'-deoxyuridine staining were employed to examine cell proliferation. Inflammatory factors were evaluated using ELISA kits and western blot analysis. Mitochondrial autophagy was examined by immunofluorescence staining, western blotting and a JC-1 assay. Western blot analysis was adopted to assess the levels of psoriasis marker proteins. PIN1 expression was markedly elevated in M5-induced HaCaT cells. Silencing of PIN1 inhibited M5-induced hyperproliferation and the inflammatory response, while it promoted mitochondrial autophagy in HaCaT cells. The addition of the mitochondrial autophagy inhibitor mitochondrial division inhibitor-1 reversed the effects of PIN1 interference on proliferation, the inflammatory response and mitochondrial autophagy in M5-induced HaCaT cells. The present study revealed that PIN1 inhibition protected HaCaT cells against M5-induced hyperproliferation and inflammatory injury through the activation of mitochondrial autophagy.
Collapse
Affiliation(s)
- Shuang Xia
- Department of Dermatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210001, P.R. China
| | - Jin Li
- Department of Dermatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210001, P.R. China
| | - Hongshan Yuan
- Department of Dermatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210001, P.R. China
| | - Wenliang Yan
- Department of Dermatology, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, Jiangsu 210001, P.R. China
| |
Collapse
|
2
|
Sridharan PS, Koh Y, Miller E, Hu D, Chakraborty S, Tripathi SJ, Kee TR, Chaubey K, Vázquez-Rosa E, Barker S, Liu H, León-Alvarado RA, Franke K, Cintrón-Pérez CJ, Dhar M, Shin MK, Flanagan ME, Castellani RJ, Gefen T, Bykova M, Dou L, Cheng F, Wilson BM, Fujioka H, Kang DE, Woo JAA, Paul BD, Qi X, Pieper AA. Acutely blocking excessive mitochondrial fission prevents chronic neurodegeneration after traumatic brain injury. Cell Rep Med 2024; 5:101715. [PMID: 39241772 PMCID: PMC11525032 DOI: 10.1016/j.xcrm.2024.101715] [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: 01/20/2024] [Revised: 06/03/2024] [Accepted: 08/13/2024] [Indexed: 09/09/2024]
Abstract
Progression of acute traumatic brain injury (TBI) into chronic neurodegeneration is a major health problem with no protective treatments. Here, we report that acutely elevated mitochondrial fission after TBI in mice triggers chronic neurodegeneration persisting 17 months later, equivalent to many human decades. We show that increased mitochondrial fission after mouse TBI is related to increased brain levels of mitochondrial fission 1 protein (Fis1) and that brain Fis1 is also elevated in human TBI. Pharmacologically preventing Fis1 from binding its mitochondrial partner, dynamin-related protein 1 (Drp1), for 2 weeks after TBI normalizes the balance of mitochondrial fission/fusion and prevents chronically impaired mitochondrial bioenergetics, oxidative damage, microglial activation and lipid droplet formation, blood-brain barrier deterioration, neurodegeneration, and cognitive impairment. Delaying treatment until 8 months after TBI offers no protection. Thus, time-sensitive inhibition of acutely elevated mitochondrial fission may represent a strategy to protect human TBI patients from chronic neurodegeneration.
Collapse
Affiliation(s)
- Preethy S Sridharan
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Yeojung Koh
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Emiko Miller
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Di Hu
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Suwarna Chakraborty
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sunil Jamuna Tripathi
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Teresa R Kee
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Molecular Medicine, USF Health College of Medicine, Tampa, FL, USA
| | - Kalyani Chaubey
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Edwin Vázquez-Rosa
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Sarah Barker
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Hui Liu
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Rose A León-Alvarado
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Earlham College Neuroscience Program, Richmond, IN, USA
| | - Kathryn Franke
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Coral J Cintrón-Pérez
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Matasha Dhar
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - Min-Kyoo Shin
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul 08226, Republic of Korea
| | - Margaret E Flanagan
- University of Texas Health Science Center at San Antonio, San Antonio, TX, USA; Glenn Bigg's Institute for Alzheimer's & Neurodegenerative Diseases, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA; Department of Pathology, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Rudolph J Castellani
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Tamar Gefen
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University Feinberg School of Medicine, Chicago, IL, USA; Department of Psychiatry and Behavioral Sciences, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Marina Bykova
- Department of Regulatory Biology, Cleveland State University, Cleveland, OH, USA; Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Lijun Dou
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Feixiong Cheng
- Genomic Medicine Institute, Lerner Research Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Brigid M Wilson
- Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Louis Stokes VA Medical Center, Cleveland, OH, USA
| | - Hisashi Fujioka
- Cryo-Electron Microscopy Core Facility, Case Western Reserve University School of Medicine, Cleveland, OH, USA
| | - David E Kang
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Molecular Medicine, USF Health College of Medicine, Tampa, FL, USA; Louis Stokes VA Medical Center, Cleveland, OH, USA
| | - Jung-A A Woo
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Molecular Medicine, USF Health College of Medicine, Tampa, FL, USA
| | - Bindu D Paul
- Department of Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Department of Psychiatry and Behavioral Sciences, Johns Hopkins University School of Medicine, Baltimore, MD, USA; The Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD, USA; Lieber Institute for Brain Development, Baltimore, MD, USA
| | - Xin Qi
- Department of Physiology and Biophysics, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| | - Andrew A Pieper
- Department of Psychiatry, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Brain Health Medicines Center, Harrington Discovery Institute, University Hospitals Cleveland Medical Center, Cleveland, OH, USA; Geriatric Psychiatry, GRECC, Louis Stokes VA Medical Center, Cleveland, OH, USA; Institute for Transformative Molecular Medicine, School of Medicine, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Neurosciences, Case Western Reserve University School of Medicine, Cleveland, OH, USA; Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, OH, USA.
| |
Collapse
|
3
|
Hao Y, Zhao L, Zhao JY, Han X, Zhou X. Unveiling the potential of mitochondrial dynamics as a therapeutic strategy for acute kidney injury. Front Cell Dev Biol 2023; 11:1244313. [PMID: 37635869 PMCID: PMC10456901 DOI: 10.3389/fcell.2023.1244313] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2023] [Accepted: 07/31/2023] [Indexed: 08/29/2023] Open
Abstract
Acute Kidney Injury (AKI), a critical clinical syndrome, has been strongly linked to mitochondrial malfunction. Mitochondria, vital cellular organelles, play a key role in regulating cellular energy metabolism and ensuring cell survival. Impaired mitochondrial function in AKI leads to decreased energy generation, elevated oxidative stress, and the initiation of inflammatory cascades, resulting in renal tissue damage and functional impairment. Therefore, mitochondria have gained significant research attention as a potential therapeutic target for AKI. Mitochondrial dynamics, which encompass the adaptive shifts of mitochondria within cellular environments, exert significant influence on mitochondrial function. Modulating these dynamics, such as promoting mitochondrial fusion and inhibiting mitochondrial division, offers opportunities to mitigate renal injury in AKI. Consequently, elucidating the mechanisms underlying mitochondrial dynamics has gained considerable importance, providing valuable insights into mitochondrial regulation and facilitating the development of innovative therapeutic approaches for AKI. This comprehensive review aims to highlight the latest advancements in mitochondrial dynamics research, provide an exhaustive analysis of existing studies investigating the relationship between mitochondrial dynamics and acute injury, and shed light on their implications for AKI. The ultimate goal is to advance the development of more effective therapeutic interventions for managing AKI.
Collapse
Affiliation(s)
- Yajie Hao
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Limei Zhao
- The Fifth Clinical Medical College of Shanxi Medical University, Taiyuan, China
| | - Jing Yu Zhao
- The Third Clinical College, Shanxi University of Chinese Medicine, Jinzhong, Shanxi, China
| | - Xiutao Han
- The Third Clinical College, Shanxi University of Chinese Medicine, Jinzhong, Shanxi, China
| | - Xiaoshuang Zhou
- Department of Nephrology, Shanxi Provincial People’s Hospital, The Fifth Clinical Medical College of Shanxi Medical University, Shanxi Kidney Disease Institute, Taiyuan, China
| |
Collapse
|
4
|
Muñoz JP, Basei FL, Rojas ML, Galvis D, Zorzano A. Mechanisms of Modulation of Mitochondrial Architecture. Biomolecules 2023; 13:1225. [PMID: 37627290 PMCID: PMC10452872 DOI: 10.3390/biom13081225] [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: 06/15/2023] [Revised: 07/27/2023] [Accepted: 08/01/2023] [Indexed: 08/27/2023] Open
Abstract
Mitochondrial network architecture plays a critical role in cellular physiology. Indeed, alterations in the shape of mitochondria upon exposure to cellular stress can cause the dysfunction of these organelles. In this scenario, mitochondrial dynamics proteins and the phospholipid composition of the mitochondrial membrane are key for fine-tuning the modulation of mitochondrial architecture. In addition, several factors including post-translational modifications such as the phosphorylation, acetylation, SUMOylation, and o-GlcNAcylation of mitochondrial dynamics proteins contribute to shaping the plasticity of this architecture. In this regard, several studies have evidenced that, upon metabolic stress, mitochondrial dynamics proteins are post-translationally modified, leading to the alteration of mitochondrial architecture. Interestingly, several proteins that sustain the mitochondrial lipid composition also modulate mitochondrial morphology and organelle communication. In this context, pharmacological studies have revealed that the modulation of mitochondrial shape and function emerges as a potential therapeutic strategy for metabolic diseases. Here, we review the factors that modulate mitochondrial architecture.
Collapse
Affiliation(s)
- Juan Pablo Muñoz
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Institut d’Investigació Biomèdica Sant Pau (IIB SANT PAU), 08041 Barcelona, Spain
| | - Fernanda Luisa Basei
- Faculdade de Ciências Farmacêuticas, Universidade Estadual de Campinas, 13083-871 Campinas, SP, Brazil
| | - María Laura Rojas
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba X5000HUA, Argentina
| | - David Galvis
- Programa de Química Farmacéutica, Universidad CES, Medellín 050031, Colombia
| | - Antonio Zorzano
- CIBER de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), 28029 Madrid, Spain
- Institute for Research in Biomedicine (IRB Barcelona), 08028 Barcelona, Spain
- Departament de Bioquímica i Biomedicina Molecular, Facultat de Biologia, Universitat de Barcelona, 08028 Barcelona, Spain
| |
Collapse
|
5
|
Hu CH, Wu SN, So EC. Tyrosine kinase inhibitors, ionic currents, and cardiac arrhythmia. Front Oncol 2023; 13:1218821. [PMID: 37554165 PMCID: PMC10405512 DOI: 10.3389/fonc.2023.1218821] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 06/19/2023] [Indexed: 08/10/2023] Open
Affiliation(s)
- Chi-Hao Hu
- Department of Physiology, National Cheng Kung University Medical College, Tainan, Taiwan
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan, Taiwan
- School of Medicine, National Sun Yat-sen University College of Medicine, Kaohsiung, Taiwan
- Department of Education and Research, An Nan Hospital, China Medical University, Tainan, Taiwan
| | - Edmund Cheung So
- Department of Anesthesia, An Nan Hospital, China Medical University, Tainan, Taiwan
| |
Collapse
|
6
|
Liu X, Guo C, Zhang Q. Novel insights into the involvement of mitochondrial fission/fusion in heart failure: From molecular mechanisms to targeted therapies. Cell Stress Chaperones 2023; 28:133-144. [PMID: 36652120 PMCID: PMC10050249 DOI: 10.1007/s12192-023-01321-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/04/2023] [Accepted: 01/08/2023] [Indexed: 01/19/2023] Open
Abstract
Mitochondria are dynamic organelles that alter their morphology through fission (fragmentation) and fusion (elongation). These morphological changes correlate highly with mitochondrial functional adaptations to stressors, such as hypoxia, pressure overload, and inflammation, and are important in the setting of heart failure. Pathological mitochondrial remodeling, characterized by increased fission and reduced fusion, is associated with impaired mitochondrial respiration, increased mitochondrial oxidative stress, abnormal cytoplasmic calcium handling, and increased cardiomyocyte apoptosis. Considering the impact of the mitochondrial morphology on mitochondrial behavior and cardiomyocyte performance, altered mitochondrial dynamics could be expected to induce or exacerbate the pathogenesis and progression of heart failure. However, whether alterations in mitochondrial fission and fusion accelerate or retard the progression of heart failure has been the subject of intense debate. In this review, we first describe the physiological processes and regulatory mechanisms of mitochondrial fission and fusion. Then, we extensively discuss the pathological contributions of mitochondrial fission and fusion to heart failure. Lastly, we examine potential therapeutic approaches targeting mitochondrial fission/fusion to treat patients with heart failure.
