1
|
Zhang R, Yang H, Guo M, Niu S, Xue Y. Mitophagy and its regulatory mechanisms in the biological effects of nanomaterials. J Appl Toxicol 2024; 44:1834-1853. [PMID: 38642013 DOI: 10.1002/jat.4609] [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: 02/18/2024] [Revised: 03/13/2024] [Accepted: 03/22/2024] [Indexed: 04/22/2024]
Abstract
Mitophagy is a selective cellular process critical for the removal of damaged mitochondria. It is essential in regulating mitochondrial number, ensuring mitochondrial functionality, and maintaining cellular equilibrium, ultimately influencing cell destiny. Numerous pathologies, such as neurodegenerative diseases, cardiovascular disorders, cancers, and various other conditions, are associated with mitochondrial dysfunctions. Thus, a detailed exploration of the regulatory mechanisms of mitophagy is pivotal for enhancing our understanding and for the discovery of novel preventive and therapeutic options for these diseases. Nanomaterials have become integral in biomedicine and various other sectors, offering advanced solutions for medical uses including biological imaging, drug delivery, and disease diagnostics and therapy. Mitophagy is vital in managing the cellular effects elicited by nanomaterials. This review provides a comprehensive analysis of the molecular mechanisms underpinning mitophagy, underscoring its significant influence on the biological responses of cells to nanomaterials. Nanoparticles can initiate mitophagy via various pathways, among which the PINK1-Parkin pathway is critical for cellular defense against nanomaterial-induced damage by promoting mitophagy. The role of mitophagy in biological effects was induced by nanomaterials, which are associated with alterations in Ca2+ levels, the production of reactive oxygen species, endoplasmic reticulum stress, and lysosomal damage.
Collapse
Affiliation(s)
- Rui Zhang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, People's Republic of China
| | - Haitao Yang
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, People's Republic of China
| | - Menghao Guo
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, People's Republic of China
| | - Shuyan Niu
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, People's Republic of China
| | - Yuying Xue
- Key Laboratory of Environmental Medicine and Engineering, Ministry of Education, School of Public Health, Southeast University, Nanjing, People's Republic of China
| |
Collapse
|
2
|
Marzook H, Gupta A, Jayakumar MN, Saleh MA, Tomar D, Qaisar R, Ahmad F. GSK-3α-BNIP3 axis promotes mitophagy in human cardiomyocytes under hypoxia. Free Radic Biol Med 2024; 221:235-244. [PMID: 38815772 DOI: 10.1016/j.freeradbiomed.2024.05.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 05/24/2024] [Accepted: 05/25/2024] [Indexed: 06/01/2024]
Abstract
Dysregulated autophagy/mitophagy is one of the major causes of cardiac injury in ischemic conditions. Glycogen synthase kinase-3alpha (GSK-3α) has been shown to play a crucial role in the pathophysiology of cardiac diseases. However, the precise role of GSK-3α in cardiac mitophagy remains unknown. Herein, we investigated the role of GSK-3α in cardiac mitophagy by employing AC16 human cardiomyocytes under the condition of acute hypoxia. We observed that the gain-of-GSK-3α function profoundly induced mitophagy in the AC16 cardiomyocytes post-hypoxia. Moreover, GSK-3α overexpression led to increased ROS generation and mitochondrial dysfunction in cardiomyocytes, accompanied by enhanced mitophagy displayed by increased mt-mKeima intensity under hypoxia. Mechanistically, we identified that GSK-3α promotes mitophagy through upregulation of BNIP3, caused by GSK-3α-mediated increase in expression of HIF-1α and FOXO3a in cardiomyocytes post-hypoxia. Moreover, GSK-3α displayed a physical interaction with BNIP3 and, inhibited PINK1 and Parkin recruitment to mitochondria was observed specifically under hypoxia. Taken together, we identified a novel mechanism of mitophagy in human cardiomyocytes. GSK-3α promotes mitochondrial dysfunction and regulates FOXO3a -mediated BNIP3 overexpression in cardiomyocytes to facilitate mitophagy following hypoxia. An interaction between GSK-3α and BNIP3 suggests a role of GSK-3α in BNIP3 recruitment to the mitochondrial membrane where it enhances mitophagy in stressed cardiomyocytes independent of the PINK1/Parkin.
Collapse
Affiliation(s)
- Hezlin Marzook
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Anamika Gupta
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Manju N Jayakumar
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Mohamed A Saleh
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates; Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Mansoura University, Mansoura, 35516, Egypt
| | - Dhanendra Tomar
- Department of Internal Medicine, Section on Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Rizwan Qaisar
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates; Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates
| | - Firdos Ahmad
- Research Institute for Medical and Health Sciences, University of Sharjah, Sharjah, 27272, United Arab Emirates; Department of Basic Medical Sciences, College of Medicine, University of Sharjah, Sharjah, 27272, United Arab Emirates.
| |
Collapse
|
3
|
Forte M, D'Ambrosio L, Schiattarella GG, Salerno N, Perrone MA, Loffredo FS, Bertero E, Pilichou K, Manno G, Valenti V, Spadafora L, Bernardi M, Simeone B, Sarto G, Frati G, Perrino C, Sciarretta S. Mitophagy modulation for the treatment of cardiovascular diseases. Eur J Clin Invest 2024; 54:e14199. [PMID: 38530070 DOI: 10.1111/eci.14199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 03/15/2024] [Accepted: 03/16/2024] [Indexed: 03/27/2024]
Abstract
BACKGROUND Defects of mitophagy, the selective form of autophagy for mitochondria, are commonly observed in several cardiovascular diseases and represent the main cause of mitochondrial dysfunction. For this reason, mitophagy has emerged as a novel and potential therapeutic target. METHODS In this review, we discuss current evidence about the biological significance of mitophagy in relevant preclinical models of cardiac and vascular diseases, such as heart failure, ischemia/reperfusion injury, metabolic cardiomyopathy and atherosclerosis. RESULTS Multiple studies have shown that cardiac and vascular mitophagy is an adaptive mechanism in response to stress, contributing to cardiovascular homeostasis. Mitophagy defects lead to cell death, ultimately impairing cardiac and vascular function, whereas restoration of mitophagy by specific compounds delays disease progression. CONCLUSIONS Despite previous efforts, the molecular mechanisms underlying mitophagy activation in response to stress are not fully characterized. A comprehensive understanding of different forms of mitophagy active in the cardiovascular system is extremely important for the development of new drugs targeting this process. Human studies evaluating mitophagy abnormalities in patients at high cardiovascular risk also represent a future challenge.
Collapse
Affiliation(s)
| | - Luca D'Ambrosio
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Gabriele G Schiattarella
- Max Rubner Center for Cardiovascular Metabolic Renal Research, Charité-Universitätsmedizin Berlin, Berlin, Germany
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
- DZHK (German Centre for Cardiovascular Research), Partner Site Berlin, Berlin, Germany
| | - Nadia Salerno
- Division of Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, Catanzaro, Italy
| | - Marco Alfonso Perrone
- Division of Cardiology and CardioLab, Department of Clinical Sciences and Translational Medicine, University of Rome Tor Vergata, Rome, Italy
- Clinical Pathways and Epidemiology Unit, Bambino Gesù Children's Hospital IRCCS, Rome, Italy
| | - Francesco S Loffredo
- Division of Cardiology, Department of Translational Medical Sciences, University of Campania "L. Vanvitelli", Naples, Italy
| | - Edoardo Bertero
- Department of Internal Medicine, University of Genova, Genoa, Italy
- Cardiovascular Disease Unit, IRCCS Ospedale Policlinico San Martino-Italian IRCCS Cardiology Network, Genoa, Italy
| | - Kalliopi Pilichou
- Department of Cardiac-Thoracic-Vascular Sciences and Public Health, University of Padova, Padova, Italy
| | - Girolamo Manno
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties (PROMISE) "G. D'Alessandro", University of Palermo, Palermo, Italy
| | - Valentina Valenti
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
- ICOT Istituto Marco Pasquali, Latina, Italy
| | | | - Marco Bernardi
- Department of Clinical, Internal Medicine, Anesthesiology and Cardiovascular Sciences, Sapienza University, Rome, Italy
| | | | | | - Giacomo Frati
- IRCCS Neuromed, Pozzilli, Italy
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| | - Cinzia Perrino
- Division of Cardiology, Department of Advanced Biomedical Sciences, Federico II University of Naples, Naples, Italy
| | - Sebastiano Sciarretta
- IRCCS Neuromed, Pozzilli, Italy
- Department of Medical-Surgical Sciences and Biotechnologies, Sapienza University of Rome, Latina, Italy
| |
Collapse
|
4
|
Watzlawik JO, Fiesel FC, Fiorino G, Bustillos BA, Baninameh Z, Markham BN, Hou X, Hayes CS, Bredenberg JM, Kurchaba NW, Fričová D, Siuda J, Wszolek ZK, Noda S, Sato S, Hattori N, Prasad AA, Kirik D, Fox HS, Stauch KL, Goldberg MS, Springer W. Basal activity of PINK1 and PRKN in cell models and rodent brain. Autophagy 2024; 20:1147-1158. [PMID: 38041584 PMCID: PMC11135862 DOI: 10.1080/15548627.2023.2286414] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 11/08/2023] [Accepted: 11/15/2023] [Indexed: 12/03/2023] Open
Abstract
The ubiquitin kinase-ligase pair PINK1-PRKN recognizes and transiently labels damaged mitochondria with ubiquitin phosphorylated at Ser65 (p-S65-Ub) to mediate their selective degradation (mitophagy). Complete loss of PINK1 or PRKN function unequivocally leads to early-onset Parkinson disease, but it is debated whether impairments in mitophagy contribute to disease later in life. While the pathway has been extensively studied in cell culture upon acute and massive mitochondrial stress, basal levels of activation under endogenous conditions and especially in vivo in the brain remain undetermined. Using rodent samples, patient-derived cells, and isogenic neurons, we here identified age-dependent, brain region-, and cell type-specific effects and determined expression levels and extent of basal and maximal activation of PINK1 and PRKN. Our work highlights the importance of defining critical risk and therapeutically relevant levels of PINK1-PRKN signaling which will further improve diagnosis and prognosis and will lead to better stratification of patients for future clinical trials.
Collapse
Affiliation(s)
| | - Fabienne C. Fiesel
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Neuroscience PhD Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA
| | - Gabriella Fiorino
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Neuroscience PhD Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA
| | | | - Zahra Baninameh
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | - Xu Hou
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | - Caleb S. Hayes
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
| | | | | | | | - Joanna Siuda
- Department of Neurology, Faculty of Medical Sciences in Katowice, Medical University of Silesia, Katowice, Poland
| | | | - Sachiko Noda
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Shigeto Sato
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Nobutaka Hattori
- Department of Neurology, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Asheeta A. Prasad
- Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
| | - Deniz Kirik
- Faculty of Medicine and Health, School of Medical Sciences, University of Sydney, Sydney, NSW, Australia
- Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - Howard S. Fox
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Kelly L. Stauch
- Department of Neurological Sciences, University of Nebraska Medical Center, Omaha, NE, USA
| | - Matthew S. Goldberg
- Center for Neurodegeneration and Experimental Therapeutics, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Wolfdieter Springer
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, USA
- Neuroscience PhD Program, Mayo Clinic Graduate School of Biomedical Sciences, Jacksonville, FL, USA
| |
Collapse
|
5
|
Marchesan E, Nardin A, Mauri S, Bernardo G, Chander V, Di Paola S, Chinellato M, von Stockum S, Chakraborty J, Herkenne S, Basso V, Schrepfer E, Marin O, Cendron L, Medina DL, Scorrano L, Ziviani E. Activation of Ca 2+ phosphatase Calcineurin regulates Parkin translocation to mitochondria and mitophagy in flies. Cell Death Differ 2024; 31:217-238. [PMID: 38238520 PMCID: PMC10850161 DOI: 10.1038/s41418-023-01251-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 11/23/2023] [Accepted: 12/05/2023] [Indexed: 02/09/2024] Open
Abstract
Selective removal of dysfunctional mitochondria via autophagy is crucial for the maintenance of cellular homeostasis. This event is initiated by the translocation of the E3 ubiquitin ligase Parkin to damaged mitochondria, and it requires the Serine/Threonine-protein kinase PINK1. In a coordinated set of events, PINK1 operates upstream of Parkin in a linear pathway that leads to the phosphorylation of Parkin, Ubiquitin, and Parkin mitochondrial substrates, to promote ubiquitination of outer mitochondrial membrane proteins. Ubiquitin-decorated mitochondria are selectively recruiting autophagy receptors, which are required to terminate the organelle via autophagy. In this work, we show a previously uncharacterized molecular pathway that correlates the activation of the Ca2+-dependent phosphatase Calcineurin to Parkin translocation and Parkin-dependent mitophagy. Calcineurin downregulation or genetic inhibition prevents Parkin translocation to CCCP-treated mitochondria and impairs stress-induced mitophagy, whereas Calcineurin activation promotes Parkin mitochondrial recruitment and basal mitophagy. Calcineurin interacts with Parkin, and promotes Parkin translocation in the absence of PINK1, but requires PINK1 expression to execute mitophagy in MEF cells. Genetic activation of Calcineurin in vivo boosts basal mitophagy in neurons and corrects locomotor dysfunction and mitochondrial respiratory defects of a Drosophila model of impaired mitochondrial functions. Our study identifies Calcineurin as a novel key player in the regulation of Parkin translocation and mitophagy.
Collapse
Affiliation(s)
| | - Alice Nardin
- Department of Biology, University of Padova, Padova, Italy
| | - Sofia Mauri
- Department of Biology, University of Padova, Padova, Italy
| | - Greta Bernardo
- Department of Biology, University of Padova, Padova, Italy
| | - Vivek Chander
- Department of Biology, University of Padova, Padova, Italy
| | - Simone Di Paola
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy
- Institute for Experimental Endocrinology and Oncology (IEOS), National Research Council (CNR), Napoli, Italy
| | | | | | | | | | | | - Emilie Schrepfer
- Department of Biology, University of Padova, Padova, Italy
- Dulbecco-Telethon Institute, Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Oriano Marin
- Department of Biomedical Sciences (DSB), University of Padova, Padova, Italy
| | - Laura Cendron
- Department of Biology, University of Padova, Padova, Italy
| | - Diego L Medina
- Telethon Institute of Genetics and Medicine (TIGEM), Pozzuoli, Naples, Italy
- Medical Genetics Unit, Department of Medical and Translational Science, Federico II University, Naples, Italy
| | - Luca Scorrano
- Department of Biology, University of Padova, Padova, Italy
- Dulbecco-Telethon Institute, Venetian Institute of Molecular Medicine (VIMM), Padova, Italy
| | - Elena Ziviani
- Department of Biology, University of Padova, Padova, Italy.
| |
Collapse
|
6
|
Jin U, Park SJ, Lee BG, Kim JB, Kim SJ, Joe EH, Woo HG, Park SM. Critical roles of parkin and PINK1 in coxsackievirus B3-induced viral myocarditis. Microbes Infect 2023; 25:105211. [PMID: 37574181 DOI: 10.1016/j.micinf.2023.105211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 08/03/2023] [Accepted: 08/08/2023] [Indexed: 08/15/2023]
Abstract
Viral myocarditis is an inflammatory disease of the myocardium, often leads to cardiac dysfunction and death. PARKIN (PRKN) and PINK1, well known as Parkinson's disease-associated genes, have been reported to be involved in innate immunity and mitochondrial damage control. Therefore, we investigated the role of parkin and PINK1 in coxsackievirus B3 (CVB3)-induced viral myocarditis because the etiology of myocarditis is related to abnormal immune response to viral infection and mitochondrial damage. After viral infection, the survival was significantly lower and myocardial damage was more severe in parkin knockout (KO) and PINK1 KO mice compared to wild-type (WT) mice. Parkin KO and PINK1 KO showed defective immune cell recruitment and impaired production of antiviral cytokines such as interferon-gamma, allowing increased viral replication. In addition, parkin KO and PINK1 KO mice were more susceptible to CVB3-induced mitochondrial damage than WT mice, resulting in susceptibility to viral-induced cardiac damage. Finally, using publicly available RNA-seq data, we found that pathogenic mutants of the PRKN gene are more common in patients with dilated cardiomyopathy and myocarditis than in controls or the general population. This study will help elucidate the molecular mechanism of CVB3-induced viral myocarditis.
