1
|
Venkatesan A, Ridilla M, Castro N, Wolosin JM, Henty-Ridilla JL, Knox BE, Ganapathy PS, Brown JS, DeVincentis Iii AF, Sieminski S, Bernstein AM. Mitochondrial and microtubule defects in Exfoliation Glaucoma. Free Radic Biol Med 2025; 233:226-239. [PMID: 40180018 PMCID: PMC12074171 DOI: 10.1016/j.freeradbiomed.2025.03.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/20/2025] [Accepted: 03/30/2025] [Indexed: 04/05/2025]
Abstract
Exfoliation Syndrome is an age-related systemic condition characterized by large aggregated fibrillar material deposition in the anterior eye tissues. This aggregate formation and deposition on the aqueous humor outflow pathway are significant risk factors for developing Exfoliation Glaucoma (XFG). XFG is a multifactorial late-onset disease that shares common features of neurodegenerative diseases, such as increased protein aggregation, impaired protein degradation, and oxidative and cellular stress. XFG patients display decreased mitochondrial membrane potential and mitochondrial DNA deletions. Here, using Tenon Capsule Fibroblasts (TFs) from patients without glaucoma (No Glaucoma, NG) and XFG patients, we found that XFG TFs have impaired mitochondrial bioenergetics and increased reactive oxygen species accumulation. These defects are associated with mitochondrial abnormalities as XFG TFs exhibit smaller mitochondria that contain dysmorphic cristae, with increased mitochondrial localization to lysosomes and slowed mitophagic flux. Mitochondrial dysfunction in the XFG TFs was associated with hyperdynamic microtubules, decreased acetylated tubulin, and increased HDAC6 activity. Treatment of XFG TFs with a mitophagy inducer, Urolithin A (UA), and a mitochondrial biogenesis inducer, Nicotinamide Ribose (NR), improved mitochondrial bioenergetics and reduced ROS accumulation. Our results demonstrate that XFG TFs have abnormal mitochondria and suggest that mitophagy inducers may represent a potential class of therapeutics for reversing mitochondrial dysfunction in XFG patients.
Collapse
Affiliation(s)
- Arunkumar Venkatesan
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, USA
| | | | - Nileyma Castro
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, USA; New York VA Health Care, Syracuse VA Medical Center, Syracuse, NY, USA
| | - J Mario Wolosin
- Eye and Vision Research Institute, Department of Ophthalmology, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Jessica L Henty-Ridilla
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Barry E Knox
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, USA; Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Preethi S Ganapathy
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, USA
| | - Jamin S Brown
- Retina-Vitreous Surgeons of Central New York, Syracuse, NY, USA
| | | | | | - Audrey M Bernstein
- Department of Ophthalmology and Visual Sciences, SUNY Upstate Medical University, Syracuse, NY, USA; New York VA Health Care, Syracuse VA Medical Center, Syracuse, NY, USA; Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, USA
| |
Collapse
|
2
|
Buckley Y, Stoll MSK, Hoppel CL, Mears JA. Fis1 regulates mitochondrial morphology, bioenergetics and removal of mitochondrial DNA damage in irradiated glioblastoma cells. J Cell Sci 2025; 138:jcs263459. [PMID: 39704270 PMCID: PMC11828467 DOI: 10.1242/jcs.263459] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2024] [Accepted: 12/05/2024] [Indexed: 12/21/2024] Open
Abstract
In response to external stress, mitochondrial dynamics is often disrupted, but the associated physiologic changes are often uncharacterized. In many cancers, including glioblastoma (GBM), mitochondrial dysfunction has been observed. Understanding how mitochondrial dynamics and physiology contribute to treatment resistance will lead to more targeted and effective therapeutics. This study aims to uncover how mitochondria in GBM cells adapt to and resist ionizing radiation (IR), a component of the standard of care for GBM. Using several approaches, we investigated how mitochondrial dynamics and physiology adapt to radiation stress, and we uncover a novel role for Fis1, a pro-fission protein, in regulating the stress response through mitochondrial DNA (mtDNA) maintenance and altered mitochondrial bioenergetics. Importantly, our data demonstrate that increased fission in response to IR leads to removal of mtDNA damage and more efficient oxygen consumption through altered electron transport chain (ETC) activities in intact mitochondria. These findings demonstrate a key role for Fis1 in targeting damaged mtDNA for degradation and regulating mitochondrial bioenergetics through altered dynamics.
Collapse
Affiliation(s)
- Yuli Buckley
- Department of Pharmacology and Center for Mitochondrial Diseases, Case Western Reserve University School of Medicine, Cleveland, OH 44016, USA
| | - Maria S. K. Stoll
- Department of Pharmacology and Center for Mitochondrial Diseases, Case Western Reserve University School of Medicine, Cleveland, OH 44016, USA
| | - Charles L. Hoppel
- Department of Pharmacology and Center for Mitochondrial Diseases, Case Western Reserve University School of Medicine, Cleveland, OH 44016, USA
| | - Jason A. Mears
- Department of Pharmacology and Center for Mitochondrial Diseases, Case Western Reserve University School of Medicine, Cleveland, OH 44016, USA
| |
Collapse
|
3
|
Özdemir M, Oeljeklaus S, Schendzielorz A, Morgenstern M, Valpadashi A, Yousefi R, Warscheid B, Dennerlein S. Definition of the human mitochondrial TOM interactome reveals TRABD as a new interacting protein. J Cell Sci 2025; 138:jcs263576. [PMID: 40105103 DOI: 10.1242/jcs.263576] [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: 09/26/2024] [Accepted: 03/03/2025] [Indexed: 03/20/2025] Open
Abstract
The mitochondrial proteome arises from dual genetic origins. Nuclear-encoded proteins need to be transported across or inserted into two distinguished membranes, and the translocase of the outer mitochondrial membrane (TOM) complex represents the main translocase in the outer mitochondrial membrane. Its composition and regulation have been extensively investigated within yeast cells. However, we have little knowledge of the TOM complex composition within human cells. Here, we have defined the TOM interactome in a comprehensive manner using biochemical approaches to isolate the TOM complex in combination with quantitative mass spectrometry analyses. With these studies, we defined the pleiotropic nature of the human TOM complex, including new interactors, such as TRABD. Our studies provide a framework to understand the various biogenesis pathways that merge at the TOM complex within human cells.
Collapse
Affiliation(s)
- Metin Özdemir
- Institute for Cellular Biochemistry, University of Goettingen, D-37073 Goettingen, Germany
| | - Silke Oeljeklaus
- Faculty of Chemistry and Pharmacy, Biochemistry II, Theodor Boveri-Institute, University of Würzburg, D-97074 Wuerzburg, Germany
| | - Alexander Schendzielorz
- Institute for Biology II, Faculty for Biology, Functional Proteomics, University Freiburg, D-79104 Freiburg, Germany
| | - Marcel Morgenstern
- Institute for Biology II, Faculty for Biology, Functional Proteomics, University Freiburg, D-79104 Freiburg, Germany
| | - Anusha Valpadashi
- Institute for Cellular Biochemistry, University of Goettingen, D-37073 Goettingen, Germany
| | - Roya Yousefi
- Institute for Cellular Biochemistry, University of Goettingen, D-37073 Goettingen, Germany
| | - Bettina Warscheid
- Faculty of Chemistry and Pharmacy, Biochemistry II, Theodor Boveri-Institute, University of Würzburg, D-97074 Wuerzburg, Germany
- Institute for Biology II, Faculty for Biology, Functional Proteomics, University Freiburg, D-79104 Freiburg, Germany
| | - Sven Dennerlein
- Institute for Cellular Biochemistry, University of Goettingen, D-37073 Goettingen, Germany
| |
Collapse
|
4
|
Hu W, Zhang J, Wu Z, Wu Y, Hu Y, Hu X, Cao J. Research progress on paternal mitochondrial inheritance: An overview. Mitochondrion 2025; 82:102019. [PMID: 40024491 DOI: 10.1016/j.mito.2025.102019] [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: 07/05/2024] [Accepted: 02/22/2025] [Indexed: 03/04/2025]
Abstract
Mitochondria are self-replicating organelles with their own DNA. They play a crucial role in biological, cellular and functional processes, such as energy production, metabolism, and signal transduction. Abnormal mitochondrial function can cause various diseases such as diabetes, tumour, Parkinson's disease, hereditary optic neuropathy, and others. Although mitochondrial functions have been extensively and widely explored, studies on mitochondrial inheritance have been limited. Mitochondrial inheritance is traditionally thought to be maternal although small amounts of paternally transmitted mitochondria have been discovered on rare occasions, and the role of paternal mitochondria transmission to offspring has been largely ignored. This review highlights the present knowledge on mitochondrial inheritance, especially the controversy and the difficulties in investigating paternal mitochondrial inheritance. More significantly, we present a comprehensive description of the physiological functions of paternal mitochondria in children and discuss the animal model to explore the mechanism of paternal mitochondrial inheritance. This review may provide a theoretical and experimental basis for improving our understanding of paternal mitochondrial inheritance, and also provide new ideas for treating mitochondrial diseases.
Collapse
Affiliation(s)
- Wen Hu
- Key Laboratory of Mitochondrial Medicine, Department of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Jiting Zhang
- Key Laboratory of Mitochondrial Medicine, Department of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Zhaoqi Wu
- Key Laboratory of Mitochondrial Medicine, Department of Basic Medicine, Gannan Medical University, Ganzhou, China
| | - Yi Wu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Ministry of Education, Gannan Medical University, Ganzhou, China
| | - Yuhui Hu
- Medical College, Jinggangshan University, Ji'an, China
| | - Xiaohui Hu
- Department of Basic Medicine, Gannan Healthcare Vocational College, Ganzhou, China.
| | - Jinguo Cao
- Key Laboratory of Mitochondrial Medicine, Department of Basic Medicine, Gannan Medical University, Ganzhou, China.
| |
Collapse
|
5
|
Zhang H, Meléndez A. Conserved components of the macroautophagy machinery in Caenorhabditis elegans. Genetics 2025; 229:iyaf007. [PMID: 40180610 PMCID: PMC12005284 DOI: 10.1093/genetics/iyaf007] [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: 03/29/2024] [Accepted: 12/13/2024] [Indexed: 04/05/2025] Open
Abstract
Macroautophagy involves the sequestration of cytoplasmic contents in a double-membrane autophagosome and its subsequent delivery to lysosomes for degradation and recycling. In Caenorhabditis elegans, autophagy participates in diverse processes such as stress resistance, cell fate specification, tissue remodeling, aging, and adaptive immunity. Genetic screens in C. elegans have identified a set of metazoan-specific autophagy genes that form the basis for our molecular understanding of steps unique to the autophagy pathway in multicellular organisms. Suppressor screens have uncovered multiple mechanisms that modulate autophagy activity under physiological conditions. C. elegans also provides a model to investigate how autophagy activity is coordinately controlled at an organismal level. In this chapter, we will discuss the molecular machinery, regulation, and physiological functions of autophagy, and also methods utilized for monitoring autophagy during C. elegans development.
Collapse
Affiliation(s)
- Hong Zhang
- National Laboratory of Biomacromolecules, New Cornerstone Science Laboratory, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, P.R. China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, P.R. China
| | - Alicia Meléndez
- Department of Biology, Queens College, City University of New York, Flushing, NY 11367, USA
- Molecular, Cellular and Developmental Biology and Biochemistry Ph.D. Programs, The Graduate Center of the City University of New York, New York, NY 10016, USA
| |
Collapse
|
6
|
Chung CY, Singh K, Sheshadri P, Valdebenito GE, Chacko AR, Costa Besada MA, Liang XF, Kabir L, Pitceathly RDS, Szabadkai G, Duchen MR. Inhibition of the PI3K-AKT-MTORC1 axis reduces the burden of the m.3243A>G mtDNA mutation by promoting mitophagy and improving mitochondrial function. Autophagy 2025; 21:881-896. [PMID: 39667405 PMCID: PMC11925111 DOI: 10.1080/15548627.2024.2437908] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 11/20/2024] [Accepted: 11/29/2024] [Indexed: 12/14/2024] Open
Abstract
Mitochondrial DNA (mtDNA) encodes genes essential for oxidative phosphorylation. The m.3243A>G mutation causes severe disease, including myopathy, lactic acidosis and stroke-like episodes (MELAS) and is the most common pathogenic mtDNA mutation in humans. We have previously shown that the mutation is associated with constitutive activation of the PI3K-AKT-MTORC1 axis. Inhibition of this pathway in patient fibroblasts reduced the mutant load, rescued mitochondrial bioenergetic function and reduced glucose dependence. We have now investigated the mechanisms that select against the mutant mtDNA under these conditions. Basal macroautophagy/autophagy and lysosomal degradation of mitochondria were suppressed in the mutant cells. Pharmacological inhibition of any step of the PI3K-AKT-MTORC1 pathway activated mitophagy and progressively reduced m.3243A>G mutant load over weeks. Inhibition of autophagy with bafilomycin A1 or chloroquine prevented the reduction in mutant load, suggesting that mitophagy was necessary to remove the mutant mtDNA. Inhibition of the pathway was associated with metabolic remodeling - mitochondrial membrane potential and respiratory rate improved even before a measurable fall in mutant load and proved crucial for mitophagy. Thus, maladaptive activation of the PI3K-AKT-MTORC1 axis and impaired autophagy play a major role in shaping the presentation and progression of disease caused by the m.3243A>G mutation. Our findings highlight a potential therapeutic target for this otherwise intractable disease.Abbreviation: ΔΨm: mitochondrial membrane potential; 2DG: 2-deoxy-D-glucose; ANOVA: analysis of variance; ARMS-qPCR: amplification-refractory mutation system quantitative polymerase chain reaction; Baf A1: bafilomycin A1; BSA: bovine serum albumin; CQ: chloroquine; Cybrid: cytoplasmic hybrid; CYCS: cytochrome c, somatic; DCA: dichloroacetic acid; DMEM: Dulbecco's modified Eagle's medium; DMSO: dimethylsulfoxide; EGFP: enhanced green fluorescent protein; LC3B-I: carboxy terminus cleaved microtubule-associated protein 1 light chain 3 beta; LC3B-II: lipidated microtubule-associated protein 1 light chain 3 beta; LY: LY290042; MAP1LC3B/LC3B: microtubule associated protein 1 light chain 3 beta; MELAS: mitochondrial encephalomyopathy, lactic acidosis and stroke-like episodes; MFC: mitochondrial fragmentation count; mt-Keima: mitochondrial-targeted mKeima; mtDNA: mitochondrial DNA/mitochondrial genome; MTOR: mechanistic target of rapamycin kinase; MTORC1: MTOR complex 1; OA: oligomycin+antimycin A; OxPhos: oxidative phosphorylation; DPBS: Dulbecco's phosphate-buffered saline; PPARGC1A/PGC-1α: PPARG coactivator 1 alpha; PPARGC1B/PGC-1β: PPARG coactivator 1 beta; PI3K: phosphoinositide 3-kinase; PINK1: PTEN induced kinase 1; qPCR: quantitative polymerase chain reaction; RNA-seq: RNA sequencing; RP: rapamycin; SQSTM1/p62: sequestosome 1; TEM: transmission electron microscopy; WT: wild-type.
