1
|
Kaur S, Khullar N, Navik U, Bali A, Bhatti GK, Bhatti JS. Multifaceted role of dynamin-related protein 1 in cardiovascular disease: From mitochondrial fission to therapeutic interventions. Mitochondrion 2024; 78:101904. [PMID: 38763184 DOI: 10.1016/j.mito.2024.101904] [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: 12/06/2023] [Revised: 05/01/2024] [Accepted: 05/15/2024] [Indexed: 05/21/2024]
Abstract
Mitochondria are central to cellular energy production and metabolic regulation, particularly in cardiomyocytes. These organelles constantly undergo cycles of fusion and fission, orchestrated by key proteins like Dynamin-related Protein 1 (Drp-1). This review focuses on the intricate roles of Drp-1 in regulating mitochondrial dynamics, its implications in cardiovascular health, and particularly in myocardial infarction. Drp-1 is not merely a mediator of mitochondrial fission; it also plays pivotal roles in autophagy, mitophagy, apoptosis, and necrosis in cardiac cells. This multifaceted functionality is often modulated through various post-translational alterations, and Drp-1's interaction with intracellular calcium (Ca2 + ) adds another layer of complexity. We also explore the pathological consequences of Drp-1 dysregulation, including increased reactive oxygen species (ROS) production and endothelial dysfunction. Furthermore, this review delves into the potential therapeutic interventions targeting Drp-1 to modulate mitochondrial dynamics and improve cardiovascular outcomes. We highlight recent findings on the interaction between Drp-1 and sirtuin-3 and suggest that understanding this interaction may open new avenues for therapeutically modulating endothelial cells, fibroblasts, and cardiomyocytes. As the cardiovascular system increasingly becomes the focal point of aging and chronic disease research, understanding the nuances of Drp-1's functionality can lead to innovative therapeutic approaches.
Collapse
Affiliation(s)
- Satinder Kaur
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda India
| | - Naina Khullar
- Department of Zoology, Mata Gujri College, Fatehgarh Sahib, Punjab, India
| | - Umashanker Navik
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Anjana Bali
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Gurjit Kaur Bhatti
- Department of Medical Lab Technology, University Institute of Applied Health Sciences, Chandigarh University, Mohali India.
| | - Jasvinder Singh Bhatti
- Laboratory of Translational Medicine and Nanotherapeutics, Department of Human Genetics and Molecular Medicine, School of Health Sciences, Central University of Punjab, Bathinda India.
| |
Collapse
|
2
|
Upadhayay S, Kumar P. Mitochondrial targeted antioxidants as potential therapy for huntington's disease. Pharmacol Rep 2024; 76:693-713. [PMID: 38982016 DOI: 10.1007/s43440-024-00619-z] [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: 03/22/2024] [Revised: 06/18/2024] [Accepted: 06/19/2024] [Indexed: 07/11/2024]
Abstract
Huntington's disease (HD) is an inherited neurodegenerative disorder caused by an expansion in CAG repeat on huntington (Htt) gene, leading to a degeneration of GABAergic medium spiny neurons (MSNs) in the striatum, resulting in the generation of reactive oxygen species, and decrease antioxidant activity. These pathophysiological alterations impair mitochondrial functions, leading to an increase in involuntary hyperkinetic movement. However, researchers investigated the neuroprotective effect of antioxidants using various animal models. Still, their impact is strictly limited to curtailing oxidative stress and increasing the antioxidant enzyme in the brain, which is less effective in HD. Meanwhile, researchers discovered Mitochondria-targeted antioxidants (MTAXs) that can improve mitochondrial functions and antioxidant activity through the modulation of mitochondrial signaling pathways, including peroxisome proliferator-activated receptor (PPAR)-coactivator 1 (PGC-1α), dynamin-related protein 1 (Drp1), mitochondrial fission protein 1 (Fis1), and Silent mating type information regulation 2 homolog 1 (SIRT-1), showing neuroprotective effects in HD. The present review discusses the clinical and preclinical studies that investigate the neuroprotective effect of MTAXs (SS31, XJB-5-131, MitoQ, bezafibrate, rosiglitazone, meldonium, coenzyme Q10, etc.) in HD. This brief literature review will help to understand the relevance of MTAXs in HD and enlighten the importance of MTAXs in future drug discovery and development.
Collapse
Affiliation(s)
- Shubham Upadhayay
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, Punjab, 151401, India.
| |
Collapse
|
3
|
Kumar Saini S, Singh D. Mitochondrial mechanisms in Cerebral Ischemia-Reperfusion Injury: Unravelling the intricacies. Mitochondrion 2024; 77:101883. [PMID: 38631511 DOI: 10.1016/j.mito.2024.101883] [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: 01/01/2024] [Revised: 04/05/2024] [Accepted: 04/13/2024] [Indexed: 04/19/2024]
Abstract
Cerebral ischemic stroke is a major contributor to physical impairments and premature death worldwide. The available reperfusion therapies for stroke in the form of mechanical thrombectomy and intravenous thrombolysis increase the risk of cerebral ischemia-reperfusion (I-R) injury due to sudden restoration of blood supply to the ischemic region. The injury is manifested by hemorrhagic transformation, worsening of neurological impairments, cerebral edema, and progression to infarction in surviving patients. A complex network of multiple pathological processes has been known to be involved in the pathogenesis of I-R injury. Primarily, 3 major contributors namely oxidative stress, neuroinflammation, and mitochondrial failure have been well studied in I-R injury. A transcription factor, Nrf2 (Nuclear factor erythroid 2-related factor 2) plays a crucial defensive role in resisting the deleterious effects of I-R injury and potentiating the cellular protective mechanisms. In this review, we delve into the critical function of mitochondria and Nrf2 in the context of cerebral I-R injury. We summarized how oxidative stress, neuroinflammation, and mitochondrial anomaly contribute to the pathophysiology of I-R injury and further elaborated the role of Nrf2 as a pivotal guardian of cellular integrity. The review further highlighted Nrf2 as a putative therapeutic target for mitochondrial dysfunction in cerebral I-R injury management.
Collapse
Affiliation(s)
- Shiv Kumar Saini
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, Himachal Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India
| | - Damanpreet Singh
- Pharmacology and Toxicology Laboratory, Dietetics and Nutrition Technology Division, CSIR-Institute of Himalayan Bioresource Technology, Palampur 176061, Himachal Pradesh, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad 201002, India.
| |
Collapse
|
4
|
Launay N, Lopez-Erauskin J, Bianchi P, Guha S, Parameswaran J, Coppa A, Torreni L, Schlüter A, Fourcade S, Paredes-Fuentes AJ, Artuch R, Casasnovas C, Ruiz M, Pujol A. Imbalanced mitochondrial dynamics contributes to the pathogenesis of X-linked adrenoleukodystrophy. Brain 2024; 147:2069-2084. [PMID: 38763511 DOI: 10.1093/brain/awae038] [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: 10/02/2023] [Revised: 12/20/2023] [Accepted: 01/21/2024] [Indexed: 05/21/2024] Open
Abstract
The peroxisomal disease adrenoleukodystrophy (X-ALD) is caused by loss of the transporter of very-long-chain fatty acids (VLCFAs), ABCD1. An excess of VLCFAs disrupts essential homeostatic functions crucial for axonal maintenance, including redox metabolism, glycolysis and mitochondrial respiration. As mitochondrial function and morphology are intertwined, we set out to investigate the role of mitochondrial dynamics in X-ALD models. Using quantitative 3D transmission electron microscopy, we revealed mitochondrial fragmentation in corticospinal axons in Abcd1- mice. In patient fibroblasts, an excess of VLCFAs triggers mitochondrial fragmentation through the redox-dependent phosphorylation of DRP1 (DRP1S616). The blockade of DRP1-driven fission by the peptide P110 effectively preserved mitochondrial morphology. Furthermore, mRNA inhibition of DRP1 not only prevented mitochondrial fragmentation but also protected axonal health in a Caenorhabditis elegans model of X-ALD, underscoring DRP1 as a potential therapeutic target. Elevated levels of circulating cell-free mtDNA in patients' CSF align this leukodystrophy with primary mitochondrial disorders. Our findings underscore the intricate interplay between peroxisomal dysfunction, mitochondrial dynamics and axonal integrity in X-ALD, shedding light on potential avenues for therapeutic intervention.
Collapse
Affiliation(s)
- Nathalie Launay
- Neurometabolic Diseases Laboratory, Institute of Neuropathology, IDIBELL, 08908 L'Hospitalet de Llobregat, Barcelona, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 28029 Madrid, Spain
| | - Jone Lopez-Erauskin
- Neurometabolic Diseases Laboratory, Institute of Neuropathology, IDIBELL, 08908 L'Hospitalet de Llobregat, Barcelona, Spain
- Department of Cellular and Molecular Medicine, Ludwig Institute for Cancer Research, University of California at San Diego, La Jolla, CA 92093, USA
| | - Patrizia Bianchi
- Neurometabolic Diseases Laboratory, Institute of Neuropathology, IDIBELL, 08908 L'Hospitalet de Llobregat, Barcelona, Spain
- Physiology and Immunology, Facultat de Medicina, Institut de Neurociències and Department of Cell Biology, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain
| | - Sanjib Guha
- Neurometabolic Diseases Laboratory, Institute of Neuropathology, IDIBELL, 08908 L'Hospitalet de Llobregat, Barcelona, Spain
- Nautilus Biotechnology, San Carlos, CA 94070, USA
| | - Janani Parameswaran
- Neurometabolic Diseases Laboratory, Institute of Neuropathology, IDIBELL, 08908 L'Hospitalet de Llobregat, Barcelona, Spain
- Department of Cell Biology, Emory University, Atlanta, GA 30322, USA
| | - Andrea Coppa
- Neurometabolic Diseases Laboratory, Institute of Neuropathology, IDIBELL, 08908 L'Hospitalet de Llobregat, Barcelona, Spain
| | - Lorenzo Torreni
- Neurometabolic Diseases Laboratory, Institute of Neuropathology, IDIBELL, 08908 L'Hospitalet de Llobregat, Barcelona, Spain
- Programa de Doctorat en Biomedicina, Universitat de Barcelona, 08193 Barcelona, Spain
| | - Agatha Schlüter
- Neurometabolic Diseases Laboratory, Institute of Neuropathology, IDIBELL, 08908 L'Hospitalet de Llobregat, Barcelona, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 28029 Madrid, Spain
| | - Stéphane Fourcade
- Neurometabolic Diseases Laboratory, Institute of Neuropathology, IDIBELL, 08908 L'Hospitalet de Llobregat, Barcelona, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 28029 Madrid, Spain
| | - Abraham J Paredes-Fuentes
- Division of Inborn Errors of Metabolism-IBC, Biochemistry and Molecular Genetics Department, Hospital Clínic de Barcelona, 08028 Barcelona, Spain
| | - Rafael Artuch
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 28029 Madrid, Spain
- Clinical Biochemistry Department, Institut de Recerca Sant Joan de Déu, Hospital Sant Joan de Déu, 08950 Barcelona, Spain
| | - Carlos Casasnovas
- Neurometabolic Diseases Laboratory, Institute of Neuropathology, IDIBELL, 08908 L'Hospitalet de Llobregat, Barcelona, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 28029 Madrid, Spain
- Neuromuscular Unit, Neurology Department, Bellvitge University Hospital, Universitat de Barcelona, 08907 Lhospitalet de Llobregat, Barcelona, Spain
| | - Montserrat Ruiz
- Neurometabolic Diseases Laboratory, Institute of Neuropathology, IDIBELL, 08908 L'Hospitalet de Llobregat, Barcelona, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 28029 Madrid, Spain
| | - Aurora Pujol
- Neurometabolic Diseases Laboratory, Institute of Neuropathology, IDIBELL, 08908 L'Hospitalet de Llobregat, Barcelona, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, 28029 Madrid, Spain
- Catalan Institution of Research and Advanced Studies (ICREA), 08010 Barcelona, Spain
| |
Collapse
|
5
|
Makvand M, Mirtorabi SD, Campbell A, Zali A, Ahangari G. Exploring neuroadaptive cellular pathways in chronic morphine exposure: An in-vitro analysis of cabergoline and Mdivi-1 co-treatment effects on the autophagy-apoptosis axis. J Cell Biochem 2024; 125:e30558. [PMID: 38577900 DOI: 10.1002/jcb.30558] [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: 09/20/2023] [Revised: 02/08/2024] [Accepted: 03/07/2024] [Indexed: 04/06/2024]
Abstract
The complex impacts of prolonged morphine exposure continue to be a significant focus in the expanding area of addiction studies. This research investigates the effectiveness of a combined treatment using Cabergoline and Mdivi-1 to counteract the neuroadaptive changes caused by in vitro morphine treatment. The impact of Methadone, Cabergoline, and a combination of Cabergoline and Mdivi-1 on the cellular and molecular responses associated with Morphine-induced changes was studied in human Neuroblastoma (SK-N-MC) and Glioblastoma (U87-MG) cell lines that were exposed to prolong Morphine treatment. Cabergoline and Mdivi-1 combined treatment effectively influenced the molecular alterations associated with neuroadaptation in chronic morphine-exposed neural cells. This combination therapy normalized autophagy and reduced oxidative stress by enhancing total-antioxidant capacity, mitigating apoptosis, restoring BDNF expression, and balancing apoptotic elements. Our research outlines morphine's dual role in modulating mitochondrial dynamics via the dysregulation of the autophagy-apoptosis axis. This emphasizes the significant involvement of DRP1 activity in neurological adaptation processes, as well as disturbances in the dopaminergic pathway during in vitro chronic exposure to morphine in neural cells. This study proposes a novel approach by recommending the potential effectiveness of combining Cabergoline and Mdivi-1 to modulate the neuroadaptations caused by morphine. Additionally, we identified BDNF and PCNA in neural cells as potential neuroprotective markers for assessing the effectiveness of drugs against opioid toxicity, emphasizing the need for further validation. The study uncovers diverse effects observed in pretreated morphine glioblastoma cells under treatment with Cabergoline and methadone. This highlights the potential for new treatments in the DRD2 pathway and underscores the importance of investigating the interplay between autophagy and apoptosis to advance research in managing cancer-related pain. The study necessitates an in-depth investigation into the relationship between autophagy and apoptosis, with a specific emphasis on protein interactions and the dynamics of cell signaling.
