1
|
Daumke O, van der Laan M. Molecular machineries shaping the mitochondrial inner membrane. Nat Rev Mol Cell Biol 2025:10.1038/s41580-025-00854-z. [PMID: 40369159 DOI: 10.1038/s41580-025-00854-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/09/2025] [Indexed: 05/16/2025]
Abstract
Mitochondria display intricately shaped deep invaginations of the mitochondrial inner membrane (MIM) termed cristae. This peculiar membrane architecture is essential for diverse mitochondrial functions, such as oxidative phosphorylation or the biosynthesis of cellular building blocks. Conserved protein nano-machineries such as F1Fo-ATP synthase oligomers and the mitochondrial contact site and cristae organizing system (MICOS) act as adaptable protein-lipid scaffolds controlling MIM biogenesis and its dynamic remodelling. Signal-dependent rearrangements of cristae architecture and MIM fusion events are governed by the dynamin-like GTPase optic atrophy 1 (OPA1). Recent groundbreaking structural insights into these nano-machineries have considerably advanced our understanding of the functional architecture of mitochondria. In this Review, we discuss how the MIM-shaping machineries cooperate to control cristae and crista junction dynamics, including MIM fusion, in response to cellular signalling pathways. We also explore how mutations affecting MIM-shaping machineries compromise mitochondrial functions.
Collapse
Affiliation(s)
- Oliver Daumke
- Structural Biology, Max Delbrück Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany.
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Berlin, Germany.
| | - Martin van der Laan
- Medical Biochemistry & Molecular Biology, Center for Molecular Signalling (PZMS), Saarland University Medical School, Homburg/Saar, Germany.
| |
Collapse
|
2
|
Ropert B, Bannwarth S, Genin EC, Vaillant-Beuchot L, Lacas-Gervais S, Madji Hounoum B, Bernardin A, Dinh N, Mauri-Crouzet A, D'Elia MA, Augé G, Lespinasse F, Di Giorgio A, Meira W, Bonnefoy N, Monassier L, Schiff M, Sago L, Kilinc D, Brau F, Redeker V, Bohl D, Tribouillard-Tanvier D, Procaccio V, Azoulay S, Ricci JE, Delahodde A, Paquis-Flucklinger V. Nifuroxazide rescues the deleterious effects due to CHCHD10-associated MICOS defects in disease models. Brain 2025; 148:1665-1679. [PMID: 39478664 DOI: 10.1093/brain/awae348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2024] [Revised: 07/25/2024] [Accepted: 10/06/2024] [Indexed: 05/15/2025] Open
Abstract
The identification of a point mutation (p.Ser59Leu) in the CHCHD10 gene was the first genetic evidence that mitochondrial dysfunction can trigger motor neuron disease. Since then, we have shown that this mutation leads to the disorganization of the MItochondrial contact site and Cristae Organizing System (MICOS) complex that maintains the mitochondrial cristae structure. Here, we generated yeast mutant strains mimicking MICOS instability and used them to test the ability of more than 1600 compounds from two repurposed libraries to rescue the growth defect of those cells. Among the hits identified, we selected nifuroxazide, a broad-spectrum antibacterial molecule. We show that nifuroxazide rescues mitochondrial network fragmentation and cristae abnormalities in CHCHD10S59L/+ patient fibroblasts. This molecule also decreases caspase-dependent death of human CHCHD10S59L/+ induced pluripotent stem cell-derived motor neurons. Its benefits involve KIF5B-mediated mitochondrial transport enhancement, evidenced by increased axonal movement and syntaphilin degradation in patient-derived motor neurons. Our findings strengthen the MICOS-mitochondrial transport connection. Nifuroxazide and analogues emerge as potential therapeutics for MICOS-related disorders like motor neuron disease. Its impact on syntaphilin hints at broader neurological disorder applicability for nifuroxazide.