Collapse
Affiliation(s)
- Xinxin Liu
- Department of First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China
| | - Chenchen Guo
- Neck, Shoulder, Waist and Leg Pain Hospital Affiliated to Shandong First Medical University, Jinan, China
| | - Qiming Zhang
- Department of First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, Shandong Province, China.
| |
Collapse
|
7
|
Maneechote C, Kerdphoo S, Jaiwongkam T, Chattipakorn SC, Chattipakorn N. Chronic Pharmacological Modulation of Mitochondrial Dynamics Alleviates Prediabetes-Induced Myocardial Ischemia-Reperfusion Injury by Preventing Mitochondrial Dysfunction and Programmed Apoptosis. Cardiovasc Drugs Ther 2023; 37:89-105. [PMID: 34515894 DOI: 10.1007/s10557-021-07250-7] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/25/2021] [Indexed: 01/14/2023]
Abstract
PURPOSE There is an increasing body of evidence to show that impairment in mitochondrial dynamics including excessive fission and insufficient fusion has been observed in the pre-diabetic condition. In pre-diabetic rats with cardiac ischemia-reperfusion (I/R) injury, acute treatment with a mitochondria fission inhibitor (Mdivi-1) and a fusion promoter (M1) showed cardioprotection. However, the potential preventive effects of chronic Mdivi-1 and M1 treatment in a pre-diabetic model of cardiac I/R have never been elucidated. METHODS Male Wistar rats (n = 40) were fed with a high-fat diet (HFD) for 12 weeks to induce prediabetes. Then, all pre-diabetic rats received the following treatments daily via intraperitoneal injection for 2 weeks: (1) HFDV (Vehicle, 0.1% DMSO); (2) HFMdivi1 (Mdivi-1 1.2 mg/kg); (3) HFM1 (M1 2 mg/kg); and (4) HFCom (Mdivi-1 + M1). At the end of treatment protocols, all rats underwent 30 min of coronary artery ligation followed by reperfusion for 120 min. RESULTS Chronic Mdivi-1, M1, and the combined treatment showed markedly improved cardiac mitochondrial function and dynamic control, leading to a decrease in cardiac arrhythmias, myocardial cell death, and infarct size (49%, 42%, and 51% reduction for HFMdivi1, HFM1, and HFCom, respectively vs HFDV). All of these treatments improved cardiac function following cardiac I/R injury in pre-diabetic rats. CONCLUSION Chronic inhibition of mitochondrial fission and promotion of fusion exerted cardioprevention in prediabetes with cardiac I/R injury through the relief of cardiac mitochondrial dysfunction and dynamic alterations, and reduction in myocardial infarction, thus improving cardiac function.
Collapse
Affiliation(s)
- Chayodom Maneechote
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Sasiwan Kerdphoo
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Thidarat Jaiwongkam
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai, 50200, Thailand.
- Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai, 50200, Thailand.
| |
Collapse
|
8
|
Rosdah AA, Abbott BM, Langendorf CG, Deng Y, Truong JQ, Waddell HMM, Ling NXY, Smiles WJ, Delbridge LMD, Liu GS, Oakhill JS, Lim SY, Holien JK. A novel small molecule inhibitor of human Drp1. Sci Rep 2022; 12:21531. [PMID: 36513726 PMCID: PMC9747717 DOI: 10.1038/s41598-022-25464-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2022] [Accepted: 11/30/2022] [Indexed: 12/15/2022] Open
Abstract
Mitochondrial dynamin-related protein 1 (Drp1) is a large GTPase regulator of mitochondrial dynamics and is known to play an important role in numerous pathophysiological processes. Despite being the most widely used Drp1 inhibitor, the specificity of Mdivi-1 towards human Drp1 has not been definitively proven and there have been numerous issues reported with its use including off-target effects. In our hands Mdivi-1 showed varying binding affinities toward human Drp1, potentially impacted by compound aggregation. Herein, we sought to identify a novel small molecule inhibitor of Drp1. From an initial virtual screening, we identified DRP1i27 as a compound which directly bound to the human isoform 3 of Drp1 via surface plasmon resonance and microscale thermophoresis. Importantly, DRP1i27 was found to have a dose-dependent increase in the cellular networks of fused mitochondria but had no effect in Drp1 knock-out cells. Further analogues of this compound were identified and screened, though none displayed greater affinity to human Drp1 isoform 3 than DRP1i27. To date, this is the first small molecule inhibitor shown to directly bind to human Drp1.
Collapse
Affiliation(s)
- Ayeshah A. Rosdah
- grid.1073.50000 0004 0626 201XSt Vincent’s Institute of Medical Research, Fitzroy, VIC Australia ,grid.108126.c0000 0001 0557 0975Faculty of Medicine, Universitas Sriwijaya, Palembang, Indonesia ,grid.1008.90000 0001 2179 088XDepartment of Surgery and Medicine, University of Melbourne, Melbourne, VIC Australia
| | - Belinda M. Abbott
- grid.1018.80000 0001 2342 0938Department of Biochemistry and Chemistry, La Trobe Institute for Molecular Science, La Trobe University, Melbourne, VIC Australia
| | | | - Yali Deng
- grid.1073.50000 0004 0626 201XSt Vincent’s Institute of Medical Research, Fitzroy, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Surgery and Medicine, University of Melbourne, Melbourne, VIC Australia
| | - Jia Q. Truong
- grid.1017.70000 0001 2163 3550School of Science, RMIT University, GPO Box 2476, Melbourne, VIC 3001 Australia
| | - Helen M. M. Waddell
- grid.1008.90000 0001 2179 088XDepartment of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3010 Australia
| | - Naomi X. Y. Ling
- grid.1073.50000 0004 0626 201XSt Vincent’s Institute of Medical Research, Fitzroy, VIC Australia
| | - William J. Smiles
- grid.1073.50000 0004 0626 201XSt Vincent’s Institute of Medical Research, Fitzroy, VIC Australia
| | - Lea M. D. Delbridge
- grid.1008.90000 0001 2179 088XDepartment of Anatomy and Physiology, The University of Melbourne, Parkville, VIC 3010 Australia
| | - Guei-Sheung Liu
- grid.1008.90000 0001 2179 088XDepartment of Surgery and Medicine, University of Melbourne, Melbourne, VIC Australia ,grid.410670.40000 0004 0625 8539Centre for Eye Research Australia, Royal Victorian Eye and Ear Hospital, East Melbourne, VIC Australia ,grid.1009.80000 0004 1936 826XMenzies Institute for Medical Research, University of Tasmania, Hobart, TAS Australia
| | - Jonathan S. Oakhill
- grid.1073.50000 0004 0626 201XSt Vincent’s Institute of Medical Research, Fitzroy, VIC Australia ,grid.411958.00000 0001 2194 1270Australian Catholic University, Fitzroy, VIC Australia
| | - Shiang Y. Lim
- grid.1073.50000 0004 0626 201XSt Vincent’s Institute of Medical Research, Fitzroy, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Surgery and Medicine, University of Melbourne, Melbourne, VIC Australia ,grid.1002.30000 0004 1936 7857Drug Discovery Biology, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Melbourne, VIC Australia ,grid.419385.20000 0004 0620 9905National Heart Centre, National Heart Research Institute Singapore, Singapore, Singapore
| | - Jessica K. Holien
- grid.1073.50000 0004 0626 201XSt Vincent’s Institute of Medical Research, Fitzroy, VIC Australia ,grid.1008.90000 0001 2179 088XDepartment of Surgery and Medicine, University of Melbourne, Melbourne, VIC Australia ,grid.1017.70000 0001 2163 3550School of Science, RMIT University, GPO Box 2476, Melbourne, VIC 3001 Australia
| |
Collapse
|
9
|
Ahmed A, Trezza A, Gentile M, Paccagnini E, Lupetti P, Spiga O, Bova S, Fusi F. The drp-1-mediated mitochondrial fission inhibitor mdivi-1 impacts the function of ion channels and pathways underpinning vascular smooth muscle tone. Biochem Pharmacol 2022; 203:115205. [DOI: 10.1016/j.bcp.2022.115205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 07/07/2022] [Accepted: 08/02/2022] [Indexed: 11/24/2022]
|
10
|
Bordt EA, Zhang N, Waddell J, Polster BM. The Non-Specific Drp1 Inhibitor Mdivi-1 Has Modest Biochemical Antioxidant Activity. Antioxidants (Basel) 2022; 11:antiox11030450. [PMID: 35326100 PMCID: PMC8944504 DOI: 10.3390/antiox11030450] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2022] [Revised: 02/16/2022] [Accepted: 02/22/2022] [Indexed: 11/16/2022] Open
Abstract
Mitochondrial division inhibitor-1 (mdivi-1), a non-specific inhibitor of Drp1-dependent mitochondrial fission, is neuroprotective in numerous preclinical disease models. These include rodent models of Alzheimer’s disease and ischemic or traumatic brain injury. Among its Drp1-independent actions, the compound was found to suppress mitochondrial Complex I-dependent respiration but with less resultant mitochondrial reactive oxygen species (ROS) emission compared with the classical Complex I inhibitor rotenone. We employed two different methods of quantifying Trolox-equivalent antioxidant capacity (TEAC) to test the prediction that mdivi-1 can directly scavenge free radicals. Mdivi-1 exhibited moderate antioxidant activity in the 2,2′-azinobis (3-ethylbenzothiazoline 6-sulfonate) (ABTS) assay. Half-maximal ABTS radical depletion was observed at ~25 μM mdivi-1, equivalent to that achieved by ~12.5 μM Trolox. Mdivi-1 also showed antioxidant activity in the α, α-diphenyl-β-picrylhydrazyl (DPPH) assay. However, mdivi-1 exhibited a reduced capacity to deplete the DPPH radical, which has a more sterically hindered radical site compared with ABTS, with 25 μM mdivi-1 displaying only 0.8 μM Trolox equivalency. Both assays indicate that mdivi-1 possesses biochemical antioxidant activity but with modest potency relative to the vitamin E analog Trolox. Future studies are needed to evaluate whether the ability of mdivi-1 to directly scavenge free radicals contributes to its mechanisms of neuroprotection.
Collapse
Affiliation(s)
- Evan A. Bordt
- Center for Shock, Trauma and Anesthesiology Research, Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
- Lurie Center for Autism, Department of Pediatrics, Massachusetts General Hospital for Children, Harvard Medical School, Boston, MA 02129, USA
- Correspondence: (E.A.B.); (B.M.P.); Tel.: +01-617-643-4351 (E.A.B.); +01-410-706-3418 (B.M.P.)
| | - Naibo Zhang
- Center for Shock, Trauma and Anesthesiology Research, Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jaylyn Waddell
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
| | - Brian M. Polster
- Center for Shock, Trauma and Anesthesiology Research, Department of Anesthesiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA;
- Program in Neuroscience, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- Correspondence: (E.A.B.); (B.M.P.); Tel.: +01-617-643-4351 (E.A.B.); +01-410-706-3418 (B.M.P.)
| |
Collapse
|
11
|
Batista AF, Rody T, Forny-Germano L, Cerdeiro S, Bellio M, Ferreira ST, Munoz DP, De Felice FG. Interleukin-1β mediates alterations in mitochondrial fusion/fission proteins and memory impairment induced by amyloid-β oligomers. J Neuroinflammation 2021; 18:54. [PMID: 33612100 PMCID: PMC7897381 DOI: 10.1186/s12974-021-02099-x] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Accepted: 02/02/2021] [Indexed: 02/06/2023] Open
Abstract
Background The lack of effective treatments for Alzheimer’s disease (AD) reflects an incomplete understanding of disease mechanisms. Alterations in proteins involved in mitochondrial dynamics, an essential process for mitochondrial integrity and function, have been reported in AD brains. Impaired mitochondrial dynamics causes mitochondrial dysfunction and has been associated with cognitive impairment in AD. Here, we investigated a possible link between pro-inflammatory interleukin-1 (IL-1), mitochondrial dysfunction, and cognitive impairment in AD models. Methods We exposed primary hippocampal cell cultures to amyloid-β oligomers (AβOs) and carried out AβO infusions into the lateral cerebral ventricle of cynomolgus macaques to assess the impact of AβOs on proteins that regulate mitochondrial dynamics. Where indicated, primary cultures were pre-treated with mitochondrial division inhibitor 1 (mdivi-1), or with anakinra, a recombinant interleukin-1 receptor (IL-1R) antagonist used in the treatment of rheumatoid arthritis. Cognitive impairment was investigated in C57BL/6 mice that received an intracerebroventricular (i.c.v.) infusion of AβOs in the presence or absence of mdivi-1. To assess the role of interleukin-1 beta (IL-1β) in AβO-induced alterations in mitochondrial proteins and memory impairment, interleukin receptor-1 knockout (Il1r1−/−) mice received an i.c.v. infusion of AβOs. Results We report that anakinra prevented AβO-induced alteration in mitochondrial dynamics proteins in primary hippocampal cultures. Altered levels of proteins involved in mitochondrial fusion and fission were observed in the brains of cynomolgus macaques that received i.c.v. infusions of AβOs. The mitochondrial fission inhibitor, mdivi-1, alleviated synapse loss and cognitive impairment induced by AβOs in mice. In addition, AβOs failed to cause alterations in expression of mitochondrial dynamics proteins or memory impairment in Il1r1−/− mice. Conclusion These findings indicate that IL-1β mediates the impact of AβOs on proteins involved in mitochondrial dynamics and that strategies aimed to prevent pathological alterations in those proteins may counteract synapse loss and cognitive impairment in AD. Supplementary Information The online version contains supplementary material available at 10.1186/s12974-021-02099-x.