Collapse
Affiliation(s)
- Uram Jin
- Department of Pharmacology, Ajou University School of Medicine, Suwon, South Korea; Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, South Korea; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, South Korea; Department of Cardiology, Ajou University School of Medicine, Suwon, South Korea
| | - Soo Jin Park
- Department of Pharmacology, Ajou University School of Medicine, Suwon, South Korea; Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, South Korea; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, South Korea; Department of Thoracic and Cardiovascular Surgery, Ajou University School of Medicine, Suwon, South Korea
| | - Byoung Gil Lee
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, South Korea; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, South Korea; Department of Physiology, Ajou University School of Medicine, Suwon, South Korea
| | - Jae-Bong Kim
- Department of Pharmacology, Ajou University School of Medicine, Suwon, South Korea; Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, South Korea; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, South Korea
| | - Soo Jeong Kim
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, South Korea
| | - Eun-Hye Joe
- Department of Pharmacology, Ajou University School of Medicine, Suwon, South Korea; Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, South Korea; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, South Korea
| | - Hyun Goo Woo
- Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, South Korea; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, South Korea; Department of Physiology, Ajou University School of Medicine, Suwon, South Korea
| | - Sang Myun Park
- Department of Pharmacology, Ajou University School of Medicine, Suwon, South Korea; Center for Convergence Research of Neurological Disorders, Ajou University School of Medicine, Suwon, South Korea; Department of Biomedical Sciences, Ajou University School of Medicine, Suwon, South Korea.
| |
Collapse
|
7
|
Titus AS, Sung EA, Zablocki D, Sadoshima J. Mitophagy for cardioprotection. Basic Res Cardiol 2023; 118:42. [PMID: 37798455 PMCID: PMC10556134 DOI: 10.1007/s00395-023-01009-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 09/13/2023] [Accepted: 09/14/2023] [Indexed: 10/07/2023]
Abstract
Mitochondrial function is maintained by several strictly coordinated mechanisms, collectively termed mitochondrial quality control mechanisms, including fusion and fission, degradation, and biogenesis. As the primary source of energy in cardiomyocytes, mitochondria are the central organelle for maintaining cardiac function. Since adult cardiomyocytes in humans rarely divide, the number of dysfunctional mitochondria cannot easily be diluted through cell division. Thus, efficient degradation of dysfunctional mitochondria is crucial to maintaining cellular function. Mitophagy, a mitochondria specific form of autophagy, is a major mechanism by which damaged or unnecessary mitochondria are targeted and eliminated. Mitophagy is active in cardiomyocytes at baseline and in response to stress, and plays an essential role in maintaining the quality of mitochondria in cardiomyocytes. Mitophagy is mediated through multiple mechanisms in the heart, and each of these mechanisms can partially compensate for the loss of another mechanism. However, insufficient levels of mitophagy eventually lead to mitochondrial dysfunction and the development of heart failure. In this review, we discuss the molecular mechanisms of mitophagy in the heart and the role of mitophagy in cardiac pathophysiology, with the focus on recent findings in the field.
Collapse
Affiliation(s)
- Allen Sam Titus
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, MSB G-609, Newark, NJ, 07103, USA
| | - Eun-Ah Sung
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, MSB G-609, Newark, NJ, 07103, USA
| | - Daniela Zablocki
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, MSB G-609, Newark, NJ, 07103, USA
| | - Junichi Sadoshima
- Department of Cell Biology and Molecular Medicine, Cardiovascular Research Institute, Rutgers New Jersey Medical School, 185 South Orange Ave, MSB G-609, Newark, NJ, 07103, USA.
| |
Collapse
|
8
|
Liang W, Sagar S, Ravindran R, Najor RH, Quiles JM, Chi L, Diao RY, Woodall BP, Leon LJ, Zumaya E, Duran J, Cauvi DM, De Maio A, Adler ED, Gustafsson ÅB. Mitochondria are secreted in extracellular vesicles when lysosomal function is impaired. Nat Commun 2023; 14:5031. [PMID: 37596294 PMCID: PMC10439183 DOI: 10.1038/s41467-023-40680-5] [Citation(s) in RCA: 46] [Impact Index Per Article: 46.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Accepted: 08/04/2023] [Indexed: 08/20/2023] Open
Abstract
Mitochondrial quality control is critical for cardiac homeostasis as these organelles are responsible for generating most of the energy needed to sustain contraction. Dysfunctional mitochondria are normally degraded via intracellular degradation pathways that converge on the lysosome. Here, we identified an alternative mechanism to eliminate mitochondria when lysosomal function is compromised. We show that lysosomal inhibition leads to increased secretion of mitochondria in large extracellular vesicles (EVs). The EVs are produced in multivesicular bodies, and their release is independent of autophagy. Deletion of the small GTPase Rab7 in cells or adult mouse heart leads to increased secretion of EVs containing ubiquitinated cargos, including intact mitochondria. The secreted EVs are captured by macrophages without activating inflammation. Hearts from aged mice or Danon disease patients have increased levels of secreted EVs containing mitochondria indicating activation of vesicular release during cardiac pathophysiology. Overall, these findings establish that mitochondria are eliminated in large EVs through the endosomal pathway when lysosomal degradation is inhibited.
Collapse
Affiliation(s)
- Wenjing Liang
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Shakti Sagar
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Rishith Ravindran
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Rita H Najor
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Justin M Quiles
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Liguo Chi
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Rachel Y Diao
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Benjamin P Woodall
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Leonardo J Leon
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Erika Zumaya
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA
| | - Jason Duran
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - David M Cauvi
- Department of Surgery, University of California San Diego, La Jolla, CA, USA
| | - Antonio De Maio
- Department of Surgery, University of California San Diego, La Jolla, CA, USA
| | - Eric D Adler
- Department of Medicine, University of California San Diego, La Jolla, CA, USA
| | - Åsa B Gustafsson
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, La Jolla, CA, USA.
- Department of Pharmacology, University of California San Diego, La Jolla, CA, USA.
| |
Collapse
|
9
|
Jankauskas SS, Mone P, Avvisato R, Varzideh F, De Gennaro S, Salemme L, Macina G, Kansakar U, Cioppa A, Frullone S, Gambardella J, Di Mauro M, Tesorio T, Santulli G. miR-181c targets Parkin and SMAD7 in human cardiac fibroblasts: Validation of differential microRNA expression in patients with diabetes and heart failure with preserved ejection fraction. Mech Ageing Dev 2023; 212:111818. [PMID: 37116731 DOI: 10.1016/j.mad.2023.111818] [Citation(s) in RCA: 19] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 04/15/2023] [Accepted: 04/19/2023] [Indexed: 04/30/2023]
Abstract
BACKGROUND Cardiac fibrosis represents a key feature in the pathophysiology of heart failure with preserved ejection fraction (HFpEF), a condition highly prevalent amongst geriatric patients, especially if diabetic. The microRNA miR-181c has been shown to be associated with the response to exercise training in HFpEF patients and has been also linked to diabetic cardiovascular complications. However, the underlying mechanisms have not been fully elucidated. OBJECTIVE To measure circulating miR-181c in elderly patients with HFpEF and DM and identify gene targets pathophysiologically relevant in HFpEF. METHODS We quantified circulating miR-181c in frail older adults with a confirmed diagnosis of HFpEF and diabetes, and, as control, we enrolled age-matched subjects without HFpEF and without diabetes. We validated in human cardiac fibroblasts the molecular mechanisms linking miR-181c to a pro-fibrotic response. RESULTS 51 frail patients were included (34 patients with diabetes and HFpEF and 17 age-matched controls. We observed that miR-181c was significantly upregulated (p<0.0001) in HFpEF patients vs controls. We confirmed in vitro that miR-181c is targeting PRKN and SMAD7. CONCLUSIONS We demonstrate that miR-181c levels are significantly increased in frail elderly adults with diabetes and HFpEF and that miR-181c targets PRKN and SMAD7 in human cardiac fibroblasts.
Collapse
Affiliation(s)
- Stanislovas S Jankauskas
- Department of Medicine, Einstein Institute for Aging Research, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Pasquale Mone
- Department of Medicine, Einstein Institute for Aging Research, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA; ASL Avellino, Avellino, 83100, Italy
| | - Roberta Avvisato
- Department of Medicine, Einstein Institute for Aging Research, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Fahimeh Varzideh
- Department of Medicine, Einstein Institute for Aging Research, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA
| | | | - Luigi Salemme
- Casa di Cura "Montevergine", Mercogliano (Avellino), 83013, Italy
| | | | - Urna Kansakar
- Department of Medicine, Einstein Institute for Aging Research, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA
| | - Angelo Cioppa
- Casa di Cura "Montevergine", Mercogliano (Avellino), 83013, Italy
| | | | - Jessica Gambardella
- Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA
| | | | - Tullio Tesorio
- Casa di Cura "Montevergine", Mercogliano (Avellino), 83013, Italy
| | - Gaetano Santulli
- Department of Medicine, Einstein Institute for Aging Research, Wilf Family Cardiovascular Research Institute, Albert Einstein College of Medicine, New York, NY 10461, USA; Department of Molecular Pharmacology, Fleischer Institute for Diabetes and Metabolism (FIDAM), Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Einstein Institute for Neuroimmunology and Inflammation (INI), Albert Einstein College of Medicine, New York, NY 10461, USA.
| |
Collapse
|
10
|
Si Z, Shen Z, Luan F, Yan J. PINK1 regulates apoptosis of osteosarcoma as the target gene of cisplatin. J Orthop Surg Res 2023; 18:132. [PMID: 36823640 PMCID: PMC9948348 DOI: 10.1186/s13018-023-03615-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2022] [Accepted: 02/14/2023] [Indexed: 02/25/2023] Open
Abstract
BACKGROUND Osteosarcoma is a common primary bone malignancy prevalent among adolescents and young adults. PTEN-induced kinase 1 (PINK1) regulates Parkinson's disease, but its role in cancers is unknown. OBJECTIVE This study was designed to analyze the mechanism by which PINK1 affects osteosarcoma using bioinformatics and cell experiments. MATERIALS AND METHODS The gene expression profiles were downloaded from the TARGET database. Several online databases were used to analyze the expression and protein‒protein interaction networks. CCK-8 cell viability assays and cisplatin treatment were used to assess cell activity with or without cisplatin treatment. Acridine orange/ethidium bromide (AO/EB) fluorescence staining was used to calculate the percentage of apoptotic cells. RESULTS Through bioinformatics analysis, we found that high expression of PINK1 was associated with poor prognosis in patients with osteosarcoma, and PINK1 inhibited apoptosis and promoted proliferation pathways. Next, we found that both PINK1 mRNA and protein levels were upregulated in osteosarcoma tissues. Additionally, we found that PTEN was reduced, while FOXO3a was markedly increased in osteosarcoma, suggesting that FOXO3a and not PTEN induced the overexpression of PINK1. CCK-8 and clonogenic assays showed that the knockdown of PINK1 decreased the growth of U2OS osteosarcoma cells. Ki67 immunofluorescence staining revealed that reduced cell proliferation in U2OS cells resulted in the depletion of PINK1. In addition, our AO/EB staining results indicated that the knockdown of PINK1 resulted in an increase in apoptotic cells and increased the levels of cleaved caspase-3. Furthermore, our experiments revealed that cisplatin promotes OS cell apoptosis by downregulating PINK1. CONCLUSION Collectively, our findings demonstrate that PINK1 is crucially involved in osteosarcoma and suggests that it can promote the apoptosis of OS cells as the downstream target gene of cisplatin.
Collapse
Affiliation(s)
- Zhenxing Si
- grid.412596.d0000 0004 1797 9737Department of Emergency Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Zilong Shen
- grid.412463.60000 0004 1762 6325Department of Orthopedic Department, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin, 150001 Heilongjiang China
| | - Feiyu Luan
- grid.412596.d0000 0004 1797 9737Department of Emergency Surgery, The First Affiliated Hospital of Harbin Medical University, Harbin, China
| | - Jinglong Yan
- Department of Orthopedic Department, The Second Affiliated Hospital of Harbin Medical University, 246 Xuefu Road, Nangang District, Harbin, 150001, Heilongjiang, China.
| |
Collapse
|
11
|
Ko TK, Tan DJY. Is Disrupted Mitophagy a Central Player to Parkinson's Disease Pathology? Cureus 2023; 15:e35458. [PMID: 36860818 PMCID: PMC9969326 DOI: 10.7759/cureus.35458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2023] [Indexed: 02/27/2023] Open
Abstract
Whilst the pathophysiology at a cellular level has been defined, the cause of Parkinson's disease (PD) remains poorly understood. This neurodegenerative disorder is associated with impaired dopamine transmission in the substantia nigra, and protein accumulations known as Lewy bodies are visible in affected neurons. Cell culture models of PD have indicated impaired mitochondrial function, so the focus of this paper is on the quality control processes involved in and around mitochondria. Mitochondrial autophagy (mitophagy) is the process through which defective mitochondria are removed from the cell by internalisation into autophagosomes which fuse with a lysosome. This process involves many proteins, notably including PINK1 and parkin, both of which are known to be coded on genes associated with PD. Normally in healthy individuals, PINK1 associates with the outer mitochondrial membrane, which then recruits parkin, activating it to attach ubiquitin proteins to the mitochondrial membrane. PINK1, parkin, and ubiquitin cooperate to form a positive feedback system which accelerates the deposition of ubiquitin on dysfunctional mitochondria, resulting in mitophagy. However, in hereditary PD, the genes encoding PINK1 and parkin are mutated, resulting in proteins that are less efficient at removing poorly performing mitochondria, leaving cells more vulnerable to oxidative stress and ubiquitinated inclusion bodies, such as Lewy bodies. Current research that looks into the connection between mitophagy and PD is promising, already yielding potentially therapeutic compounds; until now, pharmacological support for the mitophagy process has not been part of the therapeutic arsenal. Continued research in this area is warranted.
Collapse
Affiliation(s)
- Tsz Ki Ko
- Otolaryngology, College of Life Sciences, Leicester Medical School, George Davies Centre, Leicester, GBR
| | | |
Collapse
|
12
|
Heo G, Sun MH, Jiang WJ, Li XH, Lee SH, Guo J, Zhou D, Cui XS. Rotenone causes mitochondrial dysfunction and prevents maturation in porcine oocytes. PLoS One 2022; 17:e0277477. [PMID: 36441709 PMCID: PMC9704683 DOI: 10.1371/journal.pone.0277477] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Accepted: 10/26/2022] [Indexed: 11/29/2022] Open
Abstract
Rotenone is a commonly used insecticidal chemical in agriculture and it is an inhibitor of mitochondrial complex Ⅰ. Previous studies have found that rotenone induces the production of reactive oxygen species (ROS) by inhibiting electron transport in the mitochondria of somatic and germ cells. However, there is little precise information on the effects of rotenone exposure in porcine oocytes during in vitro maturation, and the mechanisms underlying these effects have not been determined. The Cumulus-oocyte complexes were supplemented with different concentrations of rotenone to elucidate the effects of rotenone exposure on the meiotic maturation of porcine oocytes during in vitro maturation for about 48 hours. First, we found that the maturation rate and expansion of cumulus cells were significantly reduced in the 3 and 5 μM rotenone-treated groups. Subsequently, the concentration of rotenone was determined to be 3 μM. Also, immunofluorescence, western blotting, and image quantification analyses were performed to test the rotenone exposure on the meiotic maturation, total and mitochondrial ROS, mitochondrial function and biogenesis, mitophagy and apoptosis in porcine oocytes. Further experiments showed that rotenone treatment induced mitochondrial dysfunction and failure of mitochondrial biogenesis by repressing the level of SIRT1 during in vitro maturation of porcine oocytes. In addition, rotenone treatment reduced the ratio of active mitochondria to total mitochondria, increased ROS production, and decreased ATP production. The levels of LC3 and active-caspase 3 were significantly increased by rotenone treatment, indicating that mitochondrial dysfunction induced by rotenone increased mitophagy but eventually led to apoptosis. Collectively, these results suggest that rotenone interferes with porcine oocyte maturation by inhibiting mitochondrial function.
Collapse
Affiliation(s)
- Geun Heo
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Ming-Hong Sun
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Wen-Jie Jiang
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Xiao-Han Li
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Song-Hee Lee
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
| | - Jing Guo
- Joint Laboratory of the Modern Agricultural Technology International Cooperation, Ministry of Education, Jilin Agricultural University, Jilin, Changchun, 130118, China
| | - Dongjie Zhou
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
- * E-mail: (DZ); (X-SC)
| | - Xiang-Shun Cui
- Department of Animal Science, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea
- * E-mail: (DZ); (X-SC)
| |
Collapse
|
13
|
Ketenci M, Zablocki D, Sadoshima J. Mitochondrial Quality Control Mechanisms during Diabetic Cardiomyopathy. JMA J 2022; 5:407-415. [PMID: 36407069 PMCID: PMC9646286 DOI: 10.31662/jmaj.2022-0155] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2022] [Accepted: 08/18/2022] [Indexed: 01/25/2023] Open
Abstract
One of the major complications of diabetes mellitus is diabetic cardiomyopathy. One of the mechanisms that initiates the irreversible deterioration of cardiac function in diabetic cardiomyopathy is mitochondrial dysfunction. Functionally impaired mitochondria result in greater levels of oxidative stress and lipotoxicity, both of which exacerbate mitochondrial damage. Mitochondrial health is constantly monitored by mitochondrial quality control mechanisms. Mitophagy selectively degrades damaged mitochondria, thereby maintaining the healthy pool of mitochondria and preserving myocardial function. Mitophagy in diabetic cardiomyopathy is mediated by multiple mechanisms in a time-dependent manner. Potential targets for the treatment of diabetic cardiomyopathy include increased oxidative stress, mitochondrial dynamics, and mitochondrial clearance. Thus, stimulation of mitophagy represents a promising strategy for the alleviation of diabetic cardiomyopathy.