Collapse
Affiliation(s)
- Chih-Yao Chung
- Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, UCL, London, UK
| | - Kritarth Singh
- Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, UCL, London, UK
| | - Preethi Sheshadri
- Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, UCL, London, UK
| | - Gabriel E Valdebenito
- Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, UCL, London, UK
| | - Anitta R. Chacko
- Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, UCL, London, UK
| | - María Alicia Costa Besada
- Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, UCL, London, UK
- Cellular and Molecular Neurobiology of Parkinson’s Disease, Research Center for Molecular Medicine and Chronic Diseases (CIMUS), University of Santiago de Compostela, Spain
| | - Xiao Fei Liang
- Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, UCL, London, UK
| | - Lida Kabir
- Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, UCL, London, UK
| | - Robert D. S. Pitceathly
- Department of Neuromuscular Diseases, UCL Queen Square Institute of Neurology, London, UK
- NHS Highly Specialised Service for Rare Mitochondrial Disorders, Queen Square Centre for Neuromuscular Diseases, The National Hospital for Neurology and Neurosurgery, London, UK
| | - Gyorgy Szabadkai
- Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, UCL, London, UK
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- The Francis Crick Institute, London, UK
| | - Michael R. Duchen
- Department of Cell and Developmental Biology and Consortium for Mitochondrial Research, UCL, London, UK
| |
Collapse
|
7
|
Yan J, Chen X, Choksi S, Liu ZG. TGFB signaling induces mitophagy via PLSCR3-mediated cardiolipin externalization in conjunction with a BNIP3L/NIX-, BNIP3-, and FUNDC1-dependent mechanism. Autophagy 2025:1-11. [PMID: 40119553 DOI: 10.1080/15548627.2025.2483441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Revised: 03/10/2025] [Accepted: 03/19/2025] [Indexed: 03/24/2025] Open
Abstract
Selective clearance of damaged mitochondria through mitophagy is crucial for the maintenance of mitochondrial homeostasis. While mitophagy can be activated by various mitochondrial toxins, the physiologically relevant signal that triggers mitophagy is less studied. TGFB/TGFβ signaling has been linked to autophagic induction, but its specific role in mitophagy is not well understood. Here, we discovered a novel mitophagy induction paradigm stimulated by TGFB1. The mitophagic response is exclusively mediated by SMAD2, SMAD3, and SMAD4 underlying the TGFB receptor signaling. The transcriptional regulation activates genes involved in the canonical autophagic pathway which is required for the TGFB1-induced mitophagy. Moreover, TGFB1 signaling promotes mitophagic flux by upregulating PLSCR3 that externalizes cardiolipin in conjunction with the MAP1LC3/LC3/GABARAPs-interacting receptor proteins (BNIP3L/NIX, BNIP3, and FUNDC1)-dependent mechanism. Overall, our study characterized the essential components engaged in the TGFB1-induced mitophagy and demonstrated that TGFB is an important signal that induces mitophagy.Abbreviations ATG5: autophagy related 5; ATG8: mammalian homolog of yeast Atg8; ATG9A: autophagy related 9A; ATG13: autophagy related 13; ATG101: autophagy related 101; BNIP3: BCL2 interacting protein 3; BNIP3L/NIX: BCL2 interacting protein 3 like; CALCOCO2/NDP52: calcium binding and coiled-coil domain 2; Cardiolipin: 1,3-bis(sn-3'-phosphatidyl)-sn-glycerol; CERS1: ceramide synthase 1; FUNDC1: FUN14 domain containing 1; GABARAP: GABA type A receptor-associated protein; GABARAPL1: GABA type A receptor-associated protein like 1; GABARAPL2: GABA type A receptor-associated protein like 2; GLS: glutaminase; KO: knockout; MAP1LC3/LC3: microtubule associated protein 1 light chain 3; MitoIP: mitochondrial immunoprecipitation; MMP: mitochondrial membrane potential; NRBF2: nuclear receptor binding factor 2; OPTN: optineurin; PINK1: PTEN induced kinase 1; PLSCR3: phospholipid scramblase 3; PRKN: parkin RBR E3 ubiquitin protein ligase; RB1CC1/FIP200: RB1 inducible coiled-coil 1; TGFB/TGFβ: transforming growth factor beta; ULK1: unc-51 like autophagy activating kinase 1.
Collapse
Affiliation(s)
- Jiong Yan
- Laboratory of Cellular and Molecular Biology (LCMB), Center for Cancer Research (CCR), National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Xin Chen
- Laboratory of Cellular and Molecular Biology (LCMB), Center for Cancer Research (CCR), National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Swati Choksi
- Laboratory of Cellular and Molecular Biology (LCMB), Center for Cancer Research (CCR), National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| | - Zheng-Gang Liu
- Laboratory of Cellular and Molecular Biology (LCMB), Center for Cancer Research (CCR), National Cancer Institute, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
8
|
Akhtar A, Shakir M, Ansari MS, Divya, Faizan MI, Chauhan V, Singh A, Alam R, Azmi I, Sharma S, Pracha M, Uddin IM, Bashir U, Shahni SN, Chaudhuri R, Albogami S, Ganguly R, Sagar S, Singh VP, Kharya G, Srivastava AK, Mabalirajan U, Roy SS, Rahman I, Ahmad T. Bioengineering the metabolic network of CAR T cells with GLP-1 and Urolithin A increases persistence and long-term anti-tumor activity. Cell Rep Med 2025; 6:102021. [PMID: 40107240 PMCID: PMC11970383 DOI: 10.1016/j.xcrm.2025.102021] [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: 05/17/2024] [Revised: 12/10/2024] [Accepted: 02/18/2025] [Indexed: 03/22/2025]
Abstract
Constant tumor antigen exposure disrupts chimeric antigen receptor (CAR) T cell metabolism, limiting their persistence and anti-tumor efficacy. To address this, we develop metabolically reprogrammed CAR (MCAR) T cells with enhanced autophagy and mitophagy. A compound screening identifies a synergy between GLP-1R agonist (semaglutide [SG]) and Urolithin A (UrA), which activate autophagy through mTOR (mechanistic target of rapamycin) inhibition and mitophagy via Atg4b activation, maintaining mitochondrial metabolism in CAR T cells (MCAR T-1). These changes increase CD8+ T memory cells (Tm), enhancing persistence and anti-tumor activity in vitro and in xenograft models. GLP-1R knockdown in CAR T cells diminishes autophagy/mitophagy induction, confirming its critical role. We further engineer GLP-1-secreting cells (MCAR T-2), which exhibited sustained memory, stemness, and long-term persistence, even under tumor re-challenge. MCAR T-2 cells also reduce cytokine release syndrome (CRS) risks while demonstrating potent anti-tumor effects. This strategy highlights the potential of metabolic reprogramming via targeting autophagy/mitophagy pathways to improve CAR T cell therapy outcomes, ensuring durability and efficacy.
Collapse
Affiliation(s)
- Areej Akhtar
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Md Shakir
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Mohammad Sufyan Ansari
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Divya
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Md Imam Faizan
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Varnit Chauhan
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Aashi Singh
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Ruquaiya Alam
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Iqbal Azmi
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Sheetal Sharma
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Mehak Pracha
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Insha Mohi Uddin
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Uzma Bashir
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Syeda Najidah Shahni
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India
| | - Rituparna Chaudhuri
- Indian Institute of Science, Centre for Brain Research, Bengaluru, Karnataka, India
| | - Sarah Albogami
- Department of Biotechnology, College of Science, Taif University, Taif, Saudi Arabia
| | - Rik Ganguly
- Department of Biotechnology & Bioinformatics, North-Eastern Hill University, Shillong, India
| | - Shakti Sagar
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, India
| | - Vijay Pal Singh
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, India
| | - Gaurav Kharya
- Centre for Bone Marrow Transplant & Cellular Therapy, Indraprastha Apollo Hospital, New Delhi, India
| | | | | | - Soumya Sinha Roy
- CSIR-Institute of Genomics & Integrative Biology, New Delhi, India
| | - Irfan Rahman
- Department of Environmental Medicine, University of Rochester Medical Center, Rochester, NY, USA
| | - Tanveer Ahmad
- Multidisciplinary Centre for Advanced Research and Studies, Jamia Millia Islamia, New Delhi, India.
| |
Collapse
|
9
|
Liu R, Zhou B. Harmine promotes axon regeneration through enhancing glucose metabolism. J Biol Chem 2025; 301:108254. [PMID: 39904483 PMCID: PMC11927705 DOI: 10.1016/j.jbc.2025.108254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 01/11/2025] [Accepted: 01/28/2025] [Indexed: 02/06/2025] Open
Abstract
Axon regeneration requires a substantial mitochondrial energy supply. However, injured mature neurons often fail to regenerate due to their inability to meet these elevated energy demands. Our findings indicate that harmine compensates for the energy deficit following axonal injury by enhancing the coupling between glucose metabolism and mitochondrial homeostasis, thereby promoting axon regeneration. Notably, harmine facilitates mitochondrial biogenesis and enhances mitophagy, ensuring efficient mitochondrial turnover, and energy supply. Thus, harmine plays a crucial role in enhancing glucose metabolism to maintain mitochondrial function, demonstrating significant potential in treating mature neuronal axon injuries and sciatic nerve injuries.
Collapse
Affiliation(s)
- Ruixuan Liu
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, China; School of Biological Science and Medical Engineering, Beihang University, Beijing, China
| | - Bing Zhou
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, Beihang University, Beijing, China; Interdisciplinary Innovation Institute of Medicine and Engineering Interdisciplinary, Beihang University, Beijing, China.
| |
Collapse
|
10
|
Wang Z, Meerod T, Cortes-Silva N, Chiang ACY, Nie Z, Di Y, Mu P, Verma A, Reid AJ, Ma H. Poldip2 promotes mtDNA elimination during Drosophila spermatogenesis to ensure maternal inheritance. EMBO J 2025; 44:1724-1748. [PMID: 39934413 PMCID: PMC11914606 DOI: 10.1038/s44318-025-00378-4] [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: 06/03/2024] [Revised: 12/08/2024] [Accepted: 01/24/2025] [Indexed: 02/13/2025] Open
Abstract
Maternal inheritance of mitochondrial DNA (mtDNA) is highly conserved in metazoans. While many species eliminate paternal mtDNA during late sperm development to foster maternal inheritance, the regulatory mechanisms governing this process remain elusive. Through a forward genetic screen in Drosophila, we identified 47 mutant lines exhibiting substantial retention of mtDNA in mature sperm. We mapped one line to poldip2, a gene predominantly expressed in the testis. Disruption of poldip2 led to substantial mtDNA retention in mature sperm and subsequent paternal transmission to progeny. Further investigation via imaging, biochemical analyses and ChIP assays revealed that Poldip2 is a mitochondrial matrix protein capable of binding mtDNA. Moreover, we showed that ClpX, the key component of a major mitochondrial protease, interacts with Poldip2 to co-regulate mtDNA elimination in Drosophila spermatids. This study sheds light on the mechanisms underlying mtDNA removal during spermatogenesis and underscores the pivotal role of this process in safeguarding maternal inheritance.
Collapse
Affiliation(s)
- Ziming Wang
- Gurdon Institute, Tennis Court Road, Cambridge, CB2 1QN, UK
- Department of Genetics, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK
| | - Tirawit Meerod
- School of Biosciences, University of Birmingham, Edgbaston, B15 2TT, UK
| | - Nuria Cortes-Silva
- Gurdon Institute, Tennis Court Road, Cambridge, CB2 1QN, UK
- Department of Genetics, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK
- Department of Pharmacology, UT Southwestern Medical Center, 6001 Forest Park Road, Dallas, TX, 75390, USA
| | - Ason C-Y Chiang
- School of Biosciences, University of Birmingham, Edgbaston, B15 2TT, UK
| | - Ziyan Nie
- Gurdon Institute, Tennis Court Road, Cambridge, CB2 1QN, UK
- Department of Genetics, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK
- Institut de Génomique Fonctionnelle de Lyon, ENS de Lyon, CNRS, Univ Lyon 1, 46 Allée d'Italie, Lyon, France
| | - Ying Di
- Gurdon Institute, Tennis Court Road, Cambridge, CB2 1QN, UK
- Department of Genetics, University of Cambridge, Downing Street, Cambridge, CB2 3EH, UK
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge, CB2 0XY, UK
| | - Peiqiang Mu
- Guangdong Provincial Key Laboratory of Protein Function and Regulation in Agricultural Organisms, South China Agricultural University, Tianhe District, Guangzhou, 510642, Guangdong, P. R. China
| | - Ankit Verma
- Gurdon Institute, Tennis Court Road, Cambridge, CB2 1QN, UK
| | | | - Hansong Ma
- School of Biosciences, University of Birmingham, Edgbaston, B15 2TT, UK.
| |
Collapse
|
11
|
Kovacheva E, Gevezova M, Mehterov N, Kazakova M, Sarafian V. The Intersection of Mitophagy and Autism Spectrum Disorder: A Systematic Review. Int J Mol Sci 2025; 26:2217. [PMID: 40076836 PMCID: PMC11899999 DOI: 10.3390/ijms26052217] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 02/27/2025] [Accepted: 02/28/2025] [Indexed: 03/14/2025] Open
Abstract
Autism spectrum disorder (ASD) is a group of neurodevelopmental and biobehavioral conditions that arises from complex interactions between environmental factors and physiological development in genetically predisposed individuals. Among the most frequently observed metabolic abnormalities in ASD is mitochondrial dysfunction. Mitochondria respond to cellular stress by altering their dynamics or initiating mitophagy. In neurons, the buildup of dysfunctional mitochondria and reactive oxygen species (ROS) poses a significant risk, as these cells cannot regenerate through division. To safeguard mitochondrial health, cells rely on an efficient "clean-up mechanism" to remove compromised organelles. Mitophagy, a specific form of autophagy, is responsible for regulating the turnover of flawed and non-functional mitochondria. Impairments in this process result in the accumulation of defective mitochondria in neurons, a characteristic of several neurodegenerative disorders associated with behavioral abnormalities. This systematic review offers an in-depth summary of the present knowledge of mitophagy and underscores its pivotal role in the pathogenesis of ASD.
Collapse
Affiliation(s)
- Eleonora Kovacheva
- Department of Medical Biology, Faculty of Medicine, Medical University—Plovdiv, 4000 Plovdiv, Bulgaria; (E.K.); (M.G.); (N.M.); (M.K.)
- Research Institute, Medical University—Plovdiv, 4000 Plovdiv, Bulgaria
| | - Maria Gevezova
- Department of Medical Biology, Faculty of Medicine, Medical University—Plovdiv, 4000 Plovdiv, Bulgaria; (E.K.); (M.G.); (N.M.); (M.K.)
- Research Institute, Medical University—Plovdiv, 4000 Plovdiv, Bulgaria
| | - Nikolay Mehterov
- Department of Medical Biology, Faculty of Medicine, Medical University—Plovdiv, 4000 Plovdiv, Bulgaria; (E.K.); (M.G.); (N.M.); (M.K.)
- Research Institute, Medical University—Plovdiv, 4000 Plovdiv, Bulgaria
| | - Maria Kazakova
- Department of Medical Biology, Faculty of Medicine, Medical University—Plovdiv, 4000 Plovdiv, Bulgaria; (E.K.); (M.G.); (N.M.); (M.K.)
- Research Institute, Medical University—Plovdiv, 4000 Plovdiv, Bulgaria
| | - Victoria Sarafian
- Department of Medical Biology, Faculty of Medicine, Medical University—Plovdiv, 4000 Plovdiv, Bulgaria; (E.K.); (M.G.); (N.M.); (M.K.)
- Research Institute, Medical University—Plovdiv, 4000 Plovdiv, Bulgaria
| |
Collapse
|
12
|
Chowdhury O, Bammidi S, Gautam P, Babu VS, Liu H, Shang P, Xin Y, Mahally E, Nemani M, Koontz V, Lathrop K, Kedziora KM, Franks J, Sun M, Smith JW, DeVine LR, Cole RN, Stepicheva N, Strizhakova A, Chattopadhyay S, Hose S, Zigler JS, Sahel JA, Qian J, Guha P, Handa JT, Ghosh S, Sinha D. Activated mTOR Signaling in the RPE Drives EMT, Autophagy, and Metabolic Disruption, Resulting in AMD-Like Pathology in Mice. Aging Cell 2025:e70018. [PMID: 39957408 DOI: 10.1111/acel.70018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 02/02/2025] [Accepted: 02/06/2025] [Indexed: 02/18/2025] Open
Abstract
The mechanistic target of rapamycin (mTOR) complexes 1 and 2 (mTORC1/2) are crucial for various physiological functions. Although the role of mTORC1 in retinal pigmented epithelium (RPE) homeostasis and age-related macular degeneration (AMD) pathogenesis is established, the function of mTORC2 remains unclear. We investigated both complexes in RPE health and disease. Therefore, in this study, we have attempted to demonstrate that the specific overexpression of mammalian lethal with Sec13 protein 8 (mLST8) in the mouse RPE activates both mTORC1 and mTORC2, inducing epithelial-mesenchymal transition (EMT)-like changes and subretinal/RPE deposits resembling early AMD-like pathogenesis. Aging in these mice leads to RPE degeneration, causing retinal damage, impaired debris clearance, and metabolic and mitochondrial dysfunction. Inhibition of mTOR with TORIN1 in vitro or βA3/A1-crystallin in vivo normalized mTORC1/2 activity and restored function, revealing a novel role for the mTOR complexes in regulating RPE function, impacting retinal health and disease.