Collapse
Affiliation(s)
- Andre F Batista
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, CCS, room H2-019, Rio de Janeiro, RJ, 21941-590, Brazil
| | - Tayná Rody
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, CCS, room H2-019, Rio de Janeiro, RJ, 21941-590, Brazil
| | - Leticia Forny-Germano
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, CCS, room H2-019, Rio de Janeiro, RJ, 21941-590, Brazil
| | - Suzana Cerdeiro
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, CCS, room H2-019, Rio de Janeiro, RJ, 21941-590, Brazil
| | - Maria Bellio
- Department of Immunology, Institute of Microbiology Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Sergio T Ferreira
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, CCS, room H2-019, Rio de Janeiro, RJ, 21941-590, Brazil.,Institute of Biophysics Carlos Chagas Filho, Federal University of Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Douglas P Munoz
- Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, K7L3N6, Canada
| | - Fernanda G De Felice
- Institute of Medical Biochemistry Leopoldo de Meis, Federal University of Rio de Janeiro, CCS, room H2-019, Rio de Janeiro, RJ, 21941-590, Brazil. .,Centre for Neuroscience Studies, Queen's University, Kingston, Ontario, K7L3N6, Canada. .,Department of Psychiatry, Queen's University, Kingston, Ontario, K7L3N6, Canada.
| |
Collapse
|
12
|
Maneechote C, Palee S, Kerdphoo S, Jaiwongkam T, Chattipakorn SC, Chattipakorn N. Pharmacological inhibition of mitochondrial fission attenuates cardiac ischemia-reperfusion injury in pre-diabetic rats. Biochem Pharmacol 2020; 182:114295. [PMID: 33080185 DOI: 10.1016/j.bcp.2020.114295] [Citation(s) in RCA: 34] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2020] [Revised: 10/15/2020] [Accepted: 10/16/2020] [Indexed: 12/23/2022]
Abstract
An increase in the number of fragmented mitochondria contributes to the pathogenesis of ischemia-reperfusion (I/R) injury. Also, mitochondrial fission has shown an increase in obese condition. However, the cardioprotective roles of a mitochondrial fission inhibitor in obesity with cardiac I/R injury are unclear. We hypothesized that a fission inhibitor (Mdivi-1) reduces cardiac dysfunction during I/R injury in pre-diabetic rats. Male Wistar rats (n = 40) were received a high-fat diet for 12 weeks to induce prediabetes. Then, rats underwent a 30-min coronary artery ligation was performed followed by reperfusion for 120 min. These I/R rats were given either: (1) vehicle or Mdivi-1 treatment at 3 time points relative to onset of ischemia: (2) pre-ischemia; (3) during ischemia; and (4) at onset of reperfusion. Cardiac function, myocardial infarct size, mitochondrial function and dynamic balance were determined. Interestingly, Mdivi-1 given at any time points effectively attenuated mitochondrial reactive oxygen species production, depolarization, swelling, and dynamic imbalance, resulting in reduced arrhythmias, myocardial cell death, infarct size and enhanced cardiac performance during I/R injury in pre-diabetic rats. Taken together, inhibition of mitochondrial fission effectively protected the heart against cardiac I/R injury regardless of the time of administration in pre-diabetic rats.
Collapse
Affiliation(s)
- Chayodom Maneechote
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siripong Palee
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Sasiwan Kerdphoo
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Thidarat Jaiwongkam
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Siriporn C Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand
| | - Nipon Chattipakorn
- Cardiac Electrophysiology Research and Training Center, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Cardiac Electrophysiology Unit, Department of Physiology, Faculty of Medicine, Chiang Mai University, Chiang Mai 50200, Thailand; Center of Excellence in Cardiac Electrophysiology Research, Chiang Mai University, Chiang Mai 50200, Thailand.
| |
Collapse
|
13
|
Yang S, Hu B, Wang Z, Zhang C, Jiao H, Mao Z, Wei L, Jia J, Zhao J. Cannabinoid CB1 receptor agonist ACEA alleviates brain ischemia/reperfusion injury via CB1-Drp1 pathway. Cell Death Discov 2020; 6:102. [PMID: 33083022 PMCID: PMC7548964 DOI: 10.1038/s41420-020-00338-3] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2020] [Accepted: 09/23/2020] [Indexed: 12/16/2022] Open
Abstract
Activation of the cannabinoid CB1 receptor induces neuroprotection against brain ischemia/reperfusion injury (IRI); however, the mechanism is still unknown. In this study, we used oxygen-glucose deprivation/reoxygenation (OGD/R)-induced injury in neuronal cells and middle cerebral artery occlusion (MCAO)-induced brain IRI in rats to mimic ischemic brain injury, and hypothesized that the CB1 receptor agonist arachidonyl-2-chloroethylamide (ACEA) would protect ischemic neurons by inhibiting mitochondrial fission via dynamin-related protein 1 (Drp1). We found that OGD/R injury reduced cell viability and mitochondrial function, increased lactate dehydrogenase (LDH) release, and increased cell apoptosis, and mitochondrial fission. Notably, ACEA significantly abolished the OGD/R-induced neuronal injuries described above. Similarly, ACEA significantly reversed MCAO-induced increases in brain infarct volume, neuronal apoptosis and mitochondrial fission, leading to the recovery of neurological functions. The neuroprotective effects of ACEA were obviously blocked by coadministration of the CB1 receptor antagonist AM251 or by the upregulation of Drp1 expression, indicating that ACEA alleviates brain IRI via the CB1-Drp1 pathway. Our findings suggest that the CB1 receptor links aberrant mitochondrial fission to brain IRI, providing a new therapeutic target for brain IRI treatment.
Collapse
Affiliation(s)
- Shuai Yang
- Department of Neurosurgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Bin Hu
- Department of Neurosurgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zongming Wang
- Department of Neurosurgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Changming Zhang
- Department of Neurosurgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Haosen Jiao
- Department of Neurosurgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Zhigang Mao
- Department of Neurosurgery, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Liguang Wei
- Department of Neurosurgery, The Second People’s Hospital of Qinzhou City, Qinzhou, China
| | - Ji Jia
- Department of Anesthesiology, General Hospital of Southern Theatre Command of PLA, Guangzhou, China
| | - Jingling Zhao
- Department of Burns, the First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
14
|
Wang J, Zhou H. Mitochondrial quality control mechanisms as molecular targets in cardiac ischemia -reperfusion injury. Acta Pharm Sin B 2020; 10:1866-1879. [PMID: 33163341 PMCID: PMC7606115 DOI: 10.1016/j.apsb.2020.03.004] [Citation(s) in RCA: 246] [Impact Index Per Article: 49.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2019] [Revised: 01/19/2020] [Accepted: 02/27/2020] [Indexed: 12/23/2022] Open
Abstract
Mitochondrial damage is a critical contributor to cardiac ischemia/reperfusion (I/R) injury. Mitochondrial quality control (MQC) mechanisms, a series of adaptive responses that preserve mitochondrial structure and function, ensure cardiomyocyte survival and cardiac function after I/R injury. MQC includes mitochondrial fission, mitochondrial fusion, mitophagy and mitochondria-dependent cell death. The interplay among these responses is linked to pathological changes such as redox imbalance, calcium overload, energy metabolism disorder, signal transduction arrest, the mitochondrial unfolded protein response and endoplasmic reticulum stress. Excessive mitochondrial fission is an early marker of mitochondrial damage and cardiomyocyte death. Reduced mitochondrial fusion has been observed in stressed cardiomyocytes and correlates with mitochondrial dysfunction and cardiac depression. Mitophagy allows autophagosomes to selectively degrade poorly structured mitochondria, thus maintaining mitochondrial network fitness. Nevertheless, abnormal mitophagy is maladaptive and has been linked to cell death. Although mitochondria serve as the fuel source of the heart by continuously producing adenosine triphosphate, they also stimulate cardiomyocyte death by inducing apoptosis or necroptosis in the reperfused myocardium. Therefore, defects in MQC may determine the fate of cardiomyocytes. In this review, we summarize the regulatory mechanisms and pathological effects of MQC in myocardial I/R injury, highlighting potential targets for the clinical management of reperfusion.
Collapse
Affiliation(s)
- Jin Wang
- Department of Cardiology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China
| | - Hao Zhou
- Department of Cardiology, Chinese PLA General Hospital, Medical School of Chinese PLA, Beijing 100853, China
| |
Collapse
|
15
|
Rosdah AA, Smiles WJ, Oakhill JS, Scott JW, Langendorf CG, Delbridge LMD, Holien JK, Lim SY. New perspectives on the role of Drp1 isoforms in regulating mitochondrial pathophysiology. Pharmacol Ther 2020; 213:107594. [PMID: 32473962 DOI: 10.1016/j.pharmthera.2020.107594] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 05/26/2020] [Indexed: 12/13/2022]
Abstract
Mitochondria are dynamic organelles constantly undergoing fusion and fission. A concerted balance between the process of mitochondrial fusion and fission is required to maintain cellular health under different physiological conditions. Mutation and dysregulation of Drp1, the major driver of mitochondrial fission, has been associated with various neurological, oncological and cardiovascular disorders. Moreover, when subjected to pathological insults, mitochondria often undergo excessive fission, generating fragmented and dysfunctional mitochondria leading to cell death. Therefore, manipulating mitochondrial fission by targeting Drp1 has been an appealing therapeutic approach for cytoprotection. However, studies have been inconsistent. Studies employing Drp1 constructs representing alternate Drp1 isoforms, have demonstrated differing impacts of these isoforms on mitochondrial fission and cell death. Furthermore, there are distinct expression patterns of Drp1 isoforms in different tissues, suggesting idiosyncratic engagement in specific cellular functions. In this review, we will discuss these inherent variations among human Drp1 isoforms and how they could affect Drp1-mediated mitochondrial fission and cell death.
