1
|
Mohamed Yusoff AA, Mohd Khair SZN. Unraveling mitochondrial dysfunction: comprehensive perspectives on its impact on neurodegenerative diseases. Rev Neurosci 2025; 36:53-90. [PMID: 39174305 DOI: 10.1515/revneuro-2024-0080] [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: 06/06/2024] [Accepted: 07/30/2024] [Indexed: 08/24/2024]
Abstract
Neurodegenerative diseases represent a significant challenge to modern medicine, with their complex etiology and progressive nature posing hurdles to effective treatment strategies. Among the various contributing factors, mitochondrial dysfunction has emerged as a pivotal player in the pathogenesis of several neurodegenerative disorders. This review paper provides a comprehensive overview of how mitochondrial impairment contributes to the development of neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, Huntington's disease, and amyotrophic lateral sclerosis, driven by bioenergetic defects, biogenesis impairment, alterations in mitochondrial dynamics (such as fusion or fission), disruptions in calcium buffering, lipid metabolism dysregulation and mitophagy dysfunction. It also covers current therapeutic interventions targeting mitochondrial dysfunction in these diseases.
Collapse
Affiliation(s)
- Abdul Aziz Mohamed Yusoff
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia
| | - Siti Zulaikha Nashwa Mohd Khair
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kelantan, Malaysia
| |
Collapse
|
2
|
Zhou Q, Meng Y, Le J, Sun Y, Dian Y, Yao L, Xiong Y, Zeng F, Chen X, Deng G. Ferroptosis: mechanisms and therapeutic targets. MedComm (Beijing) 2024; 5:e70010. [PMID: 39568772 PMCID: PMC11577302 DOI: 10.1002/mco2.70010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 11/22/2024] Open
Abstract
Ferroptosis is a nonapoptotic form of cell death characterized by iron-dependent lipid peroxidation in membrane phospholipids. Since its identification in 2012, extensive research has unveiled its involvement in the pathophysiology of numerous diseases, including cancers, neurodegenerative disorders, organ injuries, infectious diseases, autoimmune conditions, metabolic disorders, and skin diseases. Oxidizable lipids, overload iron, and compromised antioxidant systems are known as critical prerequisites for driving overwhelming lipid peroxidation, ultimately leading to plasma membrane rupture and ferroptotic cell death. However, the precise regulatory networks governing ferroptosis and ferroptosis-targeted therapy in these diseases remain largely undefined, hindering the development of pharmacological agonists and antagonists. In this review, we first elucidate core mechanisms of ferroptosis and summarize its epigenetic modifications (e.g., histone modifications, DNA methylation, noncoding RNAs, and N6-methyladenosine modification) and nonepigenetic modifications (e.g., genetic mutations, transcriptional regulation, and posttranslational modifications). We then discuss the association between ferroptosis and disease pathogenesis and explore therapeutic approaches for targeting ferroptosis. We also introduce potential clinical monitoring strategies for ferroptosis. Finally, we put forward several unresolved issues in which progress is needed to better understand ferroptosis. We hope this review will offer promise for the clinical application of ferroptosis-targeted therapies in the context of human health and disease.
Collapse
Affiliation(s)
- Qian Zhou
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Yu Meng
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Jiayuan Le
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Yuming Sun
- Department of Plastic and Cosmetic Surgery Xiangya Hospital Central South University Changsha Hunan Province China
| | - Yating Dian
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Lei Yao
- Department of General Surgery Xiangya Hospital Central South University Changsha Hunan Province China
| | - Yixiao Xiong
- Department of Dermatology Tongji Hospital Huazhong University of Science and Technology Wuhan Hubei China
| | - Furong Zeng
- Department of Oncology Xiangya Hospital Central South University Changsha Hunan Province China
| | - Xiang Chen
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| | - Guangtong Deng
- Department of Dermatology Xiangya Hospital Central South University Changsha Hunan Province China
- National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology Changsha Hunan Province China
- Furong Laboratory Changsha Hunan Province China
- Hunan Key Laboratory of Skin Cancer and Psoriasis Hunan Engineering Research Center of Skin Health and Disease Xiangya Hospital Central South University Changsha Hunan Province China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Changsha Hunan Province China
| |
Collapse
|
3
|
Padalko V, Posnik F, Adamczyk M. Mitochondrial Aconitase and Its Contribution to the Pathogenesis of Neurodegenerative Diseases. Int J Mol Sci 2024; 25:9950. [PMID: 39337438 PMCID: PMC11431987 DOI: 10.3390/ijms25189950] [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: 08/06/2024] [Revised: 08/31/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
This survey reviews modern ideas on the structure and functions of mitochondrial and cytosolic aconitase isoenzymes in eukaryotes. Cumulative experimental evidence about mitochondrial aconitases (Aco2) as one of the main targets of reactive oxygen and nitrogen species is generalized. The important role of Aco2 in maintenance of homeostasis of the intracellular iron pool and maintenance of the mitochondrial DNA is discussed. The role of Aco2 in the pathogenesis of some neurodegenerative diseases is highlighted. Inactivation or dysfunction of Aco2 as well as mutations found in the ACO2 gene appear to be significant factors in the development and promotion of various types of neurodegenerative diseases. A restoration of efficient mitochondrial functioning as a source of energy for the cell by targeting Aco2 seems to be one of the promising therapeutic directions to minimize progressive neurodegenerative disorders.
Collapse
Affiliation(s)
- Volodymyr Padalko
- Laboratory of Systems and Synthetic Biology, Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
- School of Medicine, V. N. Karazin Kharkiv National University, 61022 Kharkiv, Ukraine
| | - Filip Posnik
- Laboratory of Systems and Synthetic Biology, Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
| | - Malgorzata Adamczyk
- Laboratory of Systems and Synthetic Biology, Chair of Drug and Cosmetics Biotechnology, Faculty of Chemistry, Warsaw University of Technology, Noakowskiego 3, 00-664 Warsaw, Poland
| |
Collapse
|
4
|
Ramazi S, Dadzadi M, Darvazi M, Seddigh N, Allahverdi A. Protein modification in neurodegenerative diseases. MedComm (Beijing) 2024; 5:e674. [PMID: 39105197 PMCID: PMC11298556 DOI: 10.1002/mco2.674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 08/07/2024] Open
Abstract
Posttranslational modifications play a crucial role in governing cellular functions and protein behavior. Researchers have implicated dysregulated posttranslational modifications in protein misfolding, which results in cytotoxicity, particularly in neurodegenerative diseases such as Alzheimer disease, Parkinson disease, and Huntington disease. These aberrant posttranslational modifications cause proteins to gather in certain parts of the brain that are linked to the development of the diseases. This leads to neuronal dysfunction and the start of neurodegenerative disease symptoms. Cognitive decline and neurological impairments commonly manifest in neurodegenerative disease patients, underscoring the urgency of comprehending the posttranslational modifications' impact on protein function for targeted therapeutic interventions. This review elucidates the critical link between neurodegenerative diseases and specific posttranslational modifications, focusing on Tau, APP, α-synuclein, Huntingtin protein, Parkin, DJ-1, and Drp1. By delineating the prominent aberrant posttranslational modifications within Alzheimer disease, Parkinson disease, and Huntington disease, the review underscores the significance of understanding the interplay among these modifications. Emphasizing 10 key abnormal posttranslational modifications, this study aims to provide a comprehensive framework for investigating neurodegenerative diseases holistically. The insights presented herein shed light on potential therapeutic avenues aimed at modulating posttranslational modifications to mitigate protein aggregation and retard neurodegenerative disease progression.
Collapse
Affiliation(s)
- Shahin Ramazi
- Department of BiophysicsFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| | - Maedeh Dadzadi
- Department of BiotechnologyFaculty of Advanced Science and TechnologyTehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Mona Darvazi
- Department of BiophysicsFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| | - Nasrin Seddigh
- Department of BiochemistryFaculty of Advanced Science and TechnologyTehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Abdollah Allahverdi
- Department of BiophysicsFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| |
Collapse
|
5
|
Ferdous KA, Jansen J, Amjad E, Pray E, Bloch R, Benoit A, Callahan M, Park HA. Mitochondrial protective potential of fucoxanthin in brain disorders. J Nutr Sci 2024; 13:e21. [PMID: 39776519 PMCID: PMC11704942 DOI: 10.1017/jns.2024.16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2023] [Revised: 04/15/2024] [Accepted: 05/09/2024] [Indexed: 01/11/2025] Open
Abstract
Mitochondrial dysfunction is a common feature of brain disorders. Mitochondria play a central role in oxidative phosphorylation; thus changes in energy metabolism in the brain have been reported in conditions such as Alzheimer's disease, Parkinson's disease, and stroke. In addition, mitochondria regulate cellular responses associated with neuronal damage such as the production of reactive oxygen species (ROS), opening of the mitochondrial permeability transition pore (mPTP), and apoptosis. Therefore, interventions that aim to protect mitochondria may be effective against brain disorders. Fucoxanthin is a marine carotenoid that has recently gained recognition for its neuroprotective properties. However, the cellular mechanisms of fucoxanthin in brain disorders, particularly its role in mitochondrial function, have not been thoroughly discussed. This review summarises the current literature on the effects of fucoxanthin on oxidative stress, neuroinflammation, and apoptosis using in vitro and in vivo models of brain disorders. We further present the potential mechanisms by which fucoxanthin protects mitochondria, with the objective of developing dietary interventions for a spectrum of brain disorders. Although the studies reviewed are predominantly preclinical studies, they provide important insights into understanding the cellular and molecular functions of fucoxanthin in the brain. Future studies investigating the mechanisms of action and the molecular targets of fucoxanthin are warranted to develop translational approaches to brain disorders.
Collapse
Affiliation(s)
- Khondoker Adeba Ferdous
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, USA
| | - Joseph Jansen
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, USA
| | - Emma Amjad
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, USA
| | - Eliana Pray
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, USA
| | - Rebecca Bloch
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, USA
| | - Alex Benoit
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, USA
| | - Meredith Callahan
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, USA
| | - Han-A Park
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, USA
| |
Collapse
|
6
|
Liang H, Ma Z, Zhong W, Liu J, Sugimoto K, Chen H. Regulation of mitophagy and mitochondrial function: Natural compounds as potential therapeutic strategies for Parkinson's disease. Phytother Res 2024; 38:1838-1862. [PMID: 38356178 DOI: 10.1002/ptr.8156] [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: 09/20/2023] [Revised: 01/27/2024] [Accepted: 01/29/2024] [Indexed: 02/16/2024]
Abstract
Mitochondrial damage is associated with the development of Parkinson's disease (PD), indicating that mitochondrial-targeted treatments could hold promise as disease-modifying approaches for PD. Notably, natural compounds have demonstrated the ability to modulate mitochondrial-related processes. In this review article, we discussed the possible neuroprotective mechanisms of natural compounds against PD in modulating mitophagy and mitochondrial function. A comprehensive literature search on natural compounds related to the treatment of PD by regulating mitophagy and mitochondrial function was conducted from PubMed, Web of Science and Chinese National Knowledge Infrastructure databases from their inception until April 2023. We summarize recent advancements in mitophagy's molecular mechanisms, including upstream and downstream processes, and its relationship with PD-related genes or proteins. Importantly, we highlight how natural compounds can therapeutically regulate various mitochondrial processes through multiple targets and pathways to alleviate oxidative stress, neuroinflammation, Lewy's body aggregation and apoptosis, which are key contributors to PD pathogenesis. Unlike the single-target strategy of modern medicine, natural compounds provide neuroprotection against PD by modulating various mitochondrial-related processes, including ameliorating mitophagy by targeting the PINK1/parkin pathway, the NIX/BNIP3 pathway, and autophagosome formation (i.e., LC3 and p62). Given the prevalence of mitochondrial damage in various neurodegenerative diseases, exploring the exact mechanism of natural compounds on mitophagy and mitochondrial dysfunction could shed light on the development of highly effective disease-modifying or adjuvant therapies targeting PD and other neurodegenerative disorders.
Collapse
Affiliation(s)
- Hao Liang
- Department of Acupuncture, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| | - Zhenwang Ma
- Department of Acupuncture, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
| | - Wei Zhong
- Department of Rheumatology and Immunology, Affiliated Qiqihar Hospital, Southern Medical University, Qiqihar, China
| | - Jia Liu
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
| | - Kazuo Sugimoto
- Department of Neurology, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- Institute for Brain Disorders, Beijing University of Chinese Medicine, Beijing, China
| | - Hong Chen
- Department of Acupuncture, Heilongjiang Academy of Traditional Chinese Medicine, Harbin, China
- Department of TCM Geriatric, Southern Medical University, Guangzhou, China
| |
Collapse
|
7
|
Kuil LE, Chauhan RK, de Graaf BM, Cheng WW, Kakiailatu NJM, Lasabuda R, Verhaeghe C, Windster JD, Schriemer D, Azmani Z, Brooks AS, Edie S, Reeves RH, Eggen BJL, Shepherd IT, Burns AJ, Hofstra RMW, Melotte V, Brosens E, Alves MM. ATP5PO levels regulate enteric nervous system development in zebrafish, linking Hirschsprung disease to Down Syndrome. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166991. [PMID: 38128843 DOI: 10.1016/j.bbadis.2023.166991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2023] [Revised: 12/09/2023] [Accepted: 12/11/2023] [Indexed: 12/23/2023]
Abstract
Hirschsprung disease (HSCR) is a complex genetic disorder characterized by the absence of enteric nervous system (ENS) in the distal region of the intestine. Down Syndrome (DS) patients have a >50-fold higher risk of developing HSCR than the general population, suggesting that overexpression of human chromosome 21 (Hsa21) genes contribute to HSCR etiology. However, identification of responsible genes remains challenging. Here, we describe a genetic screening of potential candidate genes located on Hsa21, using the zebrafish. Candidate genes were located in the DS-HSCR susceptibility region, expressed in the human intestine, were known potential biomarkers for DS prenatal diagnosis, and were present in the zebrafish genome. With this approach, four genes were selected: RCAN1, ITSN1, ATP5PO and SUMO3. However, only overexpression of ATP5PO, coding for a component of the mitochondrial ATPase, led to significant reduction of ENS cells. Paradoxically, in vitro studies showed that overexpression of ATP5PO led to a reduction of ATP5PO protein levels. Impaired neuronal differentiation and reduced mitochondrial ATP production, were also detected in vitro, after overexpression of ATP5PO in a neuroblastoma cell line. Finally, epistasis was observed between ATP5PO and ret, the most important HSCR gene. Taken together, our results identify ATP5PO as the gene responsible for the increased risk of HSCR in DS patients in particular if RET variants are also present, and show that a balanced expression of ATP5PO is required for normal ENS development.