Collapse
Affiliation(s)
- Mina Makvand
- Department of Medical Genetics, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| | | | - Arezoo Campbell
- Department of Pharmaceutical Sciences, Western University of Health Sciences, Pomona, California, USA
| | - Alireza Zali
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Ghasem Ahangari
- Department of Medical Genetics, National Institute of Genetic Engineering and Biotechnology, Tehran, Iran
| |
Collapse
|
6
|
Tripathi K, Ben-Shachar D. Mitochondria in the Central Nervous System in Health and Disease: The Puzzle of the Therapeutic Potential of Mitochondrial Transplantation. Cells 2024; 13:410. [PMID: 38474374 DOI: 10.3390/cells13050410] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 02/21/2024] [Accepted: 02/23/2024] [Indexed: 03/14/2024] Open
Abstract
Mitochondria, the energy suppliers of the cells, play a central role in a variety of cellular processes essential for survival or leading to cell death. Consequently, mitochondrial dysfunction is implicated in numerous general and CNS disorders. The clinical manifestations of mitochondrial dysfunction include metabolic disorders, dysfunction of the immune system, tumorigenesis, and neuronal and behavioral abnormalities. In this review, we focus on the mitochondrial role in the CNS, which has unique characteristics and is therefore highly dependent on the mitochondria. First, we review the role of mitochondria in neuronal development, synaptogenesis, plasticity, and behavior as well as their adaptation to the intricate connections between the different cell types in the brain. Then, we review the sparse knowledge of the mechanisms of exogenous mitochondrial uptake and describe attempts to determine their half-life and transplantation long-term effects on neuronal sprouting, cellular proteome, and behavior. We further discuss the potential of mitochondrial transplantation to serve as a tool to study the causal link between mitochondria and neuronal activity and behavior. Next, we describe mitochondrial transplantation's therapeutic potential in various CNS disorders. Finally, we discuss the basic and reverse-translation challenges of this approach that currently hinder the clinical use of mitochondrial transplantation.
Collapse
Affiliation(s)
- Kuldeep Tripathi
- Laboratory of Psychobiology, Department of Neuroscience, The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, P.O. Box 9649, Haifa 31096, Israel
| | - Dorit Ben-Shachar
- Laboratory of Psychobiology, Department of Neuroscience, The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, P.O. Box 9649, Haifa 31096, Israel
| |
Collapse
|
7
|
An X, Ma X, Liu H, Song J, Wei T, Zhang R, Zhan X, Li H, Zhou J. Inhibition of PDGFRβ alleviates endothelial cell apoptotic injury caused by DRP-1 overexpression and mitochondria fusion failure after mitophagy. Cell Death Dis 2023; 14:756. [PMID: 37980402 PMCID: PMC10657461 DOI: 10.1038/s41419-023-06272-3] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 10/23/2023] [Accepted: 11/03/2023] [Indexed: 11/20/2023]
Abstract
Kawasaki disease (KD), described as "mucocutaneous lymph node syndrome", affects infants and toddlers. Patients with KD suffer from an inflammatory cascade leading to vasculitis with a predilection for coronary arteries. While the symptoms and pathogenesis of KD have received more and more attention, the precise mechanisms are still debated. Researches show that endothelial dysfunction process in KD leads to arterial damage and affect clinical outcome. In this study, we constructed a Candida albicans water soluble fraction (CAWS)-induced KD murine model and penetrated investigating the mechanisms behind endothelial dysfunction. CAWS-induced mice presented remarkably elevated vascular endothelial cell growth factor (VEGF) levels. Abundant expression of VEGF was documented in all vessels that showed edema from acute KD. It has been reported that Platelet-derived growth factor (PDGF) co-expression normalizes VEGF-induced aberrant angiogenesis. Hyperexpression of PDGFRβ was induced in the thickened medial layer and vascular endothelium of KD mice. Masitinib (Mas) is an oral tyrosine kinase inhibitor of numerous targets, which can selectively target PDGFR signaling. We set out to explore whether Mas could regulate coronary pathology in KD. Mas administration significantly reduced the VEGF-induced endothelial cells migration. NOX4 was activated in vascular endothelial cells to produce more ROS. Mitochondrial dysregulated fission and mitophagy caused by DRP-1 overexpression precipitated the arterial endothelial cells injury. Here, mitophagy seemed to work as the driving force of DRP-1/Bak/BNIP3-dependent endothelial cells apoptosis. In summary, how mitophagy is regulated by DRP-1 under pathologic status is critical and complex, which may contribute to the development of specific therapeutic interventions in cardiovascular diseases patients, for example Masatinib, the inhibitor of PDGFRβ. FACTS AND QUESTIONS: Kawasaki disease causing systemic vasculitis, affects infants and toddlers. Coronary artery injury remains the major causes of morbidity and mortality. DRP-1 overexpression induces DRP-1/Bak/BNIP3-dependent endothelial cells apoptosis. PDGFRβ was high-expressed in the thickened medial layer of CAWS-induced KD mice. Inhibition of PDGFRβ signaling alleviates arterial endothelial cells injury.
Collapse
Affiliation(s)
- Xiaohong An
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
- Yunnan Characteristic Plant Extraction Laboratory, Yunnan Yunke Characteristic Plant Extraction Laboratory Co., Ltd, Kunming, 650106, China
| | - Xiao Ma
- Yunnan Characteristic Plant Extraction Laboratory, Yunnan Yunke Characteristic Plant Extraction Laboratory Co., Ltd, Kunming, 650106, China
| | - Heng Liu
- Yunnan Provincial Key Laboratory of Entomological Biopharmaceutical R&D, Dali University, Dali, 671000, China
| | - Jing Song
- Laboratory Animal Center, Xiamen University, Xiamen, 361102, China
| | - Tiange Wei
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Rongzhan Zhang
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Xiao Zhan
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China
| | - Hongyang Li
- Institute of Dermatology, Chinese Academy of Medical Sciences and Peking Union Medical College, Nanjing, 210042, China.
| | - Jia Zhou
- School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
8
|
Song Y, Cao H, Zuo C, Gu Z, Huang Y, Miao J, Fu Y, Guo Y, Jiang Y, Wang F. Mitochondrial dysfunction: A fatal blow in depression. Biomed Pharmacother 2023; 167:115652. [PMID: 37801903 DOI: 10.1016/j.biopha.2023.115652] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2023] [Revised: 10/01/2023] [Accepted: 10/03/2023] [Indexed: 10/08/2023] Open
Abstract
Mitochondria maintain the normal physiological function of nerve cells by producing sufficient cellular energy and performing crucial roles in maintaining the metabolic balance through intracellular Ca2+ homeostasis, oxidative stress, and axonal development. Depression is a prevalent psychiatric disorder with an unclear pathophysiology. Damage to the hippocampal neurons is a key component of the plasticity regulation of synapses and plays a critical role in the mechanism of depression. There is evidence suggesting that mitochondrial dysfunction is associated with synaptic impairment. The maintenance of mitochondrial homeostasis includes quantitative maintenance and quality control of mitochondria. Mitochondrial biogenesis produces new and healthy mitochondria, and mitochondrial dynamics cooperates with mitophagy to remove damaged mitochondria. These processes maintain mitochondrial population stability and exert neuroprotective effects against early depression. In contrast, mitochondrial dysfunction is observed in various brain regions of patients with major depressive disorders. The accumulation of defective mitochondria accelerates cellular nerve dysfunction. In addition, impaired mitochondria aggravate alterations in the brain microenvironment, promoting neuroinflammation and energy depletion, thereby exacerbating the development of depression. This review summarizes the influence of mitochondrial dysfunction and the underlying molecular pathways on the pathogenesis of depression. Additionally, we discuss the maintenance of mitochondrial homeostasis as a potential therapeutic strategy for depression.
Collapse
Affiliation(s)
- Yu Song
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Huan Cao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Chengchao Zuo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Zhongya Gu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Yaqi Huang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Jinfeng Miao
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Yufeng Fu
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Yu Guo
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China
| | - Yongsheng Jiang
- Cancer Center of Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan, 430030 Hubei, China.
| | - Furong Wang
- Department of Neurology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan 430030, Hubei, China; Key Laboratory of Vascular Aging (HUST), Ministry of Education, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No.1095 Jiefang Road, Wuhan, 430030 Hubei, China.
| |
Collapse
|
9
|
Mishra E, Thakur MK. Mdivi-1 Rescues Memory Decline in Scopolamine-Induced Amnesic Male Mice by Ameliorating Mitochondrial Dynamics and Hippocampal Plasticity. Mol Neurobiol 2023; 60:5426-5449. [PMID: 37314656 DOI: 10.1007/s12035-023-03397-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Accepted: 05/22/2023] [Indexed: 06/15/2023]
Abstract
Memory loss, often known as amnesia, is common in the elderly population and refers to forgetting facts and experiences. It is associated with increased mitochondrial fragmentation, though the contribution of mitochondrial dynamics in amnesia is poorly understood. Therefore, the present study is aimed at elucidating the role of Mdivi-1 in mitochondrial dynamics, hippocampal plasticity, and memory during scopolamine (SC)-induced amnesia. The findings imply that Mdivi-1 significantly increased the expression of Arc and BDNF proteins in the hippocampus of SC-induced amnesic mice, validating improved recognition and spatial memory. Moreover, an improved mitochondrial ultrastructure was attributed to a decline in the percentage of fragmented and spherical-shaped mitochondria after Mdivi-1 treatment in SC-induced mice. The significant downregulation of p-Drp1 (S616) protein and upregulation of Mfn2, LC3BI, and LC3BII proteins in Mdivi-1-treated SC-induced mice indicated a decline in fragmented mitochondrial number and healthy mitochondrial dynamics. Mdivi-1 treatment alleviated ROS production and Caspase-3 activity and elevated mitochondrial membrane potential, Vdac1 expression, ATP production, and myelination, resulting in reduced neurodegeneration in SC mice. Furthermore, the decline of pro-apoptotic protein cytochrome-c and increase of anti-apoptotic proteins Procaspase-9 and Bcl-2 in Mdivi-1-treated SC-induced mice suggested improved neuronal health. Mdivi-1 also increased the dendritic arborization and spine density, which was further corroborated by increased expression of synaptophysin and PSD95. In conclusion, the current study suggests that Mdivi-1 treatment improves mitochondrial ultrastructure and function through the regulation of mitochondrial dynamics. These changes further improve neuronal cell density, myelination, dendritic arborization, and spine density, decrease neurodegeneration, and improve recognition and spatial memory. Schematic presentation depicts that Mdivi-1 rescues memory decline in scopolamine-induced amnesic male mice by ameliorating mitochondrial dynamics and hippocampal plasticity.
Collapse
Affiliation(s)
- Ela Mishra
- Biochemistry and Molecular Biology Laboratory, Centre of Advanced Study, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221 005, India
| | - Mahendra Kumar Thakur
- Biochemistry and Molecular Biology Laboratory, Centre of Advanced Study, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, 221 005, India.
| |
Collapse
|
10
|
Launay N, Ruiz M, Planas-Serra L, Verdura E, Rodríguez-Palmero A, Schlüter A, Goicoechea L, Guilera C, Casas J, Campelo F, Jouanguy E, Casanova JL, Boespflug-Tanguy O, Vazquez Cancela M, Gutiérrez-Solana LG, Casasnovas C, Area-Gomez E, Pujol A. RINT1 deficiency disrupts lipid metabolism and underlies a complex hereditary spastic paraplegia. J Clin Invest 2023; 133:e162836. [PMID: 37463447 DOI: 10.1172/jci162836] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Accepted: 05/26/2023] [Indexed: 07/20/2023] Open
Abstract
The Rad50 interacting protein 1 (Rint1) is a key player in vesicular trafficking between the ER and Golgi apparatus. Biallelic variants in RINT1 cause infantile-onset episodic acute liver failure (ALF). Here, we describe 3 individuals from 2 unrelated families with novel biallelic RINT1 loss-of-function variants who presented with early onset spastic paraplegia, ataxia, optic nerve hypoplasia, and dysmorphic features, broadening the previously described phenotype. Our functional and lipidomic analyses provided evidence that pathogenic RINT1 variants induce defective lipid-droplet biogenesis and profound lipid abnormalities in fibroblasts and plasma that impact both neutral lipid and phospholipid metabolism, including decreased triglycerides and diglycerides, phosphatidylcholine/phosphatidylserine ratios, and inhibited Lands cycle. Further, RINT1 mutations induced intracellular ROS production and reduced ATP synthesis, affecting mitochondria with membrane depolarization, aberrant cristae ultrastructure, and increased fission. Altogether, our results highlighted the pivotal role of RINT1 in lipid metabolism and mitochondria function, with a profound effect in central nervous system development.