Collapse
Affiliation(s)
- Ayeshah A Rosdah
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Victoria, Australia; Faculty of Medicine, Universitas Sriwijaya, Palembang, Indonesia; Department of Surgery, University of Melbourne, Victoria, Australia
| | - William J Smiles
- Metabolic Signalling Laboratory, St Vincent's Institute of Medical Research, Victoria, Australia
| | - Jonathan S Oakhill
- Metabolic Signalling Laboratory, St Vincent's Institute of Medical Research, Victoria, Australia; Mary MacKillop Institute for Health Research, Australian Catholic University, Victoria, Australia
| | - John W Scott
- Mary MacKillop Institute for Health Research, Australian Catholic University, Victoria, Australia; Protein Chemistry and Metabolism Unit, St Vincent's Institute of Medical Research, Victoria, Australia; The Florey Institute of Neuroscience and Mental Health, Victoria, Australia
| | - Christopher G Langendorf
- Protein Chemistry and Metabolism Unit, St Vincent's Institute of Medical Research, Victoria, Australia
| | - Lea M D Delbridge
- Department of Physiology, University of Melbourne, Victoria, Australia
| | - Jessica K Holien
- Department of Surgery, University of Melbourne, Victoria, Australia; Structural Bioinformatics and Drug Discovery, St Vincent's Institute of Medical Research, Victoria, Australia
| | - Shiang Y Lim
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Victoria, Australia; Department of Surgery, University of Melbourne, Victoria, Australia.
| |
Collapse
|
16
|
Wendt L, Vider J, Hoe LES, Du Toit E, Peart JN, Headrick JP. Complex Effects of Putative DRP-1 Inhibitors on Stress Responses in Mouse Heart and Rat Cardiomyoblasts. J Pharmacol Exp Ther 2020; 372:95-106. [PMID: 31704803 DOI: 10.1124/jpet.119.258897] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 09/12/2019] [Indexed: 12/11/2022] Open
Abstract
Dynamin-related protein-1 (DRP-1)-dependent mitochondrial fission may influence cardiac tolerance to ischemic or oxidative stress, presenting a potential "cardioprotective" target. Effects of dynamin inhibitors [mitochondrial division inhibitor 1 (MDIVI-1) and dynasore] on injury, mitochondrial function, and signaling proteins were assessed in distinct models: ischemia-reperfusion (I-R) in mouse hearts and oxidative stress in rat H9c2 cardiomyoblasts. Hearts exhibited substantial cell death [approx. 40 IU lactate dehydrogenase (LDH) efflux] and dysfunction (approx. 40 mmHg diastolic pressure, approx. 40% contractile recovery) following 25 minutes' ischemia. Pretreatment with 1 μM MDIVI-1 reduced dysfunction (30 mmHg diastolic pressure, approx. 55% recovery) and delayed without reducing overall cell death, whereas 5 μM MDIVI-1 reduced overall death at the same time paradoxically exaggerating dysfunction. Postischemic expression of mitochondrial DRP-1 and phospho-activation of ERK1/2 were reduced by MDIVI-1. Conversely, 1 μM dynasore worsened cell death and reduced nonmitochondrial DRP-1. Postischemic respiratory fluxes were unaltered by MDIVI-1, although a 50% fall in complex-I flux control ratio was reversed. In H9c2 myoblasts stressed with 400 μM H2O2, treatment with 50 μM MDIVI-1 preserved metabolic (MTT assay) and mitochondrial (basal respiration) function without influencing survival. This was associated with differential signaling responses, including reduced early versus increased late phospho-activation of ERK1/2, increased phospho-activation of protein kinase B (AKT), and differential changes in determinants of autophagy [reduced microtubule-associated protein 1 light chain 3b (LC3B-II/I) vs. increased Parkinson juvenile disease protein 2 (Parkin)] and apoptosis [reduced poly-(ADP-ribose) polymerase (PARP) cleavage vs. increased BCL2-associated X (BAX)/B-cell lymphoma 2 (BCL2)]. These data show MDIVI-1 (not dynasore) confers some benefit during I-R/oxidative stress. However, despite mitochondrial and metabolic preservation, MDIVI-1 exerts mixed effects on cell death versus dysfunction, potentially reflecting differential changes in survival kinase, autophagy, and apoptosis pathways. SIGNIFICANCE STATEMENT: Inhibition of mitochondrial fission is a novel approach to still elusive cardioprotection. Assessing effects of fission inhibitors on responses to ischemic or oxidative stress in hearts and cardiomyoblasts reveals mitochondrial division inhibitor 1 (MDIVI-1) and dynasore induce complex effects and limited cardioprotection. This includes differential impacts on death and dysfunction, survival kinases, and determinants of autophagy and apoptosis. Although highlighting the interconnectedness of fission and these key processes, results suggest MDIVI-1 and dynasore may be of limited value in the quest for effective cardioprotection.
Collapse
Affiliation(s)
- Lauren Wendt
- School of Medical Science, Griffith University, Southport, Australia (L.W., J.V., E.D.T., J.N.P., J.P.H.) and Critical Care Research Group, The Prince Charles Hospital and The University of Queensland, Chermside, Australia (L.E.S.H.)
| | - Jelena Vider
- School of Medical Science, Griffith University, Southport, Australia (L.W., J.V., E.D.T., J.N.P., J.P.H.) and Critical Care Research Group, The Prince Charles Hospital and The University of Queensland, Chermside, Australia (L.E.S.H.)
| | - Louise E See Hoe
- School of Medical Science, Griffith University, Southport, Australia (L.W., J.V., E.D.T., J.N.P., J.P.H.) and Critical Care Research Group, The Prince Charles Hospital and The University of Queensland, Chermside, Australia (L.E.S.H.)
| | - Eugene Du Toit
- School of Medical Science, Griffith University, Southport, Australia (L.W., J.V., E.D.T., J.N.P., J.P.H.) and Critical Care Research Group, The Prince Charles Hospital and The University of Queensland, Chermside, Australia (L.E.S.H.)
| | - Jason N Peart
- School of Medical Science, Griffith University, Southport, Australia (L.W., J.V., E.D.T., J.N.P., J.P.H.) and Critical Care Research Group, The Prince Charles Hospital and The University of Queensland, Chermside, Australia (L.E.S.H.)
| | - John P Headrick
- School of Medical Science, Griffith University, Southport, Australia (L.W., J.V., E.D.T., J.N.P., J.P.H.) and Critical Care Research Group, The Prince Charles Hospital and The University of Queensland, Chermside, Australia (L.E.S.H.)
| |
Collapse
|
17
|
Abstract
Significance: In addition to their classical role in cellular ATP production, mitochondria are of key relevance in various (patho)physiological mechanisms including second messenger signaling, neuro-transduction, immune responses and death induction. Recent Advances: Within cells, mitochondria are motile and display temporal changes in internal and external structure ("mitochondrial dynamics"). During the last decade, substantial empirical and in silico evidence was presented demonstrating that mitochondrial dynamics impacts on mitochondrial function and vice versa. Critical Issues: However, a comprehensive and quantitative understanding of the bidirectional links between mitochondrial external shape, internal structure and function ("morphofunction") is still lacking. The latter particularly hampers our understanding of the functional properties and behavior of individual mitochondrial within single living cells. Future Directions: In this review we discuss the concept of mitochondrial morphofunction in mammalian cells, primarily using experimental evidence obtained within the last decade. The topic is introduced by briefly presenting the central role of mitochondria in cell physiology and the importance of the mitochondrial electron transport chain (ETC) therein. Next, we summarize in detail how mitochondrial (ultra)structure is controlled and discuss empirical evidence regarding the equivalence of mitochondrial (ultra)structure and function. Finally, we provide a brief summary of how mitochondrial morphofunction can be quantified at the level of single cells and mitochondria, how mitochondrial ultrastructure/volume impacts on mitochondrial bioreactions and intramitochondrial protein diffusion, and how mitochondrial morphofunction can be targeted by small molecules.
Collapse
Affiliation(s)
- Elianne P. Bulthuis
- Department of Biochemistry (286), Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Merel J.W. Adjobo-Hermans
- Department of Biochemistry (286), Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Peter H.G.M. Willems
- Department of Biochemistry (286), Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
| | - Werner J.H. Koopman
- Department of Biochemistry (286), Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, Nijmegen, The Netherlands
- Address correspondence to: Dr. Werner J.H. Koopman, Department of Biochemistry (286), Radboud Institute for Molecular Life Sciences, Radboud University Medical Centre, P.O. Box 9101, Nijmegen NL-6500 HB, The Netherlands
| |
Collapse
|
18
|
Nair S, Sobotka KS, Joshi P, Gressens P, Fleiss B, Thornton C, Mallard C, Hagberg H. Lipopolysaccharide-induced alteration of mitochondrial morphology induces a metabolic shift in microglia modulating the inflammatory response in vitro and in vivo. Glia 2019; 67:1047-1061. [PMID: 30637805 DOI: 10.1002/glia.23587] [Citation(s) in RCA: 178] [Impact Index Per Article: 29.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2018] [Revised: 12/21/2018] [Accepted: 12/21/2018] [Indexed: 12/18/2022]
Abstract
Accumulating evidence suggests that changes in the metabolic signature of microglia underlie their response to inflammation. We sought to increase our knowledge of how pro-inflammatory stimuli induce metabolic changes. Primary microglia exposed to lipopolysaccharide (LPS)-expressed excessive fission leading to more fragmented mitochondria than tubular mitochondria. LPS-mediated Toll-like receptor 4 (TLR4) activation also resulted in metabolic reprogramming from oxidative phosphorylation to glycolysis. Blockade of mitochondrial fission by Mdivi-1, a putative mitochondrial division inhibitor led to the reversal of the metabolic shift. Mdivi-1 treatment also normalized the changes caused by LPS exposure, namely an increase in mitochondrial reactive oxygen species production and mitochondrial membrane potential as well as accumulation of key metabolic intermediate of TCA cycle succinate. Moreover, Mdivi-1 treatment substantially reduced LPS induced cytokine and chemokine production. Finally, we showed that Mdivi-1 treatment attenuated expression of genes related to cytotoxic, repair, and immunomodulatory microglia phenotypes in an in vivo neuroinflammation paradigm. Collectively, our data show that the activation of microglia to a classically pro-inflammatory state is associated with a switch to glycolysis that is mediated by mitochondrial fission, a process which may be a pharmacological target for immunomodulation.
Collapse
Affiliation(s)
- Syam Nair
- Centre of Perinatal Medicine and Health, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Kristina S Sobotka
- Centre of Perinatal Medicine and Health, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Pooja Joshi
- PROTECT, INSERM, Université Paris Diderot, Paris, France
| | - Pierre Gressens
- PROTECT, INSERM, Université Paris Diderot, Paris, France
- Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, United Kingdom
| | - Bobbi Fleiss
- PROTECT, INSERM, Université Paris Diderot, Paris, France
- Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, United Kingdom
- School of Health and Biomedical Sciences, RMIT University, Bundoora, Victoria, Australia
| | - Claire Thornton
- Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, United Kingdom
| | - Carina Mallard
- Centre of Perinatal Medicine and Health, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Henrik Hagberg
- Centre of Perinatal Medicine and Health, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
- Centre for the Developing Brain, Department of Division of Imaging Sciences and Biomedical Engineering, King's College London, King's Health Partners, St. Thomas' Hospital, London, United Kingdom
- Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| |
Collapse
|
19
|
Nishimura A, Shimauchi T, Tanaka T, Shimoda K, Toyama T, Kitajima N, Ishikawa T, Shindo N, Numaga-Tomita T, Yasuda S, Sato Y, Kuwahara K, Kumagai Y, Akaike T, Ide T, Ojida A, Mori Y, Nishida M. Hypoxia-induced interaction of filamin with Drp1 causes mitochondrial hyperfission-associated myocardial senescence. Sci Signal 2018; 11:11/556/eaat5185. [PMID: 30425165 DOI: 10.1126/scisignal.aat5185] [Citation(s) in RCA: 99] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Defective mitochondrial dynamics through aberrant interactions between mitochondria and actin cytoskeleton is increasingly recognized as a key determinant of cardiac fragility after myocardial infarction (MI). Dynamin-related protein 1 (Drp1), a mitochondrial fission-accelerating factor, is activated locally at the fission site through interactions with actin. Here, we report that the actin-binding protein filamin A acted as a guanine nucleotide exchange factor for Drp1 and mediated mitochondrial fission-associated myocardial senescence in mice after MI. In peri-infarct regions characterized by mitochondrial hyperfission and associated with myocardial senescence, filamin A colocalized with Drp1 around mitochondria. Hypoxic stress induced the interaction of filamin A with the GTPase domain of Drp1 and increased Drp1 activity in an actin-binding-dependent manner in rat cardiomyocytes. Expression of the A1545T filamin mutant, which potentiates actin aggregation, promoted mitochondrial hyperfission under normoxia. Furthermore, pharmacological perturbation of the Drp1-filamin A interaction by cilnidipine suppressed mitochondrial hyperfission-associated myocardial senescence and heart failure after MI. Together, these data demonstrate that Drp1 association with filamin and the actin cytoskeleton contributes to cardiac fragility after MI and suggests a potential repurposing of cilnidipine, as well as provides a starting point for innovative Drp1 inhibitor development.