Collapse
Affiliation(s)
- L E Kuil
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - R K Chauhan
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - B M de Graaf
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - W W Cheng
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - N J M Kakiailatu
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - R Lasabuda
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - C Verhaeghe
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - J D Windster
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands; Department of Pediatric Surgery, Erasmus University Medical Center Rotterdam, Sophia's Children's Hospital, Rotterdam, the Netherlands
| | - D Schriemer
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Z Azmani
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - A S Brooks
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - S Edie
- Johns Hopkins University School of Medicine, Department of Physiology and McKusick-Nathans Department of Genetic Medicine, Baltimore, MD, United States of America
| | - R H Reeves
- Johns Hopkins University School of Medicine, Department of Physiology and McKusick-Nathans Department of Genetic Medicine, Baltimore, MD, United States of America
| | - B J L Eggen
- Department of Biomedical Sciences of Cells and Systems, Section Molecular Neurobiology, University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - I T Shepherd
- Department of Biology, Emory University, Atlanta, GA, United States of America
| | - A J Burns
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands; Birth Defects Research Centre, UCL Institute of Child Health, London, United Kingdom; Gastrointestinal Drug Discovery Unit, Takeda Pharmaceuticals, Cambridge, MA, United States of America
| | - R M W Hofstra
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - V Melotte
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands; Department of Pathology, GROW-school for Oncology and Developmental Biology, Maastricht University Medical Center, Maastricht, the Netherlands
| | - E Brosens
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands
| | - M M Alves
- Department of Clinical Genetics, Erasmus University Medical Center Rotterdam - Sophia Children's Hospital, Rotterdam, the Netherlands; Department of Pediatric Surgery, Erasmus University Medical Center Rotterdam, Sophia's Children's Hospital, Rotterdam, the Netherlands.
| |
Collapse
|
8
|
Ouyang L, He T, Xing Y. Park7 protects retinal ganglion cells and promotes functional preservation after optic nerve crush via regulation of the Nrf2 signaling pathway. Graefes Arch Clin Exp Ophthalmol 2023; 261:3489-3502. [PMID: 37199801 DOI: 10.1007/s00417-023-06099-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 04/19/2023] [Accepted: 05/02/2023] [Indexed: 05/19/2023] Open
Abstract
PURPOSE We aim to investigate the effect of Park7 on mice RGC survival and function following optic nerve crush (ONC), and to explore its potential mechanism. METHODS Wild-type male C57BL/6J mice were subjected to optic nerve crush. Six weeks before ONC, mice received rAAV-shRNA (Park7)-EGFP or rAAV-EGFP intravitreally. Western blotting was used to detect Park7 levels. RGC survival was measured using immunofluorescence. Retinal cell apoptosis was detected using terminal deoxynucleotidyl transferase nick-end-labelling. An electroretinogram (ERG) and the optomotor response (OMR) were used to assess RGC function. Kelch-like ECH-associated protein 1 (Keap1), nuclear factor erythroid 2-related factor (Nrf2), and heme oxygenase 1 (HO-1) levels were assessed using western blotting. RESULTS ONC injury increased the relative expression of Park7 significantly and decreased RGC survival, the amplitude of the photopic negative response (PhNR), and OMR. Intravitreal injection of rAAV-shRNA(Park7)-EGFP downregulated Park7 expression and was clearly demonstrated by the green fluorescence protein in many retinal layers. Moreover, Park7 downregulation aggravated the decrease in RGC survival and amplitude of PhNR as well as the visual acuity after ONC. However, inhibition of Park7 significantly increased Keap1 levels, decreased the total and nuclear Nrf2 levels, and reduced HO-1 levels. CONCLUSIONS Park7 downregulation enhanced RGC injury and decreased retinal electrophysiological response and OMR after ONC in mice via the Keap1-Nrf2-HO-1 signaling pathway. Park7 may have neuroprotective effects and could represent a novel way to treat optic neuropathy.
Collapse
Affiliation(s)
- Lingyi Ouyang
- Department of Ophthalmology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei, China
| | - Tao He
- Department of Ophthalmology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei, China
| | - Yiqiao Xing
- Department of Ophthalmology, Renmin Hospital of Wuhan University, 238 Jiefang Road, Wuhan, 430060, Hubei, China.
| |
Collapse
|
9
|
Park JH, Tanaka M, Nakano T, Licastro E, Nakamura Y, Li W, Esposito E, Mandeville ET, Chou SHY, Ning M, Lo EH, Hayakawa K. O-GlcNAcylation is essential for therapeutic mitochondrial transplantation. COMMUNICATIONS MEDICINE 2023; 3:169. [PMID: 38007588 PMCID: PMC10676354 DOI: 10.1038/s43856-023-00402-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Accepted: 11/06/2023] [Indexed: 11/27/2023] Open
Abstract
BACKGROUND Transplantation of mitochondria is increasingly explored as a novel therapy in central nervous system (CNS) injury and disease. However, there are limitations in safety and efficacy because mitochondria are vulnerable in extracellular environments and damaged mitochondria can induce unfavorable danger signals. METHODS Mitochondrial O-GlcNAc-modification was amplified by recombinant O-GlcNAc transferase (OGT) and UDP-GlcNAc. O-GlcNAcylated mitochondrial proteins were identified by mass spectrometry and the antiglycation ability of O-GlcNAcylated DJ1 was determined by loss-of-function via mutagenesis. Therapeutic efficacy of O-GlcNAcylated mitochondria was assessed in a mouse model of transient focal cerebral ischemia-reperfusion. To explore translational potential, we evaluated O-GlcNAcylated DJ1 in CSF collected from patients with subarachnoid hemorrhagic stroke (SAH). RESULTS We show that isolated mitochondria are susceptible to advanced glycation end product (AGE) modification, and these glycated mitochondria induce the receptor for advanced glycation end product (RAGE)-mediated autophagy and oxidative stress when transferred into neurons. However, modifying mitochondria with O-GlcNAcylation counteracts glycation, diminishes RAGE-mediated effects, and improves viability of mitochondria recipient neurons. In a mouse model of stroke, treatment with extracellular mitochondria modified by O-GlcNAcylation reduces neuronal injury and improves neurologic deficits. In cerebrospinal fluid (CSF) samples from SAH patients, levels of O-GlcNAcylation in extracellular mitochondria correlate with better clinical outcomes. CONCLUSIONS These findings suggest that AGE-modification in extracellular mitochondria may induce danger signals, but O-GlcNAcylation can prevent glycation and improve the therapeutic efficacy of transplanted mitochondria in the CNS.
Collapse
Affiliation(s)
- Ji Hyun Park
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Masayoshi Tanaka
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Takafumi Nakano
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Ester Licastro
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Yoshihiko Nakamura
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Wenlu Li
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Elga Esposito
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Emiri T Mandeville
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Sherry Hsiang-Yi Chou
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
- Departments of Critical Care Medicine, Neurology and Neurosurgery, University of Pittsburgh, Pittsburgh, PA, USA
- Department of Neurology, Brigham and Women's Hospital and Harvard Medical School, Boston, MA, USA
| | - MingMing Ning
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
- Clinical Proteomics Research Center, Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Eng H Lo
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.
| | - Kazuhide Hayakawa
- Neuroprotection Research Laboratory, Departments of Radiology and Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.
| |
Collapse
|
10
|
MacColl Garfinkel A, Mnatsakanyan N, Patel JH, Wills AE, Shteyman A, Smith PJS, Alavian KN, Jonas EA, Khokha MK. Mitochondrial leak metabolism induces the Spemann-Mangold Organizer via Hif-1α in Xenopus. Dev Cell 2023; 58:2597-2613.e4. [PMID: 37673063 PMCID: PMC10840693 DOI: 10.1016/j.devcel.2023.08.015] [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: 06/23/2022] [Revised: 06/30/2023] [Accepted: 08/09/2023] [Indexed: 09/08/2023]
Abstract
An instructive role for metabolism in embryonic patterning is emerging, although a role for mitochondria is poorly defined. We demonstrate that mitochondrial oxidative metabolism establishes the embryonic patterning center, the Spemann-Mangold Organizer, via hypoxia-inducible factor 1α (Hif-1α) in Xenopus. Hypoxia or decoupling ATP production from oxygen consumption expands the Organizer by activating Hif-1α. In addition, oxygen consumption is 20% higher in the Organizer than in the ventral mesoderm, indicating an elevation in mitochondrial respiration. To reconcile increased mitochondrial respiration with activation of Hif-1α, we discovered that the "free" c-subunit ring of the F1Fo ATP synthase creates an inner mitochondrial membrane leak, which decouples ATP production from respiration at the Organizer, driving Hif-1α activation there. Overexpression of either the c-subunit or Hif-1α is sufficient to induce Organizer cell fates even when β-catenin is inhibited. We propose that mitochondrial leak metabolism could be a general mechanism for activating Hif-1α and Wnt signaling.
Collapse
Affiliation(s)
- Alexandra MacColl Garfinkel
- Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT 06510, USA; Section of Endocrinology, Department of Internal Medicine, Yale University, New Haven, CT 06510, USA
| | - Nelli Mnatsakanyan
- Department of Cellular and Molecular Physiology, Pennsylvania State University College of Medicine, Hershey, PA, USA
| | - Jeet H Patel
- Department of Biochemistry, University of Washington School of Medicine, Seattle, WA 98195, USA; Program in Molecular and Cellular Biology, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Andrea E Wills
- Department of Biochemistry, University of Washington School of Medicine, Seattle, WA 98195, USA; Institute for Stem Cell and Regenerative Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Amy Shteyman
- Section of Endocrinology, Department of Internal Medicine, Yale University, New Haven, CT 06510, USA
| | - Peter J S Smith
- Institute for Life Sciences, University of Southampton, Southampton SO17 1BJ, UK
| | | | - Elizabeth Ann Jonas
- Section of Endocrinology, Department of Internal Medicine, Yale University, New Haven, CT 06510, USA.
| | - Mustafa K Khokha
- Pediatric Genomics Discovery Program, Department of Pediatrics and Genetics, Yale University School of Medicine, New Haven, CT 06510, USA.
| |
Collapse
|
11
|
Magierowska K, Wójcik-Grzybek D, Korbut E, Bakalarz D, Ginter G, Danielak A, Kwiecień S, Chmura A, Torregrossa R, Whiteman M, Magierowski M. The mitochondria-targeted sulfide delivery molecule attenuates drugs-induced gastropathy. Involvement of heme oxygenase pathway. Redox Biol 2023; 66:102847. [PMID: 37597422 PMCID: PMC10458696 DOI: 10.1016/j.redox.2023.102847] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/07/2023] [Accepted: 08/11/2023] [Indexed: 08/21/2023] Open
Abstract
Hydrogen sulfide (H2S) signaling and H2S-prodrugs maintain redox balance in gastrointestinal (GI) tract. Predominant effect of any H2S-donor is mitochondrial. Non-targeted H2S-moieties were shown to decrease the non-steroidal anti-inflammatory drugs (NSAIDs)-induced gastrotoxicity but in high doses. However, direct, controlled delivery of H2S to gastric mucosal mitochondria as a molecular target improving NSAIDs-pharmacology remains overlooked. Thus, we treated Wistar rats, i.g. with vehicle, mitochondria-targeted H2S-releasing AP39 (0.004-0.5 mg/kg), AP219 (0.02 mg/kg) as structural control without H2S-releasing ability, or AP39 + SnPP (10 mg/kg) as a heme oxygenase (HMOX) inhibitor. Next, animals were administered i.g. with acetylsalicylic acid (ASA, 125 mg/kg) as NSAIDs representative or comparatively with 75% ethanol to induce translational hemorrhagic or necrotic gastric lesions, that were assessed micro-/macroscopically. Activity of mitochondrial complex IV/V, and DNA oxidation were assessed biochemically. Gastric mucosal/serum content of IL-1β, IL-10, TNF-α, TGF-β1/2, ARG1, GST-α, or phosphorylation of mTOR, NF-κB, ERK, Akt, JNK, STAT3/5 were evaluated by microbeads-fluorescent xMAP®-assay; gastric mucosal mRNA level of HMOX-1/2, COX-1/2, SOD-1/2 by real-time PCR. AP39 (but not AP219) dose-dependently (0.02 and 0.1 mg/kg) diminished NSAID- (and ethanol)-induced gastric lesions and DNA oxidation, restoring mitochondrial complexes activity, ARG1, GST-α protein levels and increasing HMOX-1 and SOD-2 expression. AP39 decreased proteins levels or phosphorylation of gastric mucosal inflammation/oxidation-sensitive markers and restored mTOR phosphorylation. Pharmacological inhibition of HMOX-1 attenuated AP39-gastroprotection. We showed that mitochondria-targeted H2S released from very low i.g. doses of AP39 improved gastric mucosal capacity to cope with NSAIDs-induced mitochondrial dysfunction and redox imbalance, mechanistically requiring the activity of HMOX-1.