Collapse
Affiliation(s)
- Nathalie Launay
- Neurometabolic Diseases Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
| | - Montserrat Ruiz
- Neurometabolic Diseases Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
| | - Laura Planas-Serra
- Neurometabolic Diseases Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
| | - Edgard Verdura
- Neurometabolic Diseases Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
| | - Agustí Rodríguez-Palmero
- Neurometabolic Diseases Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain
- Pediatric Neurology unit, Department of Pediatrics, Hospital Universitari Germans Trias i Pujol, Universitat Autònoma de Barcelona, Spain
| | - Agatha Schlüter
- Neurometabolic Diseases Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
| | - Leire Goicoechea
- Neurometabolic Diseases Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
| | - Cristina Guilera
- Neurometabolic Diseases Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
| | - Josefina Casas
- Research Unit on BioActive Molecules (RUBAM), Departament de Química Biomèdica, Institut de Química Avançada de Catalunya (IQAC-CSIC), Barcelona, Spain
- CIBEREHD, Centro de Investigación Biomédica en Red de Enfermedades heoaticas y digestivas, ISCIII, Madrid, Spain
| | - Felix Campelo
- ICFO-Institut de Ciencies Fotoniques, The Barcelona Institute of Science and Technology, Castelldefels, Spain
| | - Emmanuelle Jouanguy
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale, UMR 1163, Necker Hospital for Sick Children, Paris, France
- University of Paris, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
| | - Jean-Laurent Casanova
- Laboratory of Human Genetics of Infectious Diseases, Necker Branch, Institut National de la Santé et de la Recherche Médicale, UMR 1163, Necker Hospital for Sick Children, Paris, France
- University of Paris, Imagine Institute, Paris, France
- St. Giles Laboratory of Human Genetics of Infectious Diseases, Rockefeller Branch, The Rockefeller University, New York, New York, USA
- Pediatric Hematology-Immunology Unit, Necker Hospital for Sick Children, Paris, France
- Howard Hughes Medical Institute, New York, New York, USA
| | - Odile Boespflug-Tanguy
- CRMR Leukofrance Service de Neuropédiatrie, Hôpital Robert Debré AP-HP, Paris, France
- UMR1141 Neurodiderot Université de Paris Cité, Paris, France
| | | | - Luis González Gutiérrez-Solana
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
- Consulta de Neurodegenerativas, Sección de Neurología Pediátrica, Hospital, Infantil Universitario Niño Jesús, Madrid, Spain
| | - Carlos Casasnovas
- Neurometabolic Diseases Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
- Neuromuscular Unit, Neurology Department, Hospital Universitari de Bellvitge, Universitat de Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
| | - Estela Area-Gomez
- Department of Neurology, Columbia University, New York, New York, USA
| | - Aurora Pujol
- Neurometabolic Diseases Laboratory, Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), Hospital Duran i Reynals, L'Hospitalet de Llobregat, Barcelona, Spain
- CIBERER, Centro de Investigación Biomédica en Red de Enfermedades Raras, ISCIII, Madrid, Spain
- Catalan Institution of Research and Advanced Studies (ICREA), Barcelona, Spain
| |
Collapse
|
11
|
Pan M, Li X, Xu G, Tian X, Li Y, Fang W. Tripartite Motif Protein Family in Central Nervous System Diseases. Cell Mol Neurobiol 2023:10.1007/s10571-023-01337-5. [PMID: 36988770 DOI: 10.1007/s10571-023-01337-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2022] [Accepted: 03/13/2023] [Indexed: 03/30/2023]
Abstract
Tripartite motif (TRIM) protein superfamily is a group of E3 ubiquitin ligases characterized by the conserved RING domain, the B-box domain, and the coiled-coil domain (RBCC). It is widely involved in various physiological and pathological processes, such as intracellular signal transduction, cell cycle regulation, oncogenesis, and innate immune response. Central nervous system (CNS) diseases are composed of encephalopathy and spinal cord diseases, which have a high disability and mortality rate. Patients are often unable to take care of themselves and their life quality can be seriously declined. Initially, the function research of TRIM proteins mainly focused on cancer. However, in recent years, accumulating attention is paid to the roles they play in CNS diseases. In this review, we integrate the reported roles of TRIM proteins in the pathological process of CNS diseases and related signaling pathways, hoping to provide theoretical bases for further research in treating CNS diseases targeting TRIM proteins. TRIM proteins participated in CNS diseases. TRIM protein family is characterized by a highly conserved RBCC domain, referring to the RING domain, the B-box domain, and the coiled-coil domain. Recent research has discovered the relations between TRIM proteins and various CNS diseases, especially Alzheimer's disease, Parkinson's disease, and ischemic stroke.
Collapse
Affiliation(s)
- Mengtian Pan
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, Jiangsu, 210009, People's Republic of China
| | - Xiang Li
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, Jiangsu, 210009, People's Republic of China
| | - Guangchen Xu
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, Jiangsu, 210009, People's Republic of China
| | - Xinjuan Tian
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, Jiangsu, 210009, People's Republic of China
| | - Yunman Li
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, Jiangsu, 210009, People's Republic of China.
| | - Weirong Fang
- State Key Laboratory of Natural Medicines, School of Basic Medical Sciences and Clinical Pharmacy, China Pharmaceutical University, Tongjiaxiang 24, Nanjing, Jiangsu, 210009, People's Republic of China.
- Department of Physiology, School of Basic Medicine and Clinical Pharmacy, China Pharmaceutical University, Mailbox 207, Tongjiaxiang 24, Nanjing, Jiangsu, 210009, People's Republic of China.
| |
Collapse
|
12
|
The Drp1-Mediated Mitochondrial Fission Protein Interactome as an Emerging Core Player in Mitochondrial Dynamics and Cardiovascular Disease Therapy. Int J Mol Sci 2023; 24:ijms24065785. [PMID: 36982862 PMCID: PMC10057413 DOI: 10.3390/ijms24065785] [Citation(s) in RCA: 36] [Impact Index Per Article: 36.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/13/2023] [Accepted: 03/15/2023] [Indexed: 03/22/2023] Open
Abstract
Mitochondria, the membrane-bound cell organelles that supply most of the energy needed for cell function, are highly regulated, dynamic organelles bearing the ability to alter both form and functionality rapidly to maintain normal physiological events and challenge stress to the cell. This amazingly vibrant movement and distribution of mitochondria within cells is controlled by the highly coordinated interplay between mitochondrial dynamic processes and fission and fusion events, as well as mitochondrial quality-control processes, mainly mitochondrial autophagy (also known as mitophagy). Fusion connects and unites neighboring depolarized mitochondria to derive a healthy and distinct mitochondrion. In contrast, fission segregates damaged mitochondria from intact and healthy counterparts and is followed by selective clearance of the damaged mitochondria via mitochondrial specific autophagy, i.e., mitophagy. Hence, the mitochondrial processes encompass all coordinated events of fusion, fission, mitophagy, and biogenesis for sustaining mitochondrial homeostasis. Accumulated evidence strongly suggests that mitochondrial impairment has already emerged as a core player in the pathogenesis, progression, and development of various human diseases, including cardiovascular ailments, the leading causes of death globally, which take an estimated 17.9 million lives each year. The crucial factor governing the fission process is the recruitment of dynamin-related protein 1 (Drp1), a GTPase that regulates mitochondrial fission, from the cytosol to the outer mitochondrial membrane in a guanosine triphosphate (GTP)-dependent manner, where it is oligomerized and self-assembles into spiral structures. In this review, we first aim to describe the structural elements, functionality, and regulatory mechanisms of the key mitochondrial fission protein, Drp1, and other mitochondrial fission adaptor proteins, including mitochondrial fission 1 (Fis1), mitochondrial fission factor (Mff), mitochondrial dynamics 49 (Mid49), and mitochondrial dynamics 51 (Mid51). The core area of the review focuses on the recent advances in understanding the role of the Drp1-mediated mitochondrial fission adaptor protein interactome to unravel the missing links of mitochondrial fission events. Lastly, we discuss the promising mitochondria-targeted therapeutic approaches that involve fission, as well as current evidence on Drp1-mediated fission protein interactions and their critical roles in the pathogeneses of cardiovascular diseases (CVDs).
Collapse
|
13
|
Bai YF, Li WJ, Ji YW, An LX, Zhang L, Li JF. Sevoflurane induces neurotoxic effects on developing neurons through the WNK1/NKCC1/Ca 2+ /Drp-1 signalling pathway. Clin Exp Pharmacol Physiol 2023; 50:393-402. [PMID: 36733226 DOI: 10.1111/1440-1681.13755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 01/12/2023] [Accepted: 01/31/2023] [Indexed: 02/04/2023]
Abstract
Children repeatedly exposed to anaesthesia have a high risk of cognitive impairment, but the mechanism of its regulation in this context is unknown. The objective of this study was to investigate the possible toxic mechanism of sevoflurane through the WNK1/NKCC1/Ca2+ /Drp-1 signalling pathway. The hippocampal neuronal HT22 cell line was used in this study. The intervention group was treated with the WNK1 inhibitor WNK-463, CaN inhibitor FK506 and Drp-1 inhibitor Mdivi-1 respectively in the medium for 30 min before sevoflurane anaesthesia. The sevofluane group and all intervention group treated with 4.1% sevoflurane for 6 h. Compared with the control group, sevoflurane treatment decreased cell viability and increased cellular apoptosis. Our study found that WNK-463, FK506 and Mdivi-1 can all alleviate the sevoflurane-induced reduction in cell viability, decrease the cell apoptosis. In addition, WNK-463 pretreatment could inhibit the increase of WNK1 kinase and NKCC1 protein concentration caused by sevoflurane. Further, sevoflurane anaesthesia causes intracellular calcium overload, increases the expression of CaN and induces the dephosphorylation of Drp-1 protein at ser637, while CaN inhibitor FK506 pretreatment could reduce the dephosphorylation of Drp-1. Therefore, the WNK1/NKCC1/Ca2+ /Drp-1 signalling pathway plays an important role in sevoflurane-related neurotoxicity. Reducing intracellular calcium influx may be one of the important mechanism to ameliorate sevoflurane toxicity.
Collapse
Affiliation(s)
- Ya-Fan Bai
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Wen-Jing Li
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Yu-Wei Ji
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Li-Xin An
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Li Zhang
- Department of Anesthesiology, Beijing Friendship Hospital, Capital Medical University, Beijing, China
| | - Jun-Fa Li
- Department of Neurobiology and Beijing Institute for Brain Disorders, Capital Medical University, Beijing, China
| |
Collapse
|
14
|
Tonner H, Hunn S, Auler N, Schmelter C, Pfeiffer N, Grus FH. Dynamin-like Protein 1 (DNML1) as a Molecular Target for Antibody-Based Immunotherapy to Treat Glaucoma. Int J Mol Sci 2022; 23:ijms232113618. [PMID: 36362420 PMCID: PMC9654827 DOI: 10.3390/ijms232113618] [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: 09/01/2022] [Revised: 09/23/2022] [Accepted: 10/29/2022] [Indexed: 11/09/2022] Open
Abstract
Slow and progressive loss of retinal ganglion cells (RGCs) is the main characteristic of glaucoma, the second leading cause of blindness worldwide. Previous studies have shown that impaired mitochondrial dynamics could facilitate retinal neurodegeneration. Mitochondrial dynamics are regulated directly (fission) or more indirectly (fusion) by dynamin-like protein 1 (DNML1). Therefore, DNM1L might be a promising target for an antibody-based approach to treat glaucoma. The consequences of targeting endogenous DNM1L by antibodies in a glaucoma animal model have not been investigated yet. Here, we show that the intravitreal application of an anti-DNM1L antibody showed protective effects regarding the survival of RGCs and their axons in the retinal nerve fiber layer (RNFL). Antibody treatment also improved retinal functionality, as observed by electroretinography (Ganzfeld ERG). Western blot analysis revealed altered DNM1L phosphorylation and altered expression of proteins related to apoptosis suggesting a decreased apoptosis rate. Mass spectrometry analysis revealed 28 up-regulated and 21 down-regulated proteins (p < 0.05) in both experimental groups. Protein pathway analysis showed that many proteins interacted directly with the target protein DNM1L and could be classified into three main protein clusters: Vesicle traffic-associated (NSF, SNCA, ARF1), mitochondrion-associated (HSP9A, SLC25A5/ANT2, GLUD1) and cytoskeleton-associated (MAP1A) signaling pathway. Our results demonstrate that DNM1L is a promising target for an antibody-based approach to glaucoma therapy.
Collapse
Affiliation(s)
| | | | | | | | | | - Franz H. Grus
- Correspondence: ; Tel.: +49-6131-17-3328; Fax: +49-6131-4970563
| |
Collapse
|
15
|
Bera A, Lavanya G, Reshmi R, Dev K, Kumar R. Mechanistic and therapeutic role of Drp1 in the pathogenesis of Alzheimer's disease. Eur J Neurosci 2022; 56:5516-5531. [PMID: 35078269 DOI: 10.1111/ejn.15611] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 12/31/2021] [Accepted: 01/13/2022] [Indexed: 12/14/2022]
Abstract
Alzheimer's disease (AD), a progressive neurodegenerative disorder, has emerged as the most common form of dementia in the elderly. Two major pathological hallmarks have been identified for AD: extracellular amyloid plaques and intracellular neurofibrillary tangles (NFT). Recently, dynamin-related protein 1 (Drp1) was recognized to contribute significantly towards the pathogenesis of AD. Drp1 is primarily located in the cytosol, from where it translocates to the mitochondrial outer membrane and drives the mitochondrial fission via GTP hydrolysis. Drp1 interacts with Aβ and phosphorylated tau, leading to excessive mitochondrial fragmentation, which in turn results in synaptic dysfunction, neuronal damage and cognitive decline. Several studies suggest an increase in the level of Drp1 in the post-mortem brain specimen collected from the AD patients and murine models of AD. Interestingly, heterozygous deletion of Drp1 in the transgenic murine model of AD ameliorates the mitochondrial dysfunction, improving learning and memory. The current review article discusses the possible mechanistic pathways by which Drp1 can influence the pathogenesis of AD. Besides, it will describe various inhibitors for Drp1 and their potential role as therapeutics for AD in the future.
Collapse
Affiliation(s)
- Arpita Bera
- Department of Biotechnology, GITAM Institute of Sciences, GITAM (Deemed to be) University, Visakhapatnam, India
| | - Gantyada Lavanya
- Department of Biotechnology, GITAM Institute of Sciences, GITAM (Deemed to be) University, Visakhapatnam, India
| | - Ravada Reshmi
- Department of Biotechnology, GITAM Institute of Sciences, GITAM (Deemed to be) University, Visakhapatnam, India
| | - Kapil Dev
- Department of Biotechnology, Jamia Millia Islamia, New Delhi, India
| | - Rahul Kumar
- Department of Biotechnology, GITAM Institute of Sciences, GITAM (Deemed to be) University, Visakhapatnam, India
| |
Collapse
|
16
|
Shi W, Tan C, Liu C, Chen D. Mitochondrial fission mediated by Drp1-Fis1 pathway and neurodegenerative diseases. Rev Neurosci 2022; 34:275-294. [PMID: 36059131 DOI: 10.1515/revneuro-2022-0056] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2022] [Accepted: 07/08/2022] [Indexed: 11/15/2022]
Abstract
In recent years, the role of mitochondrial dynamics in neurodegenerative diseases has becoming increasingly important. More and more evidences have shown that in pathological conditions, abnormal mitochondrial divisions, especially Drp1-Fis1-mediated divisions, play an important role in the occurrence and development of Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, Huntington's disease, glaucoma, and other neurodegenerative diseases. This review highlights several new mechanisms of physiological fission of mitochondria and the difference/connection of physiological/pathological mitochondrial fission. In addition, we described the relationship between abnormal mitochondrial dynamics and neurodegenerative diseases in detail and emphatically summarized its detection indicators in basic experiments, trying to provide references for further mechanism exploration and therapeutic targets.