Collapse
Affiliation(s)
- Melis Ketenci
- Rutgers New Jersey Medical School, Department of Cell Biology and Molecular Medicine, Rutgers Biomedical and Health Sciences, Newark, USA
| | - Daniela Zablocki
- Rutgers New Jersey Medical School, Department of Cell Biology and Molecular Medicine, Rutgers Biomedical and Health Sciences, Newark, USA
| | - Junichi Sadoshima
- Rutgers New Jersey Medical School, Department of Cell Biology and Molecular Medicine, Rutgers Biomedical and Health Sciences, Newark, USA
| |
Collapse
|
14
|
Mauri S, Favaro M, Bernardo G, Mazzotta GM, Ziviani E. Mitochondrial autophagy in the sleeping brain. Front Cell Dev Biol 2022; 10:956394. [PMID: 36092697 PMCID: PMC9449320 DOI: 10.3389/fcell.2022.956394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Accepted: 07/13/2022] [Indexed: 11/13/2022] Open
Abstract
A significant percentage of the mitochondrial mass is replaced on a daily basis via mechanisms of mitochondrial quality control. Through mitophagy (a selective type of autophagy that promotes mitochondrial proteostasis) cells keep a healthy pool of mitochondria, and prevent oxidative stress and inflammation. Furthermore, mitophagy helps adapting to the metabolic demand of the cells, which changes on a daily basis.Core components of the mitophagy process are PINK1 and Parkin, which mutations are linked to Parkinson’s Disease. The crucial role of PINK1/Parkin pathway during stress-induced mitophagy has been extensively studied in vitro in different cell types. However, recent advances in the field allowed discovering that mitophagy seems to be only slightly affected in PINK1 KO mice and flies, putting into question the physiological relevance of this pathway in vivo in the whole organism. Indeed, several cell-specific PINK1/Parkin-independent mitophagy pathways have been recently discovered, which appear to be activated under physiological conditions such as those that promote mitochondrial proteome remodeling during differentiation or in response to specific physiological stimuli.In this Mini Review we want to summarize the recent advances in the field, and add another level of complexity by focusing attention on a potentially important aspect of mitophagy regulation: the implication of the circadian clock. Recent works showed that the circadian clock controls many aspects of mitochondrial physiology, including mitochondrial morphology and dynamic, respiratory activity, and ATP synthesis. Furthermore, one of the essential functions of sleep, which is controlled by the clock, is the clearance of toxic metabolic compounds from the brain, including ROS, via mechanisms of proteostasis. Very little is known about a potential role of the clock in the quality control mechanisms that maintain the mitochondrial repertoire healthy during sleep/wake cycles. More importantly, it remains completely unexplored whether (dys)function of mitochondrial proteostasis feedbacks to the circadian clockwork.
Collapse
Affiliation(s)
| | | | | | | | - Elena Ziviani
- *Correspondence: Gabriella M. Mazzotta, Elena Ziviani,
| |
Collapse
|
15
|
Leduc-Gaudet JP, Hussain SN, Gouspillou G. Parkin: A potential target to promote healthy aging. J Physiol 2022; 600:3405-3421. [PMID: 35691026 DOI: 10.1113/jp282567] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 06/08/2022] [Indexed: 11/08/2022] Open
Abstract
Parkin is an E3 ubiquitin ligase mostly known for its role in regulating the removal of defective mitochondria via mitophagy. However, increasing experimental evidence that Parkin regulates several other aspects of mitochondrial biology in addition to its role in mitophagy has emerged over the past two decades. Indeed, Parkin has been shown to regulate mitochondrial biogenesis and dynamics and mitochondrial-derived vesicle formation, suggesting that Parkin plays key roles in maintaining healthy mitochondria. While Parkin is commonly described as a cytosolic E3 ubiquitin ligase, Parkin was also detected in other cellular compartments, including the nucleus, where it regulates transcription factors and acts as a transcription factor itself. New evidence also suggests that Parkin overexpression can be leveraged to delay aging. In D. melanogaster, for example, Parkin overexpression extends lifespan. In mammals, Parkin overexpression delays hallmarks of aging in several tissues and cell types. Parkin overexpression also confers protection in various models of cellular senescence and neurological disorders closely associated with aging, such as Alzheimer's and Parkinson's diseases. Recently, Parkin overexpression has also been shown to suppress tumor growth. In this review, we discuss newly emerging biological roles of Parkin as a modulator of cellular homeostasis, survival, and healthy aging, and we explore potential mechanisms through which Parkin exerts its beneficial effects on cellular health. Abstract figure legend Parkin: A potential target to promote healthy aging Illustration of key aspects of Parkin biology, including Parkin function and cellular localization and key roles in the regulation of mitochondrial quality control. The organs and systems in which Parkin overexpression was shown to exert protective effects relevant to the promotion of healthy aging are highlighted in the black rectangle at the bottom of the Figure. This article is protected by copyright. All rights reserved.
Collapse
Affiliation(s)
- Jean-Philippe Leduc-Gaudet
- Department of Biomedical Sciences, Veneto Institute of Molecular Medicine, University of Padova, Padova, Italy.,Meakins-Christie Laboratories, Department of Medicine, McGill University, Montréal, QC, Canada.,Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada.,Département des sciences de l'activité physique, Faculté des sciences, Université du Québec à Montréal (UQAM), Montréal, QC, Canada
| | - Sabah Na Hussain
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montréal, QC, Canada.,Translational Research in Respiratory Diseases Program, Research Institute of the McGill University Health Centre, Montréal, QC, Canada
| | - Gilles Gouspillou
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montréal, QC, Canada.,Département des sciences de l'activité physique, Faculté des sciences, Université du Québec à Montréal (UQAM), Montréal, QC, Canada
| |
Collapse
|
16
|
Sharma S, Patel F, Ara H, Bess E, Shum A, Bhattarai S, Subedi U, Bell DS, Bhuiyan MS, Sun H, Batinic-Haberle I, Panchatcharam M, Miriyala S. Rotenone-Induced 4-HNE Aggresome Formation and Degradation in HL-1 Cardiomyocytes: Role of Autophagy Flux. Int J Mol Sci 2022; 23:ijms23094675. [PMID: 35563066 PMCID: PMC9105393 DOI: 10.3390/ijms23094675] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 04/21/2022] [Accepted: 04/22/2022] [Indexed: 11/22/2022] Open
Abstract
Reactive oxygen species (ROS) cause oxidative stress by generating reactive aldehydes known as 4-hydroxynonenal (4-HNE). 4-HNE modifies protein via covalent adduction; however, little is known about the degradation mechanism of 4-HNE-adducted proteins. Autophagy is a dynamic process that maintains cellular homeostasis by removing damaged organelles and proteins. In this study, we determined the role of a superoxide dismutase (SOD) mimetic MnTnBuOE-2-PyP5+ (MnP, BMX-001) on rotenone-induced 4-HNE aggresome degradation in HL-1 cardiomyocytes. A rotenone treatment (500 nM) given for 24 h demonstrated both increased ROS and 4-HNE aggresome accumulation in HL-1 cardiomyocytes. In addition, cardiomyocytes treated with rotenone displayed an increase in the autophagy marker LC3-II, as shown by immunoblotting and immunofluorescence. A pre-treatment with MnP (20 µM) for 24 h attenuated rotenone-induced ROS formation. An MnP pre-treatment showed decreased 4-HNE aggresomes and LC3-II formation. A rotenone-induced increase in autophagosomes was attenuated by a pre-treatment with MnP, as shown by fluorescent-tagged LC3 (tfLC3). Rotenone increased tubulin hyperacetylation through the ROS-mediated pathway, which was attenuated by MnP. The disruption of autophagy caused HL-1 cell death because a 3-methyladenine inhibitor of autophagosomes caused reduced cell death. Yet, rapamycin, an inducer of autophagy, increased cell death. These results indicated that a pre-treatment with MnP decreased rotenone-induced 4-HNE aggresomes by enhancing the degradation process.
Collapse
Affiliation(s)
- Sudha Sharma
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA; (S.S.); (F.P.); (H.A.); (E.B.); (A.S.); (S.B.); (U.S.); (D.S.B.); (H.S.)
| | - Foram Patel
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA; (S.S.); (F.P.); (H.A.); (E.B.); (A.S.); (S.B.); (U.S.); (D.S.B.); (H.S.)
| | - Hosne Ara
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA; (S.S.); (F.P.); (H.A.); (E.B.); (A.S.); (S.B.); (U.S.); (D.S.B.); (H.S.)
| | - Ezra Bess
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA; (S.S.); (F.P.); (H.A.); (E.B.); (A.S.); (S.B.); (U.S.); (D.S.B.); (H.S.)
| | - Alika Shum
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA; (S.S.); (F.P.); (H.A.); (E.B.); (A.S.); (S.B.); (U.S.); (D.S.B.); (H.S.)
| | - Susmita Bhattarai
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA; (S.S.); (F.P.); (H.A.); (E.B.); (A.S.); (S.B.); (U.S.); (D.S.B.); (H.S.)
| | - Utsab Subedi
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA; (S.S.); (F.P.); (H.A.); (E.B.); (A.S.); (S.B.); (U.S.); (D.S.B.); (H.S.)
| | - Daquonte Sanard Bell
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA; (S.S.); (F.P.); (H.A.); (E.B.); (A.S.); (S.B.); (U.S.); (D.S.B.); (H.S.)
| | - Md. Shenuarin Bhuiyan
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA;
| | - Hong Sun
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA; (S.S.); (F.P.); (H.A.); (E.B.); (A.S.); (S.B.); (U.S.); (D.S.B.); (H.S.)
| | - Ines Batinic-Haberle
- Department of Radiation Oncology, Duke University School of Medicine, Durham, NC 27710, USA;
| | - Manikandan Panchatcharam
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA; (S.S.); (F.P.); (H.A.); (E.B.); (A.S.); (S.B.); (U.S.); (D.S.B.); (H.S.)
- Correspondence: (M.P.); (S.M.); Tel.: +1-3-186-756-938 (M.P.); +1-3-186-758-326 (S.M.)
| | - Sumitra Miriyala
- Department of Cellular Biology and Anatomy, Louisiana State University Health Sciences Center, Shreveport, LA 71103, USA; (S.S.); (F.P.); (H.A.); (E.B.); (A.S.); (S.B.); (U.S.); (D.S.B.); (H.S.)
- Correspondence: (M.P.); (S.M.); Tel.: +1-3-186-756-938 (M.P.); +1-3-186-758-326 (S.M.)
| |
Collapse
|
17
|
Ma X, Rawnsley DR, Kovacs A, Islam M, Murphy JT, Zhao C, Kumari M, Foroughi L, Liu H, Qi K, Diwan A, Hyrc K, Evans S, Satoh T, French BA, Margulies KB, Javaheri A, Razani B, Mann DL, Mani K, Diwan A. TRAF2, an Innate Immune Sensor, Reciprocally Regulates Mitophagy and Inflammation to Maintain Cardiac Myocyte Homeostasis. JACC Basic Transl Sci 2022; 7:223-243. [PMID: 35411325 PMCID: PMC8993766 DOI: 10.1016/j.jacbts.2021.12.002] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/24/2021] [Revised: 12/01/2021] [Accepted: 12/02/2021] [Indexed: 12/26/2022]
Abstract
Mitochondria are essential for cardiac myocyte function, but damaged mitochondria trigger cardiac myocyte death. Although mitophagy, a lysosomal degradative pathway to remove damaged mitochondria, is robustly active in cardiac myocytes in the unstressed heart, its mechanisms and physiological role remain poorly defined. We discovered a critical role for TRAF2, an innate immunity effector protein with E3 ubiquitin ligase activity, in facilitating physiological cardiac myocyte mitophagy in the adult heart, to prevent inflammation and cell death, and maintain myocardial homeostasis.
Collapse
Key Words
- AAV9, adeno-associated virus serotype 9
- ER, endoplasmic reticulum
- FS, fractional shortening
- GFP, green fluorescent protein
- IP, intraperitoneal
- LV, left ventricular
- MAM, mitochondria-associated membranes
- MCM, MerCreMer
- MEF, murine embryonic fibroblast
- PINK1, PTEN-induced kinase 1
- RFP, red fluorescent protein
- TLR9, toll-like receptor 9
- TRAF2
- TRAF2, tumor necrosis factor receptor-associated factor-2
- TUNEL, terminal deoxynucleotidyl transferase dUTP nick end labeling
- cTnT, cardiac troponin T
- cell death
- inflammation
- mitophagy
Collapse
Affiliation(s)
- Xiucui Ma
- Center for Cardiovascular Research and Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- John Cochran VA Medical Center, St. Louis, Missouri, USA
| | - David R. Rawnsley
- Center for Cardiovascular Research and Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Attila Kovacs
- Center for Cardiovascular Research and Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Moydul Islam
- Center for Cardiovascular Research and Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - John T. Murphy
- Center for Cardiovascular Research and Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- John Cochran VA Medical Center, St. Louis, Missouri, USA
| | - Chen Zhao
- Center for Cardiovascular Research and Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Minu Kumari
- Center for Cardiovascular Research and Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Layla Foroughi
- Center for Cardiovascular Research and Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- John Cochran VA Medical Center, St. Louis, Missouri, USA
| | - Haiyan Liu
- Center for Cardiovascular Research and Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- John Cochran VA Medical Center, St. Louis, Missouri, USA
| | - Kevin Qi
- Center for Cardiovascular Research and Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Aaradhya Diwan
- Center for Cardiovascular Research and Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Krzysztof Hyrc
- Alafi Neuroimaging Laboratory, Washington University School of Medicine, St. Louis, Missouri, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Sarah Evans
- Center for Cardiovascular Research and Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Takashi Satoh
- Department of Immune Regulation, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Brent A. French
- Department of Biomedical Engineering, University of Virginia, Charlottesville, Virginia, USA
| | - Kenneth B. Margulies
- Department of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ali Javaheri
- Center for Cardiovascular Research and Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Babak Razani
- Center for Cardiovascular Research and Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- John Cochran VA Medical Center, St. Louis, Missouri, USA
| | - Douglas L. Mann
- Center for Cardiovascular Research and Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- John Cochran VA Medical Center, St. Louis, Missouri, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Kartik Mani
- Center for Cardiovascular Research and Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- John Cochran VA Medical Center, St. Louis, Missouri, USA
| | - Abhinav Diwan
- Center for Cardiovascular Research and Cardiovascular Division, Department of Internal Medicine, Washington University School of Medicine, St. Louis, Missouri, USA
- John Cochran VA Medical Center, St. Louis, Missouri, USA
- Hope Center for Neurological Disorders, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, Missouri, USA
- Department of Obstetrics and Gynecology, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
18
|
Amaral AG, da Silva CCC, Serna JDC, Honorato-Sampaio K, Freitas JA, Duarte-Neto AN, Bloise AC, Cassina L, Yoshinaga MY, Chaves-Filho AB, Qian F, Miyamoto S, Boletta A, Bordin S, Kowaltowski AJ, Onuchic LF. Disruption of polycystin-1 cleavage leads to cardiac metabolic rewiring in mice. Biochim Biophys Acta Mol Basis Dis 2022; 1868:166371. [PMID: 35218894 DOI: 10.1016/j.bbadis.2022.166371] [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: 10/04/2021] [Revised: 02/15/2022] [Accepted: 02/20/2022] [Indexed: 11/18/2022]
Abstract
Cardiovascular manifestations account for marked morbi-mortality in autosomal dominant polycystic kidney disease (ADPKD). Pkd1- and Pkd2-deficient mice develop cardiac dysfunction, however the underlying mechanisms remain largely unclear. It is unknown whether impairment of polycystin-1 cleavage at the G-protein-coupled receptor proteolysis site, a significant ADPKD mutational mechanism, is involved in this process. We analyzed the impact of polycystin-1 cleavage on heart metabolism using Pkd1V/V mice, a model unable to cleave this protein and with early cardiac dysfunction. Pkd1V/V hearts showed lower levels of glucose and amino acids and higher lipid levels than wild-types, as well as downregulation of p-AMPK, p-ACCβ, CPT1B-Cpt1b, Ppara, Nppa and Acta1. These findings suggested decreased fatty acid β-oxidation, which was confirmed by lower oxygen consumption by Pkd1V/V isolated mitochondria using palmitoyl-CoA. Pkd1V/V hearts also presented increased oxygen consumption in response to glucose, suggesting that alternative substrates may be used to generate energy. Pkd1V/V hearts displayed a higher density of decreased-size mitochondria, a finding associated with lower MFN1, Parkin and BNIP3 expression. These derangements were correlated with increased apoptosis and inflammation but not hypertrophy. Notably, Pkd1V/V neonate cardiomyocytes also displayed shifts in oxygen consumption and p-AMPK downregulation, suggesting that, at least partially, the metabolic alterations are not induced by kidney dysfunction. Our findings reveal that disruption of polycystin-1 cleavage leads to cardiac metabolic rewiring in mice, expanding the understanding of heart dysfunction associated with Pkd1 deficiency and likely with human ADPKD.