Collapse
Affiliation(s)
| | | | - Edyta Korbut
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | - Dominik Bakalarz
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland; Department of Forensic Toxicology, Institute of Forensic Research, Cracow, Poland
| | - Grzegorz Ginter
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | - Aleksandra Danielak
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | - Sławomir Kwiecień
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | - Anna Chmura
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | - Roberta Torregrossa
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - Matthew Whiteman
- University of Exeter Medical School, University of Exeter, Exeter, United Kingdom
| | - Marcin Magierowski
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland.
| |
Collapse
|
12
|
Ryan SK, Ugalde CL, Rolland AS, Skidmore J, Devos D, Hammond TR. Therapeutic inhibition of ferroptosis in neurodegenerative disease. Trends Pharmacol Sci 2023; 44:674-688. [PMID: 37657967 DOI: 10.1016/j.tips.2023.07.007] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 07/28/2023] [Accepted: 07/28/2023] [Indexed: 09/03/2023]
Abstract
Iron accumulation has been associated with the etiology and progression of multiple neurodegenerative diseases (NDDs). The exact role of iron in these diseases is not fully understood, but an iron-dependent form of regulated cell death called ferroptosis could be key. Although there is substantial preclinical and clinical evidence that ferroptosis plays a role in NDD, there are still questions regarding how to target ferroptosis therapeutically, including which proteins to target, identification of clinically relevant biomarkers, and which patients might benefit most. Clinical trials of iron- and ferroptosis-targeted therapies are beginning to provide some answers, but there is growing interest in developing new ferroptosis inhibitors. We describe newly identified ferroptosis targets, opportunities, and challenges in NDD, as well as key considerations for progressing new therapeutics to the clinic.
Collapse
Affiliation(s)
- Sean K Ryan
- Sanofi, Rare and Neurologic Diseases, Cambridge, MA, USA
| | - Cathryn L Ugalde
- The ALBORADA Drug Discovery Institute, University of Cambridge, Island Research Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0AH, UK
| | - Anne-Sophie Rolland
- Department of Medical Pharmacology, Expert Center of Parkinson's Disease, ALS, and Neurogenetics, University of Lille, LilNCog, Lille Neuroscience and Cognition, INSERM UMR S1172, CHU de Lille, LICEND, COEN, Center, NS-PARK Network, France
| | - John Skidmore
- The ALBORADA Drug Discovery Institute, University of Cambridge, Island Research Building, Cambridge Biomedical Campus, Hills Road, Cambridge CB2 0AH, UK
| | - David Devos
- Department of Medical Pharmacology, Expert Center of Parkinson's Disease, ALS, and Neurogenetics, University of Lille, LilNCog, Lille Neuroscience and Cognition, INSERM UMR S1172, CHU de Lille, LICEND, COEN, Center, NS-PARK Network, France
| | | |
Collapse
|
13
|
Romero-Carramiñana I, Esparza-Moltó PB, Domínguez-Zorita S, Nuevo-Tapioles C, Cuezva JM. IF1 promotes oligomeric assemblies of sluggish ATP synthase and outlines the heterogeneity of the mitochondrial membrane potential. Commun Biol 2023; 6:836. [PMID: 37573449 PMCID: PMC10423274 DOI: 10.1038/s42003-023-05214-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 08/04/2023] [Indexed: 08/14/2023] Open
Abstract
The coexistence of two pools of ATP synthase in mitochondria has been largely neglected despite in vitro indications for the existence of reversible active/inactive state transitions in the F1-domain of the enzyme. Herein, using cells and mitochondria from mouse tissues, we demonstrate the existence in vivo of two pools of ATP synthase: one active, the other IF1-bound inactive. IF1 is required for oligomerization and inactivation of ATP synthase and for proper cristae formation. Immunoelectron microscopy shows the co-distribution of IF1 and ATP synthase, placing the inactive "sluggish" ATP synthase preferentially at cristae tips. The intramitochondrial distribution of IF1 correlates with cristae microdomains of high membrane potential, partially explaining its heterogeneous distribution. These findings support that IF1 is the in vivo regulator of the active/inactive state transitions of the ATP synthase and suggest that local regulation of IF1-ATP synthase interactions is essential to activate the sluggish ATP synthase.
Collapse
Affiliation(s)
- Inés Romero-Carramiñana
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - Pau B Esparza-Moltó
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - Sonia Domínguez-Zorita
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - Cristina Nuevo-Tapioles
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049, Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - José M Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), 28049, Madrid, Spain.
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Madrid, Spain.
- Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain.
| |
Collapse
|
14
|
Bernardi P, Gerle C, Halestrap AP, Jonas EA, Karch J, Mnatsakanyan N, Pavlov E, Sheu SS, Soukas AA. Identity, structure, and function of the mitochondrial permeability transition pore: controversies, consensus, recent advances, and future directions. Cell Death Differ 2023; 30:1869-1885. [PMID: 37460667 PMCID: PMC10406888 DOI: 10.1038/s41418-023-01187-0] [Citation(s) in RCA: 100] [Impact Index Per Article: 50.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Revised: 06/15/2023] [Accepted: 06/23/2023] [Indexed: 07/22/2023] Open
Abstract
The mitochondrial permeability transition (mPT) describes a Ca2+-dependent and cyclophilin D (CypD)-facilitated increase of inner mitochondrial membrane permeability that allows diffusion of molecules up to 1.5 kDa in size. It is mediated by a non-selective channel, the mitochondrial permeability transition pore (mPTP). Sustained mPTP opening causes mitochondrial swelling, which ruptures the outer mitochondrial membrane leading to subsequent apoptotic and necrotic cell death, and is implicated in a range of pathologies. However, transient mPTP opening at various sub-conductance states may contribute several physiological roles such as alterations in mitochondrial bioenergetics and rapid Ca2+ efflux. Since its discovery decades ago, intensive efforts have been made to identify the exact pore-forming structure of the mPT. Both the adenine nucleotide translocase (ANT) and, more recently, the mitochondrial F1FO (F)-ATP synthase dimers, monomers or c-subunit ring alone have been implicated. Here we share the insights of several key investigators with different perspectives who have pioneered mPT research. We critically assess proposed models for the molecular identity of the mPTP and the mechanisms underlying its opposing roles in the life and death of cells. We provide in-depth insights into current controversies, seeking to achieve a degree of consensus that will stimulate future innovative research into the nature and role of the mPTP.
Collapse
Affiliation(s)
- Paolo Bernardi
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Christoph Gerle
- Laboratory of Protein Crystallography, Institute for Protein Research, Osaka University, Suita, Japan
| | - Andrew P Halestrap
- School of Biochemistry and Bristol Heart Institute, University of Bristol, Bristol, UK
| | - Elizabeth A Jonas
- Department of Internal Medicine, Section of Endocrinology, Yale University School of Medicine, New Haven, CT, USA
| | - Jason Karch
- Department of Integrative Physiology and Biophysics, Baylor College of Medicine, Houston, TX, USA
| | - Nelli Mnatsakanyan
- Department of Cellular and Molecular Physiology, College of Medicine, Penn State University, State College, PA, USA
| | - Evgeny Pavlov
- Department of Molecular Pathobiology, New York University, New York, NY, USA
| | - Shey-Shing Sheu
- Department of Medicine, Center for Translational Medicine, Sidney Kimmel Medical College, Thomas Jefferson University, Philadelphia, PA, USA.
| | - Alexander A Soukas
- Department of Medicine, Diabetes Unit and Center for Genomic Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| |
Collapse
|
15
|
Sanz FJ, Solana-Manrique C, Paricio N. Disease-Modifying Effects of Vincamine Supplementation in Drosophila and Human Cell Models of Parkinson's Disease Based on DJ-1 Deficiency. ACS Chem Neurosci 2023. [PMID: 37289979 DOI: 10.1021/acschemneuro.3c00026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/10/2023] Open
Abstract
Parkinson's disease (PD) is an incurable neurodegenerative disorder caused by the selective loss of dopaminergic neurons in the substantia nigra pars compacta. Current therapies are only symptomatic and are not able to stop or delay its progression. In order to search for new and more effective therapies, our group carried out a high-throughput screening assay, identifying several candidate compounds that are able to improve locomotor ability in DJ-1β mutant flies (a Drosophila model of familial PD) and reduce oxidative stress (OS)-induced lethality in DJ-1-deficient SH-SY5Y human cells. One of them was vincamine (VIN), a natural alkaloid obtained from the leaves of Vinca minor. Our results showed that VIN is able to suppress PD-related phenotypes in both Drosophila and human cell PD models. Specifically, VIN reduced OS levels in PD model flies. Besides, VIN diminished OS-induced lethality by decreasing apoptosis, increased mitochondrial viability, and reduced OS levels in DJ-1-deficient human cells. In addition, our results show that VIN might be exerting its beneficial role, at least partially, by the inhibition of voltage-gated sodium channels. Therefore, we propose that these channels might be a promising target in the search for new compounds to treat PD and that VIN represents a potential therapeutic treatment for the disease.
Collapse
Affiliation(s)
- Francisco José Sanz
- Departamento de Genética, Facultad de Ciencias Biológicas, Universidad de Valencia, Burjassot 46100, Spain
- Instituto Universitario de Biotecnología y Biomedicina (BIOTECMED), Universidad de Valencia, Burjassot 46100, Spain
| | - Cristina Solana-Manrique
- Departamento de Genética, Facultad de Ciencias Biológicas, Universidad de Valencia, Burjassot 46100, Spain
- Instituto Universitario de Biotecnología y Biomedicina (BIOTECMED), Universidad de Valencia, Burjassot 46100, Spain
- Departamento de Fisioterapia, Facultad de Ciencias de La Salud, Universidad Europea de Valencia, Valencia 46010, Spain
| | - Nuria Paricio
- Departamento de Genética, Facultad de Ciencias Biológicas, Universidad de Valencia, Burjassot 46100, Spain
- Instituto Universitario de Biotecnología y Biomedicina (BIOTECMED), Universidad de Valencia, Burjassot 46100, Spain
| |
Collapse
|
16
|
Banarase TA, Sammeta SS, Wankhede NL, Mangrulkar SV, Rahangdale SR, Aglawe MM, Taksande BG, Upaganlawar AB, Umekar MJ, Kale MB. Mitophagy regulation in aging and neurodegenerative disease. Biophys Rev 2023; 15:239-255. [PMID: 37124925 PMCID: PMC10133433 DOI: 10.1007/s12551-023-01057-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 03/24/2023] [Indexed: 04/07/2023] Open
Abstract
Mitochondria are the primary cellular energy generators, supplying the majority of adenosine triphosphate through oxidative phosphorylation, which is necessary for neuron function and survival. Mitophagy is the metabolic process of eliminating dysfunctional or redundant mitochondria. It is a type of autophagy and it is crucial for maintaining mitochondrial and neuronal health. Impaired mitophagy leads to an accumulation of damaged mitochondria and proteins leading to the dysregulation of mitochondrial quality control processes. Recent research shows the vital role of mitophagy in neurons and the pathogenesis of major neurodegenerative diseases. Mitophagy also plays a major role in the process of aging. This review describes the alterations that are being caused in the mitophagy process at the molecular level in aging and in neurodegenerative diseases, particularly Alzheimer's, Parkinson's, and Huntington's diseases and amyotrophic lateral sclerosis, also looks at how mitophagy can be exploited as a therapeutic target for these diseases.
Collapse
Affiliation(s)
- Trupti A. Banarase
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Shivkumar S. Sammeta
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Nitu L. Wankhede
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Shubhada V. Mangrulkar
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Sandip R. Rahangdale
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Manish M. Aglawe
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Brijesh G. Taksande
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Aman B. Upaganlawar
- SNJB’s Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra India 423101
| | - Milind J. Umekar
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| | - Mayur B. Kale
- Division of Neuroscience, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra India 441002
| |
Collapse
|
17
|
Zhang Y, Fu Q, Ruan J, Shi C, Lu W, Wu J, Zhou Z. Dexpramipexole ameliorates cognitive deficits in sepsis-associated encephalopathy through suppressing mitochondria-mediated pyroptosis and apoptosis. Neuroreport 2023; 34:220-231. [PMID: 36719835 PMCID: PMC10516177 DOI: 10.1097/wnr.0000000000001882] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Accepted: 01/09/2023] [Indexed: 02/01/2023]
Abstract
OBJECTIVES This study was aimed at evaluating the effects of dexpramipexole (DPX) - a mitochondrial protectant that sustains mitochondrial function and energy production - on cognitive function in a mouse model of sepsis-associated encephalopathy (SAE) induced by peripheral administration of lipopolysaccharide (LPS) and examining the potential mechanisms. METHODS C57BL/6 male mice were randomized into one of four treatment protocols: Con+Sal, Con+DPX, LPS+Sal or LPS+DPX. The mice were intraperitoneally (i.p.) injected with LPS or equivalent volumes of normal saline once daily for 3 consecutive days. To evaluate the protective effects of DPX, we administered DPX or normal saline i.p. to the mice once daily for 6 consecutive days. Six mice in each group were decapitated on day 7, and each brain was rapidly removed and separated into two halves for biochemical and histochemical analysis. The remaining surviving mice in each group were subjected to behavioral tests from days 7 to 10. RESULTS Peripheral administration of LPS to mice led to learning and memory deficits in behavioral tests, which were associated with mitochondrial impairment and ATP depletion in the hippocampus. Repeated DPX treatment protected the mitochondria against LPS-induced morphological and functional impairment; inhibited the activation of the Nod-like receptor pyrin domain-containing 3 (NLRP3) inflammasome-caspase-1-dependent pyroptosis pathway and cytochrome c (Cyt-c)-caspase-3-dependent apoptosis pathway; and attenuated LPS-induced neuroinflammation and cell death in the hippocampus in SAE mice. CONCLUSIONS Mitochondria-mediated pyroptosis and apoptosis are involved in the pathogenesis of cognitive deficits in a mouse model of SAE and DPX protects mitochondria and suppresses the mitochondria-medicated pyroptosis and apoptosis pathways, and ameliorates LPS-induced neuroinflammation and cognitive deficits. This study provides theoretical evidence supporting DPX for the treatment of SAE.