Collapse
Affiliation(s)
- Akiyuki Nishimura
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Aichi 444-8787, Japan.,Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Aichi 444-8787, Japan.,SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Aichi 444-8787, Japan.,Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Tsukasa Shimauchi
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Aichi 444-8787, Japan.,Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Aichi 444-8787, Japan.,Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Tomohiro Tanaka
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Aichi 444-8787, Japan.,Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Aichi 444-8787, Japan
| | - Kakeru Shimoda
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Aichi 444-8787, Japan.,Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Aichi 444-8787, Japan.,SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Aichi 444-8787, Japan
| | - Takashi Toyama
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Aichi 444-8787, Japan.,Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan.,Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan
| | - Naoyuki Kitajima
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Aichi 444-8787, Japan.,Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Tatsuya Ishikawa
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan.,EA Pharma Co. Inc., Tokyo 104-0042, Japan
| | - Naoya Shindo
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Takuro Numaga-Tomita
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Aichi 444-8787, Japan.,Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Aichi 444-8787, Japan.,SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Aichi 444-8787, Japan
| | - Satoshi Yasuda
- National Institute of Health Sciences, Kanagawa 210-9501, Japan
| | - Yoji Sato
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan.,National Institute of Health Sciences, Kanagawa 210-9501, Japan
| | | | - Yoshito Kumagai
- Faculty of Medicine, University of Tsukuba, Tsukuba 305-8575, Japan
| | - Takaaki Akaike
- Tohoku University Graduate School of Medicine, Sendai 980-8575, Japan
| | - Tomomi Ide
- Kyushu University Graduate School of Medical Sciences, Fukuoka 812-8582, Japan
| | - Akio Ojida
- Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| | - Yasuo Mori
- Graduate School of Engineering, Kyoto University, Kyoto 615-8510, Japan
| | - Motohiro Nishida
- National Institute for Physiological Sciences (NIPS), National Institutes of Natural Sciences, Aichi 444-8787, Japan. .,Exploratory Research Center on Life and Living Systems (ExCELLS), National Institutes of Natural Sciences, Aichi 444-8787, Japan.,SOKENDAI (School of Life Science, The Graduate University for Advanced Studies), Aichi 444-8787, Japan.,Graduate School of Pharmaceutical Sciences, Kyushu University, Fukuoka 812-8582, Japan
| |
Collapse
|
20
|
AKAP1 Protects from Cerebral Ischemic Stroke by Inhibiting Drp1-Dependent Mitochondrial Fission. J Neurosci 2018; 38:8233-8242. [PMID: 30093535 PMCID: PMC6146498 DOI: 10.1523/jneurosci.0649-18.2018] [Citation(s) in RCA: 100] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 07/29/2018] [Accepted: 08/01/2018] [Indexed: 12/20/2022] Open
Abstract
Mitochondrial fission and fusion impact numerous cellular functions and neurons are particularly sensitive to perturbations in mitochondrial dynamics. Here we describe that male mice lacking the mitochondrial A-kinase anchoring protein 1 (AKAP1) exhibit increased sensitivity in the transient middle cerebral artery occlusion model of focal ischemia. At the ultrastructural level, AKAP1-/- mice have smaller mitochondria and increased contacts between mitochondria and the endoplasmic reticulum in the brain. Mechanistically, deletion of AKAP1 dysregulates complex II of the electron transport chain, increases superoxide production, and impairs Ca2+ homeostasis in neurons subjected to excitotoxic glutamate. Ca2+ deregulation in neurons lacking AKAP1 can be attributed to loss of inhibitory phosphorylation of the mitochondrial fission enzyme dynamin-related protein 1 (Drp1) at the protein kinase A (PKA) site Ser637. Our results indicate that inhibition of Drp1-dependent mitochondrial fission by the outer mitochondrial AKAP1/PKA complex protects neurons from ischemic stroke by maintaining respiratory chain activity, inhibiting superoxide production, and delaying Ca2+ deregulation. They also provide the first genetic evidence that Drp1 inhibition may be of therapeutic relevance for the treatment of stroke and neurodegeneration.SIGNIFICANCE STATEMENT Previous work suggests that activation of dynamin-related protein 1 (Drp1) and mitochondrial fission contribute to ischemic injury in the brain. However, the specificity and efficacy of the pharmacological Drp1 inhibitor mdivi-1 that was used has now been discredited by several high-profile studies. Our report is timely and highly impactful because it provides the first evidence that genetic disinhibition of Drp1 via knock-out of the mitochondrial protein kinase A (PKA) scaffold AKAP1 exacerbates stroke injury in mice. Mechanistically, we show that electron transport deficiency, increased superoxide production, and Ca2+ overload result from genetic disinhibition of Drp1. In summary, our work settles current controversies regarding the role of mitochondrial fission in neuronal injury, provides mechanisms, and suggests that fission inhibitors hold promise as future therapeutic agents.
Collapse
|
21
|
Lai MC, Hung TY, Lin KM, Sung PS, Wu SJ, Yang CS, Wu YJ, Tsai JJ, Wu SN, Huang CW. Sodium Metabisulfite: Effects on Ionic Currents and Excitotoxicity. Neurotox Res 2018; 34:1-15. [PMID: 29188487 DOI: 10.1007/s12640-017-9844-4] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2017] [Revised: 11/20/2017] [Accepted: 11/21/2017] [Indexed: 12/31/2022]
Abstract
How sodium metabisulfite (SMB; Na2S2O5), a popular food preservative and antioxidant, interacts with excitable membrane and induces excitotoxicity is incompletely understood. In this study, the patch-clamp technique was used to investigate and record the electrophysiological effect of SMB on electrically excitable HL-1 cardiomyocytes and NSC-34 neurons, as well as its relationship to pilocarpine-induced seizures and neuronal excitotoxicity in rats. We used Western blotting, to analyze sodium channel expression on hippocampi after chronic SMB treatment. It was found that voltage-gated Na+ current (I Na) was stimulated, and current inactivation and deactivation were slowed in SMB-treated (30 μM) HL-1 cardiomyocytes. SMB-induced increases of I Na were attenuated in cells treated with ranolazine (10 μM) or eugenol (30 μM). The current-voltage relationship of I Na shifted to slightly more negative potentials in SMB-treated cells, the peak I Na with an EC50 value of 18 μM increased, and the steady-state inactivation curve of I Na shifted to a more positive potential. However, the tail component of the rapidly activating delayed-rectifier K+ current (I Kr) was dose-dependently inhibited. Cell-attached voltage-clamp recordings in SMB-treated cells showed that the frequency of action currents and prolonged action potential were higher. In SMB-treated NSC-34 neurons, the peak I Na was higher; however, neither the time to peak nor the inactivation time constant (I Na) changed. Pilocarpine-induced seizures were exacerbated, and acute neuronal damage and chronic mossy fiber sprouting increased in SMB-treated rats. Western blotting showed higher expression of the sodium channel in cells after chronic SMB treatment. We conclude that SMB contributes to the sodium channel-activating mechanism through which it alters cellular excitability and excitotoxicity in wide-spectrum excitable cells.
Collapse
Affiliation(s)
- Ming-Chi Lai
- Department of Pediatrics, Chi-Mei Medical Center, Tainan, Taiwan
| | - Te-Yu Hung
- Department of Pediatrics, Chi-Mei Medical Center, Tainan, Taiwan
| | - Kao-Min Lin
- Department of Pediatric Neurology, Chiayi Christian Hospital, Chiayi, Taiwan
| | - Pi-Shan Sung
- Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Shyh-Jong Wu
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Sheng Yang
- Department of Neurology, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, Taiwan
| | - Yi-Jen Wu
- Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Jing-Jane Tsai
- Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan
| | - Sheng-Nan Wu
- Department of Physiology, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
- Institute of Basic Medical Sciences, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| | - Chin-Wei Huang
- Department of Neurology, National Cheng Kung University Hospital, College of Medicine, National Cheng Kung University, Tainan, Taiwan.
| |
Collapse
|
22
|
Chang WT, Wu SN. Activation of voltage-gated sodium current and inhibition of erg-mediated potassium current caused by telmisartan, an antagonist of angiotensin II type-1 receptor, in HL-1 atrial cardiomyocytes. Clin Exp Pharmacol Physiol 2018; 45:797-807. [PMID: 29617054 DOI: 10.1111/1440-1681.12943] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 03/11/2018] [Accepted: 03/23/2018] [Indexed: 12/19/2022]
Abstract
Telmisartan (TEL) is a non-peptide blocker of angiotensin II type-1 (AT1 ) receptor. However, the mechanisms through which this drug interacts directly with ion currents in hearts remain largely unclear. Herein, we aim to investigate the effects of TEL the on ionic currents and membrane potential of murine HL-1 cardiomyocytes. In whole-cell recordings, addition of TEL stimulated the peak and late components of voltage-gated Na+ currents (INa ) with different potencies. The EC50 values required to achieve the stimulatory effect of this drug on peak and late INa were 0.2 and 1.2 μmol/L, respectively, and the current-voltage relationship of peak INa shifted toward less-depolarized potentials during exposure to TEL. Telmisartan not only increased peak INa but also prolonged the inactivation time course of late INa . Amiodarone (Amio) or ranolazine (Ran), but not angiotensin II, could reverse TEL-mediated effects. The drug enhanced the recovery rate of INa inactivation and exerted an inhibitory effect on erg-mediated K+ and L-type Ca2+ currents. In whole-cell current-clamp recordings, addition of the drug resulted in prolongation of the duration of action potentials (APs) in a dose-dependent manner in HL-1 cells; Amio or Ran could reverse this increase in AP durations. Telmisartan-mediated prolongation of AP was attenuated in KCNH2 siRNA-transfected HL-1 cells. In cultured smooth muscle cells of the human coronary artery, TEL enhanced INa amplitudes and slowed current inactivation. Stimulation by TEL of INa in HL-1 cells did not simply increase current magnitude but altered current kinetics, thereby suggesting state-dependent activation. Telmisartan may have greater affinity to the open/inactivated state than to the resting state residing in NaV channels. Collectively, TEL-mediated stimulation of INa and inhibition of IK(erg) could be an important ionic mechanism underlying the increased cell excitability of HL-1 cells; these actions, however, cannot be entirely explained by its blockade of AT1 receptor.
Collapse
Affiliation(s)
- Wei-Ting Chang
- Division of Cardiovascular Medicine, Chi-Mei Medical Center, Tainan, Taiwan.,Department of Biotechnology, Southern Taiwan University of Science and Technology, Tainan, Taiwan
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan, Taiwan
| |
Collapse
|
23
|
Ong SB, Kalkhoran SB, Hernández-Reséndiz S, Samangouei P, Ong SG, Hausenloy DJ. Mitochondrial-Shaping Proteins in Cardiac Health and Disease - the Long and the Short of It! Cardiovasc Drugs Ther 2017; 31:87-107. [PMID: 28190190 PMCID: PMC5346600 DOI: 10.1007/s10557-016-6710-1] [Citation(s) in RCA: 78] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Mitochondrial health is critically dependent on the ability of mitochondria to undergo changes in mitochondrial morphology, a process which is regulated by mitochondrial shaping proteins. Mitochondria undergo fission to generate fragmented discrete organelles, a process which is mediated by the mitochondrial fission proteins (Drp1, hFIS1, Mff and MiD49/51), and is required for cell division, and to remove damaged mitochondria by mitophagy. Mitochondria undergo fusion to form elongated interconnected networks, a process which is orchestrated by the mitochondrial fusion proteins (Mfn1, Mfn2 and OPA1), and which enables the replenishment of damaged mitochondrial DNA. In the adult heart, mitochondria are relatively static, are constrained in their movement, and are characteristically arranged into 3 distinct subpopulations based on their locality and function (subsarcolemmal, myofibrillar, and perinuclear). Although the mitochondria are arranged differently, emerging data supports a role for the mitochondrial shaping proteins in cardiac health and disease. Interestingly, in the adult heart, it appears that the pleiotropic effects of the mitochondrial fusion proteins, Mfn2 (endoplasmic reticulum-tethering, mitophagy) and OPA1 (cristae remodeling, regulation of apoptosis, and energy production) may play more important roles than their pro-fusion effects. In this review article, we provide an overview of the mitochondrial fusion and fission proteins in the adult heart, and highlight their roles as novel therapeutic targets for treating cardiac disease.