Collapse
Affiliation(s)
- Andressa G Amaral
- Disciplinas de Nefrologia e Medicina Molecular, Departamento de Clínica Médica, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP 01246903, Brazil
| | - Camille C C da Silva
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508000, Brazil
| | - Julian D C Serna
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508000, Brazil
| | - Kinulpe Honorato-Sampaio
- Faculdade de Medicina, Universidade Federal dos Vales do Jequitinhonha e Mucuri, Diamantina, MG 31270901, Brazil
| | - Jéssica A Freitas
- Disciplinas de Nefrologia e Medicina Molecular, Departamento de Clínica Médica, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP 01246903, Brazil
| | - Amaro N Duarte-Neto
- Disciplina de Emergências Clínicas, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP 01246903, Brazil
| | - Antonio C Bloise
- Departamento de Física Aplicada, Instituto de Física, Universidade de São Paulo, São Paulo, SP 05508000, Brazil
| | - Laura Cassina
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Marcos Y Yoshinaga
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508000, Brazil
| | - Adriano B Chaves-Filho
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508000, Brazil
| | - Feng Qian
- Division of Nephrology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Sayuri Miyamoto
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508000, Brazil
| | - Alessandra Boletta
- Division of Genetics and Cell Biology, San Raffaele Scientific Institute, Milan 20132, Italy
| | - Silvana Bordin
- Departamento de Fisiologia, Instituto de Ciências Biomédicas, Universidade de São Paulo, São Paulo, SP 05508000, Brazil
| | - Alicia J Kowaltowski
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, São Paulo, SP 05508000, Brazil
| | - Luiz F Onuchic
- Disciplinas de Nefrologia e Medicina Molecular, Departamento de Clínica Médica, Faculdade de Medicina da Universidade de São Paulo, São Paulo, SP 01246903, Brazil.
| |
Collapse
|
19
|
Thai PN, Miller CV, King MT, Schaefer S, Veech RL, Chiamvimonvat N, Bers DM, Dedkova EN. Ketone Ester D-β-Hydroxybutyrate-(R)-1,3 Butanediol Prevents Decline in Cardiac Function in Type 2 Diabetic Mice. J Am Heart Assoc 2021; 10:e020729. [PMID: 34583524 PMCID: PMC8649133 DOI: 10.1161/jaha.120.020729] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Background Heart failure is responsible for approximately 65% of deaths in patients with type 2 diabetes mellitus. However, existing therapeutics for type 2 diabetes mellitus have limited success on the prevention of diabetic cardiomyopathy. The aim of this study was to determine whether moderate elevation in D‐β‐hydroxybutyrate improves cardiac function in animals with type 2 diabetes mellitus. Methods and Results Type 2 diabetic (db/db) and their corresponding wild‐type mice were fed a control diet or a diet where carbohydrates were equicalorically replaced by D‐β‐hydroxybutyrate‐(R)‐1,3 butanediol monoester (ketone ester [KE]). After 4 weeks, echocardiography demonstrated that a KE diet improved systolic and diastolic function in db/db mice. A KE diet increased expression of mitochondrial succinyl‐CoA:3‐oxoacid‐CoA transferase and restored decreased expression of mitochondrial β‐hydroxybutyrate dehydrogenase, key enzymes in cardiac ketone metabolism. A KE diet significantly enhanced both basal and ADP‐mediated oxygen consumption in cardiac mitochondria from both wild‐type and db/db animals; however, it did not result in the increased mitochondrial respiratory control ratio. Additionally, db/db mice on a KE diet had increased resistance to oxidative and redox stress, with evidence of restoration of decreased expression of thioredoxin and glutathione peroxidase 4 and less permeability transition pore activity in mitochondria. Mitochondrial biogenesis, quality control, and elimination of dysfunctional mitochondria via mitophagy were significantly increased in cardiomyocytes from db/db mice on a KE diet. The increase in mitophagy was correlated with restoration of mitofusin 2 expression, which contributed to improved coupling between cytosolic E3 ubiquitin ligase translocation into mitochondria and microtubule‐associated protein 1 light chain 3–mediated autophagosome formation. Conclusions Moderate elevation in circulating D‐β‐hydroxybutyrate levels via KE supplementation enhances mitochondrial biogenesis, quality control, and oxygen consumption and increases resistance to oxidative/redox stress and mPTP opening, thus resulting in improvement of cardiac function in animals with type 2 diabetes mellitus.
Collapse
Affiliation(s)
- Phung N Thai
- Department of Internal Medicine Cardiovascular Medicine University of California Davis CA
| | | | - M Todd King
- Laboratory of Metabolic Control National Institute on Alcohol Abuse and AlcoholismNational Institutes of Health Rockville MD
| | - Saul Schaefer
- Department of Internal Medicine Cardiovascular Medicine University of California Davis CA.,Department of Veterans Affairs Northern California Health Care System Mather CA
| | - Richard L Veech
- Laboratory of Metabolic Control National Institute on Alcohol Abuse and AlcoholismNational Institutes of Health Rockville MD
| | - Nipavan Chiamvimonvat
- Department of Internal Medicine Cardiovascular Medicine University of California Davis CA.,Department of Veterans Affairs Northern California Health Care System Mather CA
| | - Donald M Bers
- Department of Pharmacology University of California Davis CA
| | - Elena N Dedkova
- Department of Pharmacology University of California Davis CA.,Department of Molecular Biosciences University of California Davis CA
| |
Collapse
|
20
|
Ramírez-Sagredo A, Quiroga C, Garrido-Moreno V, López-Crisosto C, Leiva-Navarrete S, Norambuena-Soto I, Ortiz-Quintero J, Díaz-Vesga MC, Perez W, Hendrickson T, Parra V, Pedrozo Z, Altamirano F, Chiong M, Lavandero S. Polycystin-1 regulates cardiomyocyte mitophagy. FASEB J 2021; 35:e21796. [PMID: 34324238 DOI: 10.1096/fj.202002598r] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 12/29/2022]
Abstract
Polycystin-1 (PC1) is a transmembrane protein found in different cell types, including cardiomyocytes. Alterations in PC1 expression have been linked to mitochondrial damage in renal tubule cells and in patients with autosomal dominant polycystic kidney disease. However, to date, the regulatory role of PC1 in cardiomyocyte mitochondria is not well understood. The analysis of mitochondrial morphology from cardiomyocytes of heterozygous PC1 mice (PDK1+/- ) using transmission electron microscopy showed that cardiomyocyte mitochondria were smaller with increased mitochondria density and circularity. These parameters were consistent with mitochondrial fission. We knocked-down PC1 in cultured rat cardiomyocytes and human-induced pluripotent stem cells (iPSC)-derived cardiomyocytes to evaluate mitochondrial function and morphology. The results showed that downregulation of PC1 expression results in reduced protein levels of sub-units of the OXPHOS complexes and less functional mitochondria (reduction of mitochondrial membrane potential, mitochondrial respiration, and ATP production). This mitochondrial dysfunction activates the elimination of defective mitochondria by mitophagy, assessed by an increase of autophagosome adapter protein LC3B and the recruitment of the Parkin protein to the mitochondria. siRNA-mediated PC1 knockdown leads to a loss of the connectivity of the mitochondrial network and a greater number of mitochondria per cell, but of smaller sizes, which characterizes mitochondrial fission. PC1 silencing also deregulates the AKT-FoxO1 signaling pathway, which is involved in the regulation of mitochondrial metabolism, mitochondrial morphology, and processes that are part of cell quality control, such as mitophagy. Together, these data provide new insights about the controls that PC1 exerts on mitochondrial morphology and function in cultured cardiomyocytes dependent on the AKT-FoxO1 signaling pathway.
Collapse
Affiliation(s)
- Andrea Ramírez-Sagredo
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Clara Quiroga
- Advanced Center for Chronic Diseases (ACCDiS), División de Enfermedades Cardiovasculares, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Valeria Garrido-Moreno
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Camila López-Crisosto
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Advanced Center for Chronic Diseases (ACCDiS), División de Enfermedades Cardiovasculares, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Sebastian Leiva-Navarrete
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Autophagy Research Center, Universidad de Chile, Santiago, Chile.,Network for the Study of High-lethality Cardiopulmonary Diseases (REECPAL), Universidad de Chile, Santiago, Chile
| | - Ignacio Norambuena-Soto
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Jafet Ortiz-Quintero
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Departamento de Bioanálisis e Inmunología, Escuela de Microbiología, Facultad de Ciencias, Universidad Nacional Autónoma de Honduras, Tegucigalpa, Honduras
| | - Magda C Díaz-Vesga
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Programa de Fisiología y Biofísica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Grupo de Investigación en Ciencias Básicas y Clínicas de la Salud, Pontificia Universidad Javeriana de Cali, Cali, Colombia
| | - William Perez
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA
| | - Troy Hendrickson
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA.,Texas A&M MD/PhD Program, Texas A&M Health Science Center, College Station, TX, USA
| | - Valentina Parra
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Autophagy Research Center, Universidad de Chile, Santiago, Chile.,Network for the Study of High-lethality Cardiopulmonary Diseases (REECPAL), Universidad de Chile, Santiago, Chile
| | - Zully Pedrozo
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Network for the Study of High-lethality Cardiopulmonary Diseases (REECPAL), Universidad de Chile, Santiago, Chile.,Programa de Fisiología y Biofísica, ICBM, Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Francisco Altamirano
- Department of Cardiovascular Sciences, Houston Methodist Research Institute, Houston, TX, USA.,Department of Cardiothoracic Surgery, Weill Cornell Medical College, Cornell University, Ithaca, NY, USA
| | - Mario Chiong
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile
| | - Sergio Lavandero
- Advanced Center of Chronic Diseases (ACCDiS), Facultad de Ciencias Químicas y Farmacéuticas y Facultad de Medicina, Universidad de Chile, Santiago, Chile.,Corporación Centro de Estudios Científicos de las Enfermedades Crónicas (CECEC), Santiago, Chile.,Department of Internal Medicine, Cardiology Division, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
21
|
Bhardwaj G, Penniman CM, Jena J, Suarez Beltran PA, Foster C, Poro K, Junck TL, Hinton AO, Souvenir R, Fuqua JD, Morales PE, Bravo-Sagua R, Sivitz WI, Lira VA, Abel ED, O'Neill BT. Insulin and IGF-1 receptors regulate complex-I dependent mitochondrial bioenergetics and supercomplexes via FoxOs in muscle. J Clin Invest 2021; 131:e146415. [PMID: 34343133 DOI: 10.1172/jci146415] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2020] [Accepted: 07/28/2021] [Indexed: 12/25/2022] Open
Abstract
Decreased skeletal muscle strength and mitochondrial dysfunction are characteristic of diabetes. Action of insulin and IGF-1 through insulin receptor (IR) and IGF-1 receptor (IGF1R) maintain muscle mass via suppression of FoxOs, but whether FoxO activation coordinates atrophy in concert with mitochondrial dysfunction is unknown. We show that mitochondrial respiration and complex-I activity were decreased in streptozotocin (STZ) diabetic muscle, but these defects were reversed following muscle-specific FoxO1/3/4 triple knockout in STZ-FoxO TKO. In the absence of systemic glucose or lipid abnormalities, muscle-specific IR knockout (M-IR-/-) or combined IR/IGF1R knockout (MIGIRKO) impaired mitochondrial respiration, decreased ATP production, and increased ROS. These mitochondrial abnormalities were not present in muscle-specific IR/IGF1R and FoxO1/3/4 quintuple knockout mice (M-QKO). Acute tamoxifen-inducible deletion of IR/IGF1R also decreased muscle pyruvate respiration, complex-I activity, and supercomplex assembly. Although autophagy was increased when IR/IGF1R were deleted in muscle, mitophagy was not increased. Mechanistically, RNA-seq revealed that complex-I core subunits were decreased in STZ-diabetic and MIGIRKO muscle, and these changes were not present with FoxO knockout in STZ-FoxO TKO and M-QKO. Thus, insulin-deficient diabetes or loss of insulin/IGF-1 action in muscle decreases complex-I driven mitochondrial respiration and supercomplex assembly, in part by FoxO-mediated repression of Complex-I subunit expression.
Collapse
Affiliation(s)
- Gourav Bhardwaj
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, United States of America
| | - Christie M Penniman
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, United States of America
| | - Jayashree Jena
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, United States of America
| | - Pablo A Suarez Beltran
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, United States of America
| | - Collin Foster
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, United States of America
| | - Kennedy Poro
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, United States of America
| | - Taylor L Junck
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, United States of America
| | - Antentor O Hinton
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, United States of America
| | - Rhonda Souvenir
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, United States of America
| | - Jordan D Fuqua
- Department of Health and Human Physiology, University of Iowa, Iowa City, United States of America
| | - Pablo E Morales
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago, Chile
| | - Roberto Bravo-Sagua
- Advanced Center for Chronic Diseases (ACCDiS), Universidad de Chile, Santiago, Chile
| | - William I Sivitz
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, United States of America
| | - Vitor A Lira
- Department of Health and Human Physiology, University of Iowa, Iowa City, United States of America
| | - E Dale Abel
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, United States of America
| | - Brian T O'Neill
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, United States of America
| |
Collapse
|
22
|
Leduc-Gaudet JP, Hussain SNA, Barreiro E, Gouspillou G. Mitochondrial Dynamics and Mitophagy in Skeletal Muscle Health and Aging. Int J Mol Sci 2021; 22:ijms22158179. [PMID: 34360946 PMCID: PMC8348122 DOI: 10.3390/ijms22158179] [Citation(s) in RCA: 108] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Revised: 07/23/2021] [Accepted: 07/27/2021] [Indexed: 01/18/2023] Open
Abstract
The maintenance of mitochondrial integrity is critical for muscle health. Mitochondria, indeed, play vital roles in a wide range of cellular processes, including energy supply, Ca2+ homeostasis, retrograde signaling, cell death, and many others. All mitochondria-containing cells, including skeletal muscle cells, dispose of several pathways to maintain mitochondrial health, including mitochondrial biogenesis, mitochondrial-derived vesicles, mitochondrial dynamics (fusion and fission process shaping mitochondrial morphology), and mitophagy—the process in charge of the removal of mitochondria though autophagy. The loss of skeletal muscle mass (atrophy) is a major health problem worldwide, especially in older people. Currently, there is no treatment to counteract the progressive decline in skeletal muscle mass and strength that occurs with aging, a process termed sarcopenia. There is increasing data, including our own, suggesting that accumulation of dysfunctional mitochondria contributes to the development of sarcopenia. Impairments in mitochondrial dynamics and mitophagy were recently proposed to contribute to sarcopenia. This review summarizes the current state of knowledge on the role played by mitochondrial dynamics and mitophagy in skeletal muscle health and in the development of sarcopenia. We also highlight recent studies showing that enhancing mitophagy in skeletal muscle is a promising therapeutic target to prevent or even treat skeletal muscle dysfunction in the elderly.
Collapse
Affiliation(s)
- Jean-Philippe Leduc-Gaudet
- Research Institute of the McGill University Health Centre, Department of Critical Care, Montréal, QC H4A 3J1, Canada; (S.N.A.H.); (G.G.)
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montréal, QC H4A 3J1, Canada
- Département des Sciences de l’activité Physique, Faculté des Sciences, UQAM, Montréal, QC H2X 1Y4, Canada
- Correspondence: ; Tel.: +1-514-476-6688
| | - Sabah N. A. Hussain
- Research Institute of the McGill University Health Centre, Department of Critical Care, Montréal, QC H4A 3J1, Canada; (S.N.A.H.); (G.G.)
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montréal, QC H4A 3J1, Canada
| | - Esther Barreiro
- Pulmonology Department-Muscle Wasting & Cachexia in Chronic Respiratory Diseases & Lung Cancer Research Group, IMIM-Hospital del Mar, Parc de Salut Mar, Biomedical Research Park (PRBB), C/Dr. Aiguader, 88, 08003 Barcelona, Spain;
- Centro de Investigación en Red de Enfermedades Respiratorias (CIBERES), Instituto de Salud Carlos III (ISCIII), 28029 Madrid, Spain
- Health and Experimental Sciences Department (CEXS), Pompeu Fabra University (UPF), Biomedical Research Park (PRBB), C/Dr. Aiguader, 88, 08003 Barcelona, Spain
| | - Gilles Gouspillou
- Research Institute of the McGill University Health Centre, Department of Critical Care, Montréal, QC H4A 3J1, Canada; (S.N.A.H.); (G.G.)