Collapse
Affiliation(s)
- Baptiste Ropert
- Université Côte d'Azur, Inserm U1081, CNRS UMR7284, IRCAN, CHU de Nice, Nice 06107, France
| | - Sylvie Bannwarth
- Université Côte d'Azur, Inserm U1081, CNRS UMR7284, IRCAN, CHU de Nice, Nice 06107, France
| | - Emmanuelle C Genin
- Université Côte d'Azur, Inserm U1081, CNRS UMR7284, IRCAN, CHU de Nice, Nice 06107, France
| | - Loan Vaillant-Beuchot
- Université Côte d'Azur, Inserm U1081, CNRS UMR7284, IRCAN, CHU de Nice, Nice 06107, France
| | - Sandra Lacas-Gervais
- Université Côte d'Azur, Centre Commun de Microscopie Appliquée, Nice 06108, France
| | - Blandine Madji Hounoum
- Université Côte d'Azur, Inserm U1065, C3M, Nice 06200, France
- Equipe labellisée Ligue Contre le Cancer, Nice 06204, France
| | - Aurore Bernardin
- Université Côte d'Azur, Inserm U1081, CNRS UMR7284, IRCAN, CHU de Nice, Nice 06107, France
| | - Nhu Dinh
- Université Paris Saclay, CEA, CNRS, I2BC, Gif-sur-Yvette 91190, France
| | | | | | - Gaelle Augé
- Université Côte d'Azur, Inserm U1081, CNRS UMR7284, IRCAN, CHU de Nice, Nice 06107, France
| | - Françoise Lespinasse
- Université Côte d'Azur, Inserm U1081, CNRS UMR7284, IRCAN, CHU de Nice, Nice 06107, France
| | | | - Willian Meira
- Université Côte d'Azur, Inserm U1081, CNRS UMR7284, IRCAN, CHU de Nice, Nice 06107, France
| | - Nathalie Bonnefoy
- Université Paris Saclay, CEA, CNRS, I2BC, Gif-sur-Yvette 91190, France
| | | | - Manuel Schiff
- Université Paris Descartes-Sorbonne Paris Cité, Inserm U1163, Institut Imagine, CHU Necker Enfants-Malades, APHP, Paris 75015, France
| | - Laila Sago
- Université Paris-Saclay, CNRS UMR9199, CEA MIRCen, Institut François Jacob, Fontenay-Aux-Roses 92260, France
| | - Devrim Kilinc
- Université de Lille, Inserm, CHU Lille, Institut Pasteur de Lille, U1167-RID-AGE-Facteurs de risque et déterminants moléculaires liés au vieillissement, Lille 59000, France
| | - Frédéric Brau
- Université Côte d'Azur, Institut de Pharmacologie Moléculaire et Cellulaire (IPMC), CNRS UMR7275, Inserm, Sophia Antipolis, Valbonne 06560, France
| | - Virginie Redeker
- Université Paris-Saclay, CNRS UMR9199, CEA MIRCen, Institut François Jacob, Fontenay-Aux-Roses 92260, France
| | - Delphine Bohl
- Institut du Cerveau-Paris Brain Institute-ICM, Inserm U1127, CNRS, Sorbonne University, Paris 75013, France
| | | | - Vincent Procaccio
- Université d'Angers, MitoLab, CNRS 6015, Inserm U1083, Institut MitoVasc, Angers 49100, France
| | | | - Jean-Ehrland Ricci
- Université Côte d'Azur, Inserm U1065, C3M, Nice 06200, France
- Equipe labellisée Ligue Contre le Cancer, Nice 06204, France
| | - Agnès Delahodde
- Université Paris Saclay, CEA, CNRS, I2BC, Gif-sur-Yvette 91190, France
| | | |
Collapse
|
3
|
Xu J, Zhang Q, Yang X, Tang Q, Han Y, Meng J, Zhang J, Lu X, Wang D, Liu J, Shan B, Bai X, Zhang K, Sun L, Wang L, Zhu L. Mitochondrial GCN5L1 coordinates with YME1L and MICOS to remodel mitochondrial cristae in white adipocytes and modulate obesity. Cell Rep 2025; 44:115682. [PMID: 40338741 DOI: 10.1016/j.celrep.2025.115682] [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: 10/14/2024] [Revised: 03/07/2025] [Accepted: 04/17/2025] [Indexed: 05/10/2025] Open
Abstract
The relationship between mitochondrial architecture and energy homeostasis in adipose tissues is not well understood. In this study, we utilized GCN5L1-knockout mice in white (AKO) and brown (BKO) adipose tissues to examine mitochondrial homeostasis in adipose tissues. GCN5L1, a regulator of mitochondrial metabolism and dynamics, influences resistance to high-fat-diet-induced obesity in AKO but not BKO mice. This resistance is mediated by an increase in mitochondrial cristae that stabilizes oxidative phosphorylation (OXPHOS) complexes and enhances energy expenditure. Our protein-interactome analysis reveals that GCN5L1 is associated with the mitochondrial crista complex MICOS (MIC13) and the protease YME1L, facilitating the degradation of MICOS and disassembly of cristae during obesity. This interaction results in decreased OXPHOS levels and subsequent adipocyte expansion. Accumulation of GCN5L1 in the mitochondrial intermembrane space is triggered by a high-fat diet. Our findings highlight a regulatory pathway involving YME1L/GCN5L1/MIC13 that remodels mitochondrial cristae in WAT in response to overnutrition-induced obesity.