Collapse
Affiliation(s)
- Yibao Zhang
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou
- Department of Anesthesiology, Jinling Clinical Medical College of Nanjing Medical University
| | - Qun Fu
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School
- Department of Anesthesiology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jiaping Ruan
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School
| | - Changxi Shi
- Department of Anesthesiology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School
| | - Wuguang Lu
- Department of Anesthesiology, Affiliated Hospital of Integrated Traditional Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jing Wu
- Department of Anesthesiology, Pain and Perioperative Medicine, The First Affiliated Hospital of Zhengzhou University, Zhengzhou
| | - Zhiqiang Zhou
- Department of Anesthesiology, Jinling Clinical Medical College of Nanjing Medical University
| |
Collapse
|
18
|
Bons J, Rose J, Zhang R, Burton JB, Carrico C, Verdin E, Schilling B. In-depth analysis of the Sirtuin 5-regulated mouse brain malonylome and succinylome using library-free data-independent acquisitions. Proteomics 2023; 23:e2100371. [PMID: 36479818 PMCID: PMC10363399 DOI: 10.1002/pmic.202100371] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Revised: 10/29/2022] [Accepted: 11/14/2022] [Indexed: 12/12/2022]
Abstract
Post-translational modifications (PTMs) dynamically regulate proteins and biological pathways, typically through the combined effects of multiple PTMs. Lysine residues are targeted for various PTMs, including malonylation and succinylation. However, PTMs offer specific challenges to mass spectrometry-based proteomics during data acquisition and processing. Thus, novel and innovative workflows using data-independent acquisition (DIA) ensure confident PTM identification, precise site localization, and accurate and robust label-free quantification. In this study, we present a powerful approach that combines antibody-based enrichment with comprehensive DIA acquisitions and spectral library-free data processing using directDIA (Spectronaut). Identical DIA data can be used to generate spectral libraries and comprehensively identify and quantify PTMs, reducing the amount of enriched sample and acquisition time needed, while offering a fully automated workflow. We analyzed brains from wild-type and Sirtuin 5 (SIRT5)-knock-out mice, and discovered and quantified 466 malonylated and 2211 succinylated peptides. SIRT5 regulation remodeled the acylomes by targeting 164 malonylated and 578 succinylated sites. Affected pathways included carbohydrate and lipid metabolisms, synaptic vesicle cycle, and neurodegenerative diseases. We found 48 common SIRT5-regulated malonylation and succinylation sites, suggesting potential PTM crosstalk. This innovative and efficient workflow offers deeper insights into the mouse brain lysine malonylome and succinylome.
Collapse
Affiliation(s)
- Joanna Bons
- Buck Institute for Research on Aging, Novato, California, USA
| | - Jacob Rose
- Buck Institute for Research on Aging, Novato, California, USA
| | - Ran Zhang
- Buck Institute for Research on Aging, Novato, California, USA
| | - Jordan B Burton
- Buck Institute for Research on Aging, Novato, California, USA
| | | | - Eric Verdin
- Buck Institute for Research on Aging, Novato, California, USA
| | | |
Collapse
|
19
|
García-Beltrán O, Urrutia PJ, Núñez MT. On the Chemical and Biological Characteristics of Multifunctional Compounds for the Treatment of Parkinson's Disease. Antioxidants (Basel) 2023; 12:214. [PMID: 36829773 PMCID: PMC9952574 DOI: 10.3390/antiox12020214] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2022] [Revised: 01/12/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Protein aggregation, mitochondrial dysfunction, iron dyshomeostasis, increased oxidative damage and inflammation are pathognomonic features of Parkinson's disease (PD) and other neurodegenerative disorders characterized by abnormal iron accumulation. Moreover, the existence of positive feed-back loops between these pathological components, which accelerate, and sometimes make irreversible, the neurodegenerative process, is apparent. At present, the available treatments for PD aim to relieve the symptoms, thus improving quality of life, but no treatments to stop the progression of the disease are available. Recently, the use of multifunctional compounds with the capacity to attack several of the key components of neurodegenerative processes has been proposed as a strategy to slow down the progression of neurodegenerative processes. For the treatment of PD specifically, the necessary properties of new-generation drugs should include mitochondrial destination, the center of iron-reactive oxygen species interaction, iron chelation capacity to decrease iron-mediated oxidative damage, the capacity to quench free radicals to decrease the risk of ferroptotic neuronal death, the capacity to disrupt α-synuclein aggregates and the capacity to decrease inflammatory conditions. Desirable additional characteristics are dopaminergic neurons to lessen unwanted secondary effects during long-term treatment, and the inhibition of the MAO-B and COMPT activities to increase intraneuronal dopamine content. On the basis of the published evidence, in this work, we review the molecular basis underlying the pathological events associated with PD and the clinical trials that have used single-target drugs to stop the progress of the disease. We also review the current information on multifunctional compounds that may be used for the treatment of PD and discuss the chemical characteristics that underlie their functionality. As a projection, some of these compounds or modifications could be used to treat diseases that share common pathology features with PD, such as Friedreich's ataxia, Multiple sclerosis, Huntington disease and Alzheimer's disease.
Collapse
Affiliation(s)
- Olimpo García-Beltrán
- Facultad de Ciencias Naturales y Matemáticas, Universidad de Ibagué, Carrera 22 Calle 67, Ibagué 730002, Colombia
- Centro Integrativo de Biología y Química Aplicada (CIBQA), Universidad Bernardo O’Higgins, General Gana 1702, Santiago 8370854, Chile
| | - Pamela J. Urrutia
- Faculty of Medicine and Science, Universidad San Sebastián, Lota 2465, Santiago 7510157, Chile
| | - Marco T. Núñez
- Faculty of Sciences, Universidad de Chile, Las Palmeras 3425, Santiago 7800024, Chile
| |
Collapse
|
20
|
De Lazzari F, Agostini F, Plotegher N, Sandre M, Greggio E, Megighian A, Bubacco L, Sandrelli F, Whitworth AJ, Bisaglia M. DJ-1 promotes energy balance by regulating both mitochondrial and autophagic homeostasis. Neurobiol Dis 2023; 176:105941. [PMID: 36473592 DOI: 10.1016/j.nbd.2022.105941] [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: 10/21/2022] [Revised: 11/23/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
The protein DJ-1 is mutated in rare familial forms of recessive Parkinson's disease and in parkinsonism accompanied by amyotrophic lateral sclerosis symptoms and dementia. DJ-1 is considered a multitasking protein able to confer protection under various conditions of stress. However, the precise cellular function still remains elusive. In the present work, we evaluated fruit flies lacking the expression of the DJ-1 homolog dj-1β as compared to control aged-matched individuals. Behavioral evaluations included lifespan, locomotion in an open field arena, sensitivity to oxidative insults, and resistance to starvation. Molecular analyses were carried out by analyzing the mitochondrial morphology and functionality, and the autophagic response. We demonstrated that dj-1β null mutant flies are hypoactive and display higher sensitivity to oxidative insults and food deprivation. Analysis of mitochondrial homeostasis revealed that loss of dj-1β leads to larger and more circular mitochondria, characterized by impaired complex-I-linked respiration while preserving ATP production capacity. Additionally, dj-1β null mutant flies present an impaired autophagic response, which is suppressed by treatment with the antioxidant molecule N-Acetyl-L-Cysteine. Overall, our data point to a mechanism whereby DJ-1 plays a critical role in the maintenance of energy homeostasis, by sustaining mitochondrial homeostasis and affecting the autophagic flux through the maintenance of the cellular redox state. In light of the involvement of DJ-1 in neurodegenerative diseases and considering that neurons are highly energy-demanding cells, particularly sensitive to redox stress, our study sheds light on a key role of DJ-1 in the maintenance of cellular homeostasis.
Collapse
Affiliation(s)
- Federica De Lazzari
- Department of Biology, University of Padua, Padua 35121, Italy; Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK.
| | | | | | - Michele Sandre
- Department of Neuroscience, University of Padua, Padua 35121, Italy.
| | - Elisa Greggio
- Department of Biology, University of Padua, Padua 35121, Italy; Study Center for Neurodegeneration (CESNE), Padua 35121, Italy.
| | - Aram Megighian
- Department of Biomedical Sciences, University of Padua, Padua 35121, Italy.
| | - Luigi Bubacco
- Department of Biology, University of Padua, Padua 35121, Italy; Study Center for Neurodegeneration (CESNE), Padua 35121, Italy.
| | | | - Alexander J Whitworth
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 0XY, UK.
| | - Marco Bisaglia
- Department of Biology, University of Padua, Padua 35121, Italy; Study Center for Neurodegeneration (CESNE), Padua 35121, Italy.
| |
Collapse
|
21
|
Naren P, Cholkar A, Kamble S, Khan SS, Srivastava S, Madan J, Mehra N, Tiwari V, Singh SB, Khatri DK. Pathological and Therapeutic Advances in Parkinson's Disease: Mitochondria in the Interplay. J Alzheimers Dis 2023; 94:S399-S428. [PMID: 36093711 PMCID: PMC10473111 DOI: 10.3233/jad-220682] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/08/2022] [Indexed: 11/15/2022]
Abstract
Parkinson's disease (PD) is the second most common neurodegenerative illness majorly affecting the population between the ages of 55 to 65 years. Progressive dopaminergic neuronal loss and the collective assemblage of misfolded alpha-synuclein in the substantia nigra, remain notable neuro-pathological hallmarks of the disease. Multitudes of mechanistic pathways have been proposed in attempts to unravel the pathogenesis of PD but still, it remains elusive. The convergence of PD pathology is found in organelle dysfunction where mitochondria remain a major contributor. Mitochondrial processes like bioenergetics, mitochondrial dynamics, and mitophagy are under strict regulation by the mitochondrial genome and nuclear genome. These processes aggravate neurodegenerative activities upon alteration through neuroinflammation, oxidative damage, apoptosis, and proteostatic stress. Therefore, the mitochondria have grabbed a central position in the patho-mechanistic exploration of neurodegenerative diseases like PD. The management of PD remains a challenge to physicians to date, due to the variable therapeutic response of patients and the limitation of conventional chemical agents which only offer symptomatic relief with minimal to no disease-modifying effect. This review describes the patho-mechanistic pathways involved in PD not only limited to protein dyshomeostasis and oxidative stress, but explicit attention has been drawn to exploring mechanisms like organelle dysfunction, primarily mitochondria and mitochondrial genome influence, while delineating the newer exploratory targets such as GBA1, GLP, LRRK2, and miRNAs and therapeutic agents targeting them.
Collapse
Affiliation(s)
- Padmashri Naren
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Anjali Cholkar
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Suchita Kamble
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Sabiya Samim Khan
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Saurabh Srivastava
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, India
| | - Jitender Madan
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, India
| | - Neelesh Mehra
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER)-Hyderabad, Telangana, India
| | - Vinod Tiwari
- Department of Pharmaceutical Engineering & Technology, Indian Institute of Technology (B.H.U.) Varanasi (U.P.), India
| | - Shashi Bala Singh
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| | - Dharmendra Kumar Khatri
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research, Hyderabad, Telangana, India
| |
Collapse
|
22
|
Park HA, Brown SR, Jansen J, Dunn T, Scott M, Mnatsakanyan N, Jonas EA, Kim Y. Fluid shear stress enhances proliferation of breast cancer cells via downregulation of the c-subunit of the F 1F O ATP synthase. Biochem Biophys Res Commun 2022; 632:173-180. [PMID: 36209586 PMCID: PMC10024463 DOI: 10.1016/j.bbrc.2022.09.084] [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: 09/15/2022] [Revised: 09/18/2022] [Accepted: 09/22/2022] [Indexed: 11/29/2022]
Abstract
The presence of circulating cancer cells in the bloodstream is positively correlated with metastasis. We hypothesize that fluid shear stress (FSS) occurring during circulation alters mitochondrial function, enhancing metastatic behaviors of cancer cells. MCF7 and MDA-MB-231 human breast cancer cells subjected to FSS exponentially increased proliferation. Notably, FSS-treated cells consumed more oxygen but were resistant to uncoupler-mediated ATP loss. We found that exposure to FSS downregulated the F1FO ATP synthase c-subunit and overexpression of the c-subunit arrested cancer cell migration. Approaches that regulate c-subunit abundance may reduce the likelihood of breast cancer metastasis.