- Meakins-Christie Laboratories, Department of Medicine, McGill University, Montréal, QC H4A 3J1, Canada
- Département des Sciences de l’activité Physique, Faculté des Sciences, UQAM, Montréal, QC H2X 1Y4, Canada
- Centre de Recherche de l’Institut Universitaire de Gériatrie de Montréal, Montréal, QC H3W 1W5, Canada
| |
Collapse
|
23
|
Philp AM, Saner NJ, Lazarou M, Ganley IG, Philp A. The influence of aerobic exercise on mitochondrial quality control in skeletal muscle. J Physiol 2021; 599:3463-3476. [PMID: 33369731 DOI: 10.1113/jp279411] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 12/17/2020] [Indexed: 01/12/2023] Open
Abstract
Mitochondria are dynamic organelles, intricately designed to meet cellular energy requirements. To accommodate alterations in energy demand, mitochondria have a high degree of plasticity, changing in response to transient activation of numerous stress-related pathways. This adaptive response is particularly relevant in highly metabolic tissues such as skeletal muscle, where mitochondria support numerous biological processes related to metabolism, growth and regeneration. Aerobic exercise is a potent stimulus for skeletal muscle remodelling, leading to alterations in substrate utilisation, fibre-type composition and performance. Underlying these physiological responses is a change in mitochondrial quality control (MQC), a term encompassing the co-ordination of mitochondrial synthesis (biogenesis), remodelling (dynamics) and degradation (mitophagy) pathways. Understanding of MQC in skeletal muscle and the regulatory role of aerobic exercise of this process are rapidly advancing, as are the molecular techniques allowing the study of MQC in vivo. Given the emerging link between MQC and the onset of numerous non-communicable diseases, understanding the molecular regulation of MQC, and the role of aerobic exercise in this process, will have substantial future impact on therapeutic approaches to manipulate MQC and maintain mitochondrial function across health span.
Collapse
Affiliation(s)
- Ashleigh M Philp
- Healthy Ageing Research Theme, Garvan Institute of Medical Research, 384 Victoria Street, Sydney, New South Wales, 2010, Australia
- St Vincent's Medical School, UNSW Medicine, UNSW Sydney, Sydney, New South Wales, 2010, Australia
| | - Nicholas J Saner
- Sports Cardiology, Baker Heart and Diabetes Institute, Melbourne, Australia
| | - Michael Lazarou
- Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Ian G Ganley
- Medical Research Council Protein Phosphorylation and Ubiquitylation Unit, School of Life Sciences, University of Dundee, Dundee, UK
| | - Andrew Philp
- Healthy Ageing Research Theme, Garvan Institute of Medical Research, 384 Victoria Street, Sydney, New South Wales, 2010, Australia
- St Vincent's Medical School, UNSW Medicine, UNSW Sydney, Sydney, New South Wales, 2010, Australia
| |
Collapse
|
24
|
Ji H, Wu D, Kimberlee O, Li R, Qian G. Molecular Perspectives of Mitophagy in Myocardial Stress: Pathophysiology and Therapeutic Targets. Front Physiol 2021; 12:700585. [PMID: 34276422 PMCID: PMC8279814 DOI: 10.3389/fphys.2021.700585] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 05/27/2021] [Indexed: 01/15/2023] Open
Abstract
A variety of complex risk factors and pathological mechanisms contribute to myocardial stress, which ultimately promotes the development of cardiovascular diseases, including acute cardiac insufficiency, myocardial ischemia, myocardial infarction, high-glycemic myocardial injury, and acute alcoholic cardiotoxicity. Myocardial stress is characterized by abnormal metabolism, excessive reactive oxygen species production, an insufficient energy supply, endoplasmic reticulum stress, mitochondrial damage, and apoptosis. Mitochondria, the main organelles contributing to the energy supply of cardiomyocytes, are key determinants of cell survival and death. Mitophagy is important for cardiomyocyte function and metabolism because it removes damaged and aged mitochondria in a timely manner, thereby maintaining the proper number of normal mitochondria. In this review, we first introduce the general characteristics and regulatory mechanisms of mitophagy. We then describe the three classic mitophagy regulatory pathways and their involvement in myocardial stress. Finally, we discuss the two completely opposite effects of mitophagy on the fate of cardiomyocytes. Our summary of the molecular pathways underlying mitophagy in myocardial stress may provide therapeutic targets for myocardial protection interventions.
Collapse
Affiliation(s)
- Haizhe Ji
- Department of Cardiology, The First Medical Center, Chinese People's Liberation Army Hospital, Medical School of Chinese People's Liberation Army, Beijing, China.,Department of Cardiology, The First Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Dan Wu
- Department of Cardiology, The First Medical Center, Chinese People's Liberation Army Hospital, Medical School of Chinese People's Liberation Army, Beijing, China
| | - O'Maley Kimberlee
- School of Public Health, University of California, Berkeley, Berkeley, CA, United States
| | - Ruibing Li
- Department of Clinical Laboratory Medicine, The First Medical Center, Medical School of Chinese People's Liberation Army, Beijing, China
| | - Geng Qian
- Department of Cardiology, The First Medical Center, Chinese People's Liberation Army Hospital, Medical School of Chinese People's Liberation Army, Beijing, China
| |
Collapse
|
25
|
Li S, Zhang J, Liu C, Wang Q, Yan J, Hui L, Jia Q, Shan H, Tao L, Zhang M. The Role of Mitophagy in Regulating Cell Death. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6617256. [PMID: 34113420 PMCID: PMC8154277 DOI: 10.1155/2021/6617256] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/27/2021] [Accepted: 04/24/2021] [Indexed: 02/06/2023]
Abstract
Mitochondria are multifaceted organelles that serve to power critical cellular functions, including act as power generators of the cell, buffer cytosolic calcium overload, production of reactive oxygen species, and modulating cell survival. The structure and the cellular location of mitochondria are critical for their function and depend on highly regulated activities such as mitochondrial quality control (MQC) mechanisms. The MQC is regulated by several sets of processes: mitochondrial biogenesis, mitochondrial fusion and fission, mitophagy, and other mitochondrial proteostasis mechanisms such as mitochondrial unfolded protein response (mtUPR) or mitochondrial-derived vesicles (MDVs). These processes are important for the maintenance of mitochondrial homeostasis, and alterations in the mitochondrial function and signaling are known to contribute to the dysregulation of cell death pathways. Recent studies have uncovered regulatory mechanisms that control the activity of the key components for mitophagy. In this review, we discuss how mitophagy is controlled and how mitophagy impinges on health and disease through regulating cell death.
Collapse
Affiliation(s)
- Sunao Li
- Department of Forensic Sciences, School of Basic Medicine and Biological Sciences, Affilated Guangji Hospital, Soochow University, Suzhou, China
| | - Jiaxin Zhang
- Department of Forensic Sciences, School of Basic Medicine and Biological Sciences, Affilated Guangji Hospital, Soochow University, Suzhou, China
| | - Chao Liu
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Qianliang Wang
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Jun Yan
- Department of Orthopedics, The Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Li Hui
- Department of Forensic Sciences, School of Basic Medicine and Biological Sciences, Affilated Guangji Hospital, Soochow University, Suzhou, China
| | - Qiufang Jia
- Department of Forensic Sciences, School of Basic Medicine and Biological Sciences, Affilated Guangji Hospital, Soochow University, Suzhou, China
| | - Haiyan Shan
- Department of Obstetrics and Gynecology, The Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou, China
| | - Luyang Tao
- Department of Forensic Sciences, School of Basic Medicine and Biological Sciences, Affilated Guangji Hospital, Soochow University, Suzhou, China
| | - Mingyang Zhang
- Department of Forensic Sciences, School of Basic Medicine and Biological Sciences, Affilated Guangji Hospital, Soochow University, Suzhou, China
| |
Collapse
|
26
|
Franco R, Rivas-Santisteban R, Navarro G, Pinna A, Reyes-Resina I. Genes Implicated in Familial Parkinson's Disease Provide a Dual Picture of Nigral Dopaminergic Neurodegeneration with Mitochondria Taking Center Stage. Int J Mol Sci 2021; 22:4643. [PMID: 33924963 PMCID: PMC8124903 DOI: 10.3390/ijms22094643] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Revised: 04/16/2021] [Accepted: 04/20/2021] [Indexed: 12/13/2022] Open
Abstract
The mechanism of nigral dopaminergic neuronal degeneration in Parkinson's disease (PD) is unknown. One of the pathological characteristics of the disease is the deposition of α-synuclein (α-syn) that occurs in the brain from both familial and sporadic PD patients. This paper constitutes a narrative review that takes advantage of information related to genes (SNCA, LRRK2, GBA, UCHL1, VPS35, PRKN, PINK1, ATP13A2, PLA2G6, DNAJC6, SYNJ1, DJ-1/PARK7 and FBXO7) involved in familial cases of Parkinson's disease (PD) to explore their usefulness in deciphering the origin of dopaminergic denervation in many types of PD. Direct or functional interactions between genes or gene products are evaluated using the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) database. The rationale is to propose a map of the interactions between SNCA, the gene encoding for α-syn that aggregates in PD, and other genes, the mutations of which lead to early-onset PD. The map contrasts with the findings obtained using animal models that are the knockout of one of those genes or that express the mutated human gene. From combining in silico data from STRING-based assays with in vitro and in vivo data in transgenic animals, two likely mechanisms appeared: (i) the processing of native α-syn is altered due to the mutation of genes involved in vesicular trafficking and protein processing, or (ii) α-syn mutants alter the mechanisms necessary for the correct vesicular trafficking and protein processing. Mitochondria are a common denominator since both mechanisms require extra energy production, and the energy for the survival of neurons is obtained mainly from the complete oxidation of glucose. Dopamine itself can result in an additional burden to the mitochondria of dopaminergic neurons because its handling produces free radicals. Drugs acting on G protein-coupled receptors (GPCRs) in the mitochondria of neurons may hopefully end up targeting those receptors to reduce oxidative burden and increase mitochondrial performance. In summary, the analysis of the data of genes related to familial PD provides relevant information on the etiology of sporadic cases and might suggest new therapeutic approaches.
Collapse
Affiliation(s)
- Rafael Franco
- Department Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain; (R.F.); (R.R.-S.); (I.R.-R.)
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, 28031 Madrid, Spain;
| | - Rafael Rivas-Santisteban
- Department Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain; (R.F.); (R.R.-S.); (I.R.-R.)
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, 28031 Madrid, Spain;
| | - Gemma Navarro
- Centro de Investigación Biomédica en Red Enfermedades Neurodegenerativas (CiberNed), Instituto de Salud Carlos III, 28031 Madrid, Spain;
- Department Biochemistry and Physiology, School of Pharmacy and Food Sciences, University of Barcelona, 08028 Barcelona, Spain
| | - Annalisa Pinna
- National Research Council of Italy (CNR), Neuroscience Institute–Cagliari, Cittadella Universitaria, Blocco A, SP 8, Km 0.700, 09042 Monserrato (CA), Italy
| | - Irene Reyes-Resina
- Department Biochemistry and Molecular Biomedicine, University of Barcelona, 08028 Barcelona, Spain; (R.F.); (R.R.-S.); (I.R.-R.)
| |
Collapse
|
27
|
Wu D, Lu J, Ma Y, Cao Y, Zhang T. Mitochondrial dynamics and mitophagy involved in MPA-capped CdTe quantum dots-induced toxicity in the human liver carcinoma (HepG2) cell line. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2021; 274:115681. [PMID: 33308872 DOI: 10.1016/j.envpol.2020.115681] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/19/2020] [Revised: 09/14/2020] [Accepted: 09/15/2020] [Indexed: 06/12/2023]
Abstract
Quantum dots (QDs) are nanoparticles of inorganic semiconductors and have great promise in various applications. Many studies have indicated that mitochondria are the main organelles for the distribution and toxic effects of QDs. However, the underlying mechanism of QDs interacting with mitochondria and affecting their function is unknown. Here, we report the mechanism of toxic effects of 3-mercaptopropionic acid (MPA)-capped CdTe QDs on mitochondria. Human liver carcinoma (HepG2) cells were exposed to 25, 50 and 100 μmol/L of MPA-capped CdTe QDs. The results indicated that MPA-capped CdTe QDs inhibited HepG2 cell proliferation and increased the extracellular release of LDH in a concentration-dependent manner. Furthermore, MPA-capped CdTe QDs caused reactive oxygen species (ROS) generation and cell damage through intrinsic apoptotic pathway. MPA-capped CdTe QDs can also lead to the destruction of mitochondrial cristae, elevation of intracellular Ca2+ levels, decreased mitochondrial transmembrane potential and ATP production. Finally, we showed that MPA-capped CdTe QDs inhibited mitochondrial fission, mitochondrial inner membrane fusion and mitophagy. Taken together, MPA-capped CdTe QDs induced significant mitochondrial dysfunction, which may be caused by imbalanced mitochondrial fission/fusion and mitophagy inhibition. These findings provide insights into the regulatory mechanisms involved in MPA-capped CdTe QDs-induced mitochondrial dysfunction.
Collapse
Affiliation(s)
- Daming Wu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University and Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, 210009, China
| | - Jie Lu
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University and Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, 210009, China
| | - Ying Ma
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University and Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, 210009, China
| | - Yuna Cao
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University and Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, 210009, China
| | - Ting Zhang
- Key Laboratory of Environmental Medicine Engineering, Ministry of Education, School of Public Health, Southeast University and Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, 210009, China.
| |
Collapse
|
28
|
Ng MYW, Wai T, Simonsen A. Quality control of the mitochondrion. Dev Cell 2021; 56:881-905. [PMID: 33662258 DOI: 10.1016/j.devcel.2021.02.009] [Citation(s) in RCA: 185] [Impact Index Per Article: 61.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2020] [Revised: 01/06/2021] [Accepted: 02/08/2021] [Indexed: 12/22/2022]
Abstract
Mitochondria are essential organelles that execute and coordinate various metabolic processes in the cell. Mitochondrial dysfunction severely affects cell fitness and contributes to disease. Proper organellar function depends on the biogenesis and maintenance of mitochondria and its >1,000 proteins. As a result, the cell has evolved mechanisms to coordinate protein and organellar quality control, such as the turnover of proteins via mitochondria-associated degradation, the ubiquitin-proteasome system, and mitoproteases, as well as the elimination of mitochondria through mitophagy. Specific quality control mechanisms are engaged depending upon the nature and severity of mitochondrial dysfunction, which can also feed back to elicit transcriptional or proteomic remodeling by the cell. Here, we will discuss the current understanding of how these different quality control mechanisms are integrated and overlap to maintain protein and organellar quality and how they may be relevant for cellular and organismal health.
Collapse
Affiliation(s)
- Matthew Yoke Wui Ng
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0372 Oslo, Norway; Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, 0316 Oslo, Norway
| | - Timothy Wai
- Institut Pasteur CNRS UMR 3691, 25-28 Rue du Docteur Roux, Paris, France.
| | - Anne Simonsen
- Department of Molecular Medicine, Institute of Basic Medical Sciences, University of Oslo, 0372 Oslo, Norway; Centre for Cancer Cell Reprogramming, Institute of Clinical Medicine, University of Oslo, 0316 Oslo, Norway.
| |
Collapse
|
29
|
Ebeid DE, Khalafalla FG, Broughton KM, Monsanto MM, Esquer CY, Sacchi V, Hariharan N, Korski KI, Moshref M, Emathinger J, Cottage CT, Quijada PJ, Nguyen JH, Alvarez R, Völkers M, Konstandin MH, Wang BJ, Firouzi F, Navarrete JM, Gude NA, Goumans MJ, Sussman MA. Pim1 maintains telomere length in mouse cardiomyocytes by inhibiting TGFβ signalling. Cardiovasc Res 2021; 117:201-211. [PMID: 32176281 PMCID: PMC7797214 DOI: 10.1093/cvr/cvaa066] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2019] [Accepted: 03/13/2019] [Indexed: 12/26/2022] Open
Abstract
AIMS Telomere attrition in cardiomyocytes is associated with decreased contractility, cellular senescence, and up-regulation of proapoptotic transcription factors. Pim1 is a cardioprotective kinase that antagonizes the aging phenotype of cardiomyocytes and delays cellular senescence by maintaining telomere length, but the mechanism remains unknown. Another pathway responsible for regulating telomere length is the transforming growth factor beta (TGFβ) signalling pathway where inhibiting TGFβ signalling maintains telomere length. The relationship between Pim1 and TGFβ has not been explored. This study delineates the mechanism of telomere length regulation by the interplay between Pim1 and components of TGFβ signalling pathways in proliferating A549 cells and post-mitotic cardiomyocytes. METHODS AND RESULTS Telomere length was maintained by lentiviral-mediated overexpression of PIM1 and inhibition of TGFβ signalling in A549 cells. Telomere length maintenance was further demonstrated in isolated cardiomyocytes from mice with cardiac-specific overexpression of PIM1 and by pharmacological inhibition of TGFβ signalling. Mechanistically, Pim1 inhibited phosphorylation of Smad2, preventing its translocation into the nucleus and repressing expression of TGFβ pathway genes. CONCLUSION Pim1 maintains telomere lengths in cardiomyocytes by inhibiting phosphorylation of the TGFβ pathway downstream effectors Smad2 and Smad3, which prevents repression of telomerase reverse transcriptase. Findings from this study demonstrate a novel mechanism of telomere length maintenance and provide a potential target for preserving cardiac function.