Collapse
Affiliation(s)
- Wenjia Shi
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, Hunan Province, China
| | - Cheng Tan
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, Hunan Province, China
| | - Can Liu
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, Hunan Province, China
| | - Dan Chen
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha 410013, Hunan Province, China
| |
Collapse
|
17
|
Wang M, Wei R, Li G, Bi HL, Jia Z, Zhang M, Pang M, Li X, Ma L, Tang Y. SUMOylation of SYNJ2BP-COX16 promotes breast cancer progression through DRP1-mediated mitochondrial fission. Cancer Lett 2022; 547:215871. [PMID: 35998797 DOI: 10.1016/j.canlet.2022.215871] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 08/07/2022] [Accepted: 08/08/2022] [Indexed: 11/19/2022]
Abstract
Treatments targeting oncogenic fusion proteins are notable examples of successful drug development. Abnormal splicing of genes resulting in fusion proteins is a critical driver of various tumors, but the underlying mechanism remains poorly understood. Here, we show that SUMOylation of the fusion protein Synaptojanin 2 binding protein-Cytochrome-c oxidase 16 (SYNJ2BP-COX16) at K107 induces mitochondrial fission in breast cancer and that the K107 site regulates SYNJ2BP-COX16 mitochondrial subcellular localization. Compared with a non-SUMOylated K107R mutant, wild-type SYNJ2BP-COX16 contributed to breast cancer cell proliferation and metastasis in vivo and in vitro by increasing adenosine triphosphate (ATP) production and cytochrome-c oxidase (COX) activity. SUMOylated SYNJ2BP-COX16 recruits dynamin-related protein 1 (DRP1) to the mitochondria to promote ubiquitin-conjugating enzyme 9 (UBC9) binding to DRP1, enhance SUMOylation of DRP1 and phosphorylation of DRP1 at S616, and then induce mitochondrial fission. Moreover, Mdivi-1, an inhibitor of DRP1 phosphorylation, decreased the localization of DRP1 in mitochondria, and prevents SYNJ2BP-COX16 induced mitochondrial fission, cell proliferation and metastasis. Based on these data, SYNJ2BP-COX16 promotes breast cancer progression through the phosphorylation of DRP1 and subsequent induction of mitochondrial fission, indicating that SUMOylation at the K107 residue of SYNJ2BP-COX16 is a novel potential treatment target for breast cancer.
Collapse
Affiliation(s)
- Miao Wang
- School of Bioengineering, Dalian University of Technology, No.2 Linggong Road, Dalian, Liaoning Province, 116024, China.
| | - Ranru Wei
- School of Bioengineering, Dalian University of Technology, No.2 Linggong Road, Dalian, Liaoning Province, 116024, China.
| | - Guohui Li
- School of Bioengineering, Dalian University of Technology, No.2 Linggong Road, Dalian, Liaoning Province, 116024, China; College of New Materials and Chemical Engineering, Beijing Key Laboratory of Enze Biomass Fine Chemicals, Beijing Institute of Petrochemical Technology, Beijing, China.
| | - Hai-Lian Bi
- Institute of Cardiovascular Diseases, First Affiliated Hospital of Dalian Medical University, Dalian, Liaoning Province, 116024, China.
| | - Zhaojun Jia
- College of New Materials and Chemical Engineering, Beijing Key Laboratory of Enze Biomass Fine Chemicals, Beijing Institute of Petrochemical Technology, Beijing, China.
| | - Mengjie Zhang
- School of Bioengineering, Dalian University of Technology, No.2 Linggong Road, Dalian, Liaoning Province, 116024, China.
| | - Mengyao Pang
- School of Bioengineering, Dalian University of Technology, No.2 Linggong Road, Dalian, Liaoning Province, 116024, China.
| | - Xiaona Li
- School of Environmental Science and Technology, Dalian University of Technology, No.2 Linggong Road, Dalian, Liaoning Province, 116024, China.
| | - Liming Ma
- School of Bioengineering, Dalian University of Technology, No.2 Linggong Road, Dalian, Liaoning Province, 116024, China.
| | - Ying Tang
- School of Bioengineering, Dalian University of Technology, No.2 Linggong Road, Dalian, Liaoning Province, 116024, China.
| |
Collapse
|
18
|
Cao Z, Zhao M, Sun H, Hu L, Chen Y, Fan Z. Roles of mitochondria in neutrophils. Front Immunol 2022; 13:934444. [PMID: 36081497 PMCID: PMC9447286 DOI: 10.3389/fimmu.2022.934444] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2022] [Accepted: 07/19/2022] [Indexed: 01/25/2023] Open
Abstract
Neutrophils are the most abundant leukocyte in human blood. They are critical for fighting infections and are involved in inflammatory diseases. Mitochondria are indispensable for eukaryotic cells, as they control the biochemical processes of respiration and energy production. Mitochondria in neutrophils have been underestimated since glycolysis is a major metabolic pathway for fuel production in neutrophils. However, several studies have shown that mitochondria are greatly involved in multiple neutrophil functions as well as neutrophil-related diseases. In this review, we focus on how mitochondrial components, metabolism, and related genes regulate neutrophil functions and relevant diseases.
Collapse
Affiliation(s)
- Ziming Cao
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT, United States
| | - Meng Zhao
- Arthritis and Clinical Immunology Program, Oklahoma Medical Research Foundation, Oklahoma City, OK, United States,Department of Microbiology and Immunology, University of Oklahoma Health Science Center, Oklahoma City, OK, United States
| | - Hao Sun
- Department of Medicine, University of California San Diego, La Jolla, CA, United States
| | - Liang Hu
- Academy of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yunfeng Chen
- Department of Biochemistry and Molecular Biology and Department of Pathology, University of Texas Medical Branch, Galveston, TX, United States
| | - Zhichao Fan
- Department of Immunology, School of Medicine, UConn Health, Farmington, CT, United States,*Correspondence: Zhichao Fan,
| |
Collapse
|
19
|
Ashraf GM, Chatzichronis S, Alexiou A, Firdousi G, Kamal MA, Ganash M. Dietary Alterations in Impaired Mitochondrial Dynamics Due to Neurodegeneration. Front Aging Neurosci 2022; 14:893018. [PMID: 35898328 PMCID: PMC9310440 DOI: 10.3389/fnagi.2022.893018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2022] [Accepted: 06/20/2022] [Indexed: 11/15/2022] Open
Abstract
Alzheimer's disease is still an incurable disease with significant social and economic impact globally. Nevertheless, newly FDA-approved drugs and non-pharmacological techniques may offer efficient disease treatments. Furthermore, it is widely accepted that early diagnosis or even prognosis of Alzheimer's disease using advanced computational tools could offer a compelling alternative way of management. In addition, several studies have presented an insight into the role of mitochondrial dynamics in Alzheimer's development. In combination with diverse dietary and obesity-related diseases, mitochondrial bioenergetics may be linked to neurodegeneration. Considering the probabilistic expectations of Alzheimer's disease development or progression due to specific risk factors or biomarkers, we designed a Bayesian model to formulate the impact of diet-induced obesity with an impaired mitochondrial function and altered behavior. The applied probabilities are based on clinical trials globally and are continuously subject to updating and redefinition. The proposed multiparametric model combines various data types based on uniform probabilities. The program simulates all the variables with a uniform distribution in a sample of 1000 patients. First, the program initializes the variable age (30-95) and the four different diet types ("HFO_diet," "Starvation," "HL_diet," "CR") along with the factors that are related to prodromal or mixed AD (ATP, MFN1, MFN2, DRP1, FIS1, Diabetes, Oxidative_Stress, Hypertension, Obesity, Depression, and Physical_activity). Besides the known proteins related to mitochondrial dynamics, our model includes risk factors like Age, Hypertension, Oxidative Stress, Obesity, Depression, and Physical Activity, which are associated with Prodromal Alzheimer's. The outcome is the disease progression probability corresponding to a random individual ID related to diet choices and mitochondrial dynamics parameters. The proposed model and the programming code are adjustable to different parameters and values. The program is coded and executed in Python and is fully and freely available for research purposes and testing the correlation between diet type and Alzheimer's disease progression regarding various risk factors and biomarkers.
Collapse
Affiliation(s)
- Ghulam Md Ashraf
- Pre-clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Stylianos Chatzichronis
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, Australia
| | - Athanasios Alexiou
- Department of Science and Engineering, Novel Global Community Educational Foundation, Hebersham, NSW, Australia
- AFNP Med Austria, Wien, Austria
| | - Gazala Firdousi
- Department of Health Sciences, Novel Global Community Educational Foundation, Hebersham, NSW, Australia
| | - Mohammad Amjad Kamal
- Institutes for Systems Genetics, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu, China
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
- Enzymoics, Novel Global Community Educational Foundation, Hebersham, NSW, Australia
| | - Magdah Ganash
- Department of Biology, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia
| |
Collapse
|
20
|
Liu X, Song L, Yu J, Huang F, Li Y, Ma C. Mdivi-1: a promising drug and its underlying mechanisms in the treatment of neurodegenerative diseases. Histol Histopathol 2022; 37:505-512. [PMID: 35199329 DOI: 10.14670/hh-18-443] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Mitochondria are energy-producing organelles, and neurons are high energy consumption cells. Therefore, mitochondrial dysfunction is a critical factor in neurodegenerative processes. Mitochondrial division inhibitor-1 (Mdivi-1) is a small chemical inhibitor of mitochondrial division dynamin, which plays multiple roles in mitochondrial dynamics, mitochondrial autophagy, ATP production, the immune response, and Ca²⁺ homeostasis. Mdivi-1 inhibition of excessive mitochondrial fission exerted cytoprotective effects in neurodegenerative diseases, such as Alzheimer's disease (AD), Parkinson's disease (PD), and multiple sclerosis (MS). Mdivi-1 changed the mRNA expression of the electron transport chain (ETC) and reduced Ca²⁺ overload against neuronal injury. Elucidation of the molecular mechanism of Mdivi-1 in neurodegenerative diseases will help evaluate its therapeutic potential and promote its application in clinical studies. The present article focused on the multiple effects of Mdivi-1 on mitochondrial function and its potential therapeutic effects in neurodegenerative diseases.
Collapse
Affiliation(s)
- Xiaoqin Liu
- Institute of Brain Science, Shanxi Datong University, Datong, China
| | - Lijuan Song
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, China
- Department of Physiology, Shanxi Medical University, Taiyuan, China
| | - Jiezhong Yu
- Institute of Brain Science, Shanxi Datong University, Datong, China
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, China
- Department of Neurology, The Fourth People's Hospital, Datong, China
| | - Fang Huang
- Institute of Brain Science, Shanxi Datong University, Datong, China
| | - Yanhua Li
- Institute of Brain Science, Shanxi Datong University, Datong, China
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, China.
| | - Cungen Ma
- Institute of Brain Science, Shanxi Datong University, Datong, China
- The Key Research Laboratory of Benefiting Qi for Acting Blood Circulation Method to Treat Multiple Sclerosis of State Administration of Traditional Chinese Medicine/Research Center of Neurobiology, Shanxi University of Chinese Medicine, Jinzhong, China.
| |
Collapse
|
21
|
Lee SB, Yang HO. Sinapic Acid Ameliorates REV-ERB α Modulated Mitochondrial Fission against MPTP-Induced Parkinson's Disease Model. Biomol Ther (Seoul) 2022; 30:409-417. [PMID: 35611585 PMCID: PMC9424337 DOI: 10.4062/biomolther.2022.020] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2022] [Revised: 03/29/2022] [Accepted: 04/26/2022] [Indexed: 11/15/2022] Open
Abstract
Parkinson’s disease (PD) is the second most common neurodegenerative disease worldwide, and accumulating evidence indicates that mitochondrial dysfunction is associated with progressive deterioration in PD patients. Previous studies have shown that sinapic acid has a neuroprotective effect, but its mechanisms of action remain unclear. The neuroprotective effect of sinapic acid was assayed in a PD mouse model generated by the neurotoxin 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) as well as in SH-SY5Y cells. Target protein expression was detected by western blotting. Sinapic acid treatment attenuated the behavioral defects and loss of dopaminergic neurons in the PD models. Sinapic acid also improved mitochondrial function in the PD models. MPTP treatment increased the abundance of mitochondrial fission proteins such as dynamin-related protein 1 (Drp1) and phospho-Drp1 Ser616. In addition, MPTP decreased the expression of the REV-ERB α protein. These changes were attenuated by sinapic acid treatment. We used the pharmacological REV-ERB α inhibitor SR8278 to confirmation of protective effect of sinapic acid. Treatment of SR8278 with sinapic acid reversed the protein expression of phospho-Drp1 Ser616 and REV-ERB α on MPTP-treated mice. Our findings demonstrated that sinapic acid protects against MPTP-induced PD and these effects might be related to the inhibiting abnormal mitochondrial fission through REV-ERB α.
Collapse
Affiliation(s)
- Sang-Bin Lee
- Department of Integrative Biological Sciences and Industry, Sejong University, Seoul 05006, Republic of Korea.,School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hyun Ok Yang
- Department of Integrative Biological Sciences and Industry, Sejong University, Seoul 05006, Republic of Korea
| |
Collapse
|
22
|
Brustovetsky T, Khanna R, Brustovetsky N. Involvement of CRMP2 in Regulation of Mitochondrial Morphology and Motility in Huntington's Disease. Cells 2021; 10:cells10113172. [PMID: 34831395 PMCID: PMC8619197 DOI: 10.3390/cells10113172] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 10/28/2021] [Accepted: 11/09/2021] [Indexed: 01/29/2023] Open
Abstract
Mitochondrial morphology and motility (mitochondrial dynamics) play a major role in the proper functioning of distant synapses. In Huntington’s disease (HD), mitochondria become fragmented and less motile, but the mechanisms leading to these changes are not clear. Here, we found that collapsin response mediator protein 2 (CRMP2) interacted with Drp1 and Miro 2, proteins involved in regulating mitochondrial dynamics. CRMP2 interaction with these proteins inversely correlated with CRMP2 phosphorylation. CRMP2 was hyperphosphorylated in postmortem brain tissues of HD patients, in human neurons derived from induced pluripotent stem cells from HD patients, and in cultured striatal neurons from HD mouse model YAC128. At the same time, CRMP2 interaction with Drp1 and Miro 2 was diminished in HD neurons. The CRMP2 hyperphosphorylation and dissociation from Drp1 and Miro 2 correlated with increased fission and suppressed motility. (S)-lacosamide ((S)-LCM), a small molecule that binds to CRMP2, decreased its phosphorylation at Thr 509/514 and Ser 522 and rescued CRMP2’s interaction with Drp1 and Miro 2. This was accompanied by reduced mitochondrial fission and enhanced mitochondrial motility. Additionally, (S)-LCM exerted a neuroprotective effect in YAC128 cultured neurons. Thus, our data suggest that CRMP2 may regulate mitochondrial dynamics in a phosphorylation-dependent manner and modulate neuronal survival in HD.