Collapse
Affiliation(s)
- Han-A Park
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, 35487, USA.
| | - Spenser R Brown
- Department of Chemical and Biological Engineering, College of Engineering, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Joseph Jansen
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Tracie Dunn
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Madison Scott
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Nelli Mnatsakanyan
- Department of Internal Medicine, Section of Endocrinology, Yale University, New Haven, CT, 06511, USA; Department of Cellular and Molecular Physiology, The Pennsylvania State University College of Medicine, Hershey, PA, 17033, USA
| | - Elizabeth A Jonas
- Department of Internal Medicine, Section of Endocrinology, Yale University, New Haven, CT, 06511, USA
| | - Yonghyun Kim
- Department of Chemical and Biological Engineering, College of Engineering, The University of Alabama, Tuscaloosa, AL, 35487, USA
| |
Collapse
|
23
|
Ley-Ngardigal S, Bertolin G. Approaches to monitor ATP levels in living cells: where do we stand? FEBS J 2022; 289:7940-7969. [PMID: 34437768 DOI: 10.1111/febs.16169] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 07/30/2021] [Accepted: 08/25/2021] [Indexed: 01/14/2023]
Abstract
ATP is the most universal and essential energy molecule in cells. This is due to its ability to store cellular energy in form of high-energy phosphate bonds, which are extremely stable and readily usable by the cell. This energy is key for a variety of biological functions such as cell growth and division, metabolism, and signaling, and for the turnover of biomolecules. Understanding how ATP is produced and hydrolyzed with a spatiotemporal resolution is necessary to understand its functions both in physiological and in pathological contexts. In this review, first we will describe the organization of the electron transport chain and ATP synthase, the main molecular motor for ATP production in mitochondria. Second, we will review the biochemical assays currently available to estimate ATP quantities in cells, and we will compare their readouts, strengths, and weaknesses. Finally, we will explore the palette of genetically encoded biosensors designed for microscopy-based approaches, and show how their spatiotemporal resolution opened up the possibility to follow ATP levels in living cells.
Collapse
Affiliation(s)
- Seyta Ley-Ngardigal
- CNRS, Univ Rennes, IGDR (Genetics and Development Institute of Rennes), Rennes, France.,LVMH Research Perfumes and Cosmetics, Saint-Jean-de-Braye, France
| | - Giulia Bertolin
- CNRS, Univ Rennes, IGDR (Genetics and Development Institute of Rennes), Rennes, France
| |
Collapse
|
24
|
Gallinat A, Rakovic A, Klein C, Badimon L. DJ-1 regulates mitochondrial gene expression during ischemia and reperfusion. Free Radic Biol Med 2022; 193:430-436. [PMID: 36341940 DOI: 10.1016/j.freeradbiomed.2022.10.315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/10/2022] [Accepted: 10/24/2022] [Indexed: 11/06/2022]
Abstract
The early-onset Parkinson's disease protein DJ-1 is a multifunctional protein that plays a protective role against ischemia and reperfusion (I/R) injury and oxidative stress. Despite lacking a canonical RNA-binding domain DJ-1 exhibits RNA-binding activity and multiple transcripts have been identified. However, no functional characterization has been provided to date. Here, we have investigated the DJ-1-interacting transcripts, as well as the role of DJ-1 RNA-binding activity during ischemia and reperfusion. Among the identified DJ-1-interacting transcripts, we have distinguished a significant enrichment of mRNAs encoding mitochondrial proteins. The effects of DJ-1 depletion on mitochondrial protein expression and mitochondrial morphology were investigated using a CRISPR/Cas9 generated DJ-1 knockout (DJ-1KO) cell model. DJ-1 depletion resulted in increased MTND2 protein expression in resting cells; however, after exposure to I/R, MTND2 levels were significantly reduced with respect to wild type cells. Increased mitochondrial fission was consistently found in DJ-1KO cells after I/R exposure. MTND2 transcript binding to DJ-1 was increased during ischemia. Our results indicate that the RNA-binding activity of DJ-1 shield mitochondrial transcripts from oxidative damage.
Collapse
Affiliation(s)
- Alex Gallinat
- Cardiovascular Program-ICCC, IR-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain; Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | | | - Christine Klein
- Institute of Neurogenetics, University of Lübeck, Lübeck, Germany
| | - Lina Badimon
- Cardiovascular Program-ICCC, IR-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, Barcelona, Spain; CIBERCV-Instituto de Salud Carlos III, Madrid, Spain; UAB-Chair Cardiovascular Research, Barcelona, Spain.
| |
Collapse
|
25
|
Mitochondrial ATP synthase c-subunit leak channel triggers cell death upon loss of its F 1 subcomplex. Cell Death Differ 2022; 29:1874-1887. [PMID: 35322203 PMCID: PMC9433415 DOI: 10.1038/s41418-022-00972-7] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 02/16/2022] [Accepted: 02/28/2022] [Indexed: 02/03/2023] Open
Abstract
Mitochondrial ATP synthase is vital not only for cellular energy production but also for energy dissipation and cell death. ATP synthase c-ring was suggested to house the leak channel of mitochondrial permeability transition (mPT), which activates during excitotoxic ischemic insult. In this present study, we purified human c-ring from both eukaryotic and prokaryotic hosts to biophysically characterize its channel activity. We show that purified c-ring forms a large multi-conductance, voltage-gated ion channel that is inhibited by the addition of ATP synthase F1 subcomplex. In contrast, dissociation of F1 from FO occurs during excitotoxic neuronal death suggesting that the F1 constitutes the gate of the channel. mPT is known to dissipate the osmotic gradient across the inner membrane during cell death. We show that ATP synthase c-subunit knock down (KD) prevents the osmotic change in response to high calcium and eliminates large conductance, Ca2+ and CsA sensitive channel activity of mPT. These findings elucidate the gating mechanism of the ATP synthase c-subunit leak channel (ACLC) and suggest how ACLC opening is regulated by cell stress in a CypD-dependent manner.
Collapse
|
26
|
Gallinat A, Badimon L. DJ-1 interacts with the ectopic ATP-synthase in endothelial cells during acute ischemia and reperfusion. Sci Rep 2022; 12:12753. [PMID: 35882968 PMCID: PMC9325725 DOI: 10.1038/s41598-022-16998-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2022] [Accepted: 07/19/2022] [Indexed: 01/28/2023] Open
Abstract
Endothelial cells (ECs) play a central role in ischemia. ATP-Synthase is now recognized to be ectopically expressed in the cell surface of many cell types, with putative roles described in angiogenesis, proliferation, and intracellular pH regulation. DJ-1 is a multifunctional protein, involved in cell protection against ischemia, ischemia–reperfusion (I/R), and oxidative stress, that regulates mitochondrial ATP-synthase. Here we focused on the characterization of the endothelial dynamics of DJ-1, and its implication in the regulation of the ectopic ATP-synthase (ecATP-S) activity, during acute ischemia and I/R in ECs. We found that DJ-1 is secreted from ECs, by a mechanism enhanced in ischemia and I/R. A cleaved form of DJ-1 (DJ-1∆C) was found only in the secretome of ischemic cells. The ecATP-S activity increased following acute ischemia in ECs, coinciding with DJ-1 and DJ-1∆C secretion. The inhibition of DJ-1 expression inhibited the ecATP-S response to ischemia by ∼ 50%, and its exogenous administration maximized the effect, together with an enhanced Akt phosphorylation and angiotube-formation potential at reperfusion. Immunoprecipitation studies showed direct interaction between DJ-1 and the ecATP-S. Altogether suggesting that DJ-1 is actively cleaved and released from ischemic ECs and plays an important role in the regulation of the ecATP-S activity during acute ischemia and reperfusion.
Collapse
Affiliation(s)
- Alex Gallinat
- Cardiovascular Program-ICCC, IR-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, c/Sant Antoni María Claret, 167, 08025, Barcelona, Spain.,Universitat Autònoma de Barcelona (UAB), Barcelona, Spain
| | - Lina Badimon
- Cardiovascular Program-ICCC, IR-Hospital Santa Creu i Sant Pau, IIB-Sant Pau, c/Sant Antoni María Claret, 167, 08025, Barcelona, Spain. .,CIBERCV-Instituto de Salud Carlos III, Madrid, Spain. .,UAB-Chair Cardiovascular Research, Barcelona, Spain.
| |
Collapse
|
27
|
Xue J, Li Y, Qi Y, Zhang Z, Tang X. Exogenous Otx2 protects midbrain dopaminergic neurons from MPP+ by interacting with ATP5a1 and promoting ATP synthesis. Neurotoxicology 2022; 91:211-217. [DOI: 10.1016/j.neuro.2022.05.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2022] [Revised: 04/30/2022] [Accepted: 05/20/2022] [Indexed: 11/25/2022]
|
28
|
Magierowska K, Korbut E, Wójcik-Grzybek D, Bakalarz D, Sliwowski Z, Cieszkowski J, Szetela M, Torregrossa R, Whiteman M, Magierowski M. Mitochondria-targeted hydrogen sulfide donors versus acute oxidative gastric mucosal injury. J Control Release 2022; 348:321-334. [PMID: 35654168 DOI: 10.1016/j.jconrel.2022.05.051] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 05/23/2022] [Accepted: 05/26/2022] [Indexed: 12/14/2022]
Abstract
Hydrogen sulfide (H2S) as a gaseous molecule prevents gastrointestinal (GI)-tract against various injuries. This study aimed to evaluate for the first time the detailed molecular mechanism of mitochondria-targeting H2S-prodrugs, AP39 and RT01 in gastroprotection against ischemia/reperfusion (I/R)-induced lesions. Wistar rats exposed to I/R were pretreated i.g. with vehicle, AP39 (0.004-2 mg/kg), RT01 (0.1 mg/kg), or with AP219 (0.1 mg/kg) as structural control without ability to release H2S. AP39 was also administered with mTOR1 inhibitor, rapamycin (1 mg/kg i.g.). Gastric damage area was assessed micro-/macroscopically, gastric blood flow (GBF) by laser flowmetry, mRNA level of HIF-1α, GPx, SOD1, SOD2, annexin-A1, SOCS3, IL-1RA, IL-1β, IL-1R1, IL-1R2, TNFR2, iNOS by real-time PCR. Gastric mucosal and/or serum content of IL-1β, IL-4, IL-5, IL-10, G-CSF, M-CSF, VEGFA, GRO, RANTES, MIP-1α, MCP1, TNF-α, TIMP1, FABP3, GST-α, STAT3/5 and phosphorylation of mTOR, NF-κB, ERK, Akt was evaluated by microbeads-fluorescent assay. Mitochondrial complexes activities were measured biochemically. RNA damage was assessed as 8-OHG by ELISA. AP39 and RT01 reduced micro-/macroscopic gastric I/R-injury increasing GBF. AP39-gastroprotection was accompanied by maintained activity of mitochondrial complexes, prevented RNA oxidation and enhanced mRNA/protein expression of SOCS3, IL-1RA, annexin-A1, GST-α, HIF-1α. Rapamycin reversed AP-39-gastroprotection. AP39-gastroprotection was followed by decreased NF-κB, ERK, IL-1β and enhanced Akt and mTOR proteins phosphorylation. AP39-prevented gastric mucosal damage caused by I/R-injury, partly by mitochondrial complex activity maintenance. AP39-mediated attenuation of gastric mucosal oxidation, hypoxia and inflammation involved mTOR1 and Akt pathways activity and modulation of HIF-1α, GST-α, SOCS3, IL1RA and TIMP1 molecular interplay.
Collapse
Affiliation(s)
| | - Edyta Korbut
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | | | - Dominik Bakalarz
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland; Department of Forensic Toxicology, Institute of Forensic Research, Cracow, Poland
| | - Zbigniew Sliwowski
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | - Jakub Cieszkowski
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | - Małgorzata Szetela
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland
| | | | | | - Marcin Magierowski
- Department of Physiology, Jagiellonian University Medical College, Cracow, Poland.
| |
Collapse
|
29
|
Neural stem cell secretome exerts a protective effect on damaged neuron mitochondria in Parkinson's disease model. Brain Res 2022; 1790:147978. [PMID: 35690143 DOI: 10.1016/j.brainres.2022.147978] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2022] [Revised: 05/29/2022] [Accepted: 06/06/2022] [Indexed: 11/22/2022]
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease. The main pathological changes are the loss of dopaminergic neurons and the formation of Lewy bodies. There is still no effective cure for PD, and cell replacement therapy has entered a bottleneck period due to tumorigenicity and rejection. Therefore, stem cell secretome has received widespread attention. However, the exploration of the secretome components of neural stem cells (NSCs) is still in its infancy. In this study, 6-hydroxydopamine (6-OHDA) was used to establish a PD rat model in vito and the PC12 cell-damaged model in vitro. The results indicated that the injection of neural stem cell-conditioned medium (NSC-CM) into the striatum and substantia nigra could improve the motor and non-motor deficits of PD rats and rescue the loss of dopaminergic neurons. In addition, NSC-CM alleviated 6-OHDA-induced apoptosis of PC12 cells, reduced the level of oxidative stress, and improved mitochondrial dysfunction in vitro. Parkinson disease protein 7 (Park7) was found in NSC-CM by Liquid chromatography-tandem mass spectrometry (LC-MS/MS), and it may be related to the protective effect of NSC-CM on 6-OHDA-injured neurons through Sirt1 pathway. In conclusion, NSC secretome might provide new ideas for the treatment of PD.
Collapse
|
30
|
Domínguez-Zorita S, Romero-Carramiñana I, Cuezva JM, Esparza-Moltó PB. The ATPase Inhibitory Factor 1 is a Tissue-Specific Physiological Regulator of the Structure and Function of Mitochondrial ATP Synthase: A Closer Look Into Neuronal Function. Front Physiol 2022; 13:868820. [PMID: 35620611 PMCID: PMC9128019 DOI: 10.3389/fphys.2022.868820] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Accepted: 03/02/2022] [Indexed: 12/15/2022] Open
Abstract
The ATP synthase is an essential multifunctional enzyme complex of mitochondria that produces most of cellular ATP, shapes the structure of the inner membrane into cristae and regulates the signals that control cell fate or demise. The ATPase Inhibitory Factor 1 (IF1) functions in vivo as a physiological regulator of the ATP synthase and thereby controls mitochondrial structure and function, and the retrograde signaling pathways that reprogram nuclear gene expression. However, IF1 is not ubiquitously expressed in mammals, showing tissue-restricted expression in humans and mice and large expression differences between the two species in some tissues. Herein, we summarized key regulatory functions of IF1 for tissue homeostasis, with special emphasis on the deleterious effects that its genetic ablation in neurons has in learning. The development and characterization of tissue-specific mouse models with regulated expression of IF1 will be crucial to disentangle the contribution of the ATP synthase/IF1 axis in pathophysiology.