Collapse
Affiliation(s)
- David E Ebeid
- Department of Biology, San Diego State University, North Life Sciences, 426, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Farid G Khalafalla
- Department of Biology, San Diego State University, North Life Sciences, 426, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Kathleen M Broughton
- Department of Biology, San Diego State University, North Life Sciences, 426, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Megan M Monsanto
- Department of Biology, San Diego State University, North Life Sciences, 426, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Carolina Y Esquer
- Department of Biology, San Diego State University, North Life Sciences, 426, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Veronica Sacchi
- Department of Biology, San Diego State University, North Life Sciences, 426, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Nirmala Hariharan
- Department of Biology, San Diego State University, North Life Sciences, 426, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Kelli I Korski
- Department of Biology, San Diego State University, North Life Sciences, 426, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Maryam Moshref
- Department of Biology, San Diego State University, North Life Sciences, 426, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Jacqueline Emathinger
- Department of Biology, San Diego State University, North Life Sciences, 426, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Christopher T Cottage
- Department of Biology, San Diego State University, North Life Sciences, 426, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Pearl J Quijada
- Department of Biology, San Diego State University, North Life Sciences, 426, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Jonathan H Nguyen
- Department of Biology, San Diego State University, North Life Sciences, 426, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Roberto Alvarez
- Department of Biology, San Diego State University, North Life Sciences, 426, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Mirko Völkers
- Department of Biology, San Diego State University, North Life Sciences, 426, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Mathias H Konstandin
- Department of Biology, San Diego State University, North Life Sciences, 426, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Bingyan J Wang
- Department of Biology, San Diego State University, North Life Sciences, 426, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Fareheh Firouzi
- Department of Biology, San Diego State University, North Life Sciences, 426, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Julian M Navarrete
- Department of Biology, San Diego State University, North Life Sciences, 426, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Natalie A Gude
- Department of Biology, San Diego State University, North Life Sciences, 426, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Marie-Jose Goumans
- Department of Biology, San Diego State University, North Life Sciences, 426, 5500 Campanile Drive, San Diego, CA 92182, USA
| | - Mark A Sussman
- Department of Biology, San Diego State University, North Life Sciences, 426, 5500 Campanile Drive, San Diego, CA 92182, USA
| |
Collapse
|
30
|
PIM1 Promotes Survival of Cardiomyocytes by Upregulating c-Kit Protein Expression. Cells 2020; 9:cells9092001. [PMID: 32878131 PMCID: PMC7563506 DOI: 10.3390/cells9092001] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 08/21/2020] [Accepted: 08/27/2020] [Indexed: 12/17/2022] Open
Abstract
Enhancing cardiomyocyte survival is crucial to blunt deterioration of myocardial structure and function following pathological damage. PIM1 (Proviral Insertion site in Murine leukemia virus (PIM) kinase 1) is a cardioprotective serine threonine kinase that promotes cardiomyocyte survival and antagonizes senescence through multiple concurrent molecular signaling cascades. In hematopoietic stem cells, PIM1 interacts with the receptor tyrosine kinase c-Kit upstream of the ERK (Extracellular signal-Regulated Kinase) and Akt signaling pathways involved in cell proliferation and survival. The relationship between PIM1 and c-Kit activity has not been explored in the myocardial context. This study delineates the interaction between PIM1 and c-Kit leading to enhanced protection of cardiomyocytes from stress. Elevated c-Kit expression is induced in isolated cardiomyocytes from mice with cardiac-specific overexpression of PIM1. Co-immunoprecipitation and proximity ligation assay reveal protein–protein interaction between PIM1 and c-Kit. Following treatment with Stem Cell Factor, PIM1-overexpressing cardiomyocytes display elevated ERK activity consistent with c-Kit receptor activation. Functionally, elevated c-Kit expression confers enhanced protection against oxidative stress in vitro. This study identifies the mechanistic relationship between PIM1 and c-Kit in cardiomyocytes, demonstrating another facet of cardioprotection regulated by PIM1 kinase.
Collapse
|
31
|
van der Vlag M, Havekes R, Heckman PRA. The contribution of Parkin, PINK1 and DJ-1 genes to selective neuronal degeneration in Parkinson's disease. Eur J Neurosci 2020; 52:3256-3268. [PMID: 31991026 PMCID: PMC7496448 DOI: 10.1111/ejn.14689] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Revised: 12/13/2019] [Accepted: 01/08/2020] [Indexed: 12/14/2022]
Abstract
Parkinson's disease (PD) is characterised by selective and severe degeneration of the substantia nigra pars compacta and the locus coeruleus (LC), which underlies the most prominent symptoms. Although α-synuclein accumulation has long been established to play a causal role in the disease, it alone cannot explain the selective degenerative pattern. Recent evidence shows that the selective vulnerability could arise due to the large presence of cytosolic catecholamines and Ca2+ ions in the substantia nigra pars compacta and LC specifically that can be aberrantly affected by α-synuclein accumulation. Moreover, each has its own toxic potential, and disturbance of one can exacerbate the toxic effects of the others. This presents a mechanism unique to these areas that can lead to a vicious degenerative cycle. Interestingly, in familial variants of PD, the exact same brain areas are affected, implying the underlying process is likely the same. However, the exact disease mechanisms of many of these genetic variants remain unclear. Here, we review the effects of the PD-related genes Parkin, PINK1 and DJ-1. We establish that these mutant varieties can set in motion the same degenerative process involving α-synuclein, cytosolic catecholamines and Ca2+ . Additionally, we show indications that model organisms might not accurately represent all components of this central mechanism, explaining why Parkin, PINK1 and DJ-1 model organisms often lack a convincing PD-like phenotype.
Collapse
Affiliation(s)
- Marc van der Vlag
- Neurobiology Expertise GroupGroningen Institute for Evolutionary Life Sciences (GELIFES)University of GroningenGroningenThe Netherlands
| | - Robbert Havekes
- Neurobiology Expertise GroupGroningen Institute for Evolutionary Life Sciences (GELIFES)University of GroningenGroningenThe Netherlands
| | - Pim R. A. Heckman
- Neurobiology Expertise GroupGroningen Institute for Evolutionary Life Sciences (GELIFES)University of GroningenGroningenThe Netherlands
| |
Collapse
|
32
|
Hernandez‐Resendiz S, Prunier F, Girao H, Dorn G, Hausenloy DJ. Targeting mitochondrial fusion and fission proteins for cardioprotection. J Cell Mol Med 2020; 24:6571-6585. [PMID: 32406208 PMCID: PMC7299693 DOI: 10.1111/jcmm.15384] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2020] [Revised: 04/20/2020] [Accepted: 04/22/2020] [Indexed: 01/05/2023] Open
Abstract
New treatments are needed to protect the myocardium against the detrimental effects of acute ischaemia/reperfusion (IR) injury following an acute myocardial infarction (AMI), in order to limit myocardial infarct (MI) size, preserve cardiac function and prevent the onset of heart failure (HF). Given the critical role of mitochondria in energy production for cardiac contractile function, prevention of mitochondrial dysfunction during acute myocardial IRI may provide novel cardioprotective strategies. In this regard, the mitochondrial fusion and fissions proteins, which regulate changes in mitochondrial morphology, are known to impact on mitochondrial quality control by modulating mitochondrial biogenesis, mitophagy and the mitochondrial unfolded protein response. In this article, we review how targeting these inter-related processes may provide novel treatment targets and new therapeutic strategies for reducing MI size, preventing the onset of HF following AMI.
Collapse
Affiliation(s)
- Sauri Hernandez‐Resendiz
- National Heart Research Institute SingaporeNational Heart Centre SingaporeSingaporeSingapore
- Cardiovascular & Metabolic Disorders ProgramDuke‐National University of Singapore Medical SchoolSingaporeSingapore
- Centro de Biotecnologia‐FEMSATecnologico de MonterreyNuevo LeonMexico
| | - Fabrice Prunier
- Institut MITOVASCCNRS UMR 6015 INSERM U1083University Hospital Center of AngersUniversity of AngersAngersFrance
| | - Henrique Girao
- Faculty of MedicineCoimbra Institute for Clinical and Biomedical Research (iCBR)University of CoimbraPortugal
- Center for Innovative Biomedicine and BiotechnologyUniversity of CoimbraCoimbraPortugal
- Clinical Academic Centre of Coimbra (CACC)CoimbraPortugal
| | - Gerald Dorn
- Department of Internal MedicineCenter for PharmacogenomicsWashington University School of MedicineSt. LouisMOUSA
| | - Derek J. Hausenloy
- National Heart Research Institute SingaporeNational Heart Centre SingaporeSingaporeSingapore
- Cardiovascular & Metabolic Disorders ProgramDuke‐National University of Singapore Medical SchoolSingaporeSingapore
- Yong Loo Lin School of MedicineNational University SingaporeSingaporeSingapore
- The Hatter Cardiovascular InstituteUniversity College LondonLondonUK
- Cardiovascular Research CenterCollege of Medical and Health SciencesAsia UniversityTaichungTaiwan
| | | |
Collapse
|
33
|
Xiang Q, Wu M, Zhang L, Fu W, Yang J, Zhang B, Zheng Z, Zhang H, Lao Y, Xu H. Gerontoxanthone I and Macluraxanthone Induce Mitophagy and Attenuate Ischemia/Reperfusion Injury. Front Pharmacol 2020; 11:452. [PMID: 32351391 PMCID: PMC7175665 DOI: 10.3389/fphar.2020.00452] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 03/23/2020] [Indexed: 12/26/2022] Open
Abstract
Mitophagy is a crucial process in controlling mitochondrial biogenesis. Balancing mitophagy and mitochondrial functions is required for maintaining cellular homeostasis. In this study, we found that Gerontoxanthone I (GeX1) and Macluraxanthone (McX), xanthone derivatives isolated from Garcinia bracteata C. Y. Wu ex Y. H. Li, induced Parkin puncta accumulation and promoted mitophagy. GeX1 and McX treatment induced the degradation of mitophagy-related proteins such as Tom20 and Tim23. GeX1 and McX directly stabilized PTEN-induced putative kinase 1 (PINK1) on the outer membrane of the mitochondria, and then recruited Parkin to mitochondria. This significantly induced phosphorylation and ubiquitination of Parkin, suggesting that GeX1 and McX mediate mitophagy through the PINK1-Parkin pathway. Transfecting ParkinS65A or pretreated MG132 abolished the induction effects of GeX1 and McX on mitophagy. Furthermore, GeX1 and McX treatment decreased cell death and the level of ROS in an ischemia/reperfusion (IR) injury model in H9c2 cells compared to a control group. Taken together, our data suggested that GeX1 and McX induce PINK1-Parkin–mediated mitophagy and attenuate myocardial IR injury in vitro.
Collapse
Affiliation(s)
- Qian Xiang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Man Wu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenwei Fu
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jinling Yang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Baojun Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhaoqing Zheng
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hong Zhang
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yuanzhi Lao
- School of Pharmacy, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Engineering Research Center of Shanghai Colleges for TCM New Drug Discovery, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Hongxi Xu
- Institute of Cardiovascular Disease of Integrated Traditional Chinese and Western Medicine, Shuguang Hospital affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
34
|
Neto da Silva K, Garbin Cappellaro L, Ueda CN, Rodrigues L, Pertile Remor A, Martins RDP, Latini A, Glaser V. Glyphosate-based herbicide impairs energy metabolism and increases autophagy in C6 astroglioma cell line. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART A 2020; 83:153-167. [PMID: 32085696 DOI: 10.1080/15287394.2020.1731897] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
Several investigators demonstrated that glyphosate formulations produce neurotoxicity associated with oxidative stress, alterations in glutamatergic system, inhibition of acetylcholinesterase activity and mitochondrial dysfunction. However, the underlying molecular mechanisms following exposure to this herbicide on astrocytes are unclear. Thus, the aim of the present study was to determine the activity of enzymes related to energy metabolism, in addition to oxidative stress parameters, mitochondrial mass, nuclear area, and autophagy in astrocytes treated with a glyphosate-based herbicide. Our results showed that 24 h exposure to a glyphosate-based herbicide decreased (1) cell viability, (2) activities of mitochondrial respiratory chain enzymes and creatine kinase (CK), (3) mitochondrial mass, and (4) nuclear area in rat astroglioma cell line (C6 cells). However, non-protein thiol (NPSH) levels were increased but catalase activity was not changed in cells exposed to the herbicide at non-cytotoxic concentrations. Low glyphosate concentrations elevated content of cells positive to autophagy-related proteins. Nuclear factor erythroid 2-related factor (Nrf2), NAD(P)H dehydrogenase [quinone] 1 (NQO1) and PTEN-induced kinase 1 (PINK1) labeling were not markedly altered in cells exposed to glyphosate at the same concentrations that an increase in NPSH levels and positive cells to autophagy were found. It is conceivable that mitochondria and CK may be glyphosate-based herbicides targets. Further, autophagy induction and NPSH increase may be mechanisms initiated to avoid oxidative stress and cell death. However, more studies are needed to clarify the role of autophagy in astrocytes exposed to the herbicide and which components of the formulation might be triggering the effects observed here.
Collapse
Affiliation(s)
- Katriane Neto da Silva
- Laboratório De Biologia Celular, Coordenadoria Especial De Ciências Biológicas E Agronômicas, Universidade Federal De Santa Catarina - Campus De Curitibanos, Curitibanos, Brazil
| | - Laura Garbin Cappellaro
- Laboratório De Biologia Celular, Coordenadoria Especial De Ciências Biológicas E Agronômicas, Universidade Federal De Santa Catarina - Campus De Curitibanos, Curitibanos, Brazil
| | - Caroline Naomi Ueda
- Laboratório De Biologia Celular, Coordenadoria Especial De Ciências Biológicas E Agronômicas, Universidade Federal De Santa Catarina - Campus De Curitibanos, Curitibanos, Brazil
| | - Luana Rodrigues
- Laboratório De Biologia Celular, Coordenadoria Especial De Ciências Biológicas E Agronômicas, Universidade Federal De Santa Catarina - Campus De Curitibanos, Curitibanos, Brazil
| | - Aline Pertile Remor
- Programa De Pós-graduação Em Biociências E Saúde, Universidade Do Oeste De Santa Catarina - Campus Joaçaba, Joaçaba, Brazil
| | - Roberta de Paula Martins
- Departamento De Ciências Da Saúde, Universidade Federal De Santa Catarina - Campus De Araranguá, Araranguá, Brazil
| | - Alexandra Latini
- Laboratório De Bioenergética E Estresse Oxidativo, Departamento De Bioquímica, Universidade Federal De Santa Catarina - Campus De Florianópolis, Florianópolis, Brazil
| | - Viviane Glaser
- Laboratório De Biologia Celular, Coordenadoria Especial De Ciências Biológicas E Agronômicas, Universidade Federal De Santa Catarina - Campus De Curitibanos, Curitibanos, Brazil
| |
Collapse
|
35
|
Park SY, Koh HC. FUNDC1 regulates receptor-mediated mitophagy independently of the PINK1/Parkin-dependent pathway in rotenone-treated SH-SY5Y cells. Food Chem Toxicol 2020; 137:111163. [PMID: 32001317 DOI: 10.1016/j.fct.2020.111163] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2019] [Revised: 01/09/2020] [Accepted: 01/23/2020] [Indexed: 12/21/2022]
Abstract
Upon mitochondrial stress, PINK1 and Parkin cooperatively mediate a response that removes damaged mitochondria. In addition to the PINK1/Parkin pathway, the FUNDC1, mitophagy receptor regulates mitochondrial clearance. It is not clear whether these systems coordinate to mediate mitophagy in response to stress. Rotenone caused an increase in LC3II expression, and FUNDC1-knocked down cells showed remarkably reduced LC3 expression compared to control cells. In addition, treatment of cells with autophagy flux inhibitor, chloroquine, induced further accumulation of LC3-II, suggesting that mitophagy induced by rotenone is due to involvement of mitochondrial FUNDC1. Rotenone treatment resulted in PINK1 stabilization on the outer mitochondrial membrane and a subsequent increase in recruitment of Parkin from the cytosol to the abnormal mitochondria, as well as physical interaction of PINK1 with Parkin in the mitochondria of rotenone-treated cells. Interestingly, knockdown of FUNDC1 did not alter PINK1/Parkin expression in the mitochondrial fraction of rotenone-treated cells. Our findings indicate that FUNDC1 involves in receptor-mediated mitophagy separately from PINK1/Parkin-dependent mitophagy. Furthermore, inhibiting mitophagy by FUNDC1 or PINK1 knockdown accelerated rotenone-induced cytotoxicity. Taken together, our findings suggest that rotenone can be induced both receptor-mediated and PINK1/Parkin-dependent mitophagy for mitochondrial clearance, and that mitophagy by removing damaged mitochondria, has cytoprotective effects.