Collapse
Affiliation(s)
- Olivia Chowdhury
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sridhar Bammidi
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Pooja Gautam
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Vishnu Suresh Babu
- Department of Ophthalmology, The Wilmer Eye Institute, the Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Haitao Liu
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Peng Shang
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ying Xin
- Department of Ophthalmology, The Wilmer Eye Institute, the Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Emma Mahally
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Mihir Nemani
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Victoria Koontz
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Kira Lathrop
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Katarzyna M Kedziora
- Department of Cell Biology and Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jonathan Franks
- Department of Cell Biology and Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ming Sun
- Department of Cell Biology and Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Joshua W Smith
- Johns Hopkins Mass Spectrometry and Proteomics Facility, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Lauren R DeVine
- Johns Hopkins Mass Spectrometry and Proteomics Facility, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Robert N Cole
- Johns Hopkins Mass Spectrometry and Proteomics Facility, The Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Nadezda Stepicheva
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Anastasia Strizhakova
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sreya Chattopadhyay
- Department of Physiology, University of Calcutta, Kolkata, West Bengal, India
| | - Stacey Hose
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jacob Samuel Zigler
- Department of Ophthalmology, The Wilmer Eye Institute, the Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - José-Alain Sahel
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Institut De La Vision, INSERM, CNRS, Sorbonne Université, Paris, France
| | - Jiang Qian
- Department of Ophthalmology, The Wilmer Eye Institute, the Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Prasun Guha
- Nevada Institute of Personalized Medicine, University of Nevada, Las Vegas, Nevada, USA
| | - James T Handa
- Department of Ophthalmology, The Wilmer Eye Institute, the Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| | - Sayan Ghosh
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Debasish Sinha
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
- Department of Ophthalmology, The Wilmer Eye Institute, the Johns Hopkins School of Medicine, Baltimore, Maryland, USA
| |
Collapse
|
13
|
Wang S, Xue D. Asymmetric partitioning of persistent paternal mitochondria during cell divisions safeguards embryo development and mitochondrial inheritance. Dev Cell 2025:S1534-5807(25)00033-4. [PMID: 39904343 DOI: 10.1016/j.devcel.2025.01.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 12/17/2024] [Accepted: 01/15/2025] [Indexed: 02/06/2025]
Abstract
Most eukaryotes inherit only maternal mitochondria. The reasons for paternal mitochondrial elimination and the impacts of persistent paternal mitochondria on animals remain elusive. We show that undegraded paternal mitochondria in autophagy-deficient C. elegans embryos are gradually excluded from germ blastomeres through asymmetric partitioning during cell divisions. The embryonic cortical flow drives anterior-directed movements of paternal mitochondria and contributes to their asymmetric apportioning between two daughter blastomeres. By contrast, autophagosome-enclosed paternal mitochondria cluster around and segregate with centrosomes during mitosis and are rapidly degraded through lysosomes concentrated near centrosomes. Failure to exclude persistent paternal mitochondria from the germ blastomere at first cleavage causes their enrichment in the descendant endomesodermal (EMS) blastomere, leading to elevated reactive oxygen species levels, elongated EMS lineage durations, and increased embryonic lethality, which antioxidant treatments can suppress. Thus, regulated paternal mitochondrial distribution away from germ blastomeres is a fail-safe mechanism, protecting embryo development and maternal mitochondrial inheritance.
Collapse
Affiliation(s)
- Songyun Wang
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| | - Ding Xue
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309, USA.
| |
Collapse
|
14
|
Singh A, Perez ML, Kirsanov O, Padilla-Banks E, Guardia CM. Autophagy in reproduction and pregnancy-associated diseases. iScience 2024; 27:111268. [PMID: 39628569 PMCID: PMC11613427 DOI: 10.1016/j.isci.2024.111268] [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] [Indexed: 12/06/2024] Open
Abstract
As advantageous as sexual reproduction is during progeny generation, it is also an expensive and treacherous reproductive strategy. The viviparous eukaryote has evolved to survive stress before, during, and after pregnancy. An important and conserved intracellular pathway for the control of metabolic stress is autophagy. The autophagy process occurs in multiple stages through the coordinated action of autophagy-related genes. This review summarizes the evidence that autophagy is an integral component of reproduction. Additionally, we discuss emerging in vitro techniques that will enable cellular and molecular studies of autophagy and its associated pathways in reproduction. Finally, we discuss the role of autophagy in the pathogenesis and progression of several pregnancy-related disorders such as preterm birth, preeclampsia, and intra-uterine growth restriction, and its potential as a therapeutic target.
Collapse
Affiliation(s)
- Asmita Singh
- Placental Cell Biology Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, USA
| | - Maira L. Perez
- Placental Cell Biology Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, USA
| | - Oleksandr Kirsanov
- Placental Cell Biology Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, USA
| | - Elizabeth Padilla-Banks
- Placental Cell Biology Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, USA
| | - Carlos M. Guardia
- Placental Cell Biology Group, National Institute of Environmental Health Sciences, National Institutes of Health, Research Triangle Park, Durham, NC, USA
| |
Collapse
|
15
|
Zhao A, Maple L, Jiang J, Myers KN, Jones CG, Gagg H, McGarrity-Cottrell C, Rominiyi O, Collis SJ, Wells G, Rahman M, Danson SJ, Robinson D, Smythe C, Guo C. SENP3-FIS1 axis promotes mitophagy and cell survival under hypoxia. Cell Death Dis 2024; 15:881. [PMID: 39638786 PMCID: PMC11621581 DOI: 10.1038/s41419-024-07271-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 11/23/2024] [Accepted: 11/29/2024] [Indexed: 12/07/2024]
Abstract
SUMOylation, the covalent attachment of the small ubiquitin-like modifier (SUMO) to target proteins, and its reversal, deSUMOylation by SUMO proteases like Sentrin-specific proteases (SENPs), are crucial for initiating cellular responses to hypoxia. However, their roles in subsequent adaptation processes to hypoxia such as mitochondrial autophagy (mitophagy) remain unexplored. Here, we show that general SUMOylation, particularly SUMO2/3 modification, suppresses mitophagy under both normoxia and hypoxia. Furthermore, we identify deSUMO2/3-ylation enzyme SENP3 and mitochondrial Fission protein 1 (FIS1) as key players in hypoxia-induced mitophagy (HIM), with SUMOylatable FIS1 acting as a crucial regulator for SENP3-mediated HIM regulation. Interestingly, we find that hypoxia promotes FIS1 SUMO2/3-ylation and triggers an interaction between SUMOylatable FIS1 and Rab GTPase-activating protein Tre-2/Bub2/Cdc16 domain 1 family member 17 (TBC1D17), which in turn suppresses HIM. Therefore, we propose a novel SUMOylation-dependent pathway where the SENP3-FIS1 axis promotes HIM, with TBC1D17 acting as a fine-tuning regulator. Importantly, the SENP3-FIS1 axis plays a protective role against hypoxia-induced cell death, highlighting its physiological significance, and hypoxia-inducible FIS1-TBC1D17 interaction is detectable in primary glioma stem cell-like (GSC) cultures derived from glioblastoma patients, suggesting its disease relevance. Our findings not only provide new insights into SUMOylation/deSUMOylation regulation of HIM but also suggest the potential of targeting this pathway to enhance cellular resilience under hypoxic stress.
Collapse
Affiliation(s)
- Alice Zhao
- School of Biosciences, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Laura Maple
- School of Biosciences, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Juwei Jiang
- School of Biosciences, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Katie N Myers
- Division of Clinical Medicine, University of Sheffield Medical School, Sheffield, S10 2RX, UK
| | - Callum G Jones
- Division of Clinical Medicine, University of Sheffield Medical School, Sheffield, S10 2RX, UK
| | - Hannah Gagg
- Division of Clinical Medicine, University of Sheffield Medical School, Sheffield, S10 2RX, UK
| | | | - Ola Rominiyi
- Division of Clinical Medicine, University of Sheffield Medical School, Sheffield, S10 2RX, UK
- Division of Neuroscience, University of Sheffield Medical School, Sheffield, S10 2HQ, UK
- Department of Neurosurgery, Sheffield Teaching Hospitals NHS Foundation Trust, Sheffield, S10 2JF, UK
| | - Spencer J Collis
- Division of Clinical Medicine, University of Sheffield Medical School, Sheffield, S10 2RX, UK
| | - Greg Wells
- Ex vivo Project Team, Division of Clinical Medicine, University of Sheffield Medical School, Sheffield, S10 2RX, UK
| | - Marufur Rahman
- Ex vivo Project Team, Division of Clinical Medicine, University of Sheffield Medical School, Sheffield, S10 2RX, UK
| | - Sarah J Danson
- Ex vivo Project Team, Division of Clinical Medicine, University of Sheffield Medical School, Sheffield, S10 2RX, UK
| | - Darren Robinson
- School of Biosciences, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK
| | - Carl Smythe
- School of Biosciences, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK.
| | - Chun Guo
- School of Biosciences, University of Sheffield, Firth Court, Western Bank, Sheffield, S10 2TN, UK.
| |
Collapse
|
16
|
Venkatesan A, Ridilla M, Castro N, Wolosin JM, Henty-Ridilla JL, Knox BE, Ganapathy PS, Brown JS, DeVincentis AF, Sieminski S, Bernstein AM. Mitochondrial and Microtubule Defects in Exfoliation Glaucoma. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.25.625249. [PMID: 39651300 PMCID: PMC11623661 DOI: 10.1101/2024.11.25.625249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2024]
Abstract
Exfoliation Syndrome (XFS) is an age-related systemic condition characterized by large aggregated fibrillar material deposition in the anterior eye tissues. This aggregate formation and deposition on the aqueous humor outflow pathway are significant risk factors for developing Exfoliation Glaucoma (XFG), a secondary open-angle glaucoma. XFG is a complex, multifactorial late-onset disease that shares common features of neurodegenerative diseases, such as altered cellular processes with increased protein aggregation, impaired protein degradation, and oxidative and cellular stress. XFG patients display decreased mitochondrial membrane potential and mitochondrial DNA deletions. Here, using Tenon Capsule Fibroblasts (TFs) from Normal (No Glaucoma) and XFG patients, we found that XFG TFs have impaired mitochondrial bioenergetics and increased reactive oxygen species (ROS) accumulation. These defects are associated with mitochondrial abnormalities as XFG TFs exhibit smaller mitochondria that contain dysmorphic cristae, with an increase in mitochondrial localization to lysosomes and slowed mitophagy flux. Mitochondrial dysfunction in the XFG TFs was associated with an increase in the dynamics of the microtubule cytoskeleton, decreased acetylated tubulin, and increased HDAC6 activity. Treatment of XFG TFs with a mitophagy inducer, Urolithin A, and a mitochondrial biogenesis inducer, NAD + precursor, Nicotinamide Ribose, improved mitochondrial bioenergetics and reduced ROS accumulation. Our results demonstrate abnormal mitochondria in XFG TFs and suggest that mitophagy inducers may represent a potential class of therapeutics for reversing mitochondrial dysfunction in XFG patients.
Collapse
|
17
|
Luo Y, Zeng X, Dai X, Tian Y, Li J, Zhang Q, Dong Q, Qin L, Huang G, Gu Q, Wang J, Li J. Copper Oxide Nanoparticles Impair Mouse Preimplantation Embryonic Development through Disruption of Mitophagy-Mediated Metabolism. ACS NANO 2024; 18:31244-31260. [PMID: 39487804 PMCID: PMC11562798 DOI: 10.1021/acsnano.4c09734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 10/17/2024] [Accepted: 10/23/2024] [Indexed: 11/04/2024]
Abstract
Copper oxide nanoparticles (CuONPs) have been widely applied, posing potential risks to human health. Although the toxicity of CuONPs on the liver and spleen has been reported, their effects on reproductive health remain unexplored. In this study, we investigate the effects of CuONPs on embryonic development and their potential mechanisms. Our results demonstrate that CuONPs exposure impairs mouse preimplantation embryonic development, particularly affecting the morula-to-blastocyst transition. Additionally, CuONPs were found to reduce the pluripotency of the inner cell mass (ICM) and mouse embryonic stem cells (mESCs). Mechanistically, CuONPs block autophagic flux and impair mitophagy, leading to the accumulation of damaged mitochondria. This mitochondrial dysfunction leads to reduced tricarboxylic acid (TCA) cycle activity and decreased α-ketoglutarate (α-KG) production. Insufficient α-KG induces the failure of DNA demethylation, reducing corresponding chromatin accessibility and consequently inhibiting ICM-specific genes expressions. Similar reduced development and inhibitions of pluripotency gene expression were observed in CuONPs-treated human blastocysts. Moreover, in women undergoing assisted reproductive technology (ART), a negative correlation was found between urinary Cu ion concentrations and clinical outcomes. Collectively, our study elucidates the mitophagy-mediated metabolic mechanisms of CuONPs embryotoxicity, improving our understanding of the potential reproductive toxicity associated with it.
Collapse
Affiliation(s)
- Yunyao Luo
- Chongqing
Key Laboratory of Human Embryo Engineering and Precision Medicine,
Center for Reproductive Medicine, Women
and Children’s Hospital of Chongqing Medical University, Chongqing 400013, China
- Chongqing
Clinical Research Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing 400013, China
| | - Xi Zeng
- Chongqing
Key Laboratory of Human Embryo Engineering and Precision Medicine,
Center for Reproductive Medicine, Women
and Children’s Hospital of Chongqing Medical University, Chongqing 400013, China
- Chongqing
Clinical Research Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing 400013, China
| | - Xue Dai
- Chongqing
Key Laboratory of Human Embryo Engineering and Precision Medicine,
Center for Reproductive Medicine, Women
and Children’s Hospital of Chongqing Medical University, Chongqing 400013, China
- Chongqing
Clinical Research Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing 400013, China
| | - Yin Tian
- Chongqing
Key Laboratory of Human Embryo Engineering and Precision Medicine,
Center for Reproductive Medicine, Women
and Children’s Hospital of Chongqing Medical University, Chongqing 400013, China
- Chongqing
Clinical Research Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing 400013, China
| | - Jie Li
- Institute
of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Qi Zhang
- Chongqing
Key Laboratory of Human Embryo Engineering and Precision Medicine,
Center for Reproductive Medicine, Women
and Children’s Hospital of Chongqing Medical University, Chongqing 400013, China
- Chongqing
Clinical Research Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing 400013, China
| | - Qiang Dong
- Chongqing
Key Laboratory of Human Embryo Engineering and Precision Medicine,
Center for Reproductive Medicine, Women
and Children’s Hospital of Chongqing Medical University, Chongqing 400013, China
- Chongqing
Clinical Research Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing 400013, China
| | - Lifeng Qin
- Chongqing
Key Laboratory of Human Embryo Engineering and Precision Medicine,
Center for Reproductive Medicine, Women
and Children’s Hospital of Chongqing Medical University, Chongqing 400013, China
- Chongqing
Clinical Research Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing 400013, China
| | - Guoning Huang
- Chongqing
Key Laboratory of Human Embryo Engineering and Precision Medicine,
Center for Reproductive Medicine, Women
and Children’s Hospital of Chongqing Medical University, Chongqing 400013, China
- Chongqing
Clinical Research Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing 400013, China
| | - Qi Gu
- Key
Laboratory of Organ Regeneration and Reconstruction, State Key Laboratory
of Membrane Biology, Institute of Zoology,
Chinese Academy of Sciences, Beijing 100864, China
| | - Jianyu Wang
- Institute
of Life Sciences, Chongqing Medical University, Chongqing 400016, China
| | - Jingyu Li
- Chongqing
Key Laboratory of Human Embryo Engineering and Precision Medicine,
Center for Reproductive Medicine, Women
and Children’s Hospital of Chongqing Medical University, Chongqing 400013, China
- Chongqing
Clinical Research Center for Reproductive Medicine, Chongqing Health Center for Women and Children, Chongqing 400013, China
| |
Collapse
|
18
|
Casas-Tintó S. Drosophila as a Model for Human Disease: Insights into Rare and Ultra-Rare Diseases. INSECTS 2024; 15:870. [PMID: 39590469 PMCID: PMC11594678 DOI: 10.3390/insects15110870] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 10/25/2024] [Accepted: 10/30/2024] [Indexed: 11/28/2024]
Abstract
Rare and ultra-rare diseases constitute a significant medical challenge due to their low prevalence and the limited understanding of their origin and underlying mechanisms. These disorders often exhibit phenotypic diversity and molecular complexity that represent a challenge to biomedical research. There are more than 6000 different rare diseases that affect nearly 300 million people worldwide. However, the prevalence of each rare disease is low, and in consequence, the biomedical resources dedicated to each rare disease are limited and insufficient to effectively achieve progress in the research. The use of animal models to investigate the mechanisms underlying pathogenesis has become an invaluable tool. Among the animal models commonly used in research, Drosophila melanogaster has emerged as an efficient and reliable experimental model for investigating a wide range of genetic disorders, and to develop therapeutic strategies for rare and ultra-rare diseases. It offers several advantages as a research model including short life cycle, ease of laboratory maintenance, rapid life cycle, and fully sequenced genome that make it highly suitable for studying genetic disorders. Additionally, there is a high degree of genetic conservation from Drosophila melanogaster to humans, which allows the extrapolation of findings at the molecular and cellular levels. Here, I examine the role of Drosophila melanogaster as a model for studying rare and ultra-rare diseases and highlight its significant contributions and potential to biomedical research. High-throughput next-generation sequencing (NGS) technologies, such as whole-exome sequencing and whole-genome sequencing (WGS), are providing massive amounts of information on the genomic modifications present in rare diseases and common complex traits. The sequencing of exomes or genomes of individuals affected by rare diseases has enabled human geneticists to identify rare variants and identify potential loci associated with novel gene-disease relationships. Despite these advances, the average rare disease patient still experiences significant delay until receiving a diagnosis. Furthermore, the vast majority (95%) of patients with rare conditions lack effective treatment or a cure. This scenario is enhanced by frequent misdiagnoses leading to inadequate support. In consequence, there is an urgent need to develop model organisms to explore the molecular mechanisms underlying these diseases and to establish the genetic origin of these maladies. The aim of this review is to discuss the advantages and limitations of Drosophila melanogaster, hereafter referred as Drosophila, as an experimental model for biomedical research, and the applications to study human disease. The main question to address is whether Drosophila is a valid research model to study human disease, and in particular, rare and ultra-rare diseases.