Collapse
Affiliation(s)
- Juan Xu
- Department of Pharmacology, State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Inflammatory Biology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Qiqi Zhang
- Department of Pharmacology, State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Inflammatory Biology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xinyu Yang
- Department of Pharmacology, State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Inflammatory Biology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Qiqi Tang
- Department of Physiology and Pathophysiology, Tianjin Key Laboratory of Cell Homeostasis and Major Diseases, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Yitong Han
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China
| | - Jiahui Meng
- Department of Pharmacology, State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Inflammatory Biology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jiaqi Zhang
- Department of Physiology and Pathophysiology, Tianjin Key Laboratory of Cell Homeostasis and Major Diseases, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Xin Lu
- Department of Physiology and Pathophysiology, Tianjin Key Laboratory of Cell Homeostasis and Major Diseases, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Danni Wang
- Department of Pharmacology, State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Inflammatory Biology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Jing Liu
- State Key Laboratory of Experimental Hematology, Institute of Hematology & Blood Diseases Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300020, China
| | - Bo Shan
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Institute of Translational Medicine, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xue Bai
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Kai Zhang
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China
| | - Longhao Sun
- Department of General Surgery, Tianjin Medical University General Hospital, Tianjin, China.
| | - Lingdi Wang
- Department of Physiology and Pathophysiology, Tianjin Key Laboratory of Cell Homeostasis and Major Diseases, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China.
| | - Lu Zhu
- Department of Pharmacology, State Key Laboratory of Experimental Hematology, Tianjin Key Laboratory of Inflammatory Biology, The Province and Ministry Co-sponsored Collaborative Innovation Center for Medical Epigenetics, NHC Key Laboratory of Hormones and Development, Chu Hsien-I Memorial Hospital and Tianjin Institute of Endocrinology, School of Basic Medical Sciences, Tianjin Medical University, Tianjin, China; Department of Gynecology and Obstetrics, Tianjin Medical University General Hospital, Tianjin, China.
| |
Collapse
|
4
|
Shembekar SS, Nikolaus P, Honnert U, Höring M, Attia A, Topp K, Lohmann B, Liebisch G, Bähler M. Regulation of mitochondrial cristae organization by Myo19, Miro1 and Miro2, and metaxin 3. J Cell Sci 2025; 138:JCS263637. [PMID: 39882711 PMCID: PMC11925395 DOI: 10.1242/jcs.263637] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Accepted: 01/13/2025] [Indexed: 01/31/2025] Open
Abstract
The actin-based motor myosin-19 (Myo19) exerts force on mitochondrial membrane receptors Miro1/2, influencing endoplasmic reticulum (ER)-mitochondria contact sites and mitochondrial cristae structure. The mitochondrial intermembrane bridging (MIB) complex connects the outer and inner mitochondrial membranes at the cristae junction through the mitochondrial contact site and cristae organization system (MICOS). However, the interaction between Myo19, Miro1 and Miro2 (hereafter Miro1/2), and the MIB-MICOS complex in cristae regulation remains unclear. This study investigates the roles of Miro1/2 and metaxin 3 (Mtx3), a MIB complex component, in linking Myo19 to MIB-MICOS. We show that Miro1/2 interact with Myo19 and the MIB complex but not with Mtx3. Their mitochondrial membrane anchors are not essential for MIB interaction or cristae structure. However, Mtx3 is crucial for the connection between MIB-MICOS and the Myo19 and Miro1/2 proteins. Deleting Miro1/2 mimics the effects of Myo19 deficiency on ER-mitochondria contacts and cristae structure, whereas Mtx3 deletion does not. Notably, the loss of Myo19 and Miro1/2 alters mitochondrial lipid composition, reducing cardiolipin and its precursors, suggesting Myo19 and Miro1/2 influence cristae indirectly via lipid transfer at ER-mitochondria contact sites.