Collapse
Affiliation(s)
- Si Yeon Park
- Department of Pharmacology, College of Medicine, Hanyang University, Sungdong-Gu, Haengdang-Dong 17, 133-79, Seoul, Republic of Korea; Hanyang Biomedical Research Institute, Sungdong-Gu, Haengdang-Dong 17, 133-79, Seoul, Republic of Korea
| | - Hyun Chul Koh
- Department of Pharmacology, College of Medicine, Hanyang University, Sungdong-Gu, Haengdang-Dong 17, 133-79, Seoul, Republic of Korea; Graduate School of Biomedical Science and Engineering, Hanyang University, Sungdong-Gu, Haengdang-Dong 17, 133-79, Seoul, Republic of Korea; Hanyang Biomedical Research Institute, Sungdong-Gu, Haengdang-Dong 17, 133-79, Seoul, Republic of Korea.
| |
Collapse
|
36
|
Cells deficient for Krüppel-like factor 4 exhibit mitochondrial dysfunction and impaired mitophagy. Eur J Cell Biol 2020; 99:151061. [DOI: 10.1016/j.ejcb.2019.151061] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Revised: 11/09/2019] [Accepted: 11/28/2019] [Indexed: 01/19/2023] Open
|
37
|
Morales PE, Arias-Durán C, Ávalos-Guajardo Y, Aedo G, Verdejo HE, Parra V, Lavandero S. Emerging role of mitophagy in cardiovascular physiology and pathology. Mol Aspects Med 2019; 71:100822. [PMID: 31587811 DOI: 10.1016/j.mam.2019.09.006] [Citation(s) in RCA: 106] [Impact Index Per Article: 21.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Accepted: 09/23/2019] [Indexed: 01/02/2023]
Abstract
Healthy mitochondrial function is imperative for most tissues, but especially those with a high energy demand. Robust evidence linking mitochondrial dysfunction with cardiovascular disease has demonstrated that mitochondrial activity is highly relevant to cardiac muscle performance. Mitochondrial homeostasis is maintained through coordination among the processes that comprise the so-called mitochondrial dynamics machinery. The most-studied elements of cardiac mitochondrial dynamics are mitochondrial fission and fusion, biogenesis and degradation. Selective autophagic removal of mitochondria (mitophagy) is essential for clearing away defective mitochondria but can lead to cell damage and death if not tightly controlled. In cardiovascular cells such as cardiomyocytes and cardiac fibroblasts, mitophagy is involved in metabolic activity, cell differentiation, apoptosis and other physiological processes related to major phenotypic changes. Modulation of mitophagy has detrimental and/or beneficial outcomes in various cardiovascular diseases, suggesting that a deeper understanding of the mechanisms underlying mitochondrial degradation in the heart could provide valuable clinical insights. Here, we discuss current evidence supporting the role of mitophagy in cardiac pathophysiology, with an emphasis on different research models and their interpretations; basic concepts related to this selective autophagy; and the most commonly used experimental approaches for studying this mechanism. Finally, we provide a comprehensive literature analysis on the role of mitophagy in heart failure, ischemia/reperfusion, diabetic cardiomyopathy and other cardiovascular diseases, as well as its potential biomedical applications.
Collapse
Affiliation(s)
- Pablo E Morales
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Carla Arias-Durán
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile; Autophagy Research Center, Universidad de Chile, Santiago, Chile
| | - Yáreni Ávalos-Guajardo
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Geraldine Aedo
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile
| | - Hugo E Verdejo
- Advanced Center for Chronic Diseases (ACCDiS), División de Enfermedades Cardiovasculares, Facultad de Medicina, Pontificia Universidad Católica de Chile, Santiago, Chile
| | - Valentina Parra
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile; Autophagy Research Center, Universidad de Chile, Santiago, Chile; Network for the Study of High-lethality Cardiopulmonary Diseases (REECPAL), Universidad de Chile, Santiago, Chile.
| | - Sergio Lavandero
- Advanced Center for Chronic Diseases (ACCDiS), Faculty of Chemical and Pharmaceutical Sciences and Faculty of Medicine, Universidad de Chile, Santiago, Chile; Corporación Centro de Estudios Científicos de las Enfermedades Crónicas (CECEC), Santiago, Chile; Department of Internal Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
38
|
Tan VP, Smith JM, Tu M, Yu JD, Ding EY, Miyamoto S. Dissociation of mitochondrial HK-II elicits mitophagy and confers cardioprotection against ischemia. Cell Death Dis 2019; 10:730. [PMID: 31570704 PMCID: PMC6768853 DOI: 10.1038/s41419-019-1965-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2019] [Revised: 07/11/2019] [Accepted: 09/09/2019] [Indexed: 12/23/2022]
Abstract
Preservation of mitochondrial integrity is critical for maintaining cellular homeostasis. Mitophagy is a mitochondria-specific type of autophagy which eliminates damaged mitochondria thereby contributing to mitochondrial quality control. Depolarization of the mitochondrial membrane potential is an established mechanism for inducing mitophagy, mediated through PINK1 stabilization and Parkin recruitment to mitochondria. Hexokinase-II (HK-II) which catalyzes the first step in glucose metabolism, also functions as a signaling molecule to regulate cell survival, and a significant fraction of cellular HK-II is associated with mitochondria (mitoHK-II). We demonstrate here that pharmacological interventions and adenoviral expression of a mitoHK-II dissociating peptide which reduce mitoHK-II levels lead to robust increases in mitochondrial Parkin and ubiquitination of mitochondrial proteins in cardiomyocytes and in a human glioblastoma cell line 1321N1, independent of mitochondrial membrane depolarization or PINK1 accumulation. MitoHK-II dissociation-induced mitophagy was demonstrated using Mito-Keima in cardiomyocytes and in 1321N1 cells. Subjecting cardiomyocytes or the in vivo heart to ischemia leads to modest dissociation of mitoHK-II. This response is potentiated by expression of the mitoHK-II dissociating peptide, which increases Parkin recruitment to mitochondria and, importantly, provides cardioprotection against ischemic stress. These results suggest that mitoHK-II dissociation is a physiologically relevant cellular event that is induced by ischemic stress, the enhancement of which protects against ischemic damage. The mechanism which underlies the effects of mitoHK-II dissociation can be attributed to the ability of Bcl2-associated athanogene 5 (BAG5), an inhibitor of Parkin, to localize to mitochondria and form a molecular complex with HK-II. Overexpression of BAG5 attenuates while knockdown of BAG5 sensitizes the effect of mitoHK-II dissociation on mitophagy. We suggest that HK-II, a glycolytic molecule, can function as a sensor for metabolic derangements at mitochondria to trigger mitophagy, and modulating the intracellular localization of HK-II could be a novel way of regulating mitophagy to prevent cell death induced by ischemic stress.
Collapse
Affiliation(s)
- Valerie P Tan
- Department of Pharmacology, University of California, San Diego, 9500 Gilman drive, La Jolla, CA, 92093-0636, USA
| | - Jeffrey M Smith
- Department of Pharmacology, University of California, San Diego, 9500 Gilman drive, La Jolla, CA, 92093-0636, USA
| | - Michelle Tu
- Department of Pharmacology, University of California, San Diego, 9500 Gilman drive, La Jolla, CA, 92093-0636, USA
| | - Justin D Yu
- Department of Pharmacology, University of California, San Diego, 9500 Gilman drive, La Jolla, CA, 92093-0636, USA
| | - Eric Y Ding
- Department of Pharmacology, University of California, San Diego, 9500 Gilman drive, La Jolla, CA, 92093-0636, USA
| | - Shigeki Miyamoto
- Department of Pharmacology, University of California, San Diego, 9500 Gilman drive, La Jolla, CA, 92093-0636, USA.
| |
Collapse
|
39
|
Mishra PK, Adameova A, Hill JA, Baines CP, Kang PM, Downey JM, Narula J, Takahashi M, Abbate A, Piristine HC, Kar S, Su S, Higa JK, Kawasaki NK, Matsui T. Guidelines for evaluating myocardial cell death. Am J Physiol Heart Circ Physiol 2019; 317:H891-H922. [PMID: 31418596 DOI: 10.1152/ajpheart.00259.2019] [Citation(s) in RCA: 147] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Cell death is a fundamental process in cardiac pathologies. Recent studies have revealed multiple forms of cell death, and several of them have been demonstrated to underlie adverse cardiac remodeling and heart failure. With the expansion in the area of myocardial cell death and increasing concerns over rigor and reproducibility, it is important and timely to set a guideline for the best practices of evaluating myocardial cell death. There are six major forms of regulated cell death observed in cardiac pathologies, namely apoptosis, necroptosis, mitochondrial-mediated necrosis, pyroptosis, ferroptosis, and autophagic cell death. In this article, we describe the best methods to identify, measure, and evaluate these modes of myocardial cell death. In addition, we discuss the limitations of currently practiced myocardial cell death mechanisms.
Collapse
Affiliation(s)
- Paras K Mishra
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Adriana Adameova
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Comenius University of Bratislava, Bratislava, Slovakia
| | - Joseph A Hill
- Departments of Medicine (Cardiology) and Molecular Biology, University of Texas Southwestern Medical Center, Dallas, Texas
| | - Christopher P Baines
- Department of Biomedical Sciences, Dalton Cardiovascular Research Center, University of Missouri-Columbia, Columbia, Missouri
| | - Peter M Kang
- Cardiovascular Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - James M Downey
- Department of Physiology and Cell Biology, College of Medicine, University of South Alabama, Mobile, Alabama
| | - Jagat Narula
- Mount Sinai Heart, Icahn School of Medicine at Mount Sinai Hospital, New York, New York
| | - Masafumi Takahashi
- Division of Inflammation Research, Center of Molecular Medicine, Jichi Medical University, Tochigi, Japan
| | - Antonio Abbate
- Virginia Commonwealth University, Pauley Heart Center, Richmond, Virginia
| | - Hande C Piristine
- Department of Medicine (Cardiology), University of Texas Southwestern Medical Center, Dallas, Texas
| | - Sumit Kar
- Department of Cellular and Integrative Physiology, University of Nebraska Medical Center, Omaha, Nebraska
| | - Shi Su
- Cardiovascular Division, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, Massachusetts
| | - Jason K Higa
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii
| | - Nicholas K Kawasaki
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii
| | - Takashi Matsui
- Department of Anatomy, Biochemistry, and Physiology, John A. Burns School of Medicine, University of Hawaii at Manoa, Honolulu, Hawaii
| |
Collapse
|
40
|
Mitochondrial Uncoupling: A Key Controller of Biological Processes in Physiology and Diseases. Cells 2019; 8:cells8080795. [PMID: 31366145 PMCID: PMC6721602 DOI: 10.3390/cells8080795] [Citation(s) in RCA: 260] [Impact Index Per Article: 52.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2019] [Revised: 07/26/2019] [Accepted: 07/28/2019] [Indexed: 12/27/2022] Open
Abstract
Mitochondrial uncoupling can be defined as a dissociation between mitochondrial membrane potential generation and its use for mitochondria-dependent ATP synthesis. Although this process was originally considered a mitochondrial dysfunction, the identification of UCP-1 as an endogenous physiological uncoupling protein suggests that the process could be involved in many other biological processes. In this review, we first compare the mitochondrial uncoupling agents available in term of mechanistic and non-specific effects. Proteins regulating mitochondrial uncoupling, as well as chemical compounds with uncoupling properties are discussed. Second, we summarize the most recent findings linking mitochondrial uncoupling and other cellular or biological processes, such as bulk and specific autophagy, reactive oxygen species production, protein secretion, cell death, physical exercise, metabolic adaptations in adipose tissue, and cell signaling. Finally, we show how mitochondrial uncoupling could be used to treat several human diseases, such as obesity, cardiovascular diseases, or neurological disorders.
Collapse
|
41
|
Miyai T, Vasanth S, Melangath G, Deshpande N, Kumar V, Benischke AS, Chen Y, Price MO, Price FW, Jurkunas UV. Activation of PINK1-Parkin-Mediated Mitophagy Degrades Mitochondrial Quality Control Proteins in Fuchs Endothelial Corneal Dystrophy. THE AMERICAN JOURNAL OF PATHOLOGY 2019; 189:2061-2076. [PMID: 31361992 DOI: 10.1016/j.ajpath.2019.06.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/01/2018] [Revised: 05/25/2019] [Accepted: 06/11/2019] [Indexed: 11/19/2022]
Abstract
Corneal endothelium (CE) is a monolayer of mitochondria-rich cells, critical for maintaining corneal transparency compatible with clear vision. Fuchs endothelial corneal dystrophy (FECD) is a heterogeneous, genetically complex disorder, where oxidative stress plays a key role in the rosette formation during the degenerative loss of CE. Increased mitochondrial fragmentation along with excessive mitophagy activation has been detected in FECD; however, the mechanism of aberrant mitochondrial dynamics in CE cell loss is poorly understood. Here, the role of oxidative stress in mitophagy activation in FECD is investigated. Immunoblotting of FECD ex vivo specimens revealed an accumulation of PINK1 and phospho-Parkin (Ser65) along with loss of total Parkin and total Drp1. Similarly, modeling of rosette formation with menadione (MN), led to phospho-Parkin accumulation in fragmented mitochondria resulting in mitophagy-induced mitochondrial clearance, albeit possibly in a PINK1-independent manner. Loss of PINK1, phospho-Drp1, and total Drp1 was prominent after MN-induced oxidative stress, but not after mitochondrial depolarization by carbonyl cyanide m-chlorophenyl hydrazone. Moreover, MN-induced mitophagy led to degradation of Parkin along with sequestration of Drp1 and PINK1 that was rescued by mitophagy inhibition. This study shows that in FECD, intracellular oxidative stress induces Parkin-mediated mitochondrial fragmentation where endogenous Drp1 and PINK1 are sequestered and degraded by mitophagy during degenerative loss of post-mitotic cells of ocular tissue.
Collapse
Affiliation(s)
- Takashi Miyai
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Opthalmology, Harvard Medical School, Boston, Massachusetts
| | - Shivakumar Vasanth
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Opthalmology, Harvard Medical School, Boston, Massachusetts
| | - Geetha Melangath
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Opthalmology, Harvard Medical School, Boston, Massachusetts
| | - Neha Deshpande
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Opthalmology, Harvard Medical School, Boston, Massachusetts
| | - Varun Kumar
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Opthalmology, Harvard Medical School, Boston, Massachusetts
| | - Anne-Sophie Benischke
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Opthalmology, Harvard Medical School, Boston, Massachusetts
| | - Yuming Chen
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Opthalmology, Harvard Medical School, Boston, Massachusetts
| | | | | | - Ula V Jurkunas
- Schepens Eye Research Institute, Massachusetts Eye and Ear, Department of Opthalmology, Harvard Medical School, Boston, Massachusetts.
| |
Collapse
|
42
|
Yao L, Chen H, Wu Q, Xie K. Hydrogen-rich saline alleviates inflammation and apoptosis in myocardial I/R injury via PINK-mediated autophagy. Int J Mol Med 2019; 44:1048-1062. [PMID: 31524220 PMCID: PMC6657957 DOI: 10.3892/ijmm.2019.4264] [Citation(s) in RCA: 56] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2018] [Accepted: 06/26/2019] [Indexed: 12/14/2022] Open
Abstract
Ischemia/reperfusion (I/R)-induced inflammatory reaction is one of the most important elements in myocardial I/R injury. In addition, autophagy serves an important role in normal cardiac homeostasis, and obstructions to the autophagy process lead to severe consequences for the heart. Hydrogen exerts an effective therapeutic role in numerous diseases associated with I/R injury via its anti-inflammation, anti-apoptosis and anti-oxidative properties. Therefore, the present study investigated the effect of hydrogen on the myocardial inflammation response and apoptosis in myocardial ischemic/reperfusion (MI/R) injury, and further explored the mechanism of PTEN-induced kinase 1 (PINK1)/Parkin-induced mitophagy in the protection of hydrogen on MI/R injury. MI/R injury was performed by surgical ligation of the left coronary artery in vivo and H9C2 cell injury was performed by hypoxia/reoxygenation (H/R) in vitro. Hydrogen-rich saline was administered twice through intraperitoneal injection at a daily dose of 10 ml/kg following the operation in the in vivo model, and hydrogen-rich medium culture was used for cells instead of normal medium in vitro. The infarction size of hearts, the levels of creati-nine kinase-muscle/brain (CK-MB) and cardiac troponin I (cTnI), cardiac function, cell viability and lactate dehydrogenase (LDH) release, levels of cytokines, apoptosis and the expression of autophagy-associated proteins were detected in the different treatment groups in vivo and in vitro. The results demonstrated that treatment with hydrogen improved the myocardial infarction size of hearts, cardiac function, apoptosis and cytokine release following MI/R in rats. In vitro, hydrogen improved cell viability and LDH release following hypoxia/reoxygenation in myocardial cells. In addition, it was demonstrated that hydrogen exerted an anti-inflammatory and anti-apoptotic effect in myocardial cells induced by H/R via PINK1/Parkin mediated autophagy. These results suggested that hydrogen-rich saline alleviated the inflammation response and apoptosis induced by MI/R or H/R in vivo or in vitro, and that hydrogen-rich saline contributed to the increased expression of proteins associated with autophagy. In summary, the present study indicated that treatment with hydrogen-rich saline improved the inflammatory response and apoptosis in MI/R via PINK1/Parkin-mediated mitophagy.