Collapse
Affiliation(s)
- Sergio Casas-Tintó
- Institute for Rare Diseases Research, Instituto de Salud Carlos III (ISCIII), 28222 Madrid, Spain
| |
Collapse
|
19
|
Kahlon U, Ricca FD, Pillai SJ, Olivetta M, Tharp KM, Jao LE, Dudin O, McDonald K, Aydogan MG. A mitochondrial redox switch licenses the onset of morphogenesis in animals. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.28.620733. [PMID: 39553983 PMCID: PMC11565760 DOI: 10.1101/2024.10.28.620733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Embryos undergo pre-gastrulation cleavage cycles to generate a critical cell mass before transitioning to morphogenesis. The molecular underpinnings of this transition have traditionally centered on zygotic chromatin remodeling and genome activation1,2, as their repression can prevent downstream processes of differentiation and organogenesis. Despite precedents that oxygen depletion can similarly suspend development in early embryos3-6, hinting at a pivotal role for oxygen metabolism in this transition, whether there is a bona fide chemical switch that licenses the onset of morphogenesis remains unknown. Here we discover that a mitochondrial oxidant acts as a metabolic switch to license the onset of animal morphogenesis. Concomitant with the instatement of mitochondrial membrane potential, we found a burst-like accumulation of mitochondrial superoxide (O2 -) during fly blastoderm formation. In vivo chemistry experiments revealed that an electron leak from site IIIQo at ETC Complex III is responsible for O2 - production. Importantly, depleting mitochondrial O2 - fully mimics anoxic conditions and, like anoxia, induces suspended animation prior to morphogenesis, but not after. Specifically, H2O2, and not ONOO-, NO, or HO•, can single-handedly account for this mtROS-based response. We demonstrate that depleting mitochondrial O2 - similarly prevents the onset of morphogenetic events in vertebrate embryos and ichthyosporea, close relatives of animals. We postulate that such redox-based metabolic licensing of morphogenesis is an ancient trait of holozoans that couples the availability of oxygen to development, conserved from early-diverging animal relatives to vertebrates.
Collapse
Affiliation(s)
- Updip Kahlon
- Department of Biochemistry and Biophysics, University of California, San Francisco, USA
- Touro College of Osteopathic Medicine, Touro University, USA
- These authors have contributed equally
| | - Francesco Dalla Ricca
- Department of Biochemistry and Biophysics, University of California, San Francisco, USA
- Dev. & Stem Cell Biology Graduate Program, University of California, San Francisco, USA
- These authors have contributed equally
| | - Saraswathi J. Pillai
- Department of Biochemistry and Biophysics, University of California, San Francisco, USA
- These authors have contributed equally
| | - Marine Olivetta
- Department of Biochemistry, Faculty of Sciences, University of Geneva, Switzerland
| | - Kevin M. Tharp
- Sanford Burnham Prebys Medical Discovery Institute, San Diego, USA
| | - Li-En Jao
- Department of Cell Biology and Human Anatomy, University of California, Davis, USA
| | - Omaya Dudin
- Department of Biochemistry, Faculty of Sciences, University of Geneva, Switzerland
| | - Kent McDonald
- Electron Microscope Lab, University of California, Berkeley, USA
| | - Mustafa G. Aydogan
- Department of Biochemistry and Biophysics, University of California, San Francisco, USA
- Nutrition and Obesity Research Center, University of California, San Francisco, USA
| |
Collapse
|
20
|
Zhang H, Zhu Y, Xue D. Moderate embryonic delay of paternal mitochondrial elimination impairs mating and cognition and alters behaviors of adult animals. SCIENCE ADVANCES 2024; 10:eadp8351. [PMID: 39365857 PMCID: PMC11451536 DOI: 10.1126/sciadv.adp8351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 08/30/2024] [Indexed: 10/06/2024]
Abstract
Rapid elimination of paternal mitochondria following fertilization is a conserved event in most animals, but its physiological significance remains unclear. We find that modest delay of paternal mitochondrial elimination (PME) in Caenorhabditis elegans embryos unexpectedly impairs mating and cognition of adult animals and alters their locomotion behaviors. Delayed PME causes decreased adenosine triphosphate (ATP) levels in early embryos, which lead to impaired physiological functions of adult animals through an energy-sensing pathway mediated by an adenosine monophosphate (AMP)-activated protein kinase, AAK-2, and a forkhead box class O (FOXO) transcription factor, DAF-16. Treatment of PME-delayed animals with MK-4, a subtype of vitamin K2 that can improve mitochondrial ATP production, restores ATP levels in early embryos, and rescues physiological defects of adult animals. Our results suggest that moderate PME delay during embryo development adversely affects crucial physiological functions in adults, which could be evolutionarily disadvantageous. These observations provide mechanistic explanations for the need to swiftly remove paternal mitochondria early during embryo development.
Collapse
Affiliation(s)
| | | | - Ding Xue
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, CO 80309, USA
| |
Collapse
|
21
|
Hushmandi K, Einollahi B, Aow R, Suhairi SB, Klionsky DJ, Aref AR, Reiter RJ, Makvandi P, Rabiee N, Xu Y, Nabavi N, Saadat SH, Farahani N, Kumar AP. Investigating the interplay between mitophagy and diabetic neuropathy: Uncovering the hidden secrets of the disease pathology. Pharmacol Res 2024; 208:107394. [PMID: 39233055 PMCID: PMC11934918 DOI: 10.1016/j.phrs.2024.107394] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2024] [Revised: 08/18/2024] [Accepted: 08/30/2024] [Indexed: 09/06/2024]
Abstract
Mitophagy, the cellular process of selectively eliminating damaged mitochondria, plays a crucial role in maintaining metabolic balance and preventing insulin resistance, both key factors in type 2 diabetes mellitus (T2DM) development. When mitophagy malfunctions in diabetic neuropathy, it triggers a cascade of metabolic disruptions, including reduced energy production, increased oxidative stress, and cell death, ultimately leading to various complications. Thus, targeting mitophagy to enhance the process may have emerged as a promising therapeutic strategy for T2DM and its complications. Notably, plant-derived compounds with β-cell protective and mitophagy-stimulating properties offer potential as novel therapeutic agents. This review highlights the intricate mechanisms linking mitophagy dysfunction to T2DM and its complications, particularly neuropathy, elucidating potential therapeutic interventions for this debilitating disease.
Collapse
Affiliation(s)
- Kiavash Hushmandi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran.
| | - Behzad Einollahi
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Rachel Aow
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Suhana Binte Suhairi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Daniel J Klionsky
- Life Sciences Institute, University of Michigan, Ann Arbor, MI 48109, USA
| | - Amir Reza Aref
- Department of Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Russel J Reiter
- Department of Cell Systems and Anatomy, UT Health San Antonio, Long School of Medicine, San Antonio, TX, USA
| | - Pooyan Makvandi
- Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai 600077, India; University Centre for Research & Development, Chandigarh University, Mohali, Punjab 140413, India
| | - Navid Rabiee
- Department of Biomaterials, Saveetha Dental College and Hospitals, SIMATS, Saveetha University, Chennai 600077, India
| | - Yi Xu
- Department of Science & Technology, Department of Urology, NanoBioMed Group, The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou 324000, China
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia V8V 1P7, Canada
| | - Seyed Hassan Saadat
- Nephrology and Urology Research Center, Clinical Sciences Institute, Baqiyatallah University of Medical Sciences, Tehran, Iran
| | - Najma Farahani
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore; NUS Center for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
22
|
Masahiro Hirai, Suzuki K, Kassai Y, Konishi Y. 3-Nitrotyrosine shortens axons of non-dopaminergic neurons by inhibiting mitochondrial motility. Neurochem Int 2024; 179:105832. [PMID: 39154836 DOI: 10.1016/j.neuint.2024.105832] [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: 05/09/2024] [Revised: 07/30/2024] [Accepted: 08/08/2024] [Indexed: 08/20/2024]
Abstract
3-Nitrotyrosine (3-NT), a byproduct of oxidative and nitrosative stress, is implicated in age-related neurodegenerative disorders. Current literature suggests that free 3-NT becomes integrated into the carboxy-terminal domain of α-tubulin via the tyrosination/detyrosination cycle. Independently of this integration, 3-NT has been associated with the cell death of dopaminergic neurons. Given the critical role of tyrosination/detyrosination in governing axonal morphology and function, the substitution of tyrosine with 3-NT in this process may potentially disrupt axonal homeostasis, although this aspect remains underexplored. In this study, we examined the impact of 3-NT on the axons of cerebellar granule neurons, which is used as a model for non-dopaminergic neurons. Our observations revealed axonal shortening, which correlated with the incorporation of 3-NT into α-tubulin. Importantly, this axonal effect was observed prior to the onset of cellular death. Furthermore, 3-NT was found to diminish mitochondrial motility within the axon, leading to a subsequent reduction in mitochondrial membrane potential. The suppression of syntaphilin, a protein responsible for anchoring mitochondria to microtubules, restored the mitochondrial motility and axonal elongation that were inhibited by 3-NT. These findings underscore the inhibitory role of 3-NT in axonal elongation by impeding mitochondrial movement, suggesting its potential involvement in axonal dysfunction within non-dopaminergic neurons.
Collapse
Affiliation(s)
- Masahiro Hirai
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, University of Fukui, Fukui, Japan
| | - Kohei Suzuki
- Department of Industrial Innovation Engineering, Graduate School of Engineering, University of Fukui, Fukui, Japan
| | - Yusuke Kassai
- Department of Human and Artificial Intelligence Systems, Faculty of Engineering, University of Fukui, Fukui, Japan
| | - Yoshiyuki Konishi
- Department of Applied Chemistry and Biotechnology, Graduate School of Engineering, University of Fukui, Fukui, Japan; Department of Industrial Innovation Engineering, Graduate School of Engineering, University of Fukui, Fukui, Japan; Department of Human and Artificial Intelligence Systems, Faculty of Engineering, University of Fukui, Fukui, Japan; Department of Applied Chemistry and Biotechnology, Faculty of Engineering, University of Fukui, Fukui, Japan; Life Science Innovation Center, University of Fukui, Fukui, Japan.
| |
Collapse
|
23
|
Brogyanyi T, Kejík Z, Veselá K, Dytrych P, Hoskovec D, Masařik M, Babula P, Kaplánek R, Přibyl T, Zelenka J, Ruml T, Vokurka M, Martásek P, Jakubek M. Iron chelators as mitophagy agents: Potential and limitations. Biomed Pharmacother 2024; 179:117407. [PMID: 39265234 DOI: 10.1016/j.biopha.2024.117407] [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: 06/14/2024] [Revised: 08/26/2024] [Accepted: 09/02/2024] [Indexed: 09/14/2024] Open
Abstract
Mitochondrial autophagy (mitophagy) is very important process for the maintenance of cellular homeostasis, functionality and survival. Its dysregulation is associated with high risk and progression numerous serious diseases (e.g., oncological, neurodegenerative and cardiovascular ones). Therefore, targeting mitophagy mechanisms is very hot topic in the biological and medicinal research. The interrelationships between the regulation of mitophagy and iron homeostasis are now becoming apparent. In short, mitochondria are central point for the regulation of iron homeostasis, but change in intracellular cheatable iron level can induce/repress mitophagy. In this review, relationships between iron homeostasis and mitophagy are thoroughly discussed and described. Also, therapeutic applicability of mitophagy chelators in the context of individual diseases is comprehensively and critically evaluated.
Collapse
Affiliation(s)
- Tereza Brogyanyi
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic; Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague, U Nemocnice 5, 1, Prague 28 53, Czech Republic
| | - Zdeněk Kejík
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Kateřina Veselá
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Petr Dytrych
- 1st Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital, U Nemocnice 2, Prague 121 08, Czech Republic
| | - David Hoskovec
- 1st Department of Surgery-Department of Abdominal, Thoracic Surgery and Traumatology, First Faculty of Medicine, Charles University and General University Hospital, U Nemocnice 2, Prague 121 08, Czech Republic
| | - Michal Masařik
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic; Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno CZ-625 00, Czech Republic; Department of Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno 625 00, Czech Republic
| | - Petr Babula
- Department of Pathological Physiology, Faculty of Medicine, Masaryk University, Kamenice 5, Brno CZ-625 00, Czech Republic
| | - Robert Kaplánek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Tomáš Přibyl
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Prague 166 28, Czech Republic
| | - Jaroslav Zelenka
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Prague 166 28, Czech Republic
| | - Tomáš Ruml
- Department of Biochemistry and Microbiology, University of Chemistry and Technology, Prague, Prague 166 28, Czech Republic
| | - Martin Vokurka
- Institute of Pathological Physiology, First Faculty of Medicine, Charles University in Prague, U Nemocnice 5, 1, Prague 28 53, Czech Republic
| | - Pavel Martásek
- Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic
| | - Milan Jakubek
- BIOCEV, First Faculty of Medicine, Charles University, Vestec 252 50, Czech Republic; Department of Paediatrics and Inherited Metabolic Disorders, First Faculty of Medicine, Charles University and General University Hospital, Prague 120 00, Czech Republic.
| |
Collapse
|
24
|
Wu N, Zheng W, Zhou Y, Tian Y, Tang M, Feng X, Ashrafizadeh M, Wang Y, Niu X, Tambuwala M, Wang L, Tergaonkar V, Sethi G, Klionsky D, Huang L, Gu M. Autophagy in aging-related diseases and cancer: Principles, regulatory mechanisms and therapeutic potential. Ageing Res Rev 2024; 100:102428. [PMID: 39038742 DOI: 10.1016/j.arr.2024.102428] [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: 05/18/2024] [Revised: 07/05/2024] [Accepted: 07/15/2024] [Indexed: 07/24/2024]
Abstract
Macroautophagy/autophagy is primarily accountable for the degradation of damaged organelles and toxic macromolecules in the cells. Regarding the essential function of autophagy for preserving cellular homeostasis, changes in, or dysfunction of, autophagy flux can lead to disease development. In the current paper, the complicated function of autophagy in aging-associated pathologies and cancer is evaluated, highlighting the underlying molecular mechanisms that can affect longevity and disease pathogenesis. As a natural biological process, a reduction in autophagy is observed with aging, resulting in an accumulation of cell damage and the development of different diseases, including neurological disorders, cardiovascular diseases, and cancer. The MTOR, AMPK, and ATG proteins demonstrate changes during aging, and they are promising therapeutic targets. Insulin/IGF1, TOR, PKA, AKT/PKB, caloric restriction and mitochondrial respiration are vital for lifespan regulation and can modulate or have an interaction with autophagy. The specific types of autophagy, such as mitophagy that degrades mitochondria, can regulate aging by affecting these organelles and eliminating those mitochondria with genomic mutations. Autophagy and its specific types contribute to the regulation of carcinogenesis and they are able to dually enhance or decrease cancer progression. Cancer hallmarks, including proliferation, metastasis, therapy resistance and immune reactions, are tightly regulated by autophagy, supporting the conclusion that autophagy is a promising target in cancer therapy.