Collapse
Affiliation(s)
- Samruddhi S. Shembekar
- Institute of Integrative Cell Biology and Physiology, University of Münster, 48149 Münster, Germany
| | - Petra Nikolaus
- Institute of Integrative Cell Biology and Physiology, University of Münster, 48149 Münster, Germany
| | - Ulrike Honnert
- Institute of Integrative Cell Biology and Physiology, University of Münster, 48149 Münster, Germany
| | - Marcus Höring
- Institute of Clinical Chemistry and Laboratory Medicine, Regensburg University Hospital, 93053 Regensburg, Germany
| | - Aya Attia
- Institute of Integrative Cell Biology and Physiology, University of Münster, 48149 Münster, Germany
| | - Karin Topp
- Institute of Integrative Cell Biology and Physiology, University of Münster, 48149 Münster, Germany
| | - Birgit Lohmann
- Institute of Integrative Cell Biology and Physiology, University of Münster, 48149 Münster, Germany
| | - Gerhard Liebisch
- Institute of Clinical Chemistry and Laboratory Medicine, Regensburg University Hospital, 93053 Regensburg, Germany
| | - Martin Bähler
- Institute of Integrative Cell Biology and Physiology, University of Münster, 48149 Münster, Germany
| |
Collapse
|
5
|
Venkatraman K, Lipp NF, Budin I. Origin and evolution of mitochondrial inner membrane composition. J Cell Sci 2025; 138:jcs263780. [PMID: 40265338 DOI: 10.1242/jcs.263780] [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] [Indexed: 04/24/2025] Open
Abstract
Unique membrane architectures and lipid building blocks underlie the metabolic and non-metabolic functions of mitochondria. During eukaryogenesis, mitochondria likely arose from an alphaproteobacterial symbiont of an Asgard archaea-related host cell. Subsequently, mitochondria evolved inner membrane folds known as cristae alongside a specialized lipid composition supported by metabolic and transport machinery. Advancements in phylogenetic methods and genomic and metagenomic data have suggested potential origins for cristae-shaping protein complexes, such as the mitochondrial contact site and cristae-organizing system (MICOS). MICOS protein homologs function in the formation of cristae-like intracytoplasmic membranes (ICMs) in diverse extant alphaproteobacteria. The machinery responsible for synthesizing key mitochondrial phospholipids - which cooperate with cristae-shaping proteins to establish inner membrane architecture - could have also evolved from a bacterial ancestor, but its origins have been less explored. In this Review, we examine the current understanding of mitochondrial membrane evolution, highlighting distinctions between prokaryotic and eukaryotic mitochondrial-specific proteins and lipids and their differing roles in shaping cristae and ICM architecture, and propose a model explaining the concurrent specialization of the mitochondrial lipidome and inner membrane structure in eukaryogenesis. We discuss how advancements across a range of disciplines are shedding light on how multiple membrane components co-evolved to support the central functions of eukaryotic mitochondria.
Collapse
Affiliation(s)
- Kailash Venkatraman
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
| | - Nicolas-Frédéric Lipp
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
| | - Itay Budin
- Department of Chemistry and Biochemistry, University of California San Diego, La Jolla, CA 92093, USA
| |
Collapse
|
6
|
Rigoni G, Calvo E, Glytsou C, Carro-Alvarellos M, Noguchi M, Semenzato M, Quirin C, Caicci F, Meneghetti N, Sturlese M, Ishihara T, Moro S, Rampazzo C, Ishihara N, Bezzo F, Salviati L, Vazquez J, Sales G, Romualdi C, Enriquez JA, Scorrano L, Soriano ME. MARIGOLD and MitoCIAO, two searchable compendia to visualize and functionalize protein complexes during mitochondrial remodeling. Cell Metab 2025; 37:1024-1038.e8. [PMID: 39999845 DOI: 10.1016/j.cmet.2025.01.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/15/2022] [Revised: 10/15/2024] [Accepted: 01/20/2025] [Indexed: 02/27/2025]
Abstract
Mitochondrial proteins assemble dynamically in high molecular weight complexes essential for their functions. We generated and validated two searchable compendia of these mitochondrial complexes. Following identification by mass spectrometry of proteins in complexes separated using blue-native gel electrophoresis from unperturbed, cristae-remodeled, and outer membrane-permeabilized mitochondria, we created MARIGOLD, a mitochondrial apoptotic remodeling complexome database of 627 proteins. MARIGOLD elucidates how dynamically proteins distribute in complexes upon mitochondrial membrane remodeling. From MARIGOLD, we developed MitoCIAO, a mitochondrial complexes interactome tool that, by statistical correlation, calculates the likelihood of protein cooccurrence in complexes. MitoCIAO correctly predicted biologically validated interactions among components of the mitochondrial cristae organization system (MICOS) and optic atrophy 1 (OPA1) complexes. We used MitoCIAO to functionalize two ATPase family AAA domain-containing 3A (ATAD3A) complexes: one with OPA1 that regulates mitochondrial ultrastructure and the second containing ribosomal proteins that is essential for mitoribosome stability. These compendia reveal the dynamic nature of mitochondrial complexes and enable their functionalization.