Collapse
Affiliation(s)
- Li Yao
- Sixth Department of Cardiology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Hongguang Chen
- Department of Anesthesiology, General Hospital of Tianjin Medical University, Tianjin Institute of Anesthesiology, Tianjin 300054, P.R. China
| | - Qinghua Wu
- Sixth Department of Cardiology, Cangzhou Central Hospital, Cangzhou, Hebei 061001, P.R. China
| | - Keliang Xie
- Department of Anesthesiology, General Hospital of Tianjin Medical University, Tianjin Institute of Anesthesiology, Tianjin 300054, P.R. China
| |
Collapse
|
43
|
Yang HX, Wang P, Wang NN, Li SD, Yang MH. Tongxinluo Ameliorates Myocardial Ischemia-Reperfusion Injury Mainly via Activating Parkin-Mediated Mitophagy and Downregulating Ubiquitin-Proteasome System. Chin J Integr Med 2019; 27:542-550. [PMID: 31227964 DOI: 10.1007/s11655-019-3166-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/14/2019] [Indexed: 01/29/2023]
Abstract
OBJECTIVE To investigate the protective effects and mechanism of Chinese herbal compound Tongxinluo Capsule (, TXL) on the Parkin-mediated mitophagy and the ubiquitin-proteasome system in a rat model of myocardial ischemia-reperfusion injury (MIRI). METHODS Seventy adult male Sprague-Dawley rats were randomly divided into 7 groups: sham group, MIRI group, low- and high-dose TXL (0.5 and 1 g·kg-1·d-1, respectively) groups, atorvastatin (ATV) group (7.2 g·kg-1·d-1), chloroquine (CQ) group (10 g·kg-1·d-1), and highdose TXL + CQ group. After pharmacological administration for 7 days, rats underwent left anterior descending artery ligation surgery to establish the MIRI models with 50 min ischemia followed by 4 h reperfusion. Blood was taken for cardiac troponin I (cTnI) detection and hearts were harvested for infarct staining and apoptosis detection. The autophagy or mitophagy proteins and ubiquitinated proteins were detected by Western blotting. RESULTS Compared with the sham group, the MIRI group exhibited a larger infarcted area (27.13%±0.01%, P<0.01), a higher apoptotic index (34.33%±2.03% vs.1.81%±0.03%, P<0.01), and higher cTnI expression (14.18±1.01 vs. 7.96±0.32, P<0.01). The mitochondrial integrity was damaged in the MIRI group, while TXL and ATV alleviated the damage of MIRI. More autophagosomes were observed in the high-dose TXL group than in the MIRI group (7.00±0.58 vs. 4.33±1.15, P<0.05). More amounts of PTEN-induced putative kinase protein 1 (PINK1) and Parkin translocated onto the mitochondria were detected in the high-dose TXL group than in the MIRI group (P<0.05). The ubiquitin response was signifificantly downregulated in the high-dose TXL group relative to the MIRI group (P<0.05). CQ administration abolished the activation of autophagy flux and the PINK1/ Parkin pathway induced by high-dose of TXL. CONCLUSIONS TXL ameliorates MIRI via activating Parkin-mediated mitophagy in rats. The downregulation of the ubiquitin-proteasome system is also involved.
Collapse
Affiliation(s)
- Hong-Xing Yang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Peng Wang
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100029, China
| | - Ning-Ning Wang
- Institute of Radiation Medicine, Academy of Military Medical Sciences, Beijing, 100850, China
| | - Shao-Dan Li
- Department of Traditional Chinese Medicine, Chinese People's Liberation Army General Hospital, Beijing, 100853, China
| | - Ming-Hui Yang
- Department of Traditional Chinese Medicine, Chinese People's Liberation Army General Hospital, Beijing, 100853, China.
| |
Collapse
|
44
|
Wang Y, Liu N, Lu B. Mechanisms and roles of mitophagy in neurodegenerative diseases. CNS Neurosci Ther 2019; 25:859-875. [PMID: 31050206 PMCID: PMC6566062 DOI: 10.1111/cns.13140] [Citation(s) in RCA: 107] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2019] [Revised: 03/23/2019] [Accepted: 04/06/2019] [Indexed: 12/21/2022] Open
Abstract
Mitochondria are double‐membrane‐encircled organelles existing in most eukaryotic cells and playing important roles in energy production, metabolism, Ca2+ buffering, and cell signaling. Mitophagy is the selective degradation of mitochondria by autophagy. Mitophagy can effectively remove damaged or stressed mitochondria, which is essential for cellular health. Thanks to the implementation of genetics, cell biology, and proteomics approaches, we are beginning to understand the mechanisms of mitophagy, including the roles of ubiquitin‐dependent and receptor‐dependent signals on damaged mitochondria in triggering mitophagy. Mitochondrial dysfunction and defective mitophagy have been broadly associated with neurodegenerative diseases. This review is aimed at summarizing the mechanisms of mitophagy in higher organisms and the roles of mitophagy in the pathogenesis of neurodegenerative diseases. Although many studies have been devoted to elucidating the mitophagy process, a deeper understanding of the mechanisms leading to mitophagy defects in neurodegenerative diseases is required for the development of new therapeutic interventions, taking into account the multifactorial nature of diseases and the phenotypic heterogeneity of patients.
Collapse
Affiliation(s)
- Yan Wang
- Department of Pharmacology, College of Pharmaceutical Science, Soochow University, Suzhou, China
| | - Na Liu
- Department of Pharmacology, College of Pharmaceutical Science, Soochow University, Suzhou, China
| | - Bingwei Lu
- Department of Pathology, Stanford University School of Medicine, Stanford, California
| |
Collapse
|
45
|
Affiliation(s)
- Sarah E Shires
- From the Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla (S.E.S., A.B.G.); and Departments of Medicine and Cell Biology, Wilf Family Cardiovascular Research Institute, Diabetes Research Center, Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY (R.N.K.)
| | - Richard N Kitsis
- From the Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla (S.E.S., A.B.G.); and Departments of Medicine and Cell Biology, Wilf Family Cardiovascular Research Institute, Diabetes Research Center, Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY (R.N.K.)
| | - Åsa B Gustafsson
- From the Department of Pharmacology, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla (S.E.S., A.B.G.); and Departments of Medicine and Cell Biology, Wilf Family Cardiovascular Research Institute, Diabetes Research Center, Albert Einstein Cancer Center, Albert Einstein College of Medicine, Bronx, NY (R.N.K.).
| |
Collapse
|
46
|
Bloemberg D, Quadrilatero J. Autophagy, apoptosis, and mitochondria: molecular integration and physiological relevance in skeletal muscle. Am J Physiol Cell Physiol 2019; 317:C111-C130. [PMID: 31017800 DOI: 10.1152/ajpcell.00261.2018] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Apoptosis and autophagy are processes resulting from the integration of cellular stress and death signals. Their individual importance is highlighted by the lethality of various mouse models missing apoptosis or autophagy-related genes. In addition to their independent roles, significant overlap exists with respect to the signals that stimulate these processes as well as their effector consequences. While these cellular systems exemplify the programming redundancies that underlie many fundamental biological mechanisms, their intertwined relationship means that dysfunction can promote pathology. Although both autophagic and apoptotic signaling are active in skeletal muscle during various diseases and atrophy, their specific roles here are somewhat unique. Given our growing understanding of how specific changes at the cellular level impact whole-organism physiology, there is an equally growing interest in pharmacological manipulation of apoptosis and/or autophagy for altering human physiology and health.
Collapse
Affiliation(s)
- Darin Bloemberg
- Department of Kinesiology, University of Waterloo , Waterloo, Ontario , Canada
| | - Joe Quadrilatero
- Department of Kinesiology, University of Waterloo , Waterloo, Ontario , Canada
| |
Collapse
|
47
|
Woodall BP, Orogo AM, Najor RH, Cortez MQ, Moreno ER, Wang H, Divakaruni AS, Murphy AN, Gustafsson ÅB. Parkin does not prevent accelerated cardiac aging in mitochondrial DNA mutator mice. JCI Insight 2019; 5:127713. [PMID: 30990467 DOI: 10.1172/jci.insight.127713] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
The E3 ubiquitin ligase Parkin plays an important role in regulating clearance of dysfunctional or unwanted mitochondria in tissues, including the heart. However, whether Parkin also functions to prevent cardiac aging by maintaining a healthy population of mitochondria is still unclear. Here, we have examined the role of Parkin in the context of mtDNA damage and myocardial aging using a mouse model carrying a proofreading defective mitochondrial DNA polymerase gamma (POLG). We observed both decreased Parkin protein levels and development of cardiac hypertrophy in POLG hearts with age; however, cardiac hypertrophy in POLG mice was neither rescued, nor worsened by cardiac specific overexpression or global deletion of Parkin, respectively. Unexpectedly, mitochondrial fitness did not substantially decline with age in POLG mice when compared to WT. We found that baseline mitophagy receptor-mediated mitochondrial turnover and biogenesis were enhanced in aged POLG hearts. We also observed the presence of megamitochondria in aged POLG hearts. Thus, these processes may limit the accumulation of dysfunctional mitochondria as well as the degree of cardiac functional impairment in the aging POLG heart. Overall, our results demonstrate that Parkin is dispensable for constitutive mitochondrial quality control in a mtDNA mutation model of cardiac aging.
Collapse
Affiliation(s)
| | - Amabel M Orogo
- Skaggs School of Pharmacy and Pharmaceutical Sciences and
| | - Rita H Najor
- Skaggs School of Pharmacy and Pharmaceutical Sciences and
| | | | | | - Hongxia Wang
- Skaggs School of Pharmacy and Pharmaceutical Sciences and
| | - Ajit S Divakaruni
- Department of Pharmacology, University of California, San Diego, La Jolla, California, USA
| | - Anne N Murphy
- Department of Pharmacology, University of California, San Diego, La Jolla, California, USA
| | - Åsa B Gustafsson
- Skaggs School of Pharmacy and Pharmaceutical Sciences and.,Department of Pharmacology, University of California, San Diego, La Jolla, California, USA
| |
Collapse
|
48
|
Thai PN, Seidlmayer LK, Miller C, Ferrero M, Dorn GW, Schaefer S, Bers DM, Dedkova EN. Mitochondrial Quality Control in Aging and Heart Failure: Influence of Ketone Bodies and Mitofusin-Stabilizing Peptides. Front Physiol 2019; 10:382. [PMID: 31024341 PMCID: PMC6467974 DOI: 10.3389/fphys.2019.00382] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Accepted: 03/19/2019] [Indexed: 12/12/2022] Open
Abstract
Aim: Aging and heart failure (HF) are each characterized by increased mitochondrial damage, which may contribute to further cardiac dysfunction. Mitophagy in response to mitochondrial damage can improve cardiovascular health. HF is also characterized by increased formation and consumption of ketone bodies (KBs), which may activate mitophagy and provide an endogenous mechanism to limit the adverse effects of mitochondrial damage. However, the role of KBs in activation of mitophagy in aging and HF has not been evaluated. Methods: We assessed mitophagy by measuring mitochondrial Parkin accumulation and LC3-mediated autophagosome formation in cardiomyocytes from young (2.5 months), aged (2.5 years), and aged rabbits with HF (2.5 years) induced by aortic insufficiency and stenosis. Levels of reactive oxygen species (ROS) generation and redox balance were monitored using genetically encoded sensors ORP1-roGFP2 and GRX1-roGFP2, targeted to mitochondrial or cytosolic compartments, respectively. Results: Young rabbits exhibited limited mitochondrial Parkin accumulation with small (~1 μm2) puncta. Those small Parkin puncta increased four-fold in aged rabbit hearts, accompanied by elevated LC3-mediated autophagosome formation. HF hearts exhibited fewer small puncta, but many very large Parkin-rich regions (4-5 μm2) with completely depolarized mitochondria. Parkin protein expression was barely detectable in young animals and was much higher in aged and maximal in HF hearts. Expression of mitofusin 2 (MFN2) and dynamin-related protein 1 (DRP1) was reduced by almost 50% in HF, consistent with improper fusion-fission, contributing to mitochondrial Parkin build-up. The KB β-hydroxybutyrate (β-OHB) enhanced mitophagy in young and aging myocytes, but not in HF where β-OHB further increased the number of cells with giant Parkin-rich regions. This β-OHB effect on Parkin-rich areas was prevented by cell-permeable TAT-MP1Gly peptide (thought to promote MFN2-dependent fusion). Basal levels of mitochondrial ROS were highest in HF, while cytosolic ROS was highest in aged compared to HF myocytes, suggesting that cytosolic ROS promotes Parkin recruitment to the mitochondria. Conclusion: We conclude that elevated KB levels were beneficial for mitochondrial repair in the aging heart. However, an impaired MFN2-DRP1-mediated fusion-fission process in HF reduced this benefit, as well as Parkin degradation and mitophagic signaling cascade.
Collapse
Affiliation(s)
- Phung N. Thai
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, United States
| | - Lea K. Seidlmayer
- Division of Cardiology, Department of Internal Medicine, University Hospital Würzburg, Würzburg, Germany
- Comprehensive Heart Failure Center, University of Würzburg, Würzburg, Germany
| | - Charles Miller
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Maura Ferrero
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Gerald W. Dorn
- Department of Internal Medicine, Center for Pharmacogenomics, Washington University School of Medicine, St. Louis, MO, United States
| | - Saul Schaefer
- Division of Cardiovascular Medicine, Department of Internal Medicine, University of California, Davis, Davis, CA, United States
- Department of Veterans Affairs, Northern California Health Care System, Mather, CA, United States
| | - Donald M. Bers
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| | - Elena N. Dedkova
- Department of Pharmacology, School of Medicine, University of California, Davis, Davis, CA, United States
| |
Collapse
|
49
|
Cadete VJJ, Vasam G, Menzies KJ, Burelle Y. Mitochondrial quality control in the cardiac system: An integrative view. Biochim Biophys Acta Mol Basis Dis 2018; 1865:782-796. [PMID: 30472159 DOI: 10.1016/j.bbadis.2018.11.018] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2018] [Revised: 10/29/2018] [Accepted: 11/15/2018] [Indexed: 01/26/2023]
Abstract
Recent studies have led to the discovery of multiple mitochondrial quality control (mQC) processes that operate at various scales, ranging from the degradation of proteins by mitochondrial proteases to the degradation of selected cargos or entire organelles in lysosomes. While the mechanisms governing these mQC processes are progressively being delineated, their role and importance remain unclear. Converging evidence however point to a complex system whereby multiple and partly overlapping processes are recruited to orchestrate a cell type specific mQC response that is adapted to the physiological state and level of stress encountered. Knowledge gained from basic model systems of mQC therefore need to be integrated within organ-specific (patho)physiological frameworks. Building on this notion, this article focuses on mQC in the heart, where developmental metabolic reprogramming, sustained contraction, and multiple pathophysiological conditions pose broadly different constraints. We provide an overview of current knowledge of mQC processes, and discuss their implication in cardiac mQC under normal and diseased conditions.
Collapse
Affiliation(s)
- Virgilio J J Cadete
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Sinclair Centre for Regenerative Medicine, Ottawa Hospital Research Institute, Ottawa, ON, Canada
| | - Goutham Vasam
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada
| | - Keir J Menzies
- Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada; Department of Biochemistry, Microbiology and Immunology, University of Ottawa Brain and Mind Research Institute and Centre for Neuromuscular Disease, University of Ottawa, Ottawa, ON, Canada
| | - Yan Burelle
- Department of Cellular and Molecular Medicine, Faculty of Medicine, University of Ottawa, Ottawa, ON, Canada; Interdisciplinary School of Health Sciences, Faculty of Health Sciences, University of Ottawa, Ottawa, ON, Canada.
| |
Collapse
|
50
|
Cardiac-specific Conditional Knockout of the 18-kDa Mitochondrial Translocator Protein Protects from Pressure Overload Induced Heart Failure. Sci Rep 2018; 8:16213. [PMID: 30385779 PMCID: PMC6212397 DOI: 10.1038/s41598-018-34451-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2018] [Accepted: 10/18/2018] [Indexed: 01/17/2023] Open
Abstract
Heart failure (HF) is characterized by abnormal mitochondrial calcium (Ca2+) handling, energy failure and impaired mitophagy resulting in contractile dysfunction and myocyte death. We have previously shown that the 18-kDa mitochondrial translocator protein of the outer mitochondrial membrane (TSPO) can modulate mitochondrial Ca2+ uptake. Experiments were designed to test the role of the TSPO in a murine pressure-overload model of HF induced by transverse aortic constriction (TAC). Conditional, cardiac-specific TSPO knockout (KO) mice were generated using the Cre-loxP system. TSPO-KO and wild-type (WT) mice underwent TAC for 8 weeks. TAC-induced HF significantly increased TSPO expression in WT mice, associated with a marked reduction in systolic function, mitochondrial Ca2+ uptake, complex I activity and energetics. In contrast, TSPO-KO mice undergoing TAC had preserved ejection fraction, and exhibited fewer clinical signs of HF and fibrosis. Mitochondrial Ca2+ uptake and energetics were restored in TSPO KO mice, associated with decreased ROS, improved complex I activity and preserved mitophagy. Thus, HF increases TSPO expression, while preventing this increase limits the progression of HF, preserves ATP production and decreases oxidative stress, thereby preventing metabolic failure. These findings suggest that pharmacological interventions directed at TSPO may provide novel therapeutics to prevent or treat HF.
Collapse
|