Collapse
Affiliation(s)
- Sang-Bing Ong
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore, 169857, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| | - Siavash Beikoghli Kalkhoran
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK
| | - Sauri Hernández-Reséndiz
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore, 169857, Singapore.,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore
| | - Parisa Samangouei
- The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK
| | - Sang-Ging Ong
- Stanford Cardiovascular Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Derek John Hausenloy
- Cardiovascular and Metabolic Disorders Program, Duke-National University of Singapore Medical School, 8 College Road, Singapore, 169857, Singapore. .,National Heart Research Institute Singapore, National Heart Centre Singapore, Singapore, Singapore. .,The Hatter Cardiovascular Institute, Institute of Cardiovascular Science, University College London, London, UK. .,The National Institute of Health Research, University College London Hospitals Biomedical Research Centre, London, UK.
| |
Collapse
|
24
|
Flippo KH, Strack S. Mitochondrial dynamics in neuronal injury, development and plasticity. J Cell Sci 2017; 130:671-681. [PMID: 28154157 DOI: 10.1242/jcs.171017] [Citation(s) in RCA: 172] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Mitochondria fulfill numerous cellular functions including ATP production, Ca2+ buffering, neurotransmitter synthesis and degradation, ROS production and sequestration, apoptosis and intermediate metabolism. Mitochondrial dynamics, a collective term for the processes of mitochondrial fission, fusion and transport, governs mitochondrial function and localization within the cell. Correct balance of mitochondrial dynamics is especially important in neurons as mutations in fission and fusion enzymes cause peripheral neuropathies and impaired development of the nervous system in humans. Regulation of mitochondrial dynamics is partly accomplished through post-translational modification of mitochondrial fission and fusion enzymes, in turn influencing mitochondrial bioenergetics and transport. The importance of post-translational regulation is highlighted by numerous neurodegenerative disorders associated with post-translational modification of the mitochondrial fission enzyme Drp1. Not surprisingly, mitochondrial dynamics also play an important physiological role in the development of the nervous system and synaptic plasticity. Here, we highlight recent findings underlying the mechanisms and regulation of mitochondrial dynamics in relation to neurological disease, as well as the development and plasticity of the nervous system.
Collapse
Affiliation(s)
- Kyle H Flippo
- Department of Pharmacology, University of Iowa, Iowa City, USA
| | - Stefan Strack
- Department of Pharmacology, University of Iowa, Iowa City, USA
| |
Collapse
|
25
|
Rowlands DJ. Mitochondria dysfunction: A novel therapeutic target in pathological lung remodeling or bystander? Pharmacol Ther 2016; 166:96-105. [PMID: 27373853 DOI: 10.1016/j.pharmthera.2016.06.019] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2016] [Accepted: 06/14/2016] [Indexed: 11/19/2022]
Abstract
The renascence in mitochondrial research has fueled breakthroughs in our understanding of mitochondrial biology identifying major roles in biological processes ranging from cellular oxygen sensing and regulation of intracellular calcium levels through to initiation of apoptosis or a shift in cell phenotype. Chronic respiratory diseases are no exception to the resurgent interest in mitochondrial biology. Microscopic observations of lungs from patients with chronic respiratory diseases such as pulmonary arterial hypertension, asthma and COPD show accumulation of dysmorphic mitochondria and provide the first evidence of mitochondrial dysfunction in diseased lungs. Recent mechanistic insights have established links between mitochondrial dysfunction or aberrant biogenesis and the pathogenesis of chronic respiratory diseases through playing a causative role in structural remodeling of the lung. The aim here is to discuss the case for a mitochondrial basis of lung remodeling in patients with chronic respiratory diseases. The present article will focus on the question of whether currently available data supports mitochondrial mechanisms as a viable point of therapeutic intervention in respiratory diseases and suggestions for future avenues of research in this rapidly evolving field.
Collapse
Affiliation(s)
- David J Rowlands
- Novartis Institutes for Biomedical Research, 250 Massachusetts Avenue, Cambridge, MA 02139, USA.
| |
Collapse
|
26
|
Rosdah AA, K Holien J, Delbridge LMD, Dusting GJ, Lim SY. Mitochondrial fission - a drug target for cytoprotection or cytodestruction? Pharmacol Res Perspect 2016; 4:e00235. [PMID: 27433345 PMCID: PMC4876145 DOI: 10.1002/prp2.235] [Citation(s) in RCA: 101] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 03/24/2016] [Indexed: 01/18/2023] Open
Abstract
Mitochondria are morphologically dynamic organelles constantly undergoing processes of fission and fusion that maintain integrity and bioenergetics of the organelle: these processes are vital for cell survival. Disruption in the balance of mitochondrial fusion and fission is thought to play a role in several pathological conditions including ischemic heart disease. Proteins involved in regulating the processes of mitochondrial fusion and fission are therefore potential targets for pharmacological therapies. Mdivi‐1 is a small molecule inhibitor of the mitochondrial fission protein Drp1. Inhibiting mitochondrial fission with Mdivi‐1 has proven cytoprotective benefits in several cell types involved in a wide array of cardiovascular injury models. On the other hand, Mdivi‐1 can also exert antiproliferative and cytotoxic effects, particularly in hyperproliferative cells. In this review, we discuss these divergent effects of Mdivi‐1 on cell survival, as well as the potential and limitations of Mdivi‐1 as a therapeutic agent.
Collapse
Affiliation(s)
- Ayeshah A Rosdah
- O'Brien Institute Department St Vincent's Institute of Medical Research Victoria Australia; Department of Physiology University of Melbourne Victoria Australia; Faculty of Medicine Sriwijaya University Palembang Indonesia
| | - Jessica K Holien
- ACRF Rational Drug Discovery Centre St Vincent's Institute of Medical Research Victoria Australia
| | | | - Gregory J Dusting
- O'Brien Institute Department St Vincent's Institute of Medical Research Victoria Australia; Centre for Eye Research Australia Royal Victorian Eye and Ear Hospital Victoria Australia; Department of Surgery University of Melbourne Victoria Australia
| | - Shiang Y Lim
- O'Brien Institute Department St Vincent's Institute of Medical Research Victoria Australia; Department of Surgery University of Melbourne Victoria Australia
| |
Collapse
|
27
|
Abstract
In addition to oxidative phosphorylation (OXPHOS), mitochondria perform other functions such as heme biosynthesis and oxygen sensing and mediate calcium homeostasis, cell growth, and cell death. They participate in cell communication and regulation of inflammation and are important considerations in aging, drug toxicity, and pathogenesis. The cell's capacity to maintain its mitochondria involves intramitochondrial processes, such as heme and protein turnover, and those involving entire organelles, such as fusion, fission, selective mitochondrial macroautophagy (mitophagy), and mitochondrial biogenesis. The integration of these processes exemplifies mitochondrial quality control (QC), which is also important in cellular disorders ranging from primary mitochondrial genetic diseases to those that involve mitochondria secondarily, such as neurodegenerative, cardiovascular, inflammatory, and metabolic syndromes. Consequently, mitochondrial biology represents a potentially useful, but relatively unexploited area of therapeutic innovation. In patients with genetic OXPHOS disorders, the largest group of inborn errors of metabolism, effective therapies, apart from symptomatic and nutritional measures, are largely lacking. Moreover, the genetic and biochemical heterogeneity of these states is remarkably similar to those of certain acquired diseases characterized by metabolic and oxidative stress and displaying wide variability. This biologic variability reflects cell-specific and repair processes that complicate rational pharmacological approaches to both primary and secondary mitochondrial disorders. However, emerging concepts of mitochondrial turnover and dynamics along with new mitochondrial disease models are providing opportunities to develop and evaluate mitochondrial QC-based therapies. The goals of such therapies extend beyond amelioration of energy insufficiency and tissue loss and entail cell repair, cell replacement, and the prevention of fibrosis. This review summarizes current concepts of mitochondria as disease elements and outlines novel strategies to address mitochondrial dysfunction through the stimulation of mitochondrial biogenesis and quality control.
Collapse
Affiliation(s)
- Hagir B Suliman
- Departments of Medicine (C.A.P.), Anesthesiology (H.B.S.), Duke Cancer Institute (H.B.S.), and Pathology (C.A.P.), Duke University Medical Center, Durham North Carolina
| | - Claude A Piantadosi
- Departments of Medicine (C.A.P.), Anesthesiology (H.B.S.), Duke Cancer Institute (H.B.S.), and Pathology (C.A.P.), Duke University Medical Center, Durham North Carolina
| |
Collapse
|
28
|
Stimers JR, Song L, Rusch NJ, Rhee SW. Overexpression of the Large-Conductance, Ca2+-Activated K+ (BK) Channel Shortens Action Potential Duration in HL-1 Cardiomyocytes. PLoS One 2015; 10:e0130588. [PMID: 26091273 PMCID: PMC4474436 DOI: 10.1371/journal.pone.0130588] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2014] [Accepted: 05/22/2015] [Indexed: 12/29/2022] Open
Abstract
Long QT syndrome is characterized by a prolongation of the interval between the Q wave and the T wave on the electrocardiogram. This abnormality reflects a prolongation of the ventricular action potential caused by a number of genetic mutations or a variety of drugs. Since effective treatments are unavailable, we explored the possibility of using cardiac expression of the large-conductance, Ca2+-activated K+ (BK) channel to shorten action potential duration (APD). We hypothesized that expression of the pore-forming α subunit of human BK channels (hBKα) in HL-1 cells would shorten action potential duration in this mouse atrial cell line. Expression of hBKα had minimal effects on expression levels of other ion channels with the exception of a small but significant reduction in Kv11.1. Patch-clamped hBKα expressing HL-1 cells exhibited an outward voltage- and Ca2+-sensitive K+ current, which was inhibited by the BK channel blocker iberiotoxin (100 nM). This BK current phenotype was not detected in untransfected HL-1 cells or in HL-1 null cells sham-transfected with an empty vector. Importantly, APD in hBKα-expressing HL-1 cells averaged 14.3 ± 2.8 ms (n = 10), which represented a 53% reduction in APD compared to HL-1 null cells lacking BKα expression. APD in the latter cells averaged 31.0 ± 5.1 ms (n = 13). The shortened APD in hBKα-expressing cells was restored to normal duration by 100 nM iberiotoxin, suggesting that a repolarizing K+ current attributed to BK channels accounted for action potential shortening. These findings provide initial proof-of-concept that the introduction of hBKα channels into a cardiac cell line can shorten APD, and raise the possibility that gene-based interventions to increase hBKα channels in cardiac cells may hold promise as a therapeutic strategy for long QT syndrome.
Collapse
Affiliation(s)
- Joseph R. Stimers
- Department of Pharmacology & Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
- * E-mail:
| | - Li Song
- Department of Pharmacology & Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Nancy J. Rusch
- Department of Pharmacology & Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| | - Sung W. Rhee
- Department of Pharmacology & Toxicology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, United States of America
| |
Collapse
|
29
|
Huang S, Wang Y, Gan X, Fang D, Zhong C, Wu L, Hu G, Sosunov AA, McKhann GM, Yu H, Yan SS. Drp1-mediated mitochondrial abnormalities link to synaptic injury in diabetes model. Diabetes 2015; 64:1728-42. [PMID: 25412623 PMCID: PMC4407851 DOI: 10.2337/db14-0758] [Citation(s) in RCA: 119] [Impact Index Per Article: 11.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2014] [Accepted: 11/15/2014] [Indexed: 02/05/2023]
Abstract
Diabetes has adverse effects on the brain, especially the hippocampus, which is particularly susceptible to synaptic injury and cognitive dysfunction. The underlying mechanisms and strategies to rescue such injury and dysfunction are not well understood. Using a mouse model of type 2 diabetes (db/db mice) and a human neuronal cell line treated with high concentration of glucose, we demonstrate aberrant mitochondrial morphology, reduced ATP production, and impaired activity of complex I. These mitochondrial abnormalities are induced by imbalanced mitochondrial fusion and fission via a glycogen synthase kinase 3β (GSK3β)/dynamin-related protein-1 (Drp1)-dependent mechanism. Modulation of the Drp1 pathway or inhibition of GSK3β activity restores hippocampal long-term potentiation that is impaired in db/db mice. Our results point to a novel role for mitochondria in diabetes-induced synaptic impairment. Exploration of the mechanisms behind diabetes-induced synaptic deficit may provide a novel treatment for mitochondrial and synaptic injury in patients with diabetes.