Collapse
Affiliation(s)
- Na Wu
- Department of Infectious Diseases, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Wenhui Zheng
- Department of Anesthesiology, The Shengjing Hospital of China Medical University, Shenyang, Liaoning 110001, China
| | - Yundong Zhou
- Department of Thoracic Surgery, Ningbo Medical Center Lihuili Hospital, Ningbo University, Ningbo, Zhejiang 315040, China
| | - Yu Tian
- School of Public Health, Benedictine University, No.5700 College Road, Lisle, IL 60532, USA; Research Center, the Huizhou Central People's Hospital, Guangdong Medical University, Huizhou, Guangdong, China
| | - Min Tang
- Department of Oncology, Chongqing General Hospital, Chongqing University, Chongqing 401120, China
| | - Xiaoqiang Feng
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou, Guangdong 525200, China
| | - Milad Ashrafizadeh
- Department of Radiation Oncology, Shandong Provincial Key Laboratory of Radiation Oncology, Shandong Cancer Hospital and Institute, Shandong First Medical University, Shandong Academy of Medical Sciences, Jinan, Shandong 250000, China; Shanghai Institute of Cardiovascular Diseases, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yuzhuo Wang
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H3Z6, Canada
| | - Xiaojia Niu
- Department of Urologic Sciences and Vancouver Prostate Centre, University of British Columbia, Vancouver, BC V6H3Z6, Canada
| | - Murtaza Tambuwala
- Lincoln Medical School, University of Lincoln, Brayford Pool Campus, Lincoln LN6 7TS, UK
| | - Lingzhi Wang
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117600, Singapore
| | - Vinay Tergaonkar
- Laboratory of NF-κB Signalling, Institute of Molecular and Cell Biology (IMCB), Agency for Science, Technology and Research (A⁎STAR), 61 Biopolis Drive, Proteos, Singapore 138673, Singapore
| | - Gautam Sethi
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, 16 Medical Drive, Singapore 117600, Singapore; NUS Centre for Cancer Research (N2CR), Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore.
| | - Daniel Klionsky
- Life Sciences Institute and Department of Molecular, Cellular and Developmental Biology, University of Michigan, Ann Arbor, MI, USA.
| | - Li Huang
- Center of Stem Cell and Regenerative Medicine, Gaozhou People's Hospital, Gaozhou, Guangdong 525200, China.
| | - Ming Gu
- Department of Breast Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| |
Collapse
|
25
|
Novelli G, Calcaterra G, Casciani F, Pecorelli S, Mehta JL. 'Exerkines': A Comprehensive Term for the Factors Produced in Response to Exercise. Biomedicines 2024; 12:1975. [PMID: 39335489 PMCID: PMC11429193 DOI: 10.3390/biomedicines12091975] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2024] [Revised: 08/23/2024] [Accepted: 08/27/2024] [Indexed: 09/30/2024] Open
Abstract
Regular exercise and physical activity are now considered lifestyle factors with positive effects on human health. Physical activity reduces disease burden, protects against the onset of pathologies, and improves the clinical course of disease. Unlike pharmacological therapies, the effects mediated by exercise are not limited to a specific target organ but act in multiple biological systems simultaneously. Despite the substantial health benefits of physical training, the precise molecular signaling processes that lead to structural and functional tissue adaptation remain largely unknown. Only recently, several bioactive molecules have been discovered that are produced following physical exercise. These molecules are collectively called "exerkines". Exerkines are released from various tissues in response to exercise, and play a crucial role in mediating the beneficial effects of exercise on the body. Major discoveries involving exerkines highlight their diverse functions and health implications, particularly in metabolic regulation, neuroprotection, and muscle adaptation. These molecules, including peptides, nucleic acids, lipids, and microRNAs, act through paracrine, endocrine, and autocrine pathways to exert their effects on various organs and tissues. Exerkines represent a complex network of signaling molecules that mediate the multiple benefits of exercise. Their roles in metabolic regulation, neuroprotection, and muscle adaptation highlight the importance of physical activity in maintaining health and preventing disease.
Collapse
Affiliation(s)
- Giuseppe Novelli
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00173 Rome, Italy
- Giovanni Lorenzini Medical Foundation, 20129 Milan, Italy
- Giovanni Lorenzini Medical Foundation New York, Woodcliff Lake, NJ 07677, USA
- Italian Federation of Sports Medicine, 00196 Rome, Italy
| | - Giuseppe Calcaterra
- Postgraduate Medical School of Cardiology, University of Palermo, 90127 Palermo, Italy
| | - Federico Casciani
- Department of Biomedicine and Prevention, Tor Vergata University of Rome, 00173 Rome, Italy
| | - Sergio Pecorelli
- Giovanni Lorenzini Medical Foundation, 20129 Milan, Italy
- Giovanni Lorenzini Medical Foundation New York, Woodcliff Lake, NJ 07677, USA
- Italian Federation of Sports Medicine, 00196 Rome, Italy
- School of Medicine, University of Brescia, 25123 Brescia, Italy
| | - Jawahar L Mehta
- Giovanni Lorenzini Medical Foundation, 20129 Milan, Italy
- Giovanni Lorenzini Medical Foundation New York, Woodcliff Lake, NJ 07677, USA
- Department of Medicine (Cardiology), University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| |
Collapse
|
26
|
Wong HTC, Lang AE, Stein C, Drerup CM. ALS-Linked VapB P56S Mutation Alters Neuronal Mitochondrial Turnover at the Synapse. J Neurosci 2024; 44:e0879242024. [PMID: 39054069 PMCID: PMC11358610 DOI: 10.1523/jneurosci.0879-24.2024] [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: 05/09/2024] [Revised: 07/11/2024] [Accepted: 07/19/2024] [Indexed: 07/27/2024] Open
Abstract
Mitochondrial population maintenance in neurons is essential for neuron function and survival. Contact sites between mitochondria and the endoplasmic reticulum (ER) are poised to regulate mitochondrial homeostasis in neurons. These contact sites can facilitate transfer of calcium and lipids between the organelles and have been shown to regulate aspects of mitochondrial dynamics. Vesicle-associated membrane protein-associated protein B (VapB) is an ER membrane protein present at a subset of ER-mitochondrial contact sites. A proline-to-serine mutation in VapB at amino acid 56 (P56S) correlates with susceptibility to amyotrophic lateral sclerosis (ALS) type 8. Given the relationship between failed mitochondrial health and neurodegenerative disease, we investigated the function of VapB in mitochondrial population maintenance. We demonstrated that transgenic expression of VapBP56S in zebrafish larvae (sex undetermined) increased mitochondrial biogenesis, causing increased mitochondrial population size in the axon terminal. Expression of wild-type VapB did not alter biogenesis but, instead, increased mitophagy in the axon terminal. Using genetic manipulations to independently increase mitochondrial biogenesis, we show that biogenesis is normally balanced by mitophagy to maintain a constant mitochondrial population size. VapBP56S transgenics fail to increase mitophagy to compensate for the increase in mitochondrial biogenesis, suggesting an impaired mitophagic response. Finally, using a synthetic ER-mitochondrial tether, we show that VapB's function in mitochondrial turnover is likely independent of ER-mitochondrial tethering by contact sites. Our findings demonstrate that VapB can control mitochondrial turnover in the axon terminal, and this function is altered by the P56S ALS-linked mutation.
Collapse
Affiliation(s)
- Hiu-Tung C Wong
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - Angelica E Lang
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53706
- Genetics Training Program, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - Chris Stein
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53706
| | - Catherine M Drerup
- Department of Integrative Biology, University of Wisconsin-Madison, Madison, Wisconsin 53706
- Genetics Training Program, University of Wisconsin-Madison, Madison, Wisconsin 53706
| |
Collapse
|
27
|
Read TA, Cisterna BA, Skruber K, Ahmadieh S, Liu TM, Vitriol JA, Shi Y, Black JB, Butler MT, Lindamood HL, Lefebvre AE, Cherezova A, Ilatovskaya DV, Bear JE, Weintraub NL, Vitriol EA. The actin binding protein profilin 1 localizes inside mitochondria and is critical for their function. EMBO Rep 2024; 25:3240-3262. [PMID: 39026010 PMCID: PMC11316047 DOI: 10.1038/s44319-024-00209-3] [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: 08/17/2023] [Revised: 06/16/2024] [Accepted: 06/28/2024] [Indexed: 07/20/2024] Open
Abstract
The monomer-binding protein profilin 1 (PFN1) plays a crucial role in actin polymerization. However, mutations in PFN1 are also linked to hereditary amyotrophic lateral sclerosis, resulting in a broad range of cellular pathologies which cannot be explained by its primary function as a cytosolic actin assembly factor. This implies that there are important, undiscovered roles for PFN1 in cellular physiology. Here we screened knockout cells for novel phenotypes associated with PFN1 loss of function and discovered that mitophagy was significantly upregulated. Indeed, despite successful autophagosome formation, fusion with the lysosome, and activation of additional mitochondrial quality control pathways, PFN1 knockout cells accumulate depolarized, dysmorphic mitochondria with altered metabolic properties. Surprisingly, we also discovered that PFN1 is present inside mitochondria and provide evidence that mitochondrial defects associated with PFN1 loss are not caused by reduced actin polymerization in the cytosol. These findings suggest a previously unrecognized role for PFN1 in maintaining mitochondrial integrity and highlight new pathogenic mechanisms that can result from PFN1 dysregulation.
Collapse
Affiliation(s)
- Tracy-Ann Read
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| | - Bruno A Cisterna
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Kristen Skruber
- Department of Cellular and Molecular Pharmacology, University of California San Francisco, San Francisco, CA, USA
| | - Samah Ahmadieh
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Tatiana M Liu
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Josefine A Vitriol
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Yang Shi
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
- Department of Population Health Sciences, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Joseph B Black
- Division of Urologic Surgery, Beth Israel Deaconess Medical Center, Boston, MA, USA
| | - Mitchell T Butler
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Halli L Lindamood
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | | | - Alena Cherezova
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Daria V Ilatovskaya
- Department of Physiology, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - James E Bear
- Department of Cell Biology and Physiology, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
- Lineberger Comprehensive Cancer Center, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, NC, USA
| | - Neal L Weintraub
- Vascular Biology Center, Medical College of Georgia, Augusta University, Augusta, GA, USA
- Department of Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA
| | - Eric A Vitriol
- Department of Neuroscience and Regenerative Medicine, Medical College of Georgia at Augusta University, Augusta, GA, USA.
| |
Collapse
|
28
|
Restrepo LJ, Baehrecke EH. Regulation and Functions of Autophagy During Animal Development. J Mol Biol 2024; 436:168473. [PMID: 38311234 PMCID: PMC11260256 DOI: 10.1016/j.jmb.2024.168473] [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: 12/12/2023] [Revised: 01/29/2024] [Accepted: 01/30/2024] [Indexed: 02/10/2024]
Abstract
Autophagy is used to degrade cytoplasmic materials, and is critical to maintain cell and organismal health in diverse animals. Here we discuss the regulation, utilization and impact of autophagy on development, including roles in oogenesis, spermatogenesis and embryogenesis in animals. We also describe how autophagy influences postembryonic development in the context of neuronal and cardiac development, wound healing, and tissue regeneration. We describe recent studies of selective autophagy during development, including mitochondria-selective autophagy and endoplasmic reticulum (ER)-selective autophagy. Studies of developing model systems have also been used to discover novel regulators of autophagy, and we explain how studies of autophagy in these physiologically relevant systems are advancing our understanding of this important catabolic process.
Collapse
Affiliation(s)
- Lucas J Restrepo
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605 USA
| | - Eric H Baehrecke
- Department of Molecular, Cell and Cancer Biology, University of Massachusetts Chan Medical School, Worcester, MA 01605 USA.
| |
Collapse
|
29
|
Xie X, Li J, Zhang X, Mo S, Li A, Sun TY, Xie FY, Luo SM, Wang G, Ou XH, Sun QY, Zhou Q. Endonuclease G is dispensable for sperm mitochondrial DNA elimination during spermatogenesis in mice. Biol Open 2024; 13:bio061730. [PMID: 39373150 PMCID: PMC11554256 DOI: 10.1242/bio.061730] [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: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 10/08/2024] Open
Abstract
Maternal inheritance of mitochondrial DNA (mtDNA) is a widespread phenomenon in eukaryotes. Our earlier research indicated that sperm mtDNA is removed prior to fertilization in mice, and Endonuclease G (ENDOG) orchestrates the degradation of sperm mitochondria in Caenorhabditis elegans. However, the mechanisms underlying sperm mtDNA disposal in mammals remain poorly understood. To investigate the potential role of ENDOG in sperm mtDNA elimination, we created Endog knockout (Endog-/-) mice. Our findings revealed that Endog-/- mice maintained normal spermatogenesis and fertility. Most strikingly, we detected no substantial discrepancy in sperm mtDNA copy number between Endog-/- and control mice. Furthermore, we noted that sperm mtDNA copy numbers were unchanged in both less motile and motile sperm isolated by Percoll gradient centrifugation from Endog-/- and control mice. Taken together, our results indicate that ENDOG is not essential for spermatogenesis or the elimination of sperm mtDNA in mice.
Collapse
Affiliation(s)
- Xuefeng Xie
- Guangdong Second Provincial General Hospital,Postdoctoral Research Station of Basic Medicine, School of Medicine,Jinan University, Guangzhou, 510317, Guangdong, China
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China
| | - Jianshuang Li
- Department of Orthopaedics, Guangzhou Red Cross Hospital, Faculty of Medical Science, Jinan University, Guangzhou, 510220, Guangdong, China
- The College of Life Science and Technology,Jinan University, Guangzhou, 510632, Guangdong, China
| | - Xue Zhang
- Guangdong Second Provincial General Hospital,Postdoctoral Research Station of Basic Medicine, School of Medicine,Jinan University, Guangzhou, 510317, Guangdong, China
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China
| | - Shaomei Mo
- Guangdong Second Provincial General Hospital,Postdoctoral Research Station of Basic Medicine, School of Medicine,Jinan University, Guangzhou, 510317, Guangdong, China
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China
| | - Ang Li
- Guangdong Second Provincial General Hospital,Postdoctoral Research Station of Basic Medicine, School of Medicine,Jinan University, Guangzhou, 510317, Guangdong, China
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China
| | - Tian-Yi Sun
- Guangdong Second Provincial General Hospital,Postdoctoral Research Station of Basic Medicine, School of Medicine,Jinan University, Guangzhou, 510317, Guangdong, China
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China
| | - Feng-Yun Xie
- Guangdong Second Provincial General Hospital,Postdoctoral Research Station of Basic Medicine, School of Medicine,Jinan University, Guangzhou, 510317, Guangdong, China
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China
| | - Shi-Ming Luo
- Guangdong Second Provincial General Hospital,Postdoctoral Research Station of Basic Medicine, School of Medicine,Jinan University, Guangzhou, 510317, Guangdong, China
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China
| | - Guang Wang
- Guangdong Second Provincial General Hospital,Postdoctoral Research Station of Basic Medicine, School of Medicine,Jinan University, Guangzhou, 510317, Guangdong, China
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China
- International Joint Laboratory for Embryonic Development & Prenatal Medicine, Division of Histology and Embryology, Medical College, Jinan University, Guangzhou, 510632, Guangdong, China
| | - Xiang-Hong Ou
- Guangdong Second Provincial General Hospital,Postdoctoral Research Station of Basic Medicine, School of Medicine,Jinan University, Guangzhou, 510317, Guangdong, China
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China
| | - Qing-Yuan Sun
- Guangdong Second Provincial General Hospital,Postdoctoral Research Station of Basic Medicine, School of Medicine,Jinan University, Guangzhou, 510317, Guangdong, China
- Guangzhou Key Laboratory of Metabolic Diseases and Reproductive Health, Guangdong-Hong Kong Metabolism & Reproduction Joint Laboratory, Reproductive Medicine Center, Guangdong Second Provincial General Hospital, Guangzhou, 510317, Guangdong, China
| | - Qinghua Zhou
- Department of Orthopaedics, Guangzhou Red Cross Hospital, Faculty of Medical Science, Jinan University, Guangzhou, 510220, Guangdong, China
- The College of Life Science and Technology,Jinan University, Guangzhou, 510632, Guangdong, China
| |
Collapse
|
30
|
Nakai A, Fukushima Y, Yamamoto A, Amatsu Y, Chen X, Nishigori M, Yoshioka Y, Kaneko M, Koshiba T, Watanabe T. Increased ROS levels in mitochondrial outer membrane protein Mul1-deficient oocytes result in abnormal preimplantation embryogenesis. FEBS Lett 2024; 598:1740-1752. [PMID: 38639871 DOI: 10.1002/1873-3468.14876] [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: 11/03/2023] [Revised: 02/15/2024] [Accepted: 03/22/2024] [Indexed: 04/20/2024]
Abstract
Reactive oxygen species (ROS) are associated with oocyte maturation inhibition, and N-acetyl-l-cysteine (NAC) partially reduces their harmful effects. Mitochondrial E3 ubiquitin ligase 1 (Mul1) localizes to the mitochondrial outer membrane. We found that female Mul1-deficient mice are infertile, and their oocytes contain high ROS concentrations. After fertilization, Mul1-deficient embryos showed a DNA damage response (DDR) and abnormal preimplantation embryogenesis, which was rescued by NAC addition and ROS depletion. These observations clearly demonstrate that loss of Mul1 in oocytes increases ROS concentrations and triggers DDR, resulting in abnormal preimplantation embryogenesis. We conclude that manipulating the mitochondrial ROS levels in oocytes may be a potential therapeutic approach to target infertility.