Collapse
Affiliation(s)
- Tatiana Brustovetsky
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
| | - Rajesh Khanna
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, AZ 85721, USA;
- Center for Innovation in Brain Sciences, University of Arizona, Tucson, AZ 85721, USA
| | - Nickolay Brustovetsky
- Department of Pharmacology and Toxicology, Indiana University School of Medicine, Indianapolis, IN 46202, USA;
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Correspondence: ; Tel.: +1-317-278-9229; Fax: +1-317-274-7714
| |
Collapse
|
23
|
Halcrow PW, Lynch ML, Geiger JD, Ohm JE. Role of endolysosome function in iron metabolism and brain carcinogenesis. Semin Cancer Biol 2021; 76:74-85. [PMID: 34139350 PMCID: PMC8627927 DOI: 10.1016/j.semcancer.2021.06.013] [Citation(s) in RCA: 21] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/10/2021] [Accepted: 06/11/2021] [Indexed: 02/07/2023]
Abstract
Iron, the most abundant metal in human brain, is an essential microelement that regulates numerous cellular mechanisms. Some key physiological roles of iron include oxidative phosphorylation and ATP production, embryonic neuronal development, formation of iron-sulfur clusters, and the regulation of enzymes involved in DNA synthesis and repair. Because of its physiological and pathological importance, iron homeostasis must be tightly regulated by balancing its uptake, transport, and storage. Endosomes and lysosomes (endolysosomes) are acidic organelles known to contain readily releasable stores of various cations including iron and other metals. Increased levels of ferrous (Fe2+) iron can generate reactive oxygen species (ROS) via Fenton chemistry reactions and these increases can damage mitochondria and genomic DNA as well as promote carcinogenesis. Accumulation of iron in the brain has been linked with aging, diet, disease, and cerebral hemorrhage. Further, deregulation of brain iron metabolism has been implicated in carcinogenesis and may be a contributing factor to the increased incidence of brain tumors around the world. Here, we provide insight into mechanisms by which iron accumulation in endolysosomes is altered by pH and lysosome membrane permeabilization. Such events generate excess ROS resulting in mitochondrial DNA damage, fission, and dysfunction, as well as DNA oxidative damage in the nucleus; all of which promote carcinogenesis. A better understanding of the roles that endolysosome iron plays in carcinogenesis may help better inform the development of strategic therapeutic options for cancer treatment and prevention.
Collapse
Affiliation(s)
- Peter W Halcrow
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| | - Miranda L Lynch
- Hauptman-Woodward Medical Research Institute, Buffalo, NY, United States
| | - Jonathan D Geiger
- Department of Biomedical Sciences, University of North Dakota School of Medicine and Health Sciences, Grand Forks, ND, United States
| | - Joyce E Ohm
- Department of Cancer Genetics and Genomics, Roswell Park Cancer Institute, Buffalo, NY, United States.
| |
Collapse
|
24
|
Banerjee R, Mukherjee A, Nagotu S. Mitochondrial dynamics and its impact on human health and diseases: inside the DRP1 blackbox. J Mol Med (Berl) 2021; 100:1-21. [PMID: 34657190 DOI: 10.1007/s00109-021-02150-7] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Revised: 08/24/2021] [Accepted: 10/06/2021] [Indexed: 01/01/2023]
Abstract
Mitochondria are essential organelles that play a significant role in various cellular processes apart from providing energy in eukaryotic cells. An intricate link between mitochondrial structure and function is now unequivocally accepted. Several molecular players have been identified, which are important in maintaining the structure of the organelle. Dynamin-related protein 1 (DRP1) is one such conserved protein that is a vital regulator of mitochondrial dynamics. Multidisciplinary studies have helped elucidate the structure of the protein and its mechanism of action in great detail. Mutations in various domains of the protein have been identified that are associated with debilitating conditions in patients. The involvement of the protein in disease conditions such as neurodegeneration, cancer, and cardiovascular disorders is also gaining attention. The purpose of this review is to highlight recent findings on the role of DRP1 in human disease conditions and address its importance as a therapeutic target.
Collapse
Affiliation(s)
- Riddhi Banerjee
- Organelle Biology and Cellular Ageing Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati-781039, Assam, India
| | - Agradeep Mukherjee
- Organelle Biology and Cellular Ageing Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati-781039, Assam, India
| | - Shirisha Nagotu
- Organelle Biology and Cellular Ageing Lab, Department of Biosciences and Bioengineering, Indian Institute of Technology Guwahati, Guwahati-781039, Assam, India.
| |
Collapse
|
25
|
Current Concepts on Genetic Aspects of Mitochondrial Dysfunction in Amyotrophic Lateral Sclerosis. Int J Mol Sci 2021; 22:ijms22189832. [PMID: 34575995 PMCID: PMC8469731 DOI: 10.3390/ijms22189832] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2021] [Revised: 09/08/2021] [Accepted: 09/09/2021] [Indexed: 12/12/2022] Open
Abstract
Amyotrophic Lateral Sclerosis (ALS), neurodegenerative motor neuron disorder is characterized as multisystem disease with important contribution of genetic factors. The etiopahogenesis of ALS is not fully elucidate, but the dominant theory at present relates to RNA processing, as well as protein aggregation and miss-folding, oxidative stress, glutamate excitotoxicity, inflammation and epigenetic dysregulation. Additionally, as mitochondria plays a leading role in cellular homeostasis maintenance, a rising amount of evidence indicates mitochondrial dysfunction as a substantial contributor to disease onset and progression. The aim of this review is to summarize most relevant findings that link genetic factors in ALS pathogenesis with different mechanisms with mitochondrial involvement (respiratory chain, OXPHOS control, calcium buffering, axonal transport, inflammation, mitophagy, etc.). We highlight the importance of a widening perspective for better understanding overlapping pathophysiological pathways in ALS and neurodegeneration in general. Finally, current and potentially novel therapies, especially gene specific therapies, targeting mitochondrial dysfunction are discussed briefly.
Collapse
|
26
|
Zeng S, Zhao Z, Zheng S, Wu M, Song X, Li Y, Zheng Y, Liu B, Chen L, Gao C, Liu H. The E3 ubiquitin ligase TRIM31 is involved in cerebral ischemic injury by promoting degradation of TIGAR. Redox Biol 2021; 45:102058. [PMID: 34218200 PMCID: PMC8260875 DOI: 10.1016/j.redox.2021.102058] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 06/23/2021] [Accepted: 06/24/2021] [Indexed: 11/19/2022] Open
Abstract
Tripartite motif (TRIM) 31 has been implicated in diverse biological and pathological conditions. However, whether TRIM31 plays a role in ischemic stroke progression is not clarified. Here we demonstrated that TRIM31 was significantly downregulated in the ischemic brain and the deficiency of TRIM31 alleviated brain injury induced by middle cerebral artery occlusion by reducing reactive oxygen species production and maintaining mitochondrial homeostasis. Mechanistically, we found that TRIM31 is an E3 ubiquitin ligase for TP53-induced glycolysis and apoptosis regulator (TIGAR), which confers protection against brain ischemia by increasing the pentose phosphate pathway flux and preserving mitochondria function. TRIM31 interacted with TIGAR and promoted the polyubiquitination of TIGAR, consequently facilitated its degradation in a proteasome-dependent pathway. Furthermore, TIGAR knockdown effectively abolished the protective effect of TRIM31 deficiency after cerebral ischemia. In conclusion, we identified that TRIM31 was a novel E3 ubiquitin ligase for TIGAR, played a critical role in regulating its protein level, and subsequently involved in the ischemic brain injury, suggesting TRIM31 as a potential therapeutic target for ischemic stroke.
Collapse
Affiliation(s)
- Shenglan Zeng
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Ze Zhao
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Shengnan Zheng
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Mengting Wu
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Xiaomeng Song
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Yiquan Li
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Yi Zheng
- Key Laboratory of Infection and Immunity of Shandong Province & Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Bingyu Liu
- Key Laboratory of Infection and Immunity of Shandong Province & Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Lin Chen
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, PR China
| | - Chengjiang Gao
- Key Laboratory of Infection and Immunity of Shandong Province & Department of Immunology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, PR China.
| | - Huiqing Liu
- Department of Pharmacology, School of Basic Medical Sciences, Shandong University, Jinan, Shandong, 250012, PR China.
| |
Collapse
|
27
|
Pascucci B, Spadaro F, Pietraforte D, Nuccio CD, Visentin S, Giglio P, Dogliotti E, D’Errico M. DRP1 Inhibition Rescues Mitochondrial Integrity and Excessive Apoptosis in CS-A Disease Cell Models. Int J Mol Sci 2021; 22:ijms22137123. [PMID: 34281194 PMCID: PMC8268695 DOI: 10.3390/ijms22137123] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 06/25/2021] [Accepted: 06/28/2021] [Indexed: 11/16/2022] Open
Abstract
Cockayne syndrome group A (CS-A) is a rare recessive progeroid disorder characterized by sun sensitivity and neurodevelopmental abnormalities. Cells derived from CS-A patients present as pathological hallmarks excessive oxidative stress, mitochondrial fragmentation and apoptosis associated with hyperactivation of the mitochondrial fission dynamin related protein 1 (DRP1). In this study, by using human cell models we further investigated the interplay between DRP1 and CSA and we determined whether pharmacological or genetic inhibition of DRP1 affects disease progression. Both reactive oxygen and nitrogen species are in excess in CS-A cells and when the mitochondrial translocation of DRP1 is inhibited a reduction of these species is observed together with a recovery of mitochondrial integrity and a significant decrease of apoptosis. This study indicates that the CSA-driven modulation of DRP1 pathway is key to control mitochondrial homeostasis and apoptosis and suggests DRP1 as a potential target in the treatment of CS patients.
Collapse
Affiliation(s)
- Barbara Pascucci
- Institute of Crystallography, Consiglio Nazionale delle Ricerche, 00015 Rome, Italy;
- Department of Environment and Health, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Francesca Spadaro
- Core Facilities, Istituto Superiore di Sanità, 00161 Rome, Italy; (F.S.); (D.P.)
| | | | - Chiara De Nuccio
- Research Coordination and Support Service, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Sergio Visentin
- National Center for Research and Preclinical and Clinical Evaluation of Drugs, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Paola Giglio
- Department of Biology, Tor Vergata University, 00133 Rome, Italy;
| | - Eugenia Dogliotti
- Department of Environment and Health, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Mariarosaria D’Errico
- Department of Environment and Health, Istituto Superiore di Sanità, 00161 Rome, Italy;
- Correspondence:
| |
Collapse
|
28
|
Kedra J, Lin S, Pacheco A, Gallo G, Smith GM. Axotomy Induces Drp1-Dependent Fragmentation of Axonal Mitochondria. Front Mol Neurosci 2021; 14:668670. [PMID: 34149354 PMCID: PMC8209475 DOI: 10.3389/fnmol.2021.668670] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 05/07/2021] [Indexed: 02/02/2023] Open
Abstract
It is well established that CNS axons fail to regenerate, undergo retrograde dieback, and form dystrophic growth cones due to both intrinsic and extrinsic factors. We sought to investigate the role of axonal mitochondria in the axonal response to injury. A viral vector (AAV) containing a mitochondrially targeted fluorescent protein (mitoDsRed) as well as fluorescently tagged LC3 (GFP-LC3), an autophagosomal marker, was injected into the primary motor cortex, to label the corticospinal tract (CST), of adult rats. The axons of the CST were then injured by dorsal column lesion at C4-C5. We found that mitochondria in injured CST axons near the injury site are fragmented and fragmentation of mitochondria persists for 2 weeks before returning to pre-injury lengths. Fragmented mitochondria have consistently been shown to be dysfunctional and detrimental to cellular health. Inhibition of Drp1, the GTPase responsible for mitochondrial fission, using a specific pharmacological inhibitor (mDivi-1) blocked fragmentation. Additionally, it was determined that there is increased mitophagy in CST axons following Spinal cord injury (SCI) based on increased colocalization of mitochondria and LC3. In vitro models revealed that mitochondrial divalent ion uptake is necessary for injury-induced mitochondrial fission, as inhibiting the mitochondrial calcium uniporter (MCU) using RU360 prevented injury-induced fission. This phenomenon was also observed in vivo. These studies indicate that following the injury, both in vivo and in vitro, axonal mitochondria undergo increased fission, which may contribute to the lack of regeneration seen in CNS neurons.
Collapse
Affiliation(s)
- Joseph Kedra
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Shen Lin
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Almudena Pacheco
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - Gianluca Gallo
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States.,Department of Anatomy and Cell Biology, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| | - George M Smith
- Shriners Hospitals Pediatric Research Center, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States.,Department of Neuroscience, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, United States
| |
Collapse
|
29
|
Drp1-dependent mitochondrial fission in cardiovascular disease. Acta Pharmacol Sin 2021; 42:655-664. [PMID: 32913266 DOI: 10.1038/s41401-020-00518-y] [Citation(s) in RCA: 173] [Impact Index Per Article: 57.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2020] [Accepted: 08/19/2020] [Indexed: 12/21/2022] Open
Abstract
Mitochondria are highly dynamic organelles undergoing cycles of fusion and fission to modulate their morphology, distribution, and function, which are referred as 'mitochondrial dynamics'. Dynamin-related protein 1 (Drp1) is known as the major pro-fission protein whose activity is tightly regulated to clear the damaged mitochondria via mitophagy, ensuring a strict control over the intricate process of cellular and organ dynamics in heart. Various posttranslational modifications (PTMs) of Drp1 have been identified including phosphorylation, SUMOylation, palmitoylation, ubiquitination, S-nitrosylation, and O-GlcNAcylation, which implicate a role in the regulation of mitochondrial dynamics. An intact mitochondrial homeostasis is critical for heart to fuel contractile function and cardiomyocyte metabolism, while defects in mitochondrial dynamics constitute an essential part of the pathophysiology underlying various cardiovascular diseases (CVDs). In this review, we summarize current knowledge on the critical role of Drp1 in the pathogenesis of CVDs including endothelial dysfunction, smooth muscle remodeling, cardiac hypertrophy, pulmonary arterial hypertension, myocardial ischemia-reperfusion, and myocardial infarction. We also highlight how the targeting of Drp1 could potentially contribute to CVDs treatments.