Collapse
Affiliation(s)
- Giovanni Rigoni
- Department of Biology, University of Padova, Via U. Bassi 58B, 35121 Padova, Italy
| | - Enrique Calvo
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain; CIBER de Enfermedades Cardiovasculares, 28029 Madrid, Spain
| | - Christina Glytsou
- Department of Biology, University of Padova, Via U. Bassi 58B, 35121 Padova, Italy; Veneto Institute of Molecular Medicine, 35129 Padova, Italy
| | | | - Masafumi Noguchi
- Department of Biology, University of Padova, Via U. Bassi 58B, 35121 Padova, Italy; Veneto Institute of Molecular Medicine, 35129 Padova, Italy
| | - Martina Semenzato
- Department of Biology, University of Padova, Via U. Bassi 58B, 35121 Padova, Italy; Veneto Institute of Molecular Medicine, 35129 Padova, Italy
| | - Charlotte Quirin
- Department of Biology, University of Padova, Via U. Bassi 58B, 35121 Padova, Italy; Veneto Institute of Molecular Medicine, 35129 Padova, Italy
| | - Federico Caicci
- Department of Biology, University of Padova, Via U. Bassi 58B, 35121 Padova, Italy
| | - Natascia Meneghetti
- CAPE-Lab, Department of Industrial Engineering, University of Padova, Padova, Italy
| | - Mattia Sturlese
- Department of Pharmaceutical Sciences, University of Padova, 35131 Padova, Italy
| | - Takaya Ishihara
- Department of Biological Sciences, Graduate School of Science, Osaka University, 560-0043 Toyonaka, Japan
| | - Stefano Moro
- Department of Pharmaceutical Sciences, University of Padova, 35131 Padova, Italy
| | - Chiara Rampazzo
- Department of Biology, University of Padova, Via U. Bassi 58B, 35121 Padova, Italy
| | - Naotada Ishihara
- Department of Biological Sciences, Graduate School of Science, Osaka University, 560-0043 Toyonaka, Japan
| | - Fabrizio Bezzo
- CAPE-Lab, Department of Industrial Engineering, University of Padova, Padova, Italy
| | - Leonardo Salviati
- Department of Women's and Children's health, University of Padova and IRP Città della Speranza, 35127 Padova, Italy
| | - Jesùs Vazquez
- Centro Nacional de Investigaciones Cardiovasculares Carlos III, 28029 Madrid, Spain; CIBER de Enfermedades Cardiovasculares, 28029 Madrid, Spain
| | - Gabriele Sales
- Department of Biology, University of Padova, Via U. Bassi 58B, 35121 Padova, Italy
| | - Chiara Romualdi
- Department of Biology, University of Padova, Via U. Bassi 58B, 35121 Padova, Italy
| | | | - Luca Scorrano
- Department of Biology, University of Padova, Via U. Bassi 58B, 35121 Padova, Italy; Veneto Institute of Molecular Medicine, 35129 Padova, Italy.
| | | |
Collapse
|
7
|
Macuada J, Molina-Riquelme I, Eisner V. How are mitochondrial nucleoids trafficked? Trends Cell Biol 2025; 35:194-204. [PMID: 39984359 DOI: 10.1016/j.tcb.2024.12.007] [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: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 02/23/2025]
Abstract
Mitochondria harbor their own DNA (mtDNA), which codifies essential proteins of the oxidative phosphorylation (OXPHOS) system and locally feeds them to their surrounding inner mitochondrial membrane (IMM), according to the 'sphere of influence' theory. mtDNA is compacted into nucleoids, which are tethered to the IMM and distributed throughout the mitochondrial network. Some nucleoid subpopulations present distinct intramitochondrial positioning during fission and their correct positioning is associated with mtDNA segregation and selective degradation. This opinion article focuses on different mechanisms that could control nucleoid positioning through intramitochondrial trafficking, either by cristae reshaping or by intercompartment-driven mechanisms involving the mitochondrial membranes and extramitochondrial elements. Understanding nucleoid trafficking promises insights into mitochondrial dysfunction in pathologies with mtDNA distribution and segregation issues.