Collapse
Affiliation(s)
- Ann Nakai
- Department of Biological Science, Graduate School of Humanities and Sciences, Nara Women's University, Japan
| | - Yuki Fukushima
- Department of Biological Science, Graduate School of Humanities and Sciences, Nara Women's University, Japan
| | - Ayaka Yamamoto
- Department of Biological Science, Graduate School of Humanities and Sciences, Nara Women's University, Japan
| | - Yuki Amatsu
- Department of Biological Science, Graduate School of Humanities and Sciences, Nara Women's University, Japan
| | - Xiaoyan Chen
- Department of Biological Science, Graduate School of Humanities and Sciences, Nara Women's University, Japan
| | | | - Yukino Yoshioka
- Department of Biological Science, Graduate School of Humanities and Sciences, Nara Women's University, Japan
| | - Mari Kaneko
- Laboratory for Animal Resources and Genetic Engineering, RIKEN Center for Biosystems Dynamics Research, Kobe, Japan
| | - Takumi Koshiba
- Department of Chemistry, Faculty of Science, Fukuoka University, Japan
| | - Toshio Watanabe
- Department of Biological Science, Graduate School of Humanities and Sciences, Nara Women's University, Japan
| |
Collapse
|
31
|
Hu Y, Zhang L, Tian C, Chen F, Li P, Zhang A, Wang W. Molecular crosstalk and putative mechanisms underlying mitochondrial quality control: The hidden link with methylmercury-induced cognitive impairment. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 278:116360. [PMID: 38678690 DOI: 10.1016/j.ecoenv.2024.116360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/04/2024] [Accepted: 04/19/2024] [Indexed: 05/01/2024]
Abstract
Methylmercury (MeHg) is a neurotoxin associated with foetal neurodevelopmental and adult cognitive deficits. Neurons are highly dependent on the tricarboxylic acid cycle and oxidative phosphorylation to produce ATP and meet their high energy demands. Therefore, mitochondrial quality control (MQC) is critical for neuronal homeostasis. While existing studies have generated a wealth of data on the toxicity of MeHg, the complex cascades and molecular pathways governing the mitochondrial network remain to be elucidated. Here, 0.6, 1.2 and 2.4 mg/kg body weight of MeHg were administered intragastrically to pregnant Sprague Dawley rats to model maternal MeHg exposure. The results of the in vivo study revealed that MeHg-treated rats tended to perform more directionless repetitive strategies in the Morris Water Maze and fewer target-orientation strategies than control offspring. Moreover, pathological injury and synaptic toxicity were observed in the hippocampus. Transmission electron microscopy (TEM) demonstrated that the autophagosomes encapsulated damaged mitochondria, while showing a typical mitochondrial fission phenotype, which was supported by the activation of PINK1-dependent key regulators of mitophagy. Moreover, there was upregulation of DRP1 and FIS1. Additionally, MeHg compensation promoted mitochondrial biogenesis, as evidenced by the activation of the mitochondrial PGC1-α-NRF1-TFAM signalling pathway. Notably, SIRT3/AMPK was activated by MeHg, and the expression and activity of p-AMPK, p-LKB1 and SIRT3 were consistently coordinated. Collectively, these findings provide new insights into the potential molecular mechanisms regulating MeHg-induced cognitive deficits through SIRT3/AMPK MQC network coordination.
Collapse
Affiliation(s)
- Yi Hu
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, China
| | - Li Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, China
| | - Changsong Tian
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, China
| | - Fang Chen
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, China
| | - Ping Li
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, China; State Key Laboratory of Environmental Geochemistry, Institute of Geochemistry, Chinese Academy of Sciences, Guiyang 550081, China
| | - Aihua Zhang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, China; Collaborative Innovation Centre for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang 550025, China.
| | - Wenjuan Wang
- The Key Laboratory of Environmental Pollution Monitoring and Disease Control, Ministry of Education, School of Public Health, Guizhou Medical University, Guiyang 550025, China; Collaborative Innovation Centre for Prevention and Control of Endemic and Ethnic Regional Diseases Co-constructed by the Province and Ministry, Guizhou Medical University, Guiyang 550025, China.
| |
Collapse
|
32
|
Yildirim RM, Seli E. The role of mitochondrial dynamics in oocyte and early embryo development. Semin Cell Dev Biol 2024; 159-160:52-61. [PMID: 38330625 DOI: 10.1016/j.semcdb.2024.01.007] [Citation(s) in RCA: 31] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Revised: 01/09/2024] [Accepted: 01/25/2024] [Indexed: 02/10/2024]
Abstract
Mitochondrial dysfunction is widely implicated in various human diseases, through mechanisms that go beyond mitochondria's well-established role in energy generation. These dynamic organelles exert vital control over numerous cellular processes, including calcium regulation, phospholipid synthesis, innate immunity, and apoptosis. While mitochondria's importance is acknowledged in all cell types, research has revealed the exceptionally dynamic nature of the mitochondrial network in oocytes and embryos, finely tuned to meet unique needs during gamete and pre-implantation embryo development. Within oocytes, both the quantity and morphology of mitochondria can significantly change during maturation and post-fertilization. These changes are orchestrated by fusion and fission processes (collectively known as mitochondrial dynamics), crucial for energy production, content exchange, and quality control as mitochondria adjust to the shifting energy demands of oocytes and embryos. The roles of proteins that regulate mitochondrial dynamics in reproductive processes have been primarily elucidated through targeted deletion studies in animal models. Notably, impaired mitochondrial dynamics have been linked to female reproductive health, affecting oocyte quality, fertilization, and embryo development. Dysfunctional mitochondria can lead to fertility problems and can have an impact on the success of pregnancy, particularly in older reproductive age women.
Collapse
Affiliation(s)
- Raziye Melike Yildirim
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
| | - Emre Seli
- Department of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA.
| |
Collapse
|
33
|
Tomar A, Gomez-Velazquez M, Gerlini R, Comas-Armangué G, Makharadze L, Kolbe T, Boersma A, Dahlhoff M, Burgstaller JP, Lassi M, Darr J, Toppari J, Virtanen H, Kühnapfel A, Scholz M, Landgraf K, Kiess W, Vogel M, Gailus-Durner V, Fuchs H, Marschall S, Hrabě de Angelis M, Kotaja N, Körner A, Teperino R. Epigenetic inheritance of diet-induced and sperm-borne mitochondrial RNAs. Nature 2024; 630:720-727. [PMID: 38839949 PMCID: PMC11186758 DOI: 10.1038/s41586-024-07472-3] [Citation(s) in RCA: 55] [Impact Index Per Article: 55.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2023] [Accepted: 04/26/2024] [Indexed: 06/07/2024]
Abstract
Spermatozoa harbour a complex and environment-sensitive pool of small non-coding RNAs (sncRNAs)1, which influences offspring development and adult phenotypes1-7. Whether spermatozoa in the epididymis are directly susceptible to environmental cues is not fully understood8. Here we used two distinct paradigms of preconception acute high-fat diet to dissect epididymal versus testicular contributions to the sperm sncRNA pool and offspring health. We show that epididymal spermatozoa, but not developing germ cells, are sensitive to the environment and identify mitochondrial tRNAs (mt-tRNAs) and their fragments (mt-tsRNAs) as sperm-borne factors. In humans, mt-tsRNAs in spermatozoa correlate with body mass index, and paternal overweight at conception doubles offspring obesity risk and compromises metabolic health. Sperm sncRNA sequencing of mice mutant for genes involved in mitochondrial function, and metabolic phenotyping of their wild-type offspring, suggest that the upregulation of mt-tsRNAs is downstream of mitochondrial dysfunction. Single-embryo transcriptomics of genetically hybrid two-cell embryos demonstrated sperm-to-oocyte transfer of mt-tRNAs at fertilization and suggested their involvement in the control of early-embryo transcription. Our study supports the importance of paternal health at conception for offspring metabolism, shows that mt-tRNAs are diet-induced and sperm-borne and demonstrates, in a physiological setting, father-to-offspring transfer of sperm mitochondrial RNAs at fertilization.
Collapse
MESH Headings
- Animals
- Female
- Humans
- Male
- Mice
- Body Mass Index
- Diet, High-Fat/adverse effects
- Embryo, Mammalian/cytology
- Embryo, Mammalian/embryology
- Embryo, Mammalian/metabolism
- Epididymis/cytology
- Epigenesis, Genetic/genetics
- Fertilization/genetics
- Gene Expression Profiling
- Gene Expression Regulation, Developmental
- Mice, Inbred C57BL
- Mitochondria/genetics
- Mitochondria/metabolism
- Mitochondria/pathology
- Obesity/genetics
- Obesity/metabolism
- Obesity/etiology
- Oocytes/metabolism
- Overweight/genetics
- Overweight/metabolism
- Paternal Inheritance/genetics
- RNA, Mitochondrial/genetics
- RNA, Mitochondrial/metabolism
- RNA, Small Untranslated/genetics
- RNA, Small Untranslated/metabolism
- RNA, Transfer/genetics
- RNA, Transfer/metabolism
- Spermatozoa/metabolism
- Testis/cytology
- Transcription, Genetic
Collapse
Affiliation(s)
- A Tomar
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - M Gomez-Velazquez
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - R Gerlini
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - G Comas-Armangué
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - L Makharadze
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - T Kolbe
- Unit of in vivo and in vitro Models, Center for Biological Sciences, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
- IFA-Tulln, University of Natural Resources and Life Sciences, Vienna, Austria
| | - A Boersma
- Unit of in vivo and in vitro Models, Center for Biological Sciences, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - M Dahlhoff
- Unit of in vivo and in vitro Models, Center for Biological Sciences, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
| | - J P Burgstaller
- Institute of Animal Breeding and Genetics, Department of Biological Sciences and Pathobiology, University of Veterinary Medicine Vienna, Vienna, Austria
- Group Molecular Reproduction, IFA-Tulln, Tulln, Austria
| | - M Lassi
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - J Darr
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
| | - J Toppari
- Institute of Biomedicine, Integrative Physiology and Pharmacology Unit, University of Turku, Turku, Finland
- Center for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - H Virtanen
- Institute of Biomedicine, Integrative Physiology and Pharmacology Unit, University of Turku, Turku, Finland
- Center for Population Health Research, University of Turku and Turku University Hospital, Turku, Finland
| | - A Kühnapfel
- University of Leipzig, Medical Faculty, Institute for Medical Informatics, Statistics and Epidemiology, Leipzig, Germany
| | - M Scholz
- University of Leipzig, Medical Faculty, Institute for Medical Informatics, Statistics and Epidemiology, Leipzig, Germany
| | - K Landgraf
- Center for Pediatric Research Leipzig (CPL), Hospital for Children & Adolescents, University of Leipzig, Leipzig, Germany
| | - W Kiess
- Center for Pediatric Research Leipzig (CPL), Hospital for Children & Adolescents, University of Leipzig, Leipzig, Germany
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - M Vogel
- Center for Pediatric Research Leipzig (CPL), Hospital for Children & Adolescents, University of Leipzig, Leipzig, Germany
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
| | - V Gailus-Durner
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - H Fuchs
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - S Marschall
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
| | - M Hrabě de Angelis
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- German Center for Diabetes Research (DZD), Neuherberg, Germany
- German Mouse Clinic, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany
- Chair of Experimental Genetics, TUM School of Life Sciences, Technische Universität München, Freising, Germany
| | - N Kotaja
- Institute of Biomedicine, Integrative Physiology and Pharmacology Unit, University of Turku, Turku, Finland
| | - A Körner
- Center for Pediatric Research Leipzig (CPL), Hospital for Children & Adolescents, University of Leipzig, Leipzig, Germany
- LIFE Leipzig Research Center for Civilization Diseases, University of Leipzig, Leipzig, Germany
- Helmholtz Institute for Metabolic Obesity and Vascular Research (HI-MAG), Helmholtz Zentrum München at the University of Leipzig and University Hospital Leipzig, Leipzig, Germany
| | - R Teperino
- Institute of Experimental Genetics, Helmholtz Zentrum München, German Research Center for Environmental Health (GmbH), Neuherberg, Germany.
- German Center for Diabetes Research (DZD), Neuherberg, Germany.
| |
Collapse
|
34
|
Tower J. Selectively advantageous instability in biotic and pre-biotic systems and implications for evolution and aging. FRONTIERS IN AGING 2024; 5:1376060. [PMID: 38818026 PMCID: PMC11137231 DOI: 10.3389/fragi.2024.1376060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 04/15/2024] [Indexed: 06/01/2024]
Abstract
Rules of biology typically involve conservation of resources. For example, common patterns such as hexagons and logarithmic spirals require minimal materials, and scaling laws involve conservation of energy. Here a relationship with the opposite theme is discussed, which is the selectively advantageous instability (SAI) of one or more components of a replicating system, such as the cell. By increasing the complexity of the system, SAI can have benefits in addition to the generation of energy or the mobilization of building blocks. SAI involves a potential cost to the replicating system for the materials and/or energy required to create the unstable component, and in some cases, the energy required for its active degradation. SAI is well-studied in cells. Short-lived transcription and signaling factors enable a rapid response to a changing environment, and turnover is critical for replacement of damaged macromolecules. The minimal gene set for a viable cell includes proteases and a nuclease, suggesting SAI is essential for life. SAI promotes genetic diversity in several ways. Toxin/antitoxin systems promote maintenance of genes, and SAI of mitochondria facilitates uniparental transmission. By creating two distinct states, subject to different selective pressures, SAI can maintain genetic diversity. SAI of components of synthetic replicators favors replicator cycling, promoting emergence of replicators with increased complexity. Both classical and recent computer modeling of replicators reveals SAI. SAI may be involved at additional levels of biological organization. In summary, SAI promotes replicator genetic diversity and reproductive fitness, and may promote aging through loss of resources and maintenance of deleterious alleles.
Collapse
Affiliation(s)
- John Tower
- Molecular and Computational Biology Section, Department of Biological Sciences, University of Southern California, Los Angeles, CA, United States
| |
Collapse
|
35
|
Ma L, Li J, Zhang X, Zhang W, Jiang C, Yang B, Yang H. Chinese botanical drugs targeting mitophagy to alleviate diabetic kidney disease, a comprehensive review. Front Pharmacol 2024; 15:1360179. [PMID: 38803440 PMCID: PMC11128677 DOI: 10.3389/fphar.2024.1360179] [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: 01/15/2024] [Accepted: 04/29/2024] [Indexed: 05/29/2024] Open
Abstract
Diabetic kidney disease (DKD) is one of the chronic microvascular complications caused by diabetes, which is characterized by persistent albuminuria and/or progressive decline of estimated glomerular filtration rate (eGFR), and has been the major cause of dialysis around the world. At present, although the treatments for DKD including lifestyle modification, glycemic control and even using of Sodium-glucose cotransporter 2 (SGLT2) inhibitors can relieve kidney damage caused to a certain extent, there is still a lack of effective treatment schemes that can prevent DKD progressing to ESRD. It is urgent to find new complementary and effective therapeutic agents. Growing animal researches have shown that mitophagy makes a great difference to the pathogenesis of DKD, therefore, exploration of new drugs that target the restoration of mitophagy maybe a potential perspective treatment for DKD. The use of Chinese botanical drugs (CBD) has been identified to be an effective treatment option for DKD. There is growing concern on the molecular mechanism of CBD for treatment of DKD by regulating mitophagy. In this review, we highlight the current findings regarding the function of mitophagy in the pathological damages and progression of DKD and summarize the contributions of CBD that ameliorate renal injuries in DKD by interfering with mitophagy, which will help us further explain the mechanism of CBD in treatment for DKD and explore potential therapeutic strategies for DKD.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Hongtao Yang
- Department of Nephrology, First Teaching Hospital of Tianjin University of Traditional Chinese Medicine, National Clinical Research Center for Chinese Medicine Acupuncture and Moxibustion, Tianjin, China
| |
Collapse
|
36
|
Yamashita SI, Sugiura Y, Matsuoka Y, Maeda R, Inoue K, Furukawa K, Fukuda T, Chan DC, Kanki T. Mitophagy mediated by BNIP3 and NIX protects against ferroptosis by downregulating mitochondrial reactive oxygen species. Cell Death Differ 2024; 31:651-661. [PMID: 38519771 PMCID: PMC11094013 DOI: 10.1038/s41418-024-01280-y] [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: 08/22/2023] [Revised: 03/12/2024] [Accepted: 03/12/2024] [Indexed: 03/25/2024] Open
Abstract
Mitophagy plays an important role in the maintenance of mitochondrial homeostasis and can be categorized into two types: ubiquitin-mediated and receptor-mediated pathways. During receptor-mediated mitophagy, mitophagy receptors facilitate mitophagy by tethering the isolation membrane to mitochondria. Although at least five outer mitochondrial membrane proteins have been identified as mitophagy receptors, their individual contribution and interrelationship remain unclear. Here, we show that HeLa cells lacking BNIP3 and NIX, two of the five receptors, exhibit a complete loss of mitophagy in various conditions. Conversely, cells deficient in the other three receptors show normal mitophagy. Using BNIP3/NIX double knockout (DKO) cells as a model, we reveal that mitophagy deficiency elevates mitochondrial reactive oxygen species (mtROS), which leads to activation of the Nrf2 antioxidant pathway. Notably, BNIP3/NIX DKO cells are highly sensitive to ferroptosis when Nrf2-driven antioxidant enzymes are compromised. Moreover, the sensitivity of BNIP3/NIX DKO cells is fully rescued upon the introduction of wild-type BNIP3 and NIX, but not the mutant forms incapable of facilitating mitophagy. Consequently, our results demonstrate that BNIP3 and NIX-mediated mitophagy plays a role in regulating mtROS levels and protects cells from ferroptosis.