Collapse
|
30
|
Depletion of mitochondrial inorganic polyphosphate (polyP) in mammalian cells causes metabolic shift from oxidative phosphorylation to glycolysis. Biochem J 2021; 478:1631-1646. [PMID: 33843973 DOI: 10.1042/bcj20200975] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 04/08/2021] [Accepted: 04/12/2021] [Indexed: 12/11/2022]
Abstract
Inorganic polyphosphate (polyP) is a linear polymer composed of up to a few hundred orthophosphates linked together by high-energy phosphoanhydride bonds, identical with those found in ATP. In mammalian mitochondria, polyP has been implicated in multiple processes, including energy metabolism, ion channels function, and the regulation of calcium signaling. However, the specific mechanisms of all these effects of polyP within the organelle remain poorly understood. The central goal of this study was to investigate how mitochondrial polyP participates in the regulation of the mammalian cellular energy metabolism. To accomplish this, we created HEK293 cells depleted of mitochondrial polyP, through the stable expression of the polyP hydrolyzing enzyme (scPPX). We found that these cells have significantly reduced rates of oxidative phosphorylation (OXPHOS), while their rates of glycolysis were elevated. Consistent with this, metabolomics assays confirmed increased levels of metabolites involved in glycolysis in these cells, compared with the wild-type samples. At the same time, key respiratory parameters of the isolated mitochondria were unchanged, suggesting that respiratory chain activity is not affected by the lack of mitochondrial polyP. However, we detected that mitochondria from cells that lack mitochondrial polyP are more fragmented when compared with those from wild-type cells. Based on these results, we propose that mitochondrial polyP plays an important role as a regulator of the metabolic switch between OXPHOS and glycolysis.
Collapse
|
31
|
Machine learning-based classification of mitochondrial morphology in primary neurons and brain. Sci Rep 2021; 11:5133. [PMID: 33664336 PMCID: PMC7933342 DOI: 10.1038/s41598-021-84528-8] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2020] [Accepted: 02/17/2021] [Indexed: 01/31/2023] Open
Abstract
The mitochondrial network continually undergoes events of fission and fusion. Under physiologic conditions, the network is in equilibrium and is characterized by the presence of both elongated and punctate mitochondria. However, this balanced, homeostatic mitochondrial profile can change morphologic distribution in response to various stressors. Therefore, it is imperative to develop a method that robustly measures mitochondrial morphology with high accuracy. Here, we developed a semi-automated image analysis pipeline for the quantitation of mitochondrial morphology for both in vitro and in vivo applications. The image analysis pipeline was generated and validated utilizing images of primary cortical neurons from transgenic mice, allowing genetic ablation of key components of mitochondrial dynamics. This analysis pipeline was further extended to evaluate mitochondrial morphology in vivo through immunolabeling of brain sections as well as serial block-face scanning electron microscopy. These data demonstrate a highly specific and sensitive method that accurately classifies distinct physiological and pathological mitochondrial morphologies. Furthermore, this workflow employs the use of readily available, free open-source software designed for high throughput image processing, segmentation, and analysis that is customizable to various biological models.
Collapse
|
32
|
Song N, Zhu H, Xu R, Liu J, Fang Y, Zhang J, Ding J, Hu G, Lu M. Induced Expression of kir6.2 in A1 Astrocytes Propagates Inflammatory Neurodegeneration via Drp1-dependent Mitochondrial Fission. Front Pharmacol 2021; 11:618992. [PMID: 33584303 PMCID: PMC7876245 DOI: 10.3389/fphar.2020.618992] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 12/24/2020] [Indexed: 12/20/2022] Open
Abstract
Glia-mediated inflammatory processes are crucial in the pathogenesis of Parkinson’s disease (PD). As the most abundant cells of the brain and active participants in neuroinflammatory responses, astrocytes largely propagate inflammatory signals and amplify neuronal loss. Hence, intensive control of astrocytic activation is necessary to prevent neurodegeneration. In this study, we report that the astrocytic kir6.2, as a abnormal response after inflammatory stimuli, promotes the reactivity of A1 neurotoxic astrocytes. Using kir6.2 knockout (KO) mice, we find reversal effects of kir6.2 deficiency on A1-like astrocyte activation and death of dopaminergic neurons in lipopolysaccharide (LPS)-induced mouse models for PD. Further in vitro experiments show that aberrant kir6.2 expression induced by inflammatory irritants in astrocytes mediates the dynamin-related protein 1 (Drp1)-dependent excessive mitochondrial fragmentation and results in mitochondrial malfunctions. By deleting kir6.2, astrocytic activation is reduced and astrocytes-derived neuronal injury is prevented. We therefore conclude that astrocytic kir6.2 can potentially elucidate the pathology of PD and promote the development of therapeutic strategies for PD.
Collapse
Affiliation(s)
- Nanshan Song
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Hong Zhu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Rong Xu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Jiaqi Liu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Yinquan Fang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Jing Zhang
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Jianhua Ding
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| | - Gang Hu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China.,Department of Pharmacology, Nanjing University of Chinese Medicine, Nanjing, China
| | - Ming Lu
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology, Nanjing Medical University, Nanjing, China.,Neuroprotective Drug Discovery Key Laboratory, Department of Pharmacology, Nanjing Medical University, Nanjing, China
| |
Collapse
|
33
|
Clark AJ, Parikh SM. Targeting energy pathways in kidney disease: the roles of sirtuins, AMPK, and PGC1α. Kidney Int 2020; 99:828-840. [PMID: 33307105 DOI: 10.1016/j.kint.2020.09.037] [Citation(s) in RCA: 39] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2020] [Revised: 09/03/2020] [Accepted: 09/09/2020] [Indexed: 12/16/2022]
Abstract
The kidney has extraordinary metabolic demands to sustain the active transport of solutes that is critical to renal filtration and clearance. Mitochondrial health is vital to meet those demands and maintain renal fitness. Decades of studies have linked poor mitochondrial health to kidney disease. Key regulators of mitochondrial health-adenosine monophosphate kinase, sirtuins, and peroxisome proliferator-activated receptor γ coactivator-1α-have all been shown to play significant roles in renal resilience against disease. This review will summarize the latest research into the activities of those regulators and evaluate the roles and therapeutic potential of targeting those regulators in acute kidney injury, glomerular kidney disease, and renal fibrosis.
Collapse
Affiliation(s)
- Amanda J Clark
- Division of Nephrology, Boston Children's Hospital, Boston, Massachusetts, USA; Harvard Medical School, Boston, Massachusetts, USA
| | - Samir M Parikh
- Harvard Medical School, Boston, Massachusetts, USA; Division of Nephrology and Center for Vascular Biology Research, Beth Israel Deaconess Medical Center, Boston, Massachusetts, USA.
| |
Collapse
|
34
|
Mitochondrial dysfunction in the development and progression of neurodegenerative diseases. Arch Biochem Biophys 2020; 702:108698. [PMID: 33259796 DOI: 10.1016/j.abb.2020.108698] [Citation(s) in RCA: 120] [Impact Index Per Article: 30.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/11/2020] [Accepted: 11/21/2020] [Indexed: 02/07/2023]
Abstract
In addition to ATP synthesis, mitochondria are highly dynamic organelles that modulate apoptosis, ferroptosis, and inflammasome activation. Through executing these varied functions, the mitochondria play critical roles in the development and progression of neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, Huntington's disease, and Friedreich ataxia, among others. Impaired mitochondrial biogenesis and abnormal mitochondrial dynamics contribute to mitochondrial dysfunction in these diseases. Additionally, dysfunctional mitochondria play critical roles in signaling for both inflammasome activation and ferroptosis. Therapeutics are being developed to circumvent inflammasome activation and ferroptosis in dysfunctional mitochondria. Targeting these aspects of mitochondrial dysfunction may present viable therapeutic strategies for combatting the neurodegenerative diseases. This review aims to summarize the role of the mitochondria in the development and progression of neurodegenerative diseases and to present current therapeutic approaches that target mitochondrial dysfunction in these diseases.
Collapse
|
35
|
Chen Y, Guo S, Tang Y, Mou C, Hu X, Shao F, Yan W, Wu Q. Mitochondrial Fusion and Fission in Neuronal Death Induced by Cerebral Ischemia-Reperfusion and Its Clinical Application: A Mini-Review. Med Sci Monit 2020; 26:e928651. [PMID: 33156817 PMCID: PMC7654336 DOI: 10.12659/msm.928651] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Mitochondria are highly dynamic organelles which are joined by mitochondrial fusion and divided by mitochondrial fission. The balance of mitochondrial fusion and fission plays a critical role in maintaining the normal function of neurons, of which the processes are both mediated by several proteins activated by external stimulation. Cerebral ischemia-reperfusion (I/R) injury can disrupt the balance of mitochondrial fusion and fission through regulating the expression and post-translation modification of fusion- and fission-related proteins, thereby destroying homeostasis of the intracellular environment and causing neuronal death. Furthermore, human intervention in fusion- and fission-related proteins can influence the function of neurons and change the outcomes of cerebral I/R injury. In recent years, researchers have found that mitochondrial dysfunction was one of the main factors involved in I/R, and mitochondria is an attractive target in I/R neuroprotection. Therefore, mitochondrial-targeted therapy of the nervous system for I/R gradually started from basic study to clinical application. In the present review, we highlight recent progress in mitochondria fusion and fission in neuronal death induced by cerebral I/R to help understanding the regulatory factors and signaling networks of aberrant mitochondrial fusion and fission contributing to neuronal death during I/R, as well as the potential neuroprotective therapeutics targeting mitochondrial dynamics, which may help clinical treatment and development of relevant dugs.
Collapse
Affiliation(s)
- Yike Chen
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| | - Songxue Guo
- Department of Plastic Surgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| | - Yajuan Tang
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| | - Chaohui Mou
- Department of Neurosurgery, Taizhou First People's Hospital, Taizhou, Zhejiang, China (mainland)
| | - Xinben Hu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| | - Fangjie Shao
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| | - Wei Yan
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| | - Qun Wu
- Department of Neurosurgery, Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China (mainland)
| |
Collapse
|
36
|
Choi SY, Lee JH, Chung AY, Jo Y, Shin JH, Park HC, Kim H, Lopez-Gonzalez R, Ryu JR, Sun W. Prevention of mitochondrial impairment by inhibition of protein phosphatase 1 activity in amyotrophic lateral sclerosis. Cell Death Dis 2020; 11:888. [PMID: 33087694 PMCID: PMC7578657 DOI: 10.1038/s41419-020-03102-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 12/11/2022]
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal neurodegenerative disease caused by progressive loss of motor neurons (MNs) and subsequent muscle weakness. These pathological features are associated with numerous cellular changes, including alteration in mitochondrial morphology and function. However, the molecular mechanisms associating mitochondrial structure with ALS pathology are poorly understood. In this study, we found that Dynamin-related protein 1 (Drp1) was dephosphorylated in several ALS models, including those with SOD1 and TDP-43 mutations, and the dephosphorylation was mediated by the pathological induction of protein phosphatase 1 (PP1) activity in these models. Suppression of the PP1-Drp1 cascade effectively prevented ALS-related symptoms, including mitochondrial fragmentation, mitochondrial complex I impairment, axonal degeneration, and cell death, in primary neuronal culture models, iPSC-derived human MNs, and zebrafish models in vivo. These results suggest that modulation of PP1-Drp1 activity may be a therapeutic target for multiple pathological features of ALS.
Collapse
Affiliation(s)
- So Yoen Choi
- Department of Anatomy, Korea University College of Medicine, Brain Korea 21 plus, Seoul, 02841, Republic of Korea
- Department of Neurology, University of Massachusetts Medical school, Worcester, MA, USA
| | - Ju-Hyun Lee
- Department of Anatomy, Korea University College of Medicine, Brain Korea 21 plus, Seoul, 02841, Republic of Korea
| | - Ah-Young Chung
- Graduate School of Medicine, Korea University, Ansan, Gyeonggido, Republic of Korea
| | - Youhwa Jo
- Department of Anatomy, Korea University College of Medicine, Brain Korea 21 plus, Seoul, 02841, Republic of Korea
| | - Joo-Ho Shin
- Division of Pharmacology, Department of Molecular Cell Biology, Samsung Biomedical Research Institute, Sungkyunkwan University School of Medicine, Suwon, Gyeonggi-do, 440-746, Republic of Korea
| | - Hae-Chul Park
- Graduate School of Medicine, Korea University, Ansan, Gyeonggido, Republic of Korea
| | - Hyun Kim
- Department of Anatomy, Korea University College of Medicine, Brain Korea 21 plus, Seoul, 02841, Republic of Korea
| | | | - Jae Ryun Ryu
- Department of Anatomy, Korea University College of Medicine, Brain Korea 21 plus, Seoul, 02841, Republic of Korea
| | - Woong Sun
- Department of Anatomy, Korea University College of Medicine, Brain Korea 21 plus, Seoul, 02841, Republic of Korea.
| |
Collapse
|
37
|
Finelli MJ. Redox Post-translational Modifications of Protein Thiols in Brain Aging and Neurodegenerative Conditions-Focus on S-Nitrosation. Front Aging Neurosci 2020; 12:254. [PMID: 33088270 PMCID: PMC7497228 DOI: 10.3389/fnagi.2020.00254] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/24/2020] [Indexed: 12/14/2022] Open
Abstract
Reactive oxygen species and reactive nitrogen species (RONS) are by-products of aerobic metabolism. RONS trigger a signaling cascade that can be transduced through oxidation-reduction (redox)-based post-translational modifications (redox PTMs) of protein thiols. This redox signaling is essential for normal cellular physiology and coordinately regulates the function of redox-sensitive proteins. It plays a particularly important role in the brain, which is a major producer of RONS. Aberrant redox PTMs of protein thiols can impair protein function and are associated with several diseases. This mini review article aims to evaluate the role of redox PTMs of protein thiols, in particular S-nitrosation, in brain aging, and in neurodegenerative diseases. It also discusses the potential of using redox-based therapeutic approaches for neurodegenerative conditions.