Collapse
Affiliation(s)
- Josefa Macuada
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile
| | | | - Verónica Eisner
- Facultad de Ciencias Biológicas, Pontificia Universidad Católica de Chile, Santiago, Chile.
| |
Collapse
|
8
|
Adams R, Afzal N, Jafri MS, Mannella CA. How the Topology of the Mitochondrial Inner Membrane Modulates ATP Production. Cells 2025; 14:257. [PMID: 39996730 PMCID: PMC11853683 DOI: 10.3390/cells14040257] [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: 12/30/2024] [Revised: 01/31/2025] [Accepted: 02/06/2025] [Indexed: 02/26/2025] Open
Abstract
Cells in heart muscle need to generate ATP at or near peak capacity to meet their energy demands. Over 90% of this ATP comes from mitochondria, strategically located near myofibrils and densely packed with cristae to concentrate ATP generation per unit volume. However, a consequence of dense inner membrane (IM) packing is that restricted metabolite diffusion inside mitochondria may limit ATP production. Under physiological conditions, the flux of ATP synthase is set by ADP levels in the matrix, which in turn depends on diffusion-dependent concentration of ADP inside cristae. Computer simulations show how ADP diffusion and consequently rates of ATP synthesis are modulated by IM topology, in particular (i) number, size, and positioning of crista junctions that connect cristae to the IM boundary region, and (ii) branching of cristae. Predictions are compared with the actual IM topology of a cardiomyocyte mitochondrion in which cristae vary systematically in length and morphology. The analysis indicates that this IM topology decreases but does not eliminate the "diffusion penalty" on ATP output. It is proposed that IM topology normally attenuates mitochondrial ATP output under conditions of low workload and can be regulated by the cell to better match ATP supply to demand.
Collapse
Affiliation(s)
- Raquel Adams
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA; (R.A.); (N.A.)
| | - Nasrin Afzal
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA; (R.A.); (N.A.)
| | - Mohsin Saleet Jafri
- School of Systems Biology, George Mason University, Fairfax, VA 22030, USA; (R.A.); (N.A.)
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 20201, USA
| | - Carmen A. Mannella
- Center for Biomedical Engineering and Technology, University of Maryland School of Medicine, Baltimore, MD 20201, USA
- Department of Pharmacology, Physiology and Drug Development, University of Maryland School of Medicine, Baltimore, MD 20201, USA
| |
Collapse
|
9
|
Ravi R, Routray D, Mahalakshmi R. Mitochondrial Sorting and Assembly Machinery: Chaperoning a Moonlighting Role? Biochemistry 2025; 64:312-328. [PMID: 39754567 DOI: 10.1021/acs.biochem.4c00727] [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: 01/06/2025]
Abstract
The mitochondrial outer membrane (OMM) β-barrel proteins link the mitochondrion with the cytosol, endoplasmic reticulum, and other cellular membranes, establishing cellular homeostasis. Their active insertion and assembly in the outer mitochondrial membrane is achieved in an energy-independent yet highly effective manner by the Sorting and Assembly Machinery (SAM) of the OMM. The core SAM constituent is the 16-stranded transmembrane β-barrel Sam50. For over two decades, the primary role of Sam50 has been linked to its function as a chaperone in the OMM, wherein it assembles all β-barrels through a lateral gating and β-barrel switching mechanism. Interestingly, recent studies have demonstrated that despite its low copy number, Sam50 performs various diverse functions beyond assembling β-barrels. This includes maintaining cristae morphology, bidirectional lipid shuttling between the ER and mitochondrial inner membrane, import of select proteins, regulation of PINK1-Parkin function, and timed trigger of cell death. Given these multifaceted critical regulatory functions of SAM across all eukaryotes, we now reason that SAM merely moonlights as the hub for β-barrel biogenesis and has indeed evolved a diverse array of primary roles in maintaining mitochondrial function and cellular homeostasis.