Collapse
Affiliation(s)
- Shun-Ichi Yamashita
- Department of Cellular Physiology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 950-8510, Japan.
| | - Yuki Sugiura
- Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan
| | - Yuta Matsuoka
- Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan
| | - Rae Maeda
- Center for Cancer Immunotherapy and Immunobiology, Kyoto University Graduate School of Medicine, Kyoto, 606-8501, Japan
| | - Keiichi Inoue
- Department of Cellular Physiology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 950-8510, Japan
| | - Kentaro Furukawa
- Department of Cellular Physiology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 950-8510, Japan
| | - Tomoyuki Fukuda
- Department of Cellular Physiology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 950-8510, Japan
| | - David C Chan
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125, USA
| | - Tomotake Kanki
- Department of Cellular Physiology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, 950-8510, Japan.
| |
Collapse
|
37
|
Cilenti L, Di Gregorio J, Mahar R, Liu F, Ambivero CT, Periasamy M, Merritt ME, Zervos AS. Inactivation of mitochondrial MUL1 E3 ubiquitin ligase deregulates mitophagy and prevents diet-induced obesity in mice. Front Mol Biosci 2024; 11:1397565. [PMID: 38725872 PMCID: PMC11079312 DOI: 10.3389/fmolb.2024.1397565] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 04/05/2024] [Indexed: 05/12/2024] Open
Abstract
Obesity is a growing epidemic affecting millions of people worldwide and a major risk factor for a multitude of chronic diseases and premature mortality. Accumulating evidence suggests that mitochondria have a profound role in diet-induced obesity and the associated metabolic changes, but the molecular mechanisms linking mitochondria to obesity remain poorly understood. Our studies have identified a new function for mitochondrial MUL1 E3 ubiquitin ligase, a protein known to regulate mitochondrial dynamics and mitophagy, in the control of energy metabolism and lipogenesis. Genetic deletion of Mul1 in mice impedes mitophagy and presents a metabolic phenotype that is resistant to high-fat diet (HFD)-induced obesity and metabolic syndrome. Several metabolic and lipidomic pathways are perturbed in the liver and white adipose tissue (WAT) of Mul1(-/-) animals on HFD, including the one driven by Stearoyl-CoA Desaturase 1 (SCD1), a pivotal regulator of lipid metabolism and obesity. In addition, key enzymes crucial for lipogenesis and fatty acid oxidation such as ACC1, FASN, AMPK, and CPT1 are also modulated in the absence of MUL1. The concerted action of these enzymes, in the absence of MUL1, results in diminished fat storage and heightened fatty acid oxidation. Our findings underscore the significance of MUL1-mediated mitophagy in regulating lipogenesis and adiposity, particularly in the context of HFD. Consequently, our data advocate the potential of MUL1 as a therapeutic target for drug development in the treatment of obesity, insulin resistance, NAFLD, and cardiometabolic diseases.
Collapse
Affiliation(s)
- Lucia Cilenti
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Jacopo Di Gregorio
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Rohit Mahar
- Department of Chemistry, Hemvati Nandan Bahuguna Garhwal University (A Central University), Srinagar Garhwal, Uttarakhand, India
| | - Fei Liu
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Camilla T. Ambivero
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Muthu Periasamy
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, United States
| | - Matthew E. Merritt
- Department of Biochemistry and Molecular Biology, University of Florida, Gainesville, FL, United States
| | - Antonis S. Zervos
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, United States
| |
Collapse
|
38
|
Cao J, Luo Y, Chen Y, Wu Z, Zhang J, Wu Y, Hu W. Maternal mitochondrial function affects paternal mitochondrial inheritance in Drosophila. Genetics 2024; 226:iyae014. [PMID: 38290047 PMCID: PMC10990420 DOI: 10.1093/genetics/iyae014] [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: 12/06/2023] [Revised: 01/14/2024] [Accepted: 01/17/2024] [Indexed: 02/01/2024] Open
Abstract
The maternal inheritance of mitochondria is a widely accepted paradigm, and mechanisms that prevent paternal mitochondria transmission to offspring during spermatogenesis and postfertilization have been described. Although certain species do retain paternal mitochondria, the factors affecting paternal mitochondria inheritance in these cases are unclear. More importantly, the evolutionary benefit of retaining paternal mitochondria and their ultimate fate are unknown. Here we show that transplanted exogenous paternal D. yakuba mitochondria can be transmitted to offspring when maternal mitochondria are dysfunctional in D. melanogaster. Furthermore, we show that the preserved paternal mitochondria are functional, and can be stably inherited, such that the proportion of paternal mitochondria increases gradually in subsequent generations. Our work has important implications that paternal mitochondria inheritance should not be overlooked as a genetic phenomenon in evolution, especially when paternal mitochondria are of significant differences from the maternal mitochondria or the maternal mitochondria are functionally abnormal. Our results improve the understanding of mitochondrial inheritance and provide a new model system for its study.
Collapse
Affiliation(s)
- Jinguo Cao
- Department of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
- Key Laboratory of Mitochondrial Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Yuying Luo
- Department of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Yonghe Chen
- Department of Public Health and Health Management, Gannan Medical University, Ganzhou 341000, China
| | - Zhaoqi Wu
- Department of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Jiting Zhang
- Department of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Yi Wu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Gannan Medical University, Ministry of Education, Ganzhou 341000, China
- Key Laboratory of Genetic and Developmental Related Diseases, Gannan Medical University, Ganzhou 341000, China
| | - Wen Hu
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases, Gannan Medical University, Ministry of Education, Ganzhou 341000, China
| |
Collapse
|
39
|
Sasaki T, Kushida Y, Norizuki T, Kosako H, Sato K, Sato M. ALLO-1- and IKKE-1-dependent positive feedback mechanism promotes the initiation of paternal mitochondrial autophagy. Nat Commun 2024; 15:1460. [PMID: 38368448 PMCID: PMC10874384 DOI: 10.1038/s41467-024-45863-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 02/06/2024] [Indexed: 02/19/2024] Open
Abstract
Allophagy is responsible for the selective removal of paternally inherited organelles, including mitochondria, in Caenorhabditis elegans embryos, thereby facilitating the maternal inheritance of mitochondrial DNA. We previously identified two key factors in allophagy: an autophagy adaptor allophagy-1 (ALLO-1) and TBK1/IKKε family kinase IKKE-1. However, the precise mechanisms by which ALLO-1 and IKKE-1 regulate local autophagosome formation remain unclear. In this study, we identify two ALLO-1 isoforms with different substrate preferences during allophagy. Live imaging reveals a stepwise mechanism of ALLO-1 localization with rapid cargo recognition, followed by ALLO-1 accumulation around the cargo. In the ikke-1 mutant, the accumulation of ALLO-1, and not the recognition of cargo, is impaired, resulting in the failure of isolation membrane formation. Our results also suggest a feedback mechanism for ALLO-1 accumulation via EPG-7/ATG-11, a worm homolog of FIP200, which is a candidate for IKKE-1-dependent phosphorylation. This feedback mechanism may underlie the ALLO-1-dependent initiation and progression of autophagosome formation around paternal organelles.
Collapse
Affiliation(s)
- Taeko Sasaki
- Laboratory of Molecular Membrane Biology, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, 371-8512, Japan
- Laboratory of Molecular Traffic, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, 371-8512, Japan
| | - Yasuharu Kushida
- Laboratory of Molecular Traffic, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, 371-8512, Japan
| | - Takuya Norizuki
- Laboratory of Molecular Membrane Biology, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, 371-8512, Japan
| | - Hidetaka Kosako
- Division of Cell Signaling, Fujii Memorial Institute of Medical Sciences, Institute of Advanced Medical Sciences, Tokushima University, Tokushima, 770-8503, Japan
| | - Ken Sato
- Laboratory of Molecular Traffic, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, 371-8512, Japan.
| | - Miyuki Sato
- Laboratory of Molecular Membrane Biology, Institute for Molecular and Cellular Regulation, Gunma University, Maebashi, Gunma, 371-8512, Japan.
| |
Collapse
|
40
|
Sprason C, Tucker T, Clancy D. MtDNA deletions and aging. FRONTIERS IN AGING 2024; 5:1359638. [PMID: 38425363 PMCID: PMC10902006 DOI: 10.3389/fragi.2024.1359638] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 01/29/2024] [Indexed: 03/02/2024]
Abstract
Aging is the major risk factor in most of the leading causes of mortality worldwide, yet its fundamental causes mostly remain unclear. One of the clear hallmarks of aging is mitochondrial dysfunction. Mitochondria are best known for their roles in cellular energy generation, but they are also critical biosynthetic and signaling organelles. They also undergo multiple changes with organismal age, including increased genetic errors in their independent, circular genome. A key group of studies looking at mice with increased mtDNA mutations showed that premature aging phenotypes correlated with increased deletions but not point mutations. This generated an interest in mitochondrial deletions as a potential fundamental cause of aging. However, subsequent studies in different models have yielded diverse results. This review summarizes the research on mitochondrial deletions in various organisms to understand their possible roles in causing aging while identifying the key complications in quantifying deletions across all models.
Collapse
Affiliation(s)
| | | | - David Clancy
- Biomedical and Life Sciences, Lancaster University, Lancaster, United Kingdom
| |
Collapse
|
41
|
Udagawa O. Oocyte Health and Quality: Implication of Mitochondria-related Organelle Interactions. Results Probl Cell Differ 2024; 73:25-42. [PMID: 39242373 DOI: 10.1007/978-3-031-62036-2_2] [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] [Indexed: 09/09/2024]
Abstract
Among factors like hormonal imbalance and uterine condition, oocyte quality is regarded as one of the key factors involved in age-related decline in the reproductive capacity. Here, are discussions about the functions played by organelles within the oocyte in forming the next generation that is more suitable for survival. Many insights on the adaptation to aging and maintenance of quality can be obtained from: interactions between mitochondria and other organelles that enable the long life of primordial oocytes; characteristics of organelle interactions after breaking dormancy from primary oocytes to mature oocytes; and characteristics of interactions between mitochondria and other organelles of aged oocytes collected during the ovulatory cycle from elderly individuals and animals. This information would potentially be beneficial to the development of future therapeutic methods or agents.
Collapse
Affiliation(s)
- Osamu Udagawa
- Center for Health and Environmental Risk Research, National Institute for Environmental Studies, Tsukuba, Ibaraki, Japan.
| |
Collapse
|
42
|
Lenz LS, Torgo D, Buss JH, Pereira LC, Bueno M, Filippi-Chiela EC, Lenz G. Mitochondrial response of glioma cells to temozolomide. Exp Cell Res 2023; 433:113825. [PMID: 37866459 DOI: 10.1016/j.yexcr.2023.113825] [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: 05/19/2023] [Revised: 10/03/2023] [Accepted: 10/16/2023] [Indexed: 10/24/2023]
Abstract
Metabolic adaptations are central for carcinogenesis and response to therapy, but little is known about the contribution of mitochondrial dynamics to the response of glioma cells to the standard treatment with temozolomide (TMZ). Glioma cells responded to TMZ with mitochondrial mass increased and the production of round structures of dysfunctional mitochondria. At single-cell level, asymmetric mitosis contributed to the heterogeneity of mitochondrial levels. It affected the fitness of cells in control and treated condition, indicating that the mitochondrial levels are relevant for glioma cell fitness in the presence of TMZ.
Collapse
Affiliation(s)
- Luana Suéling Lenz
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil; Departamento de Biofísica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Daphne Torgo
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil; Departamento de Biofísica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Julieti Huch Buss
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil; Departamento de Biofísica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Luiza Cherobini Pereira
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil; Departamento de Biofísica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Mardja Bueno
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil
| | - Eduardo Cremonese Filippi-Chiela
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil; Serviço de Pesquisa Experimental, Hospital de Clínicas de Porto Alegre, Porto Alegre 90035-903, Rio Grande do Sul, Brazil; Departamento de Ciências Morfológicas, Universidade Federal do Rio Grande do Sul, Porto Alegre 90050-170, Rio Grande do Sul, Brazil
| | - Guido Lenz
- Centro de Biotecnologia, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil; Departamento de Biofísica, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Rio Grande do Sul, Brazil.
| |
Collapse
|
43
|
Goyon V, Besse‐Patin A, Zunino R, Ignatenko O, Nguyen M, Coyaud É, Lee JM, Nguyen BN, Raught B, McBride HM. MAPL loss dysregulates bile and liver metabolism in mice. EMBO Rep 2023; 24:e57972. [PMID: 37962001 PMCID: PMC10702803 DOI: 10.15252/embr.202357972] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 10/10/2023] [Accepted: 10/17/2023] [Indexed: 11/15/2023] Open
Abstract
Mitochondrial and peroxisomal anchored protein ligase (MAPL) is a dual ubiquitin and small ubiquitin-like modifier (SUMO) ligase with roles in mitochondrial quality control, cell death and inflammation in cultured cells. Here, we show that MAPL function in the organismal context converges on metabolic control, as knockout mice are viable, insulin-sensitive, and protected from diet-induced obesity. MAPL loss leads to liver-specific activation of the integrated stress response, inducing secretion of stress hormone FGF21. MAPL knockout mice develop fully penetrant spontaneous hepatocellular carcinoma. Mechanistically, the peroxisomal bile acid transporter ABCD3 is a primary MAPL interacting partner and SUMOylated in a MAPL-dependent manner. MAPL knockout leads to increased bile acid production coupled with defective regulatory feedback in liver in vivo and in isolated primary hepatocytes, suggesting cell-autonomous function. Together, our findings establish MAPL function as a regulator of bile acid synthesis whose loss leads to the disruption of bile acid feedback mechanisms. The consequences of MAPL loss in liver, along with evidence of tumor suppression through regulation of cell survival pathways, ultimately lead to hepatocellular carcinogenesis.
Collapse
Affiliation(s)
- Vanessa Goyon
- Montreal Neurological InstituteMcGill UniversityMontrealQCCanada
| | | | - Rodolfo Zunino
- Montreal Neurological InstituteMcGill UniversityMontrealQCCanada
| | - Olesia Ignatenko
- Montreal Neurological InstituteMcGill UniversityMontrealQCCanada
| | - Mai Nguyen
- Montreal Neurological InstituteMcGill UniversityMontrealQCCanada
| | - Étienne Coyaud
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoONCanada
- Department of Medical BiophysicsUniversity of TorontoTorontoONCanada
| | - Jonathan M Lee
- Biochemistry, Microbiology & ImmunologyUniversity of OttawaOttawaONCanada
| | - Bich N Nguyen
- Department of Pathology and Cell BiologyUniversity of MontrealMontrealQCCanada
- University of Montreal Health NetworkMontrealQCCanada
| | - Brian Raught
- Princess Margaret Cancer CentreUniversity Health NetworkTorontoONCanada
- Department of Medical BiophysicsUniversity of TorontoTorontoONCanada
| | - Heidi M McBride
- Montreal Neurological InstituteMcGill UniversityMontrealQCCanada
| |
Collapse
|
44
|
Samare-Najaf M, Neisy A, Samareh A, Moghadam D, Jamali N, Zarei R, Zal F. The constructive and destructive impact of autophagy on both genders' reproducibility, a comprehensive review. Autophagy 2023; 19:3033-3061. [PMID: 37505071 PMCID: PMC10621263 DOI: 10.1080/15548627.2023.2238577] [Citation(s) in RCA: 33] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 07/08/2023] [Accepted: 07/14/2023] [Indexed: 07/29/2023] Open
Abstract
Reproduction is characterized by a series of massive renovations at molecular, cellular, and tissue levels. Recent studies have strongly tended to reveal the involvement of basic molecular pathways such as autophagy, a highly conserved eukaryotic cellular recycling, during reproductive processes. This review comprehensively describes the current knowledge, updated to September 2022, of autophagy contribution during reproductive processes in males including spermatogenesis, sperm motility and viability, and male sex hormones and females including germ cells and oocytes viability, ovulation, implantation, fertilization, and female sex hormones. Furthermore, the consequences of disruption in autophagic flux on the reproductive disorders including oligospermia, azoospermia, asthenozoospermia, teratozoospermia, globozoospermia, premature ovarian insufficiency, polycystic ovarian syndrome, endometriosis, and other disorders related to infertility are discussed as well.Abbreviations: AKT/protein kinase B: AKT serine/threonine kinase; AMPK: AMP-activated protein kinase; ATG: autophagy related; E2: estrogen; EDs: endocrine disruptors; ER: endoplasmic reticulum; FSH: follicle stimulating hormone; FOX: forkhead box; GCs: granulosa cells; HIF: hypoxia inducible factor; IVF: in vitro fertilization; IVM: in vitro maturation; LCs: Leydig cells; LDs: lipid droplets; LH: luteinizing hormone; LRWD1: leucine rich repeats and WD repeat domain containing 1; MAP1LC3: microtubule associated protein 1 light chain 3; MAPK: mitogen-activated protein kinase; MTOR: mechanistic target of rapamycin kinase; NFKB/NF-kB: nuclear factor kappa B; P4: progesterone; PCOS: polycystic ovarian syndrome; PDLIM1: PDZ and LIM domain 1; PI3K: phosphoinositide 3-kinase; PtdIns3P: phosphatidylinositol-3-phosphate; PtdIns3K: class III phosphatidylinositol 3-kinase; POI: premature ovarian insufficiency; ROS: reactive oxygen species; SCs: Sertoli cells; SQSTM1/p62: sequestosome 1; TSGA10: testis specific 10; TST: testosterone; VCP: vasolin containing protein.