Collapse
Affiliation(s)
- Shengbin Huang
- Department of Pharmacology & Toxicology and Higuchi Biosciences Center, School of Pharmacy, University of Kansas, Lawrence, KS State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Yongfu Wang
- Department of Pharmacology & Toxicology and Higuchi Biosciences Center, School of Pharmacy, University of Kansas, Lawrence, KS
| | - Xueqi Gan
- Department of Pharmacology & Toxicology and Higuchi Biosciences Center, School of Pharmacy, University of Kansas, Lawrence, KS State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Du Fang
- Department of Pharmacology & Toxicology and Higuchi Biosciences Center, School of Pharmacy, University of Kansas, Lawrence, KS
| | - Changjia Zhong
- Department of Pharmacology & Toxicology and Higuchi Biosciences Center, School of Pharmacy, University of Kansas, Lawrence, KS
| | - Long Wu
- Department of Pharmacology & Toxicology and Higuchi Biosciences Center, School of Pharmacy, University of Kansas, Lawrence, KS
| | - Gang Hu
- Department of Pharmacology & Toxicology and Higuchi Biosciences Center, School of Pharmacy, University of Kansas, Lawrence, KS
| | - Alexander A Sosunov
- Department of Neurosurgery, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Guy M McKhann
- Department of Neurosurgery, College of Physicians and Surgeons, Columbia University, New York, NY
| | - Haiyang Yu
- State Key Laboratory of Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shirley ShiDu Yan
- Department of Pharmacology & Toxicology and Higuchi Biosciences Center, School of Pharmacy, University of Kansas, Lawrence, KS
| |
Collapse
|
30
|
Givvimani S, Pushpakumar SB, Metreveli N, Veeranki S, Kundu S, Tyagi SC. Role of mitochondrial fission and fusion in cardiomyocyte contractility. Int J Cardiol 2015; 187:325-33. [PMID: 25841124 DOI: 10.1016/j.ijcard.2015.03.352] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2014] [Revised: 03/12/2015] [Accepted: 03/22/2015] [Indexed: 11/28/2022]
Abstract
BACKGROUND Mitochondria constitute 30% of cell volume and are engaged in two dynamic processes called fission and fusion, regulated by Drp-1 (dynamin related protein) and mitofusin 2 (Mfn2). Previously, we showed that Drp-1 inhibition attenuates cardiovascular dysfunction following pressure overload in aortic banding model and myocardial infarction. As dynamic organelles, mitochondria are capable of changing their morphology in response to stress. However, whether such changes can alter their function and in turn cellular function is unknown. Further, a direct role of fission and fusion in cardiomyocyte contractility has not yet been studied. In this study, we hypothesize that disrupted fission and fusion balance by increased Drp-1 and decreased Mfn2 expression in cardiomyocytes affects their contractility through alterations in the calcium and potassium concentrations. METHODS To verify this, we used freshly isolated ventricular myocytes from wild type mouse and transfected them with either siRNA to Drp-1 or Mfn2. Myocyte contractility studies were performed by IonOptix using a myopacer. Intracellular calcium and potassium measurements were done using flow cytometry. Immunocytochemistry (ICC) was done to evaluate live cell mitochondria and its membrane potential. Protein expression was done by western blot and immunocytochemistry. RESULTS We found that silencing mitochondrial fission increased the myocyte contractility, while fusion inhibition decreased contractility with simultaneous changes in calcium and potassium. Also, we observed that increase in fission prompted decrease in Serca-2a and increase in cytochrome c leakage leading to mitophagy. CONCLUSION Our results suggested that regulating mitochondrial fission and fusion have direct effects on overall cardiomyocyte contractility and thus function.
Collapse
Affiliation(s)
- S Givvimani
- Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, KY 40202, United States.
| | - S B Pushpakumar
- Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, KY 40202, United States
| | - N Metreveli
- Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, KY 40202, United States
| | - S Veeranki
- Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, KY 40202, United States
| | - S Kundu
- Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, KY 40202, United States
| | - S C Tyagi
- Department of Physiology and Biophysics, University of Louisville School of Medicine, Louisville, KY 40202, United States
| |
Collapse
|
31
|
Liu JM, Yi Z, Liu SZ, Chang JH, Dang XB, Li QY, Zhang YL. The mitochondrial division inhibitor mdivi-1 attenuates spinal cord ischemia-reperfusion injury both in vitro and in vivo: Involvement of BK channels. Brain Res 2015; 1619:155-65. [PMID: 25818100 DOI: 10.1016/j.brainres.2015.03.033] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2014] [Revised: 03/13/2015] [Accepted: 03/16/2015] [Indexed: 11/29/2022]
Abstract
Mitochondrial division inhibitor (mdivi-1), a selective inhibitor of a mitochondrial fission protein dynamin-related protein 1 (Drp1), has been shown to exert protective effects in heart and cerebral ischemia-reperfusion models. The present study was designed to investigate the beneficial effects of mdivi-1 against spinal cord ischemia-reperfusion (SCIR) injury and its associated mechanisms. SCIR injury was induced by glutamate treatment in cultured spinal cord neurons and by descending thoracic aorta occlusion for 20 min in rats. We found that mdivi-1 (10 μM) significantly attenuated glutamate induced neuronal injury and apoptosis in spinal cord neurons. This neuroprotective effect was accompanied by decreased expression of oxidative stress markers, inhibited mitochondrial dysfunction and preserved activities of antioxidant enzymes. In addition, mdivi-1 significantly increased the expression of the large-conductance Ca(2+)- and voltage-activated K(+) (BK) channels, and blocking BK channels by paxilline partly ablated mdivi-1 induced protection. The in vivo experiments showed that mdivi-1 treatment (1 mg/kg) overtly mitigated SCIR injury induced spinal cord edema and neurological dysfunction with no organ-related toxicity in rats. Moreover, mdivi-1 increased the expression of BK channels in spinal cord tissues, and paxilline pretreatment nullified mdivi-1 induced protection after SCIR injury in rats. Thus, mdivi-1 may be an effective therapeutic agent for SCIR injury via activation of BK channels as well as reduction of oxidative stress, mitochondrial dysfunction and neuronal apoptosis. This article is part of a Special Issue entitled SI: Spinal cord injury.
Collapse
Affiliation(s)
- Jian-Min Liu
- Department of Emergency, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710068, Shaanxi, China
| | - Zhi Yi
- Department of Orthopaedics, Shaanxi Provincial People's Hospital, The Third affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710068, Shaanxi, China
| | - Shi-Zhang Liu
- Department of Orthopaedics, Shaanxi Provincial People's Hospital, The Third affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710068, Shaanxi, China
| | - Jian-Hua Chang
- Department of Anesthesiology, Shaanxi Provincial People's Hospital, The Third affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710068, Shaanxi, China
| | - Xing-Bo Dang
- Department of Emergency, Shaanxi Provincial People's Hospital, The Third Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710068, Shaanxi, China
| | - Quan-Yi Li
- Department of Orthopaedics, Shaanxi Provincial People's Hospital, The Third affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710068, Shaanxi, China
| | - Yue-Lin Zhang
- Department of Neurosurgery, Shaanxi Provincial People's Hospital, The Third affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an 710068, Shaanxi, China.
| |
Collapse
|
32
|
Ikeda Y, Shirakabe A, Brady C, Zablocki D, Ohishi M, Sadoshima J. Molecular mechanisms mediating mitochondrial dynamics and mitophagy and their functional roles in the cardiovascular system. J Mol Cell Cardiol 2015; 78:116-22. [PMID: 25305175 PMCID: PMC4268018 DOI: 10.1016/j.yjmcc.2014.09.019] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/05/2014] [Revised: 09/12/2014] [Accepted: 09/16/2014] [Indexed: 02/05/2023]
Abstract
Mitochondria are essential organelles that produce the cellular energy source, ATP. Dysfunctional mitochondria are involved in the pathophysiology of heart disease, which is associated with reduced levels of ATP and excessive production of reactive oxygen species. Mitochondria are dynamic organelles that change their morphology through fission and fusion in order to maintain their function. Fusion connects neighboring depolarized mitochondria and mixes their contents to maintain membrane potential. In contrast, fission segregates damaged mitochondria from intact ones, where the damaged part of mitochondria is subjected to mitophagy whereas the intact part to fusion. It is generally believed that mitochondrial fusion is beneficial for the heart, especially under stress conditions, because it consolidates the mitochondria's ability to supply energy. However, both excessive fusion and insufficient fission disrupt the mitochondrial quality control mechanism and potentiate cell death. In this review, we discuss the role of mitochondrial dynamics and mitophagy in the heart and the cardiomyocytes therein, with a focus on their roles in cardiovascular disease. This article is part of a Special Issue entitled "Mitochondria: From Basic Mitochondrial Biology to Cardiovascular Disease".
Collapse
Affiliation(s)
- Yoshiyuki Ikeda
- Department of Cell Biology & Molecular Medicine, NJ Medical School, Rutgers University, USA; Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan
| | - Akihiro Shirakabe
- Department of Cell Biology & Molecular Medicine, NJ Medical School, Rutgers University, USA
| | - Christopher Brady
- Department of Cell Biology & Molecular Medicine, NJ Medical School, Rutgers University, USA
| | - Daniela Zablocki
- Department of Cell Biology & Molecular Medicine, NJ Medical School, Rutgers University, USA
| | - Mitsuru Ohishi
- Department of Cardiovascular Medicine and Hypertension, Graduate School of Medical and Dental Science, Kagoshima University, Japan
| | - Junichi Sadoshima
- Department of Cell Biology & Molecular Medicine, NJ Medical School, Rutgers University, USA.
| |
Collapse
|
33
|
Yeon JY, Min SH, Park HJ, Kim JW, Lee YH, Park SY, Jeong PS, Park H, Lee DS, Kim SU, Chang KT, Koo DB. Mdivi-1, mitochondrial fission inhibitor, impairs developmental competence and mitochondrial function of embryos and cells in pigs. J Reprod Dev 2014; 61:81-9. [PMID: 25501014 PMCID: PMC4410306 DOI: 10.1262/jrd.2014-070] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Mitochondria are highly dynamic organelles that undergo constant fusion/fission as well as activities orchestrated by large dynamin-related GTPases. These dynamic mitochondrial processes influence mitochondrial morphology, size and function. Therefore, this study was conducted to evaluate the effects of mitochondrial fission inhibitor, mdivi-1, on developmental competence and mitochondrial function of porcine embryos and primary cells. Presumptive porcine embryos were cultured in PZM-3 medium supplemented with mdivi-1 (0, 10 and 50 μM) for 6 days. Porcine fibroblast cells were cultured in growth medium with mdivi-1 (0 and 50 μM) for 2 days. Our results showed that the rate of blastocyst production and cell growth in the mdivi-1 (50 μM) treated group was lower than that of the control group (P < 0.05). Moreover, loss of mitochondrial membrane potential in the mdivi-1 (50 μM) treated group was increased relative to the control group (P < 0.05). Subsequent evaluation
revealed that the intracellular levels of reactive oxygen species (ROS) and the apoptotic index were increased by mdivi-1 (50 μM) treatment (P < 0.05). Finally, the expression of mitochondrial fission-related protein (Drp 1) was lower in the embryos and cells in the mdivi-1-treated group than the control group. Taken together, these results indicate that mdivi-1 treatment may inhibit developmental competence and mitochondrial function in porcine embryos and primary cells.