Collapse
Affiliation(s)
- Roshika Ravi
- Molecular Biophysics Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal 462066, India
| | - Deepsikha Routray
- Molecular Biophysics Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal 462066, India
| | - Radhakrishnan Mahalakshmi
- Molecular Biophysics Laboratory, Department of Biological Sciences, Indian Institute of Science Education and Research, Bhopal 462066, India
| |
Collapse
|
10
|
Benning FMC, Bell TA, Nguyen TH, Syau D, Connell LB, Coughlin M, Nordstrom AEH, Ericsson M, daCosta CJB, Chao LH. Ancestral sequence reconstruction of the Mic60 Mitofilin domain reveals residues supporting respiration in yeast. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.04.26.591372. [PMID: 38746426 PMCID: PMC11092495 DOI: 10.1101/2024.04.26.591372] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
In eukaryotes, cellular respiration takes place in the cristae of mitochondria. The mitochondrial inner membrane protein Mic60, a core component of the mitochondrial contact site and cristae organizing system (MICOS), is crucial for the organization and stabilization of crista junctions and its associated functions. While the C-terminal Mitofilin domain of Mic60 is necessary for cellular respiration, the sequence determinants for this function have remained unclear. Here, we used ancestral sequence reconstruction to generate Mitofilin ancestors up to and including the last opisthokont common ancestor (LOCA). We found that yeast-lineage derived Mitofilin ancestors as far back as the LOCA rescue respiration. By comparing Mitofilin ancestors, we identified four residues sufficient to explain the respiratory difference between yeast- and animal-derived Mitofilin ancestors. Our results provide a foundation for investigating the conservation of Mic60-mediated cristae junction interactions.
Collapse
Affiliation(s)
- Friederike M. C. Benning
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| | - Tristan A. Bell
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Present address: Generate Biomedicines, Somerville, MA 02143, USA
| | - Tran H. Nguyen
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
- Present address: Medical Scientist Training Program, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Della Syau
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
- Present address: Biological and Biomedical Sciences Program, Harvard Medical School, Boston, MA 02115
| | - Louise B. Connell
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Margaret Coughlin
- Electron Microscopy Core Facility, Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Anja E. H. Nordstrom
- Electron Microscopy Core Facility, Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Maria Ericsson
- Electron Microscopy Core Facility, Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA
| | - Corrie J. B. daCosta
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, Ottawa, Ontario K1N 6N5, Canada
| | - Luke H. Chao
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
- Department of Genetics, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
11
|
Zerbes RM, Colina-Tenorio L, Bohnert M, von der Malsburg K, Peikert CD, Mehnert CS, Perschil I, Klar RFU, de Boer R, Kram A, van der Klei I, Oeljeklaus S, Warscheid B, Rampelt H, van der Laan M. Coordination of cytochrome bc 1 complex assembly at MICOS. EMBO Rep 2025; 26:353-384. [PMID: 39623166 PMCID: PMC11772845 DOI: 10.1038/s44319-024-00336-x] [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: 04/15/2024] [Revised: 11/04/2024] [Accepted: 11/15/2024] [Indexed: 01/29/2025] Open
Abstract
The boundary and cristae domains of the mitochondrial inner membrane are connected by crista junctions. Most cristae membrane proteins are nuclear-encoded and inserted by the mitochondrial protein import machinery into the inner boundary membrane. Thus, they must overcome the diffusion barrier imposed by crista junctions to reach their final location. Here, we show that respiratory chain complexes and assembly intermediates are physically connected to the mitochondrial contact site and cristae organizing system (MICOS) that is essential for the formation and stability of crista junctions. We identify the inner membrane protein Mar26 (Fmp10) as a determinant in the biogenesis of the cytochrome bc1 complex (complex III). Mar26 couples a Rieske Fe/S protein-containing assembly intermediate to MICOS. Our data indicate that Mar26 maintains an assembly-competent Rip1 pool at crista junctions where complex III maturation likely occurs. MICOS facilitates efficient Rip1 assembly by recruiting complex III assembly intermediates to crista junctions. We propose that MICOS, via interaction with assembly factors such as Mar26, contributes to the spatial and temporal coordination of respiratory chain biogenesis.