Collapse
Affiliation(s)
- Sonia Domínguez-Zorita
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, (CSIC-UAM), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - Inés Romero-Carramiñana
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, (CSIC-UAM), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - José M Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, (CSIC-UAM), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain
| | - Pau B Esparza-Moltó
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, (CSIC-UAM), Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER) ISCIII, Instituto de Investigación Hospital 12 de Octubre, Universidad Autónoma de Madrid, Madrid, Spain.,Salk Institute for Biological Studies, La Jolla, CA, United States
| |
Collapse
|
31
|
Ali MZ, Dholaniya PS. Oxidative phosphorylation mediated pathogenesis of Parkinson's disease and its implication via Akt signaling. Neurochem Int 2022; 157:105344. [PMID: 35483538 DOI: 10.1016/j.neuint.2022.105344] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2021] [Revised: 04/19/2022] [Accepted: 04/21/2022] [Indexed: 12/21/2022]
Abstract
Substantia Nigra Pars-compacta (SNpc), in the basal ganglion region, is a primary source of dopamine release. These dopaminergic neurons require more energy than other neurons, as they are highly arborized and redundant. Neurons meet most of their energy demand (∼90%) from mitochondria. Oxidative phosphorylation (OxPhos) is the primary pathway for energy production. Many genes involved in Parkinson's disease (PD) have been associated with OxPhos, especially complex I. Abrogation in complex I leads to reduced ATP formation in these neurons, succumbing to death by inducing apoptosis. This review discusses the interconnection between complex I-associated PD genes and specific mitochondrial metabolic factors (MMFs) of OxPhos. Interestingly, all the complex I-associated PD genes discussed here have been linked to the Akt signaling pathway; thus, neuron survival is promoted and smooth mitochondrial function is ensured. Any changes in these genes disrupt the Akt pathway, which hampers the opening of the permeability transition pore (PTP) via GSK3β dephosphorylation; promotes destabilization of OxPhos; and triggers the release of pro-apoptotic factors.
Collapse
Affiliation(s)
- Md Zainul Ali
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500 046, India
| | - Pankaj Singh Dholaniya
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana, 500 046, India.
| |
Collapse
|
32
|
Ženíšková K, Grechnikova M, Sutak R. Copper Metabolism in Naegleria gruberi and Its Deadly Relative Naegleria fowleri. Front Cell Dev Biol 2022; 10:853463. [PMID: 35478954 PMCID: PMC9035749 DOI: 10.3389/fcell.2022.853463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Accepted: 03/18/2022] [Indexed: 12/04/2022] Open
Abstract
Although copper is an essential nutrient crucial for many biological processes, an excessive concentration can be toxic and lead to cell death. The metabolism of this two-faced metal must be strictly regulated at the cell level. In this study, we investigated copper homeostasis in two related unicellular organisms: nonpathogenic Naegleria gruberi and the “brain-eating amoeba” Naegleria fowleri. We identified and confirmed the function of their specific copper transporters securing the main pathway of copper acquisition. Adjusting to different environments with varying copper levels during the life cycle of these organisms requires various metabolic adaptations. Using comparative proteomic analyses, measuring oxygen consumption, and enzymatic determination of NADH dehydrogenase, we showed that both amoebas respond to copper deprivation by upregulating the components of the branched electron transport chain: the alternative oxidase and alternative NADH dehydrogenase. Interestingly, analysis of iron acquisition indicated that this system is copper-dependent in N. gruberi but not in its pathogenic relative. Importantly, we identified a potential key protein of copper metabolism of N. gruberi, the homolog of human DJ-1 protein, which is known to be linked to Parkinson’s disease. Altogether, our study reveals the mechanisms underlying copper metabolism in the model amoeba N. gruberi and the fatal pathogen N. fowleri and highlights the differences between the two amoebas.
Collapse
|
33
|
Garone C, Pietra A, Nesci S. From the Structural and (Dys)Function of ATP Synthase to Deficiency in Age-Related Diseases. Life (Basel) 2022; 12:401. [PMID: 35330152 PMCID: PMC8949411 DOI: 10.3390/life12030401] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2022] [Revised: 02/25/2022] [Accepted: 03/08/2022] [Indexed: 12/21/2022] Open
Abstract
The ATP synthase is a mitochondrial inner membrane complex whose function is essential for cell bioenergy, being responsible for the conversion of ADP into ATP and playing a role in mitochondrial cristae morphology organization. The enzyme is composed of 18 protein subunits, 16 nuclear DNA (nDNA) encoded and two mitochondrial DNA (mtDNA) encoded, organized in two domains, FO and F1. Pathogenetic variants in genes encoding structural subunits or assembly factors are responsible for fatal human diseases. Emerging evidence also underlines the role of ATP-synthase in neurodegenerative diseases as Parkinson's, Alzheimer's, and motor neuron diseases such as Amyotrophic Lateral Sclerosis. Post-translational modification, epigenetic modulation of ATP gene expression and protein level, and the mechanism of mitochondrial transition pore have been deemed responsible for neuronal cell death in vivo and in vitro models for neurodegenerative diseases. In this review, we will explore ATP synthase assembly and function in physiological and pathological conditions by referring to the recent cryo-EM studies and by exploring human disease models.
Collapse
Affiliation(s)
- Caterina Garone
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40137 Bologna, Italy;
- Center for Applied Biomedical Research, Alma Mater Studiorum University of Bologna, 40137 Bologna, Italy
- UOC Neuropsichiatria dell’età Pediatrica, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40137 Bologna, Italy
| | - Andrea Pietra
- Department of Medical and Surgical Sciences, Alma Mater Studiorum University of Bologna, 40137 Bologna, Italy;
- UO Genetica Medica, IRCCS Azienda Ospedaliero-Universitaria di Bologna, 40137 Bologna, Italy
| | - Salvatore Nesci
- Department of Veterinary Medical Sciences, Alma Mater Studiorum University of Bologna, 40064 Ozzano Emilia, Italy
| |
Collapse
|
34
|
Park HA, Crowe-White KM, Ciesla L, Scott M, Bannerman S, Davis AU, Adhikari B, Burnett G, Broman K, Ferdous KA, Lackey KH, Licznerski P, Jonas EA. Alpha-Tocotrienol Enhances Arborization of Primary Hippocampal Neurons via Upregulation of Bcl-xL. Nutr Res 2022; 101:31-42. [PMID: 35366596 PMCID: PMC9081260 DOI: 10.1016/j.nutres.2022.02.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2021] [Revised: 02/18/2022] [Accepted: 02/28/2022] [Indexed: 11/28/2022]
Abstract
Alpha-tocotrienol (α-TCT) is a member of the vitamin E family. It has been reported to protect the brain against various pathologies including cerebral ischemia and neurodegeneration. However, it is still unclear if α-TCT exhibits beneficial effects during brain development. We hypothesized that treatment with α-TCT improves intracellular redox homeostasis supporting normal development of neurons. We found that primary hippocampal neurons isolated from rat feti grown in α-TCT-containing media achieved greater levels of neurite complexity compared to ethanol-treated control neurons. Neurons were treated with 1 μM α-TCT for 3 weeks, and media were replaced with fresh α-TCT every week. Treatment with α-TCT increased α-TCT levels (26 pmol/mg protein) in the cells, whereas the control neurons did not contain α-TCT. α-TCT-treated neurons produced adenosine triphosphate (ATP) at a higher rate and increased ATP retention at neurites, supporting formation of neurite branches. Although treatment with α-TCT alone did not change neuronal viability, neurons grown in α-TCT were more resistant to death at maturity. We further found that messenger RNA and protein levels of B-cell lymphoma-extra large (Bcl-xL) are increased by α-TCT treatment without inducing posttranslational cleavage of Bcl-xL. Bcl-xL is known to enhance mitochondrial energy production, which improves neuronal function including neurite outgrowth and neurotransmission. Therefore α-TCT-mediated Bcl-xL upregulation may be the central mechanism of neuroprotection seen in the α-TCT-treated group. In summary, treatment with α-TCT upregulates Bcl-xL and increases ATP levels at neurites. This correlates with increased neurite branching during development and with protection of mature neurons against oxidative stress.
Collapse
Affiliation(s)
- Han-A Park
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, 35487, USA.
| | - Kristi M Crowe-White
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Lukasz Ciesla
- Department of Biological Sciences, College of Arts and Sciences, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Madison Scott
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Sydni Bannerman
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Abigail U Davis
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Bishnu Adhikari
- Department of Biological Sciences, College of Arts and Sciences, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Garrett Burnett
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Katheryn Broman
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Khondoker Adeba Ferdous
- Department of Human Nutrition and Hospitality Management, College of Human Environmental Sciences, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Kimberly H Lackey
- Department of Biological Sciences, College of Arts and Sciences, The University of Alabama, Tuscaloosa, AL, 35487, USA
| | - Pawel Licznerski
- Department of Internal Medicine, Section of Endocrinology, Yale University, New Haven, CT, 06511, USA
| | - Elizabeth A Jonas
- Department of Internal Medicine, Section of Endocrinology, Yale University, New Haven, CT, 06511, USA
| |
Collapse
|
35
|
Karayel-Basar M, Uras I, Kiris I, Sahin B, Akgun E, Baykal AT. Spatial proteomic alterations detected via MALDI-MS imaging implicate neuronal loss in a Huntington's disease mouse (YAC128) brain. Mol Omics 2022; 18:336-347. [PMID: 35129568 DOI: 10.1039/d1mo00440a] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder that occurs with the increase of CAG trinucleotide repeats in the huntingtin gene. To understand the mechanisms of HD, powerful proteomics techniques, such as liquid chromatography-tandem mass spectrometry (LC-MS/MS) were employed. However, one major drawback of these methods is loss of the region-specific quantitative information of the proteins due to analysis of total tissue lysates. Matrix-assisted laser desorption/ionization mass spectrometry imaging (MALDI-MSI) is a MS-based label-free technique that works directly on tissue sections and gathers m/z values with their respective regional information. In this study, we established a data processing protocol that includes several software programs and methods to determine spatial protein alterations between the brain samples of a 12 month-old YAC128 HD mouse model and their non-transgenic littermates. 22 differentially expressed proteins were revealed with their respective regional information, and possible relationships of several proteins were discussed. As a validation of the MALDI-MSI analysis, a differentially expressed protein (GFAP) was verified using immunohistochemical staining. Furthermore, since several proteins detected in this study have previously been associated with neuronal loss, neuronal loss in the cortical region was demonstrated using an anti-NeuN immunohistochemical staining method. In conclusion, the findings of this research have provided insights into the spatial proteomic changes between HD transgenic and non-transgenic littermates and therefore, we suggest that MALDI-MSI is a powerful technique to determine spatial proteomic alterations between biological samples, and the data processing that we present here can be employed as a complementary tool for the data analysis.
Collapse
Affiliation(s)
- Merve Karayel-Basar
- Department of Medical Biochemistry and Molecular Biology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Irep Uras
- Department of Medical Biochemistry and Molecular Biology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Irem Kiris
- Department of Medical Biochemistry and Molecular Biology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Betul Sahin
- Acibadem Labmed Clinical Laboratories, R&D Center, Istanbul, Turkey
| | - Emel Akgun
- Department of Medical Biochemistry and Molecular Biology, Institute of Health Sciences, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| | - Ahmet Tarik Baykal
- Department of Medical Biochemistry, Faculty of Medicine, Acibadem Mehmet Ali Aydinlar University, Istanbul, Turkey
| |
Collapse
|
36
|
Li JL, Lin TY, Chen PL, Guo TN, Huang SY, Chen CH, Lin CH, Chan CC. Mitochondrial Function and Parkinson's Disease: From the Perspective of the Electron Transport Chain. Front Mol Neurosci 2021; 14:797833. [PMID: 34955747 PMCID: PMC8695848 DOI: 10.3389/fnmol.2021.797833] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2021] [Accepted: 11/18/2021] [Indexed: 12/21/2022] Open
Abstract
Parkinson’s disease (PD) is known as a mitochondrial disease. Some even regarded it specifically as a disorder of the complex I of the electron transport chain (ETC). The ETC is fundamental for mitochondrial energy production which is essential for neuronal health. In the past two decades, more than 20 PD-associated genes have been identified. Some are directly involved in mitochondrial functions, such as PRKN, PINK1, and DJ-1. While other PD-associate genes, such as LRRK2, SNCA, and GBA1, regulate lysosomal functions, lipid metabolism, or protein aggregation, some have been shown to indirectly affect the electron transport chain. The recent identification of CHCHD2 and UQCRC1 that are critical for functions of complex IV and complex III, respectively, provide direct evidence that PD is more than just a complex I disorder. Like UQCRC1 in preventing cytochrome c from release, functions of ETC proteins beyond oxidative phosphorylation might also contribute to the pathogenesis of PD.