Collapse
Affiliation(s)
- Mattéa J Finelli
- School of Medicine, Biodiscovery Institute, University of Nottingham, Nottingham, United Kingdom
| |
Collapse
|
38
|
Dai CQ, Guo Y, Chu XY. Neuropathic Pain: the Dysfunction of Drp1, Mitochondria, and ROS Homeostasis. Neurotox Res 2020; 38:553-563. [PMID: 32696439 DOI: 10.1007/s12640-020-00257-2] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2020] [Revised: 07/09/2020] [Accepted: 07/13/2020] [Indexed: 12/17/2022]
Abstract
Neuropathic pain affects the physical and mental health status of patients. Due to its complex pathogenesis and the adverse reactions to medicines, its treatment remains challenging. Among all the etiologies, increasing evidence has pointed to mitochondrial dysfunction. Dynamin-related protein 1 (Drp1)-mediated mitochondrial fragmentation leads to excess ROS generation, which is implicated in the pathogenesis of neuropathic pain. However, the exact mechanism remains unclear. Studies aiming to clarify the possible pathway and relationship between Drp1, mitochondria, ROS, and neuropathic pain may identify a good treatment for neuropathic pain in the clinic. As shown in this review, dysfunction of Drp1 and ROS homeostasis plays essential roles in neuropathic pain. We summarized a Drp1-mitochondrial fission-ROS cycle that potentially functions in neuropathic pain and is regulated by posttranslational modifications and Ca2+. Additionally, we further enumerated six Drp1 inhibitors, including Mdivi-1, P110, Drp1 antisense oligodeoxynucleotides, hyperbaric oxygen, melatonin, and β-hydroxybutyrate, as potential treatments, with the aim of providing guidance for novel molecules to be used in the clinic.
Collapse
Affiliation(s)
- Chun-Qiu Dai
- Third Medical District, Lintong Rehabilitation and Convalescent Centre, Xi'an, 710600, People's Republic of China
| | - Yu Guo
- Third Medical District, Lintong Rehabilitation and Convalescent Centre, Xi'an, 710600, People's Republic of China
| | - Xue-Yan Chu
- Third Medical District, Lintong Rehabilitation and Convalescent Centre, Xi'an, 710600, People's Republic of China.
| |
Collapse
|
39
|
Luo TT, Dai CQ, Wang JQ, Wang ZM, Yang Y, Zhang KL, Wu FF, Yang YL, Wang YY. Drp1 is widely, yet heterogeneously, distributed in the mouse central nervous system. Mol Brain 2020; 13:90. [PMID: 32522292 PMCID: PMC7288424 DOI: 10.1186/s13041-020-00628-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2020] [Accepted: 05/28/2020] [Indexed: 02/08/2023] Open
Abstract
Objectives Drp1 is widely expressed in the mouse central nervous system and plays a role in inducing the mitochondrial fission process. Many diseases are associated with Drp1 and mitochondria. However, since the exact distribution of Drp1 has not been specifically observed, it is difficult to determine the impact of anti-Drp1 molecules on the human body. Clarifying the specific Drp1 distribution could be a good approach to targeted treatment or prognosis. Methods We visualized the distribution of Drp1 in different brain regions and explicated the relationship between Drp1 and mitochondria. GAD67-GFP knock-in mice were utilized to detect the expression patterns of Drp1 in GABAergic neurons. We also further analyzed Drp1 expression in human malignant glioma tissue. Results Drp1 was widely but heterogeneously distributed in the central nervous system. Further observation indicated that Drp1 was highly and heterogeneously expressed in inhibitory neurons. Under transmission electron microscopy, the distribution of Drp1 was higher in dendrites than other areas in neurons, and only a small amount of Drp1 was localized in mitochondria. In human malignant glioma, the fluorescence intensity of Drp1 increased from grade I-III, while grade IV showed a declining trend. Conclusion In this study, we observed a wide heterogeneous distribution of Drp1 in the central nervous system, which might be related to the occurrence and development of neurologic disease. We hope that the relationship between Drp1 and mitochondria may will to therapeutic guidance in the clinic.
Collapse
Affiliation(s)
- Ting-Ting Luo
- National Demonstration Center for Experimental Preclinical Medicine Education, Air Force Medical University (The Fourth Military Medical University), Xi'an, 710032, China.,Mental Health Center, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Chun-Qiu Dai
- National Demonstration Center for Experimental Preclinical Medicine Education, Air Force Medical University (The Fourth Military Medical University), Xi'an, 710032, China.,Third Medical District, Lintong Rehabilitation and Convalescent Centre, Xi'an, 710600, China
| | - Jia-Qi Wang
- National Demonstration Center for Experimental Preclinical Medicine Education, Air Force Medical University (The Fourth Military Medical University), Xi'an, 710032, China
| | - Zheng-Mei Wang
- National Demonstration Center for Experimental Preclinical Medicine Education, Air Force Medical University (The Fourth Military Medical University), Xi'an, 710032, China.,Medical College of Yan'an University, Yan'an, 716000, China
| | - Yi Yang
- National Demonstration Center for Experimental Preclinical Medicine Education, Air Force Medical University (The Fourth Military Medical University), Xi'an, 710032, China.,Medical College of Yan'an University, Yan'an, 716000, China
| | - Kun-Long Zhang
- National Demonstration Center for Experimental Preclinical Medicine Education, Air Force Medical University (The Fourth Military Medical University), Xi'an, 710032, China.,Department of Rehabilitation Physiotherapy, Xi-Jing Hospital, Air Force Medical University (The Fourth Military Medical University), Xi'an, 710032, China
| | - Fei-Fei Wu
- National Demonstration Center for Experimental Preclinical Medicine Education, Air Force Medical University (The Fourth Military Medical University), Xi'an, 710032, China
| | - Yan-Ling Yang
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Air Force Medical University (The Fourth Military Medical University), Xi'an, 710032, China.
| | - Ya-Yun Wang
- National Demonstration Center for Experimental Preclinical Medicine Education, Air Force Medical University (The Fourth Military Medical University), Xi'an, 710032, China.
| |
Collapse
|
40
|
Protective Effects of Hyperbaric Oxygen Therapy on Brain Injury by Regulating the Phosphorylation of Drp1 Through ROS/PKC Pathway in Heatstroke Rats. Cell Mol Neurobiol 2020; 40:1253-1269. [PMID: 32043174 DOI: 10.1007/s10571-020-00811-8] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 02/04/2020] [Indexed: 12/14/2022]
Abstract
This study aimed to elucidate the neurotherapeutic effect of hyperbaric oxygen (HBO) on brain injury and the potential role of dynamin-related protein 1 (Drp1) and its regulatory pathway in heatstroke (HS) rats. In in vivo experiments, rats were exposed to HBO after the onset of HS, or the same pressure but normal air as a control. The results indicated that HBO decreased the mortality and thermoregulatory dysfunction and prolonged the survival time of HS rats. Neurological dysfunction induced by HS was attenuated by HBO through assessment of modified neurological severity score and Morris water maze. HBO also alleviated histopathologic changes and oxidative injury (malondialdehyde and 8-hydroxyguanine), increased activities of superoxide dismutase (SOD) and glutathione/oxidized glutathione and ameliorated apoptotic parameters (caspase-3/6 activities and the number of apoptotic cells) of the hippocampus, hypothalamus and brain stem in rats compared to the HS group. Phosphorylation of DrpSer616 was increased by HS but decreased by HBO in the brains of rats determined by Western blot and immunohistochemical staining. In experiments in vitro, rat hippocampal neurons were used as a heat stress (HS) cellular model to examine the effects of HBO. As the results, HBO attenuated HS-induced cytotoxicity, oxidative injury (malondialdehyde), reactive oxygen species (ROS) generation, decreasing SOD activity and apoptosis. Drp1 inhibitor (Mdivi-1) treatment produced the same effects and had a trend to decrease oxidative injury. But the difference is not statistically significant. HBO and Mdivi-1decreased the phosphorylation of DrpSer616 induced by HS and HBO decreased the phosphorylation of protein kinase C (PKC) induced by HS. Moreover, both PKC inhibitor and ROS scavenger inhibited HS-induced p-DrpSer616. In conclusion, HBO may alleviate the brain injury caused by HS by decreasing ROS/PKC-regulated p-DrpSer616.
Collapse
|
41
|
Li Y, He Y, Miao K, Zheng Y, Deng C, Liu TM. Imaging of macrophage mitochondria dynamics in vivo reveals cellular activation phenotype for diagnosis. Am J Cancer Res 2020; 10:2897-2917. [PMID: 32194843 PMCID: PMC7053213 DOI: 10.7150/thno.40495] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2019] [Accepted: 01/14/2020] [Indexed: 12/17/2022] Open
Abstract
Highly plastic macrophages are pivotal players in the body's homeostasis and pathogenesis. Grasping the molecular or cellular factors that drive and support the macrophage activation will help to develop diagnostics and manipulate their functions in these contexts. However, the lack of in vivo characterization methods to reveal the dynamic activation of macrophages impedes these studies in various disease contexts. Methods: Here, in vitro bone marrow-derived macrophages (BMDMs) and in vivo Matrigel plug were used to evaluate how mitochondria dynamics supports cellular activation and functions. We conducted macrophage repolarization in vitro to track mitochondria dynamics during the shift of activation status. For in vivo diagnosis, a novel MitoTracker-loaded liposome was first developed to label macrophage mitochondria in mice before/after inflammatory stimulation. Results: Based on the typical activation of in vitro BMDMs, we found glycolysis based macrophages have punctate and discrete mitochondria, while OXPHOS active macrophages have elongated and interconnected mitochondria. M1, M2a, M2b, and M2c activated BMDMs showed clustered and differentiable features in mitochondrial morphology. These features also hold for Matrigel plug-recruited macrophages in mice. Furthermore, with the interventions on M2a macrophages in vitro, we demonstrated that mitochondria morphology could be a metabolic index to evaluate macrophage activation status under drug manipulation. Using the MitoTracker-loaded liposomes, we further achieved subcellular imaging of macrophage mitochondria in vivo. Their organization dynamics revealed the dynamic change from anti-inflammatory macrophages to inflammatory ones in vivo under the lipopolysaccharide (LPS) challenge. Conclusion: These results reveal that subcellular imaging of mitochondria organization can characterize the activation status of macrophage in vitro and in vivo at a single-cell level, which is critical for the studies of noninvasive diagnosis and therapeutic drug monitoring.
Collapse
|
42
|
Yu M, Yang J, Gao X, Sun W, Liu S, Han Y, Lu X, Jin C, Wu S, Cai Y. Lanthanum chloride impairs spatial learning and memory by inducing [Ca2+]m overload, mitochondrial fission–fusion disorder and excessive mitophagy in hippocampal nerve cells of rats. Metallomics 2020; 12:592-606. [DOI: 10.1039/c9mt00291j] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Lanthanum chloride damages hippocampal nerve cells of rats through inducing [Ca2+]m overload, mitochondrial fission–fusion disorder, and excessive mitophagy.
Collapse
|
43
|
Suresh K, Servinsky L, Jiang H, Bigham Z, Zaldumbide J, Huetsch JC, Kliment C, Acoba MG, Kirsch BJ, Claypool SM, Le A, Damarla M, Shimoda LA. Regulation of mitochondrial fragmentation in microvascular endothelial cells isolated from the SU5416/hypoxia model of pulmonary arterial hypertension. Am J Physiol Lung Cell Mol Physiol 2019; 317:L639-L652. [PMID: 31461316 PMCID: PMC6879901 DOI: 10.1152/ajplung.00396.2018] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2018] [Revised: 08/16/2019] [Accepted: 08/17/2019] [Indexed: 01/10/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a morbid disease characterized by progressive right ventricle (RV) failure due to elevated pulmonary artery pressures (PAP). In PAH, histologically complex vaso-occlusive lesions in the pulmonary vasculature contribute to elevated PAP. However, the mechanisms underlying dysfunction of the microvascular endothelial cells (MVECs) that comprise a significant portion of these lesions are not well understood. We recently showed that MVECs isolated from the Sugen/hypoxia (SuHx) rat experimental model of PAH (SuHx-MVECs) exhibit increases in migration/proliferation, mitochondrial reactive oxygen species (ROS; mtROS) production, intracellular calcium levels ([Ca2+]i), and mitochondrial fragmentation. Furthermore, quenching mtROS with the targeted antioxidant MitoQ attenuated basal [Ca2+]i, migration and proliferation; however, whether increased mtROS-induced [Ca2+]i entry affected mitochondrial morphology was not clear. In this study, we sought to better understand the relationship between increased ROS, [Ca2+]i, and mitochondrial morphology in SuHx-MVECs. We measured changes in mitochondrial morphology at baseline and following inhibition of mtROS, with the targeted antioxidant MitoQ, or transient receptor potential vanilloid-4 (TRPV4) channels, which we previously showed were responsible for mtROS-induced increases in [Ca2+]i in SuHx-MVECs. Quenching mtROS or inhibiting TRPV4 attenuated fragmentation in SuHx-MVECs. Conversely, inducing mtROS production in MVECs from normoxic rats (N-MVECs) increased fragmentation. Ca2+ entry induced by the TRPV4 agonist GSK1017920A was significantly increased in SuHx-MVECs and was attenuated with MitoQ treatment, indicating that mtROS contributes to increased TRPV4 activity in SuHx-MVECs. Basal and maximal respiration were depressed in SuHx-MVECs, and inhibiting mtROS, but not TRPV4, improved respiration in these cells. Collectively, our data show that, in SuHx-MVECs, mtROS production promotes TRPV4-mediated increases in [Ca2+]i, mitochondrial fission, and decreased mitochondrial respiration. These results suggest an important role for mtROS in driving MVEC dysfunction in PAH.