Collapse
Affiliation(s)
- Mohammad Samare-Najaf
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
- Blood Transfusion Research Center, High Institute for Research and Education in Transfusion Medicine, Kerman Regional Blood Transfusion Center, Kerman, Iran
| | - Asma Neisy
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Samareh
- Department of Biochemistry, School of Medicine, Kerman University of Medical Sciences, Kerman, Iran
| | - Delaram Moghadam
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Department of Medicinal Chemistry, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Navid Jamali
- Department of Laboratory Sciences, Sirjan School of Medical Sciences, Sirjan, Iran
| | - Reza Zarei
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Fatemeh Zal
- Department of Biochemistry, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
- Infertility Research Centre, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
45
|
Smith CH, Mejia-Trujillo R, Breton S, Pinto BJ, Kirkpatrick M, Havird JC. Mitonuclear Sex Determination? Empirical Evidence from Bivalves. Mol Biol Evol 2023; 40:msad240. [PMID: 37935058 PMCID: PMC10653589 DOI: 10.1093/molbev/msad240] [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: 06/28/2023] [Revised: 10/04/2023] [Accepted: 10/26/2023] [Indexed: 11/09/2023] Open
Abstract
Genetic elements encoded in nuclear DNA determine the sex of an individual in many animals. In certain bivalve lineages that possess doubly uniparental inheritance (DUI), mitochondrial DNA (mtDNA) has been hypothesized to contribute to sex determination. In these cases, females transmit a female mtDNA to all offspring, while male mtDNA (M mtDNA) is transmitted only from fathers to sons. Because M mtDNA is inherited in the same way as Y chromosomes, it has been hypothesized that mtDNA may be responsible for sex determination. However, the role of mitochondrial and nuclear genes in sex determination has yet to be validated in DUI bivalves. In this study, we used DNA, RNA, and mitochondrial short noncoding RNA (sncRNA) sequencing to explore the role of mitochondrial and nuclear elements in the sexual development pathway of the freshwater mussel Potamilus streckersoni (Bivalvia: Unionida). We found that the M mtDNA sheds a sncRNA partially within a male-specific mitochondrial gene that targets a pathway hypothesized to be involved in female development and mitophagy. RNA-seq confirmed the gene target was significantly upregulated in females, supporting a direct role of mitochondrial sncRNAs in gene silencing. These findings support the hypothesis that M mtDNA inhibits female development. Genome-wide patterns of genetic differentiation and heterozygosity did not support a nuclear sex-determining region, although we cannot reject that nuclear factors are involved with sex determination. Our results provide further evidence that mitochondrial loci contribute to diverse, nonrespiratory functions and additional insights into an unorthodox sex-determining system.
Collapse
Affiliation(s)
- Chase H Smith
- Department of Integrative Biology, University of Texas at Austin, Austin, TX, USA
| | | | - Sophie Breton
- Department of Biological Sciences, University of Montreal, Montreal, Canada
| | - Brendan J Pinto
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
- Center for Evolution and Medicine, Arizona State University, Tempe, AZ, USA
- Department of Zoology, Milwaukee Public Museum, Milwaukee, WI, USA
| | - Mark Kirkpatrick
- Department of Integrative Biology, University of Texas at Austin, Austin, TX, USA
| | - Justin C Havird
- Department of Integrative Biology, University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
46
|
Zhang R, Fang J, Qi T, Zhu S, Yao L, Fang G, Li Y, Zang X, Xu W, Hao W, Liu S, Yang D, Chen D, Yang J, Ma X, Wu L. Maternal aging increases offspring adult body size via transmission of donut-shaped mitochondria. Cell Res 2023; 33:821-834. [PMID: 37500768 PMCID: PMC10624822 DOI: 10.1038/s41422-023-00854-8] [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: 12/09/2022] [Accepted: 06/21/2023] [Indexed: 07/29/2023] Open
Abstract
Maternal age at childbearing has continued to increase in recent decades. However, whether and how it influences offspring adult traits are largely unknown. Here, using adult body size as the primary readout, we reveal that maternal rather than paternal age has an evolutionarily conserved effect on offspring adult traits in humans, Drosophila, and Caenorhabditis elegans. Elucidating the mechanisms of such effects in humans and other long-lived animals remains challenging due to their long life course and difficulties in conducting in vivo studies. We thus employ the short-lived and genetically tractable nematode C. elegans to explore the mechanisms underlying the regulation of offspring adult trait by maternal aging. By microscopic analysis, we find that old worms transmit aged mitochondria with a donut-like shape to offspring. These mitochondria are rejuvenated in the offspring's early life, with their morphology fully restored before adulthood in an AMPK-dependent manner. Mechanistically, we demonstrate that early-life mitochondrial dysfunction activates AMPK, which in turn not only alleviates mitochondrial abnormalities but also activates TGFβ signaling to increase offspring adult size. Together, our findings provide mechanistic insight into the ancient role of maternal aging in shaping the traits of adult offspring.
Collapse
Affiliation(s)
- Runshuai Zhang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Jinan Fang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
| | - Ting Qi
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Shihao Zhu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Luxia Yao
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Guicun Fang
- Microscopy Core Facility, Westlake University, Hangzhou, Zhejiang, China
| | - Yunsheng Li
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Xiao Zang
- Model Animal Research Center of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Weina Xu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Wanyu Hao
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China
| | - Shouye Liu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
- Institute for Molecular Bioscience, University of Queensland, St Lucia, Brisbane, QLD, Australia
| | - Dan Yang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China
| | - Di Chen
- Model Animal Research Center of Medical School, Nanjing University, Nanjing, Jiangsu, China
| | - Jian Yang
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
| | - Xianjue Ma
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China.
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China.
| | - Lianfeng Wu
- Westlake Laboratory of Life Sciences and Biomedicine, Hangzhou, Zhejiang, China.
- School of Life Sciences, Westlake University, Hangzhou, Zhejiang, China.
- Key Laboratory of Growth Regulation and Translational Research of Zhejiang Province, Hangzhou, Zhejiang, China.
- Research Center for Industries of the Future, Westlake University, Hangzhou, Zhejiang, China.
- Institute of Basic Medical Sciences, Westlake Institute for Advanced Study, Hangzhou, Zhejiang, China.
| |
Collapse
|
47
|
Chen X, Wang Z, Zheng P, Dongol A, Xie Y, Ge X, Zheng M, Dang X, Seyhan ZB, Nagaratnam N, Yu Y, Huang X. Impaired mitophagosome-lysosome fusion mediates olanzapine-induced aging. Aging Cell 2023; 22:e14003. [PMID: 37828862 PMCID: PMC10652317 DOI: 10.1111/acel.14003] [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: 03/29/2023] [Revised: 09/23/2023] [Accepted: 09/25/2023] [Indexed: 10/14/2023] Open
Abstract
The lifespan of schizophrenia patients is significantly shorter than the general population. Olanzapine is one of the most commonly used antipsychotic drugs (APDs) for treating patients with psychosis, including schizophrenia and bipolar disorder. Despite their effectiveness in treating positive and negative symptoms, prolonged exposure to APDs may lead to accelerated aging and cognitive decline, among other side effects. Here we report that dysfunctional mitophagy is a fundamental mechanism underlying accelerated aging induced by olanzapine, using in vitro and in vivo (Caenorhabditis elegans) models. We showed that the aberrant mitophagy caused by olanzapine was via blocking mitophagosome-lysosome fusion. Furthermore, olanzapine can induce mitochondrial damage and hyperfragmentation of the mitochondrial network. The mitophagosome-lysosome fusion in olanzapine-induced aging models can be restored by a mitophagy inducer, urolithin A, which alleviates defective mitophagy, mitochondrial damage, and fragmentation of the mitochondrial network. Moreover, the mitophagy inducer ameliorated behavioral changes induced by olanzapine, including shortened lifespan, and impaired health span, learning, and memory. These data indicate that olanzapine impairs mitophagy, leading to the shortened lifespan, impaired health span, and cognitive deficits. Furthermore, this study suggests the potential application of mitophagy inducers as therapeutic strategies to reverse APD-induced adverse effects associated with accelerated aging.
Collapse
Affiliation(s)
- Xi Chen
- School of Medical, Indigenous and Health SciencesUniversity of WollongongWollongongNew South WalesAustralia
| | - Zhizhen Wang
- School of Medical, Indigenous and Health SciencesUniversity of WollongongWollongongNew South WalesAustralia
| | - Peng Zheng
- School of Medical, Indigenous and Health SciencesUniversity of WollongongWollongongNew South WalesAustralia
| | - Anjila Dongol
- School of Medical, Indigenous and Health SciencesUniversity of WollongongWollongongNew South WalesAustralia
| | - Yuanyi Xie
- School of Medical, Indigenous and Health SciencesUniversity of WollongongWollongongNew South WalesAustralia
| | - Xing Ge
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and ImmunologyXuzhou Medical UniversityXuzhouJiangsuChina
| | - Mingxuan Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and ImmunologyXuzhou Medical UniversityXuzhouJiangsuChina
| | - Xuemei Dang
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and ImmunologyXuzhou Medical UniversityXuzhouJiangsuChina
| | - Zehra Boz Seyhan
- School of Medical, Indigenous and Health SciencesUniversity of WollongongWollongongNew South WalesAustralia
| | - Nathan Nagaratnam
- School of Medical, Indigenous and Health SciencesUniversity of WollongongWollongongNew South WalesAustralia
| | - Yinghua Yu
- School of Medical, Indigenous and Health SciencesUniversity of WollongongWollongongNew South WalesAustralia
- Jiangsu Key Laboratory of Immunity and Metabolism, Jiangsu International Laboratory of Immunity and Metabolism, Department of Pathogen Biology and ImmunologyXuzhou Medical UniversityXuzhouJiangsuChina
| | - Xu‐Feng Huang
- School of Medical, Indigenous and Health SciencesUniversity of WollongongWollongongNew South WalesAustralia
| |
Collapse
|
48
|
Yazdankhah M, Ghosh S, Liu H, Hose S, Zigler JS, Sinha D. Mitophagy in Astrocytes Is Required for the Health of Optic Nerve. Cells 2023; 12:2496. [PMID: 37887340 PMCID: PMC10605486 DOI: 10.3390/cells12202496] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2023] [Revised: 10/17/2023] [Accepted: 10/18/2023] [Indexed: 10/28/2023] Open
Abstract
Mitochondrial dysfunction in astrocytes has been implicated in the development of various neurological disorders. Mitophagy, mitochondrial autophagy, is required for proper mitochondrial function by preventing the accumulation of damaged mitochondria. The importance of mitophagy, specifically in the astrocytes of the optic nerve (ON), has been little studied. We introduce an animal model in which two separate mutations act synergistically to produce severe ON degeneration. The first mutation is in Cryba1, which encodes βA3/A1-crystallin, a lens protein also expressed in astrocytes, where it regulates lysosomal pH. The second mutation is in Bckdk, which encodes branched-chain ketoacid dehydrogenase kinase, which is ubiquitously expressed in the mitochondrial matrix and involved in the catabolism of the branched-chain amino acids. BCKDK is essential for mitochondrial function and the amelioration of oxidative stress. Neither of the mutations in isolation has a significant effect on the ON, but animals homozygous for both mutations (DM) exhibit very serious ON degeneration. ON astrocytes from these double-mutant (DM) animals have lysosomal defects, including impaired mitophagy, and dysfunctional mitochondria. Urolithin A can rescue the mitophagy impairment in DM astrocytes and reduce ON degeneration. These data demonstrate that efficient mitophagy in astrocytes is required for ON health and functional integrity.
Collapse
Affiliation(s)
- Meysam Yazdankhah
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (S.G.); (H.L.); (S.H.); (D.S.)
| | - Sayan Ghosh
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (S.G.); (H.L.); (S.H.); (D.S.)
| | - Haitao Liu
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (S.G.); (H.L.); (S.H.); (D.S.)
| | - Stacey Hose
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (S.G.); (H.L.); (S.H.); (D.S.)
| | - J. Samuel Zigler
- Department of Ophthalmology, The Wilmer Eye Institute, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA;
| | - Debasish Sinha
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (S.G.); (H.L.); (S.H.); (D.S.)
- Department of Ophthalmology, The Wilmer Eye Institute, The Johns Hopkins School of Medicine, Baltimore, MD 21205, USA;
| |
Collapse
|
49
|
Socodato R, Rodrigues-Santos A, Tedim-Moreira J, Almeida TO, Canedo T, Portugal CC, Relvas JB. RhoA balances microglial reactivity and survival during neuroinflammation. Cell Death Dis 2023; 14:690. [PMID: 37863874 PMCID: PMC10589285 DOI: 10.1038/s41419-023-06217-w] [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: 10/07/2022] [Revised: 09/29/2023] [Accepted: 10/13/2023] [Indexed: 10/22/2023]
Abstract
Microglia are the largest myeloid cell population in the brain. During injury, disease, or inflammation, microglia adopt different functional states primarily involved in restoring brain homeostasis. However, sustained or exacerbated microglia inflammatory reactivity can lead to brain damage. Dynamic cytoskeleton reorganization correlates with alterations of microglial reactivity driven by external cues, and proteins controlling cytoskeletal reorganization, such as the Rho GTPase RhoA, are well positioned to refine or adjust the functional state of the microglia during injury, disease, or inflammation. Here, we use multi-biosensor-based live-cell imaging approaches and tissue-specific conditional gene ablation in mice to understand the role of RhoA in microglial response to inflammation. We found that a decrease in RhoA activity is an absolute requirement for microglial metabolic reprogramming and reactivity to inflammation. However, without RhoA, inflammation disrupts Ca2+ and pH homeostasis, dampening mitochondrial function, worsening microglial necrosis, and triggering microglial apoptosis. Our results suggest that a minimum level of RhoA activity is obligatory to concatenate microglia inflammatory reactivity and survival during neuroinflammation.
Collapse
Affiliation(s)
- Renato Socodato
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal.
| | - Artur Rodrigues-Santos
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
| | - Joana Tedim-Moreira
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
- Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal
| | - Tiago O Almeida
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
- ICBAS - School of Medicine and Biomedical Sciences, Porto, Portugal
| | - Teresa Canedo
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
| | - Camila C Portugal
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal
| | - João B Relvas
- Institute of Research and Innovation in Health (i3S) and Institute for Molecular and Cell Biology (IBMC), University of Porto, Porto, Portugal.
- Faculty of Medicine of the University of Porto (FMUP), Porto, Portugal.
| |
Collapse
|
50
|
Liu L, Li Y, Chen G, Chen Q. Crosstalk between mitochondrial biogenesis and mitophagy to maintain mitochondrial homeostasis. J Biomed Sci 2023; 30:86. [PMID: 37821940 PMCID: PMC10568841 DOI: 10.1186/s12929-023-00975-7] [Citation(s) in RCA: 82] [Impact Index Per Article: 41.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 09/13/2023] [Indexed: 10/13/2023] Open
Abstract
Mitochondrial mass and quality are tightly regulated by two essential and opposing mechanisms, mitochondrial biogenesis (mitobiogenesis) and mitophagy, in response to cellular energy needs and other cellular and environmental cues. Great strides have been made to uncover key regulators of these complex processes. Emerging evidence has shown that there exists a tight coordination between mitophagy and mitobiogenesis, and their defects may cause many human diseases. In this review, we will first summarize the recent advances made in the discovery of molecular regulations of mitobiogenesis and mitophagy and then focus on the mechanism and signaling pathways involved in the simultaneous regulation of mitobiogenesis and mitophagy in the response of tissue or cultured cells to energy needs, stress, or pathophysiological conditions. Further studies of the crosstalk of these two opposing processes at the molecular level will provide a better understanding of how the cell maintains optimal cellular fitness and function under physiological and pathophysiological conditions, which holds promise for fighting aging and aging-related diseases.
Collapse
Affiliation(s)
- Lei Liu
- Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China.
- Institute for Stem Cell and Regenerative Medicine, Beijing, China.
| | - Yanjun Li
- Center of Cell Response, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Guo Chen
- Center of Cell Response, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China
| | - Quan Chen
- Center of Cell Response, State Key Laboratory of Medicinal Chemical Biology, College of Life Sciences, Nankai University, Tianjin, China.
| |
Collapse
|