Collapse
Affiliation(s)
- Jeng-Lin Li
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan.,Division of Neurology, Department of Internal Medicine, Lo-Hsu Medical Foundation, Lotung Poh-Ai Hospital, Yilan County, Taiwan
| | - Tai-Yi Lin
- College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Po-Lin Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan
| | - Ting-Ni Guo
- Graduate Institute of Physiology, National Taiwan University, Taipei, Taiwan
| | - Shu-Yi Huang
- Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Chun-Hong Chen
- National Institute of Infectious Diseases and Vaccinology, National Health Research Institutes, Miaoli County, Taiwan
| | - Chin-Hsien Lin
- Department of Neurology, National Taiwan University Hospital, Taipei, Taiwan.,Department of Medical Research, National Taiwan University Hospital, Taipei, Taiwan
| | - Chih-Chiang Chan
- Graduate Institute of Physiology, National Taiwan University, Taipei, Taiwan
| |
Collapse
|
37
|
Sanz FJ, Solana-Manrique C, Torres J, Masiá E, Vicent MJ, Paricio N. A High-Throughput Chemical Screen in DJ-1β Mutant Flies Identifies Zaprinast as a Potential Parkinson's Disease Treatment. Neurotherapeutics 2021; 18:2565-2578. [PMID: 34697772 PMCID: PMC8804136 DOI: 10.1007/s13311-021-01134-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2021] [Indexed: 12/12/2022] Open
Abstract
Dopamine replacement represents the standard therapy for Parkinson's disease (PD), a common, chronic, and incurable neurological disorder; however, this approach only treats the symptoms of this devastating disease. In the search for novel disease-modifying therapies that target other relevant molecular and cellular mechanisms, Drosophila has emerged as a valuable tool to study neurodegenerative diseases due to the presence of a complex central nervous system, the blood-brain barrier, and a similar neurotransmitter profile to humans. Human PD-related genes also display conservation in flies; DJ-1β is the fly ortholog of DJ-1, a gene for which mutations prompt early-onset recessive PD. Interestingly, flies mutant for DJ-1β exhibit PD-related phenotypes, including motor defects, high oxidative stress (OS) levels and metabolic alterations. To identify novel therapies for PD, we performed an in vivo high-throughput screening assay using DJ-1β mutant flies and compounds from the Prestwick® chemical library. Drugs that improved motor performance in DJ-1ß mutant flies were validated in DJ-1-deficient human neural-like cells, revealing that zaprinast displayed the most significant ability to suppress OS-induced cell death. Zaprinast inhibits phosphodiesterases and activates GPR35, an orphan G-protein-coupled receptor not previously associated with PD. We found that zaprinast exerts its beneficial effect in both fly and human PD models through several disease-modifying mechanisms, including reduced OS levels, attenuated apoptosis, increased mitochondrial viability, and enhanced glycolysis. Therefore, our results support zaprinast as a potential therapeutic for PD in future clinical trials.
Collapse
Affiliation(s)
- Francisco José Sanz
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100, Burjassot, Spain
- Instituto Universitario de Biotecnología Y Biomedicina (BIOTECMED), Universidad de Valencia, 46100, Burjassot, Spain
| | - Cristina Solana-Manrique
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100, Burjassot, Spain
- Instituto Universitario de Biotecnología Y Biomedicina (BIOTECMED), Universidad de Valencia, 46100, Burjassot, Spain
| | - Josema Torres
- Departamento de Biología Celular, Biología Funcional Y Antropología Física, Facultad CC Biológicas, Universidad de Valencia, 46100, Burjassot, Spain
| | - Esther Masiá
- Polymer Therapeutics Lab and Screening Platform, Centro de Investigación Príncipe Felipe, 46012, Valencia, Spain
| | - María J Vicent
- Polymer Therapeutics Lab and Screening Platform, Centro de Investigación Príncipe Felipe, 46012, Valencia, Spain
| | - Nuria Paricio
- Departamento de Genética, Facultad CC Biológicas, Universidad de Valencia, 46100, Burjassot, Spain.
- Instituto Universitario de Biotecnología Y Biomedicina (BIOTECMED), Universidad de Valencia, 46100, Burjassot, Spain.
| |
Collapse
|
38
|
Pantner Y, Polavarapu R, Chin LS, Li L, Shimizu Y, Calvert JW. DJ-1 attenuates the glycation of mitochondrial complex I and complex III in the post-ischemic heart. Sci Rep 2021; 11:19408. [PMID: 34593886 PMCID: PMC8484662 DOI: 10.1038/s41598-021-98722-1] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Accepted: 09/09/2021] [Indexed: 02/02/2023] Open
Abstract
DJ-1 is a ubiquitously expressed protein that protects cells from stress through its conversion into an active protease. Recent work found that the active form of DJ-1 was induced in the ischemic heart as an endogenous mechanism to attenuate glycative stress-the non-enzymatic glycosylation of proteins. However, specific proteins protected from glycative stress by DJ-1 are not known. Given that mitochondrial electron transport proteins have a propensity for being targets of glycative stress, we investigated if DJ-1 regulates the glycation of Complex I and Complex III after myocardial ischemia-reperfusion (I/R) injury. Initial studies found that DJ-1 localized to the mitochondria and increased its interaction with Complex I and Complex III 3 days after the onset of myocardial I/R injury. Next, we investigated the role DJ-1 plays in modulating glycative stress in the mitochondria. Analysis revealed that compared to wild-type control mice, mitochondria from DJ-1 deficient (DJ-1 KO) hearts showed increased levels of glycative stress following I/R. Additionally, Complex I and Complex III glycation were found to be at higher levels in DJ-1 KO hearts. This corresponded with reduced complex activities, as well as reduced mitochondrial oxygen consumption ant ATP synthesis in the presence of pyruvate and malate. To further determine if DJ-1 influenced the glycation of the complexes, an adenoviral approach was used to over-express the active form of DJ-1(AAV9-DJ1ΔC). Under I/R conditions, the glycation of Complex I and Complex III were attenuated in hearts treated with AAV9-DJ1ΔC. This was accompanied by improvements in complex activities, oxygen consumption, and ATP production. Together, this data suggests that cardiac DJ-1 maintains Complex I and Complex III efficiency and mitochondrial function during the recovery from I/R injury. In elucidating a specific mechanism for DJ-1's role in the post-ischemic heart, these data break new ground for potential therapeutic strategies using DJ-1 as a target.
Collapse
Affiliation(s)
- Yvanna Pantner
- Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center, Emory University School of Medicine, 101 Woodruff Circle, Atlanta, GA, 30322, USA
| | - Rohini Polavarapu
- Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center, Emory University School of Medicine, 101 Woodruff Circle, Atlanta, GA, 30322, USA
| | - Lih-Shen Chin
- Department Pharmacology, Emory University School of Medicine, Atlanta, GA, USA
| | - Lian Li
- Department Pharmacology, Emory University School of Medicine, Atlanta, GA, USA
| | - Yuuki Shimizu
- Department of Cardiology, Nagoya University Graduate School of Medicine, Nagoya, 466-8550, Japan
| | - John W Calvert
- Department of Surgery, Division of Cardiothoracic Surgery, Carlyle Fraser Heart Center, Emory University School of Medicine, 101 Woodruff Circle, Atlanta, GA, 30322, USA.
| |
Collapse
|
39
|
Park HA, Stumpf A, Broman K, Jansen J, Dunn T, Scott M, Crowe-White KM. Role of lycopene in mitochondrial protection during differential levels of oxidative stress in primary cortical neurons. BRAIN DISORDERS 2021. [DOI: 10.1016/j.dscb.2021.100016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023] Open
|
40
|
Bcl-xL Is Required by Primary Hippocampal Neurons during Development to Support Local Energy Metabolism at Neurites. BIOLOGY 2021; 10:biology10080772. [PMID: 34440004 PMCID: PMC8389656 DOI: 10.3390/biology10080772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2021] [Revised: 08/01/2021] [Accepted: 08/06/2021] [Indexed: 11/17/2022]
Abstract
B-cell lymphoma-extra large (Bcl-xL) is a mitochondrial protein known to inhibit mitochondria-dependent intrinsic apoptotic pathways. An increasing number of studies have demonstrated that Bcl-xL is critical in regulating neuronal energy metabolism and has a protective role in pathologies associated with an energy deficit. However, it is less known how Bcl-xL regulates physiological processes of the brain. In this study, we hypothesize that Bcl-xL is required for neurite branching and maturation during neuronal development by improving local energy metabolism. We found that the absence of Bcl-xL in rat primary hippocampal neurons resulted in mitochondrial dysfunction. Specifically, the ATP/ADP ratio was significantly decreased in the neurites of Bcl-xL depleted neurons. We further found that neurons transduced with Bcl-xL shRNA or neurons treated with ABT-263, a pharmacological inhibitor of Bcl-xL, showed impaired mitochondrial motility. Neurons lacking Bcl-xL had significantly decreased anterograde and retrograde movement of mitochondria and an increased stationary mitochondrial population when Bcl-xL was depleted by either means. These mitochondrial defects, including loss of ATP, impaired normal neurite development. Neurons lacking Bcl-xL showed significantly decreased neurite arborization, growth and complexity. Bcl-xL depleted neurons also showed impaired synapse formation. These neurons showed increased intracellular calcium concentration and were more susceptible to excitotoxic challenge. Bcl-xL may support positioning of mitochondria at metabolically demanding regions of neurites like branching points. Our findings suggest a role for Bcl-xL in physiological regulation of neuronal growth and development.
Collapse
|
41
|
Murali Mahadevan H, Hashemiaghdam A, Ashrafi G, Harbauer AB. Mitochondria in Neuronal Health: From Energy Metabolism to Parkinson's Disease. Adv Biol (Weinh) 2021; 5:e2100663. [PMID: 34382382 DOI: 10.1002/adbi.202100663] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 06/30/2021] [Indexed: 01/01/2023]
Abstract
Mitochondria are the main suppliers of neuronal adenosine triphosphate and play a critical role in brain energy metabolism. Mitochondria also serve as Ca2+ sinks and anabolic factories and are therefore essential for neuronal function and survival. Dysregulation of neuronal bioenergetics is increasingly implicated in neurodegenerative disorders, particularly Parkinson's disease. This review describes the role of mitochondria in energy metabolism under resting conditions and during synaptic transmission, and presents evidence for the contribution of neuronal mitochondrial dysfunction to Parkinson's disease.
Collapse
Affiliation(s)
| | - Arsalan Hashemiaghdam
- Department of Cell Biology and Physiology, Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Ghazaleh Ashrafi
- Department of Cell Biology and Physiology, Department of Genetics, Washington University School of Medicine in St. Louis, St. Louis, MO, 63110, USA
| | - Angelika Bettina Harbauer
- Max-Planck-Institute for Neurobiology, 82152, Martinsried, Germany.,Technical University of Munich, Institute of Neuronal Cell Biology, 80333, Munich, Germany.,Munich Cluster for Systems Neurology, Munich, Germany
| |
Collapse
|
42
|
Effect of Novel Antipsychotics on Energy Metabolism - In Vitro Study in Pig Brain Mitochondria. Mol Neurobiol 2021; 58:5548-5563. [PMID: 34365585 DOI: 10.1007/s12035-021-02498-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 07/15/2021] [Indexed: 10/20/2022]
Abstract
The identification and quantification of mitochondrial effects of novel antipsychotics (brexpiprazole, cariprazine, loxapine, and lurasidone) were studied in vitro in pig brain mitochondria. Selected parameters of mitochondrial metabolism, electron transport chain (ETC) complexes, citrate synthase (CS), malate dehydrogenase (MDH), monoamine oxidase (MAO), mitochondrial respiration, and total ATP and reactive oxygen species (ROS) production were evaluated and associated with possible adverse effects of drugs. All tested antipsychotics decreased the ETC activities (except for complex IV, which increased in activity after brexpiprazole and loxapine addition). Both complex I- and complex II-linked respiration were dose-dependently inhibited, and significant correlations were found between complex I-linked respiration and both complex I activity (positive correlation) and complex IV activity (negative correlation). All drugs significantly decreased mitochondrial ATP production at higher concentrations. Hydrogen peroxide production was significantly increased at 10 µM brexpiprazole and lurasidone and at 100 µM cariprazine and loxapine. All antipsychotics acted as partial inhibitors of MAO-A, brexpiprazole and loxapine partially inhibited MAO-B. Based on our results, novel antipsychotics probably lacked oxygen uncoupling properties. The mitochondrial effects of novel antipsychotics might contribute on their adverse effects, which are mostly related to decreased ATP production and increased ROS production, while MAO-A inhibition might contribute to their antidepressant effect, and brexpiprazole- and loxapine-induced MAO-B inhibition might likely promote neuroplasticity and neuroprotection. The assessment of drug-induced mitochondrial dysfunctions is important in development of new drugs as well as in the understanding of molecular mechanism of adverse or side drug effects.
Collapse
|
43
|
Red Light Irradiation In Vivo Upregulates DJ-1 in the Retinal Ganglion Cell Layer and Protects against Axotomy-Related Dendritic Pruning. Int J Mol Sci 2021; 22:ijms22168380. [PMID: 34445085 PMCID: PMC8395066 DOI: 10.3390/ijms22168380] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 07/30/2021] [Accepted: 07/30/2021] [Indexed: 12/26/2022] Open
Abstract
Retinal ganglion cells (RGCs) undergo dendritic pruning in a variety of neurodegenerative diseases, including glaucoma and autosomal dominant optic atrophy (ADOA). Axotomising RGCs by severing the optic nerve generates an acute model of RGC dendropathy, which can be utilized to assess the therapeutic potential of treatments for RGC degeneration. Photobiomodulation (PBM) with red light provided neuroprotection to RGCs when administered ex vivo to wild-type retinal explants. In the current study, we used aged (13–15-month-old) wild-type and heterozygous B6;C3-Opa1Q285STOP (Opa1+/−) mice, a model of ADOA exhibiting RGC dendropathy. These mice were pre-treated with 4 J/cm2 of 670 nm light for five consecutive days before the eyes were enucleated and the retinas flat-mounted into explant cultures for 0-, 8- or 16-h ex vivo. RGCs were imaged by confocal microscopy, and their dendritic architecture was quantified by Sholl analysis. In vivo 670 nm light pretreatment inhibited the RGC dendropathy observed in untreated wild-type retinas over 16 h ex vivo and inhibited dendropathy in ON-center RGCs in wild-type but not Opa1+/− retinas. Immunohistochemistry revealed that aged Opa1+/− RGCs exhibited increased nitrosative damage alongside significantly lower activation of NF-κB and upregulation of DJ-1. PBM restored NF-κB activation in Opa1+/− RGCs and enhanced DJ-1 expression in both genotypes, indicating a potential molecular mechanism priming the retina to resist future oxidative insult. These data support the potential of PBM as a treatment for diseases involving RGC degeneration.