Collapse
Affiliation(s)
- Karthik Suresh
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Laura Servinsky
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Haiyang Jiang
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Zahna Bigham
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Joel Zaldumbide
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - John C Huetsch
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Corrine Kliment
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Michelle G Acoba
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Brian J Kirsch
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Steven M Claypool
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Anne Le
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Mahendra Damarla
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| | - Larissa A Shimoda
- Division of Pulmonary and Critical Care Medicine, Department of Medicine, Johns Hopkins University School of Medicine, Baltimore, Maryland
| |
Collapse
|
44
|
Yu L, Xiao Z, Tu H, Tong B, Chen S. The expression and prognostic significance of Drp1 in lung cancer: A bioinformatics analysis and immunohistochemistry. Medicine (Baltimore) 2019; 98:e18228. [PMID: 31770286 PMCID: PMC6890372 DOI: 10.1097/md.0000000000018228] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
BACKGROUND Dynamin-related protein 1 (Drp1) plays important roles in tumorigenesis, including lung cancer. However, the effect of Drp1 in lung cancer remains unclear. The present study was aimed to investigate the clinical significance and effect of Drp1 on prognosis of lung cancer. METHODS Oncomine and The Cancer Genome Atlas (TCGA) databases were selected to predict the differential expression levels of Drp1 in lung cancer. Then, 70 cases of lung cancer and normal tissues were collected and immunohistochemistry was used to detect the expression of Drp1. In addition, Kaplan-Meier Plotter database and TCGA database were used to verify the correlation between Drp1 expression and the clinical prognosis in lung cancer patients. RESULTS Drp1 was significantly overexpressed in lung cancer tissues based on Oncomine and TCGA databases (P < .05). Moreover, results from immunohistochemistry showed that Drp1 protein level in lung cancer was also significantly higher than that in the matched normal tissues (P < .05). Prognostic analysis from Kaplan-Meier Plotter database with the chosen probe IDs of 203105_s_at suggested that Drp1 was negatively correlated to overall survival (OS) of lung cancer patients (HR = 1.16, 95% CI: 1.02-1.31; P = .025), but not in the probe IDs of 226154_at (HR = 0.86, 95% CI: 0.73-1.01; P = .069). However, prognosis from TCGA database showed inconsistent results in which high expression of Drp1 was correlated with worse survival probability of all, male, female in lung adenocarcinoma (P < .05), but not in LUSC (P > .05). CONCLUSION Drp1 was highly expressed in lung cancer based on bioinformatics analysis and tissue microarray, but there was a lot of inconsistency in prognosis depending on different levels of Drp1 from the bioinformatics analysis.
Collapse
Affiliation(s)
- Lingling Yu
- Department of Rehabilitation Medicine, the Second Affiliated Hospital of Nanchang University
| | - Zuke Xiao
- Department of Pulmonary and Critical Care Medicine, Jiangxi Provincial People's Hospital, Nanchang of Jiangxi, China
| | - Hongying Tu
- Department of Pulmonary and Critical Care Medicine, Jiangxi Provincial People's Hospital, Nanchang of Jiangxi, China
| | - Bo Tong
- Department of Pulmonary and Critical Care Medicine, Jiangxi Provincial People's Hospital, Nanchang of Jiangxi, China
| | - Shengsong Chen
- Department of Pulmonary and Critical Care Medicine, Jiangxi Provincial People's Hospital, Nanchang of Jiangxi, China
| |
Collapse
|
45
|
Peroxiredoxin 5 Inhibits Glutamate-Induced Neuronal Cell Death through the Regulation of Calcineurin-Dependent Mitochondrial Dynamics in HT22 Cells. Mol Cell Biol 2019; 39:MCB.00148-19. [PMID: 31358548 DOI: 10.1128/mcb.00148-19] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2019] [Accepted: 07/21/2019] [Indexed: 11/20/2022] Open
Abstract
Glutamate is an essential neurotransmitter in the central nervous system (CNS). However, high glutamate concentrations can lead to neurodegenerative diseases. A hallmark of glutamate toxicity is high levels of reactive oxygen species (ROS), which can trigger Ca2+ influx and dynamin-related protein 1 (Drp1)-mediated mitochondrial fission. Peroxiredoxin 5 (Prx5) is a well-known cysteine-dependent peroxidase enzyme. However, the precise effects of Prx5 on glutamate toxicity are still unclear. In this study, we investigated the role of Prx5 in glutamate-induced neuronal cell death. We found that glutamate treatment induces endogenous Prx5 expression and Ca2+/calcineurin-dependent dephosphorylation of Drp1, resulting in mitochondrial fission and neuronal cell death. Our results indicate that Prx5 inhibits glutamate-induced mitochondrial fission through the regulation of Ca2+/calcineurin-dependent dephosphorylation of Drp1, and it does so by scavenging cytosolic and mitochondrial ROS. Therefore, we suggest that Ca2+/calcineurin-dependent mitochondrial dynamics are deeply associated with glutamate-induced neurotoxicity. Consequently, Prx5 may be used as a potential agent for developing therapies against glutamate-induced neurotoxicity and neurodegenerative diseases where it plays a key role.
Collapse
|
46
|
Lee DG, Kim KM, Lee HS, Bae YC, Huh JW, Lee SR, Lee DS. Peroxiredoxin 5 prevents diethylhexyl phthalate-induced neuronal cell death by inhibiting mitochondrial fission in mouse hippocampal HT-22 cells. Neurotoxicology 2019; 74:242-251. [DOI: 10.1016/j.neuro.2019.08.003] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Revised: 07/29/2019] [Accepted: 08/05/2019] [Indexed: 02/07/2023]
|
47
|
Tseng HC, Lin CC, Hsiao LD, Yang CM. Lysophosphatidylcholine-induced mitochondrial fission contributes to collagen production in human cardiac fibroblasts. J Lipid Res 2019; 60:1573-1589. [PMID: 31363041 PMCID: PMC6718437 DOI: 10.1194/jlr.ra119000141] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Revised: 07/26/2019] [Indexed: 12/12/2022] Open
Abstract
Lysophosphatidylcholine (LPC) may accumulate in the heart to cause fibrotic events, which is mediated through fibroblast activation and collagen accumulation. Here, we evaluated the mechanisms underlying LPC-mediated collagen induction via mitochondrial events in human cardiac fibroblasts (HCFs), coupling application of the pharmacologic cyclooxygenase-2 (COX-2) inhibitor, celecoxib, and genetic mutations in FOXO1 on the fibrosis pathway. In HCFs, LPC caused prostaglandin E2 (PGE2)/PGE2 receptor 4 (EP4)-dependent collagen induction via activation of transcriptional activity of forkhead box protein O1 (FoxO1) on COX-2 gene expression. These responses were mediated through LPC-induced generation of mitochondrial reactive oxygen species (mitoROS), as confirmed by ex vivo studies, which indicated that LPC increased COX-2 expression and oxidative stress. LPC-induced mitoROS mediated the activation of protein kinase C (PKC)α, which interacted with and phosphorylated dynamin-related protein 1 (Drp1) at Ser616, thereby increasing Drp1-mediated mitochondrial fission and mitochondrial depolarization. Furthermore, inhibition of PKCα and Drp1 reduced FoxO1-mediated phosphorylation at Ser256 and nuclear accumulation, which suppressed COX-2/PGE2 expression and collagen production. Moreover, pretreatment with celecoxib or COX-2 siRNA suppressed WT FoxO1; mutated Ser256-to-Asp256 FoxO1-enhanced collagen induction, which was reversed by addition of PGE2 Our results demonstrate that LPC-induced generation of mitoROS regulates PKCα-mediated Drp1-dependent mitochondrial fission and COX-2 expression via a PKCα/Drp1/FoxO1 cascade, leading to PGE2/EP4-mediated collagen induction. These findings provide new insights about the role of LPC in the pathway of fibrotic injury in HCFs.
Collapse
Affiliation(s)
- Hui-Ching Tseng
- Graduate Institute of Biomedical Sciences, College of Medicine, and Department of Physiology and Pharmacology and Health Ageing Research Center, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Chih-Chung Lin
- Department of Anesthetics Chang Gung Memorial Hospital, Linkuo, Taiwan and Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Li-Der Hsiao
- Department of Anesthetics Chang Gung Memorial Hospital, Linkuo, Taiwan and Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | - Chuen-Mao Yang
- Graduate Institute of Biomedical Sciences, College of Medicine, and Department of Physiology and Pharmacology and Health Ageing Research Center, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan; Department of Anesthetics Chang Gung Memorial Hospital, Linkuo, Taiwan and Chang Gung University, Kwei-San, Tao-Yuan, Taiwan; Research Center for Chinese Herbal Medicine and Research Center for Food and Cosmetic Safety College of Human Ecology, Chang Gung University of Science and Technology, Tao-Yuan, Taiwan; Department of Pharmacology, College of Medicine, China Medical University, Taichung, Taiwan.
| |
Collapse
|
48
|
Bae JE, Park SJ, Hong Y, Jo DS, Lee H, Park NY, Kim JB, Park HJ, Bunch H, Chang JH, Lee EK, Cho DH. Loss of RNA binding protein, human antigen R enhances mitochondrial elongation by regulating Drp1 expression in SH-SY5Y cells. Biochem Biophys Res Commun 2019; 516:713-718. [DOI: 10.1016/j.bbrc.2019.06.091] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2019] [Accepted: 06/16/2019] [Indexed: 01/10/2023]
|
49
|
Chandra R, Engeln M, Schiefer C, Patton MH, Martin JA, Werner CT, Riggs LM, Francis TC, McGlincy M, Evans B, Nam H, Das S, Girven K, Konkalmatt P, Gancarz AM, Golden SA, Iñiguez SD, Russo SJ, Turecki G, Mathur BN, Creed M, Dietz DM, Lobo MK. Drp1 Mitochondrial Fission in D1 Neurons Mediates Behavioral and Cellular Plasticity during Early Cocaine Abstinence. Neuron 2019; 96:1327-1341.e6. [PMID: 29268097 DOI: 10.1016/j.neuron.2017.11.037] [Citation(s) in RCA: 68] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2016] [Revised: 09/12/2017] [Accepted: 11/17/2017] [Indexed: 02/07/2023]
Abstract
Altered brain energy homeostasis is a key adaptation occurring in the cocaine-addicted brain, but the effect of cocaine on the fundamental source of energy, mitochondria, is unknown. We demonstrate an increase of dynamin-related protein-1 (Drp1), the mitochondrial fission mediator, in nucleus accumbens (NAc) after repeated cocaine exposure and in cocaine-dependent individuals. Mdivi-1, a demonstrated fission inhibitor, blunts cocaine seeking and locomotor sensitization, while blocking c-Fos induction and excitatory input onto dopamine receptor-1 (D1) containing NAc medium spiny neurons (MSNs). Drp1 and fission promoting Drp1 are increased in D1-MSNs, consistent with increased smaller mitochondria in D1-MSN dendrites after repeated cocaine. Knockdown of Drp1 in D1-MSNs blocks drug seeking after cocaine self-administration, while enhancing the fission promoting Drp1 enhances seeking after long-term abstinence from cocaine. We demonstrate a role for altered mitochondrial fission in the NAc, during early cocaine abstinence, suggesting potential therapeutic treatment of disrupting mitochondrial fission in cocaine addiction.
Collapse
Affiliation(s)
- Ramesh Chandra
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Michel Engeln
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Christopher Schiefer
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Mary H Patton
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Jennifer A Martin
- Department of Pharmacology and Toxicology, The Research Institution on Addictions, State University of New York at Buffalo, Buffalo, NY, USA
| | - Craig T Werner
- Department of Pharmacology and Toxicology, The Research Institution on Addictions, State University of New York at Buffalo, Buffalo, NY, USA
| | - Lace M Riggs
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - T Chase Francis
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Madeleine McGlincy
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Brianna Evans
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Hyungwoo Nam
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Shweta Das
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Kasey Girven
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Prasad Konkalmatt
- Division of Renal Diseases and Hypertension, The George Washington University, Washington, D.C., USA
| | - Amy M Gancarz
- Department of Pharmacology and Toxicology, The Research Institution on Addictions, State University of New York at Buffalo, Buffalo, NY, USA
| | - Sam A Golden
- Fishberg Department of Neuroscience and Friedman Brain Institute, Graduate School of Biomedical Sciences at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Sergio D Iñiguez
- Department of Psychology, University of Texas at El Paso, El Paso, TX, USA
| | - Scott J Russo
- Fishberg Department of Neuroscience and Friedman Brain Institute, Graduate School of Biomedical Sciences at the Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Gustavo Turecki
- McGill Group for Suicide Studies, Douglas Mental Health University Institute, McGill University, Montréal, QC, Canada
| | - Brian N Mathur
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - Meaghan Creed
- Department of Pharmacology, University of Maryland School of Medicine, Baltimore, MD, USA
| | - David M Dietz
- Department of Pharmacology and Toxicology, The Research Institution on Addictions, State University of New York at Buffalo, Buffalo, NY, USA
| | - Mary Kay Lobo
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
50
|
Sehrawat A, Samanta SK, Hahm ER, St Croix C, Watkins S, Singh SV. Withaferin A-mediated apoptosis in breast cancer cells is associated with alterations in mitochondrial dynamics. Mitochondrion 2019; 47:282-293. [PMID: 30685490 PMCID: PMC6599725 DOI: 10.1016/j.mito.2019.01.003] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2018] [Accepted: 01/21/2019] [Indexed: 12/18/2022]
Abstract
Withaferin A (WA), a steroidal lactone derived from a medicinal plant (Withania somnifera), inhibits cancer development in transgenic and chemically-induced rodent models of breast cancer but the underlying mechanism is not fully grasped. We have shown previously that WA treatment causes apoptotic cell death in human breast cancer cells that is preceded by inhibition of complex III of the mitochondrial electron transport chain. This study extends these observations to now demonstrate alterations in mitochondrial dynamics in WA-induced apoptosis. Assembly of complex III was decreased in MCF-7 and SUM159 cells but not in MDA-MB-231 as determined by native blue gel electrophoresis. Because WA is a Michael acceptor (electrophile), we explored the possibility of whether it covalently modifies cysteine residue(s) in ubiquinol-cytochrome c reductase, Rieske iron-sulfur polypeptide 1 (UQCRFS1). Covalent modification of cysteine in UQCRFS1 was not observed after WA treatment. Instead, WA treatment inhibited chemically-induced mitochondrial fusion and decreased the mitochondrial volume, and this effect was accompanied by a decrease in the expression of proteins involved in fusion process, including mitofusin1, mitofusin2, and full-length optic atrophy protein 1 (OPA1). A loss of volume in fragmented mitochondria also occurred in WA-exposed cells when compared to vehicle-treated control. WA treatment also caused a decrease in protein level of mitochondrial fission-regulating protein dynamin-related protein 1 (DRP1). Functional studies revealed that DRP1 deficiency and OPA1 knockdown attenuated apoptotic potential of WA. Taken together, these results indicate that WA not only alters Complex III assembly but also inhibits mitochondrial dynamics in breast cancer cells.
Collapse
Affiliation(s)
- Anuradha Sehrawat
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Suman K Samanta
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; Life Sciences Division, Institute of Advanced Study in Science and Technology, Guwahati, India
| | - Eun-Ryeong Hahm
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Claudette St Croix
- Department of Cell Biology, University of Pittsburgh, Pittsburgh, PA, USA
| | - Simon Watkins
- Department of Cell Biology and Physiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; UPMC Hillman Cancer Center, Pittsburgh, PA, USA
| | - Shivendra V Singh
- Department of Pharmacology & Chemical Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA; UPMC Hillman Cancer Center, Pittsburgh, PA, USA.
| |
Collapse
|