Collapse
Affiliation(s)
- Ji-Yeong Yeon
- Department of Biotechnology, College of Engineering, Daegu University, Gyeongbuk 712-714, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
So EC, Wu KC, Kao FC, Wu SN. Effects of midazolam on ion currents and membrane potential in differentiated motor neuron-like NSC-34 and NG108-15 cells. Eur J Pharmacol 2014; 724:152-160. [PMID: 24374009 DOI: 10.1016/j.ejphar.2013.12.034] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 11/22/2013] [Accepted: 12/16/2013] [Indexed: 11/25/2022]
Abstract
Midazolam (MDL) was known to act through stimulation of benzodiazepine receptors (GABA). Whether midazolam affects ion currents and membrane potential in neurons remains largely unclear. Electrophysiological studies of midazolam actions were performed in differentiated motor neuron-like (NSC-34 and NG108-15) cells. Midazolam suppressed the amplitude of delayed rectifier K(+) current (IK(DR)) in a time- and concentration-dependent manner with an IC50 value of 10.4 µM. Addition of midazolam was noted to enhance the rate of IK(DR) inactivation. On the basis of minimal binding scheme, midazolam-induced block of IK(DR) was quantitatively provided with a dissociation constant of 9.8 µM. Recovery of IK(DR) from inactivation in the presence of midazolam was fitted by a single exponential. midazolam had no effect on M-type or erg-mediated K(+) current in these cells. Midazaolam (30 µM) suppressed the peak amplitude of voltage-gated Na(+) current (INa) with no change in the current-voltage relationships of this current. Inactivation kinetics of INa remained unaltered in the presence of this agent. In current-clamp configuration, midazolam (30 µM) prolonged the duration of action potentials (APs) and reduce AP amplitude. Similarly, in differentiated NG108-15 cells, the exposure to midazolam also suppressed IK(DR) with a concomitant increase in current inactivation. Midazolam can act as an open-channel blocker of delayed-rectifier K(+) channels in these cells. The synergistic blocking effects on IK(DR) and INa may contribute to the underlying mechanisms through which midazolam affects neuronal function in vivo.
Collapse
Affiliation(s)
- Edmund Cheung So
- Department of Anesthesia, Tainan Municipal An-Nan Hospital, China Medical University, 66# Cheung Ho Road, An-Nan District, Tainan, Taiwan; Department of Anesthesia, China Medical University, 91# Hsueh-Shih Road, Taichung, Taiwan; Department of Cell Biology and Anatomy, Cheng Kung University, 1# University Road, Tainan City, Taiwan; Department of Anesthesia, Taishan Medical University, 619 Chang Cheng Road, Taian 271016, Shandong Province, PR China; Department of Anesthesia, Nan Shan branch of Gilu Hospital, Shandong University, Shandong Province, PR China.
| | - King Chuen Wu
- Department of Anesthesiology, Eda-Hospital/I-Shou University, 1# Yida Road, Jiaosu Village, Yanchao District, Kaohsiung, Taiwan.
| | - Feng Chen Kao
- Department of Orthopedic Surgery, E-Da Hospital/I-Shou University, 1# Yida Road, Jiaosu Village, Yanchao District, Kaohsiung, Taiwan.
| | - Sheng Nan Wu
- Department of Physiology, National Cheng Kung University, 1# University Road, Tainan City, Taiwan.
| |
Collapse
|
35
|
Attenuation of doxorubicin-induced cardiotoxicity by mdivi-1: a mitochondrial division/mitophagy inhibitor. PLoS One 2013; 8:e77713. [PMID: 24147064 PMCID: PMC3798380 DOI: 10.1371/journal.pone.0077713] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2013] [Accepted: 09/12/2013] [Indexed: 11/19/2022] Open
Abstract
Doxorubicin is one of the most effective anti-cancer agents. However, its use is associated with adverse cardiac effects, including cardiomyopathy and progressive heart failure. Given the multiple beneficial effects of the mitochondrial division inhibitor (mdivi-1) in a variety of pathological conditions including heart failure and ischaemia and reperfusion injury, we investigated the effects of mdivi-1 on doxorubicin-induced cardiac dysfunction in naïve and stressed conditions using Langendorff perfused heart models and a model of oxidative stress was used to assess the effects of drug treatments on the mitochondrial depolarisation and hypercontracture of cardiac myocytes. Western blot analysis was used to measure the levels of p-Akt and p-Erk 1/2 and flow cytometry analysis was used to measure the levels p-Drp1 and p-p53 upon drug treatment. The HL60 leukaemia cell line was used to evaluate the effects of pharmacological inhibition of mitochondrial division on the cytotoxicity of doxorubicin in a cancer cell line. Doxorubicin caused a significant impairment of cardiac function and increased the infarct size to risk ratio in both naïve conditions and during ischaemia/reperfusion injury. Interestingly, co-treatment of doxorubicin with mdivi-1 attenuated these detrimental effects of doxorubicin. Doxorubicin also caused a reduction in the time taken to depolarisation and hypercontracture of cardiac myocytes, which were reversed with mdivi-1. Finally, doxorubicin caused a significant elevation in the levels of signalling proteins p-Akt, p-Erk 1/2, p-Drp1 and p-p53. Co-incubation of mdivi-1 with doxorubicin did not reduce the cytotoxicity of doxorubicin against HL-60 cells. These data suggest that the inhibition of mitochondrial fission protects the heart against doxorubicin-induced cardiac injury and identify mitochondrial fission as a new therapeutic target in ameliorating doxorubicin-induced cardiotoxicity without affecting its anti-cancer properties.
Collapse
|
36
|
Disatnik MH, Ferreira JCB, Campos JC, Gomes KS, Dourado PMM, Qi X, Mochly-Rosen D. Acute inhibition of excessive mitochondrial fission after myocardial infarction prevents long-term cardiac dysfunction. J Am Heart Assoc 2013; 2:e000461. [PMID: 24103571 PMCID: PMC3835263 DOI: 10.1161/jaha.113.000461] [Citation(s) in RCA: 264] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Background Ischemia and reperfusion (IR) injury remains a major cause of morbidity and mortality and multiple molecular and cellular pathways have been implicated in this injury. We determined whether acute inhibition of excessive mitochondrial fission at the onset of reperfusion improves mitochondrial dysfunction and cardiac contractility postmyocardial infarction in rats. Methods and Results We used a selective inhibitor of the fission machinery, P110, which we have recently designed. P110 treatment inhibited the interaction of fission proteins Fis1/Drp1, decreased mitochondrial fission, and improved bioenergetics in three different rat models of IR, including primary cardiomyocytes, ex vivo heart model, and an in vivo myocardial infarction model. Drp1 transiently bound to the mitochondria following IR injury and P110 treatment blocked this Drp1 mitochondrial association. Compared with control treatment, P110 (1 μmol/L) decreased infarct size by 28±2% and increased adenosine triphosphate levels by 70+1% after IR relative to control IR in the ex vivo model. Intraperitoneal injection of P110 (0.5 mg/kg) at the onset of reperfusion in an in vivo model resulted in improved mitochondrial oxygen consumption by 68% when measured 3 weeks after ischemic injury, improved cardiac fractional shortening by 35%, reduced mitochondrial H2O2 uncoupling state by 70%, and improved overall mitochondrial functions. Conclusions Together, we show that excessive mitochondrial fission at reperfusion contributes to long‐term cardiac dysfunction in rats and that acute inhibition of excessive mitochondrial fission at the onset of reperfusion is sufficient to result in long‐term benefits as evidenced by inhibiting cardiac dysfunction 3 weeks after acute myocardial infarction.
Collapse
Affiliation(s)
- Marie-Hélène Disatnik
- Department of Chemical and Systems Biology, Stanford University School of Medicine, Stanford, 94305, CA
| | | | | | | | | | | | | |
Collapse
|
37
|
Qian W, Wang J, Van Houten B. The role of dynamin-related protein 1 in cancer growth: a promising therapeutic target? Expert Opin Ther Targets 2013; 17:997-1001. [PMID: 23888838 DOI: 10.1517/14728222.2013.823160] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Mitochondrial functions are altered in many human diseases including cancer. Development of mitochondria-targeted therapies, either through restoring normal mitochondrial function or promoting mitochondrial-induced cell death, is one of the attractive strategies to improve the outcome of cancer treatment. Recent advances have revealed the important functional involvement of mitochondrial dynamics in cancer biology. Dynamin-related protein 1 (Drp1), a member of the dynamin family of GTPases required for mitochondrial fission, has been found upregulated in certain types of cancers, such as lung and breast cancers. In addition, the roles of Drp1 in cell cycle progression, genome instability, cell migration and apoptosis in cancer cells have also been recently uncovered. These findings raise the possibility of targeting Drp1-mediated mitochondrial fission as an effective therapy for treating cancer. This article explores the function of Drp1 in cancer cells and discusses the theoretical basis for the development of potential targeted therapy.
Collapse
|
38
|
Chalmers S, Saunter C, Wilson C, Coats P, Girkin JM, McCarron JG. Mitochondrial motility and vascular smooth muscle proliferation. Arterioscler Thromb Vasc Biol 2012; 32:3000-11. [PMID: 23104850 DOI: 10.1161/atvbaha.112.255174] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
OBJECTIVE Mitochondria are widely described as being highly dynamic and adaptable organelles, and their movement is thought to be vital for cell function. Yet, in various native cells, including those of heart and smooth muscle, mitochondria are stationary and rigidly structured. The significance of the differences in mitochondrial behavior to the physiological function of cells is unclear and was studied in single myocytes and intact resistance-sized cerebral arteries. We hypothesized that mitochondrial dynamics is controlled by the proliferative status of the cells. METHODS AND RESULTS High-speed fluorescence imaging of mitochondria in live vascular smooth muscle cells shows that the organelle undergoes significant reorganization as cells become proliferative. In nonproliferative cells, mitochondria are individual (≈ 2 μm by 0.5 μm), stationary, randomly dispersed, fixed structures. However, on entering the proliferative state, mitochondria take on a more diverse architecture and become small spheres, short rod-shaped structures, long filamentous entities, and networks. When cells proliferate, mitochondria also continuously move and change shape. In the intact pressurized resistance artery, mitochondria are largely immobile structures, except in a small number of cells in which motility occurred. When proliferation of smooth muscle was encouraged in the intact resistance artery, in organ culture, the majority of mitochondria became motile and the majority of smooth muscle cells contained moving mitochondria. Significantly, restriction of mitochondrial motility using the fission blocker mitochondrial division inhibitor prevented vascular smooth muscle proliferation in both single cells and the intact resistance artery. CONCLUSIONS These results show that mitochondria are adaptable and exist in intact tissue as both stationary and highly dynamic entities. This mitochondrial plasticity is an essential mechanism for the development of smooth muscle proliferation and therefore presents a novel therapeutic target against vascular disease.
Collapse
Affiliation(s)
- Susan Chalmers
- Strathclyde Institute of Pharmacy and Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| | | | | | | | | | | |
Collapse
|
39
|
Wondergem R, Graves BM, Li C, Williams DL. Lipopolysaccharide prolongs action potential duration in HL-1 mouse cardiomyocytes. Am J Physiol Cell Physiol 2012; 303:C825-33. [PMID: 22895260 DOI: 10.1152/ajpcell.00173.2012] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Sepsis has deleterious effects on cardiac function including reduced contractility. We have shown previously that lipopolysaccharides (LPS) directly affect HL-1 cardiac myocytes by inhibiting Ca(2+) regulation and by impairing pacemaker "funny" current, I(f). We now explore further cellular mechanisms whereby LPS inhibits excitability in HL-1 cells. LPS (1 μg/ml) derived from Salmonella enteritidis decreased rate of firing of spontaneous action potentials in HL-1 cells, and it increased their pacemaker potential durations and decreased their rates of depolarization, all measured by whole cell current clamp. LPS also increased action potential durations and decreased their amplitude in cells paced at 1 Hz with 0.1 nA, and 20 min were necessary for maximal effect. LPS decreased the amplitude of a rapidly inactivating inward current attributed to Na(+) and of an outward current attributed to K(+); both were measured by whole cell voltage clamp. The K(+) currents displayed a resurgent outward tail current, which is characteristic of the rapid delayed-rectifier K(+) current, I(Kr). LPS accordingly reduced outward currents measured with pipette Cs(+) substituted for K(+) to isolate I(Kr). E-4031 (1 μM) markedly inhibited I(Kr) in HL-1 cells and also increased action potential duration; however, the direct effects of E-4031 occurred minutes faster than the slow effects of LPS. We conclude that LPS increases action potential duration in HL-1 mouse cardiomyocytes by inhibition of I(Kr) and decreases their rate of firing by inhibition of I(Na.) This protracted time course points toward an intermediary metabolic event, which either decreases available mouse ether-a-go-go (mERG) and Na(+) channels or potentiates their inactivation.
Collapse
Affiliation(s)
- Robert Wondergem
- Department of Biomedical Science, James H. Quillen College of Medicine, East Tennessee State University, Johnson City, Tennessee 37614-1708, USA.
| | | | | | | |
Collapse
|