Collapse
|
44
|
Cen X, Zhang M, Zhou M, Ye L, Xia H. Mitophagy Regulates Neurodegenerative Diseases. Cells 2021; 10:1876. [PMID: 34440645 PMCID: PMC8392649 DOI: 10.3390/cells10081876] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 07/19/2021] [Accepted: 07/21/2021] [Indexed: 12/12/2022] Open
Abstract
Mitochondria play an essential role in supplying energy for the health and survival of neurons. Mitophagy is a metabolic process that removes dysfunctional or redundant mitochondria. This process preserves mitochondrial health. However, defective mitophagy triggers the accumulation of damaged mitochondria, causing major neurodegenerative disorders. This review introduces molecular mechanisms and signaling pathways behind mitophagy regulation. Furthermore, we focus on the recent advances in understanding the potential role of mitophagy in the pathogenesis of major neurodegenerative diseases (Parkinson's, Alzheimer's, Huntington's, etc.) and aging. The findings will help identify the potential interventions of mitophagy regulation and treatment strategies of neurodegenerative diseases.
Collapse
Affiliation(s)
- Xufeng Cen
- Department of Biochemistry & Research Center of Clinical Pharmacy of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; (X.C.); (M.Z.); (M.Z.); (L.Y.)
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, China
| | - Manke Zhang
- Department of Biochemistry & Research Center of Clinical Pharmacy of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; (X.C.); (M.Z.); (M.Z.); (L.Y.)
| | - Mengxin Zhou
- Department of Biochemistry & Research Center of Clinical Pharmacy of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; (X.C.); (M.Z.); (M.Z.); (L.Y.)
| | - Lingzhi Ye
- Department of Biochemistry & Research Center of Clinical Pharmacy of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; (X.C.); (M.Z.); (M.Z.); (L.Y.)
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, China
| | - Hongguang Xia
- Department of Biochemistry & Research Center of Clinical Pharmacy of The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou 310058, China; (X.C.); (M.Z.); (M.Z.); (L.Y.)
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou 311121, China
| |
Collapse
|
45
|
Buneeva OA, Medvedev AE. DJ-1 Protein and Its Role in the Development of Parkinson's Disease: Studies on Experimental Models. BIOCHEMISTRY (MOSCOW) 2021; 86:627-640. [PMID: 34225587 DOI: 10.1134/s000629792106002x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
DJ-1, also known as Parkinson's disease protein 7, is a multifunctional protein ubiquitously expressed in cells and tissues. Interacting with proteins of various intracellular compartments, DJ-1 plays an important role in maintaining different cellular functions. Mutant DJ-1 forms containing amino acid substitutions (especially L166P), typical of Parkinson's disease, are characterized by impaired dimerization, stability, and folding. DJ-1 exhibits several types of catalytic activity; however, in the enzyme classification it exists as protein deglycase (EC 3.5.1.124). Apparently, in different cell compartments DJ-1 exhibits catalytic and non-catalytic functions, and their ratio still remains unknown. Oxidative stress promotes dissociation of cytoplasmic DJ-1 dimers into monomers, which are translocated to the nucleus, where this protein acts as a coactivator of various signaling pathways, preventing cell death. In mitochondria, DJ-1 is found in the synthasome, where it interacts with the β ATP synthase subunit. Downregulation of the DJ-1 gene under conditions of experimental PD increases sensitivity of the cells to neurotoxins, and introduction of the recombinant DJ-1 protein attenuates manifestation of this pathology. The thirteen-membered fragment of the DJ-1 amino acid sequence attached to the heptapeptide of the TAT protein penetrating into the cells exhibited neuroprotective properties in various PD models both in cell cultures and after administration to animals. Low molecular weight DJ-1 ligands also demonstrate therapeutic potential, providing neuroprotective effects seen during their incubation with cells and administration to animals.
Collapse
Affiliation(s)
- Olga A Buneeva
- Institute of Biomedical Chemistry, Moscow, 119121, Russia
| | | |
Collapse
|
46
|
Jadiya P, Garbincius JF, Elrod JW. Reappraisal of metabolic dysfunction in neurodegeneration: Focus on mitochondrial function and calcium signaling. Acta Neuropathol Commun 2021; 9:124. [PMID: 34233766 PMCID: PMC8262011 DOI: 10.1186/s40478-021-01224-4] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 06/27/2021] [Indexed: 02/06/2023] Open
Abstract
The cellular and molecular mechanisms that drive neurodegeneration remain poorly defined. Recent clinical trial failures, difficult diagnosis, uncertain etiology, and lack of curative therapies prompted us to re-examine other hypotheses of neurodegenerative pathogenesis. Recent reports establish that mitochondrial and calcium dysregulation occur early in many neurodegenerative diseases (NDDs), including Alzheimer's disease, Parkinson's disease, Huntington's disease, and others. However, causal molecular evidence of mitochondrial and metabolic contributions to pathogenesis remains insufficient. Here we summarize the data supporting the hypothesis that mitochondrial and metabolic dysfunction result from diverse etiologies of neuropathology. We provide a current and comprehensive review of the literature and interpret that defective mitochondrial metabolism is upstream and primary to protein aggregation and other dogmatic hypotheses of NDDs. Finally, we identify gaps in knowledge and propose therapeutic modulation of mCa2+ exchange and mitochondrial function to alleviate metabolic impairments and treat NDDs.
Collapse
Affiliation(s)
- Pooja Jadiya
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, 3500 N Broad St, MERB 949, Philadelphia, PA, 19140, USA
| | - Joanne F Garbincius
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, 3500 N Broad St, MERB 949, Philadelphia, PA, 19140, USA
| | - John W Elrod
- Center for Translational Medicine, Lewis Katz School of Medicine at Temple University, 3500 N Broad St, MERB 949, Philadelphia, PA, 19140, USA.
| |
Collapse
|
47
|
Huang M, Chen S. DJ-1 in neurodegenerative diseases: Pathogenesis and clinical application. Prog Neurobiol 2021; 204:102114. [PMID: 34174373 DOI: 10.1016/j.pneurobio.2021.102114] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Revised: 05/22/2021] [Accepted: 06/21/2021] [Indexed: 12/23/2022]
Abstract
Neurodegenerative diseases (NDs) are one of the major health threats to human characterized by selective and progressive neuronal loss. The mechanisms of NDs are still not fully understood. The study of genetic defects and disease-related proteins offers us a window into the mystery of it, and the extension of knowledge indicates that different NDs share similar features, mechanisms, and even genetic or protein abnormalities. Among these findings, PARK7 and its production DJ-1 protein, which was initially found implicated in PD, have also been found altered in other NDs. PARK7 mutations, altered expression and posttranslational modification (PTM) cause DJ-1 abnormalities, which in turn lead to downstream mechanisms shared by most NDs, such as mitochondrial dysfunction, oxidative stress, protein aggregation, autophagy defects, and so on. The knowledge of DJ-1 derived from PD researches might apply to other NDs in both basic research and clinical application, and might yield novel insights into and alternative approaches for dealing with NDs.
Collapse
Affiliation(s)
- Maoxin Huang
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China
| | - Shengdi Chen
- Department of Neurology and Institute of Neurology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, 200025, Shanghai, China; Lab for Translational Research of Neurodegenerative Diseases, Institute of Immunochemistry, Shanghai Tech University, 201210, Shanghai, China.
| |
Collapse
|
48
|
Esparza-Moltó PB, Romero-Carramiñana I, Núñez de Arenas C, Pereira MP, Blanco N, Pardo B, Bates GR, Sánchez-Castillo C, Artuch R, Murphy MP, Esteban JA, Cuezva JM. Generation of mitochondrial reactive oxygen species is controlled by ATPase inhibitory factor 1 and regulates cognition. PLoS Biol 2021; 19:e3001252. [PMID: 33983919 PMCID: PMC8148373 DOI: 10.1371/journal.pbio.3001252] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2021] [Revised: 05/25/2021] [Accepted: 04/29/2021] [Indexed: 11/18/2022] Open
Abstract
The mitochondrial ATP synthase emerges as key hub of cellular functions controlling the production of ATP, cellular signaling, and fate. It is regulated by the ATPase inhibitory factor 1 (IF1), which is highly abundant in neurons. Herein, we ablated or overexpressed IF1 in mouse neurons to show that IF1 dose defines the fraction of active/inactive enzyme in vivo, thereby controlling mitochondrial function and the production of mitochondrial reactive oxygen species (mtROS). Transcriptomic, proteomic, and metabolomic analyses indicate that IF1 dose regulates mitochondrial metabolism, synaptic function, and cognition. Ablation of IF1 impairs memory, whereas synaptic transmission and learning are enhanced by IF1 overexpression. Mechanistically, quenching the IF1-mediated increase in mtROS production in mice overexpressing IF1 reduces the increased synaptic transmission and obliterates the learning advantage afforded by the higher IF1 content. Overall, IF1 plays a key role in neuronal function by regulating the fraction of ATP synthase responsible for mitohormetic mtROS signaling.
Collapse
Affiliation(s)
- Pau B. Esparza-Moltó
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | - Inés Romero-Carramiñana
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | - Cristina Núñez de Arenas
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | - Marta P. Pereira
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - Noelia Blanco
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| | - Beatriz Pardo
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
| | - Georgina R. Bates
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - Carla Sánchez-Castillo
- Unidad de Neuropatología Molecular, Centro de Biología Molecular Severo Ochoa, Madrid, Spain
| | - Rafael Artuch
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
- Departamento de Bioquímica Clínica, Institut de Recerca Sant Joan de Déu, Barcelona, Spain
| | - Michael P. Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, United Kingdom
- Department of Medicine, University of Cambridge, Addenbrooke’s Hospital, Cambridge, United Kingdom
| | - José A. Esteban
- Unidad de Neuropatología Molecular, Centro de Biología Molecular Severo Ochoa, Madrid, Spain
| | - José M. Cuezva
- Departamento de Biología Molecular, Centro de Biología Molecular Severo Ochoa, Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), ISCIII, Madrid, Spain
- Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain
| |
Collapse
|
49
|
Galber C, Carissimi S, Baracca A, Giorgio V. The ATP Synthase Deficiency in Human Diseases. Life (Basel) 2021; 11:life11040325. [PMID: 33917760 PMCID: PMC8068106 DOI: 10.3390/life11040325] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/01/2021] [Accepted: 04/03/2021] [Indexed: 11/29/2022] Open
Abstract
Human diseases range from gene-associated to gene-non-associated disorders, including age-related diseases, neurodegenerative, neuromuscular, cardiovascular, diabetic diseases, neurocognitive disorders and cancer. Mitochondria participate to the cascades of pathogenic events leading to the onset and progression of these diseases independently of their association to mutations of genes encoding mitochondrial protein. Under physiological conditions, the mitochondrial ATP synthase provides the most energy of the cell via the oxidative phosphorylation. Alterations of oxidative phosphorylation mainly affect the tissues characterized by a high-energy metabolism, such as nervous, cardiac and skeletal muscle tissues. In this review, we focus on human diseases caused by altered expressions of ATP synthase genes of both mitochondrial and nuclear origin. Moreover, we describe the contribution of ATP synthase to the pathophysiological mechanisms of other human diseases such as cardiovascular, neurodegenerative diseases or neurocognitive disorders.
Collapse
Affiliation(s)
- Chiara Galber
- Consiglio Nazionale delle Ricerche, Institute of Neuroscience, I-35121 Padova, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, I-40126 Bologna, Italy
| | - Stefania Carissimi
- Consiglio Nazionale delle Ricerche, Institute of Neuroscience, I-35121 Padova, Italy
| | - Alessandra Baracca
- Department of Biomedical and Neuromotor Sciences, University of Bologna, I-40126 Bologna, Italy
| | - Valentina Giorgio
- Consiglio Nazionale delle Ricerche, Institute of Neuroscience, I-35121 Padova, Italy
- Department of Biomedical and Neuromotor Sciences, University of Bologna, I-40126 Bologna, Italy
| |
Collapse
|
50
|
PGC-1s in the Spotlight with Parkinson's Disease. Int J Mol Sci 2021; 22:ijms22073487. [PMID: 33800548 PMCID: PMC8036867 DOI: 10.3390/ijms22073487] [Citation(s) in RCA: 49] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 03/24/2021] [Accepted: 03/25/2021] [Indexed: 12/12/2022] Open
Abstract
Parkinson's disease is one of the most common neurodegenerative disorders worldwide, characterized by a progressive loss of dopaminergic neurons mainly localized in the substantia nigra pars compacta. In recent years, the detailed analyses of both genetic and idiopathic forms of the disease have led to a better understanding of the molecular and cellular pathways involved in PD, pointing to the centrality of mitochondrial dysfunctions in the pathogenic process. Failure of mitochondrial quality control is now considered a hallmark of the disease. The peroxisome proliferator-activated receptor gamma coactivator 1 (PGC-1) family acts as a master regulator of mitochondrial biogenesis. Therefore, keeping PGC-1 level in a proper range is fundamental to guarantee functional neurons. Here we review the major findings that tightly bond PD and PGC-1s, raising important points that might lead to future investigations.
Collapse
|