Collapse
Affiliation(s)
- Ralf M Zerbes
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Lilia Colina-Tenorio
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Maria Bohnert
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
- Institute of Cell Dynamics and Imaging, Cells in Motion Interfaculty Centre (CiM), University of Münster, 48149, Münster, Germany
| | - Karina von der Malsburg
- Medical Biochemistry and Molecular Biology, Saarland University, 66421, Homburg, Germany
- Center for Molecular Signaling, PZMS, Saarland University, 66421, Homburg, Germany
| | - Christian D Peikert
- BIOSS Centre for Biological Signalling Studies, Universität Freiburg, 79104, Freiburg, Germany
- Bioinformatics Research & Development, BioNTech SE, 55131, Mainz, Germany
| | - Carola S Mehnert
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Inge Perschil
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Rhena F U Klar
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
- Institute of Molecular Medicine and Cell Research (IMMZ), University of Freiburg, 79104, Freiburg, Germany
| | - Rinse de Boer
- Molecular Cell Biology, University of Groningen, 9700 CC, Groningen, The Netherlands
| | - Anita Kram
- Molecular Cell Biology, University of Groningen, 9700 CC, Groningen, The Netherlands
| | - Ida van der Klei
- Molecular Cell Biology, University of Groningen, 9700 CC, Groningen, The Netherlands
| | - Silke Oeljeklaus
- Faculty of Chemistry and Pharmacy, Biochemistry II, Theodor Boveri-Institute, University of Würzburg, 97074, Würzburg, Germany
| | - Bettina Warscheid
- BIOSS Centre for Biological Signalling Studies, Universität Freiburg, 79104, Freiburg, Germany
- Faculty of Chemistry and Pharmacy, Biochemistry II, Theodor Boveri-Institute, University of Würzburg, 97074, Würzburg, Germany
| | - Heike Rampelt
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany.
- CIBSS Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany.
| | - Martin van der Laan
- Medical Biochemistry and Molecular Biology, Saarland University, 66421, Homburg, Germany.
- Center for Molecular Signaling, PZMS, Saarland University, 66421, Homburg, Germany.
| |
Collapse
|
12
|
Latorre-Muro P, Vitale T, Ravichandran M, Zhang K, Palozzi JM, Bennett CF, Lamas-Paz A, Sohn JH, Jackson TD, Jedrychowski M, Gygi SP, Kajimura S, Schmoker A, Jeon H, Eck MJ, Puigserver P. Chaperone-mediated insertion of mitochondrial import receptor TOM70 protects against diet-induced obesity. Nat Cell Biol 2025; 27:130-140. [PMID: 39753947 DOI: 10.1038/s41556-024-01555-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 10/04/2024] [Indexed: 01/18/2025]
Abstract
Outer mitochondrial membrane (OMM) proteins communicate with the cytosol and other organelles, including the endoplasmic reticulum. This communication is important in thermogenic adipocytes to increase the energy expenditure that controls body temperature and weight. However, the regulatory mechanisms of OMM protein insertion are poorly understood. Here the stress-induced cytosolic chaperone PPID (peptidyl-prolyl isomerase D/cyclophilin 40/Cyp40) drives OMM insertion of the mitochondrial import receptor TOM70 that regulates body temperature and weight in obese mice, and respiratory/thermogenic function in brown adipocytes. PPID PPIase activity and C-terminal tetratricopeptide repeats, which show specificity towards TOM70 core and C-tail domains, facilitate OMM insertion. Our results provide an unprecedented role for endoplasmic-reticulum-stress-activated chaperones in controlling energy metabolism through a selective OMM protein insertion mechanism with implications in adaptation to cold temperatures and high-calorie diets.
Collapse
Affiliation(s)
- Pedro Latorre-Muro
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Tevis Vitale
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | | | - Katherine Zhang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jonathan M Palozzi
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Christopher F Bennett
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Arantza Lamas-Paz
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Jee Hyung Sohn
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Thomas D Jackson
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | | | - Steven P Gygi
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Shingo Kajimura
- Division of Endocrinology, Diabetes, and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
- Howard Hughes Medical Institute, Chevy Chase, MD, USA
| | - Anna Schmoker
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Hyesung Jeon
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Michael J Eck
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
| | - Pere Puigserver
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA.
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
Vassallo N. Poration of mitochondrial membranes by amyloidogenic peptides and other biological toxins. J Neurochem 2025; 169:e16213. [PMID: 39213385 DOI: 10.1111/jnc.16213] [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: 06/26/2024] [Revised: 08/09/2024] [Accepted: 08/12/2024] [Indexed: 09/04/2024]
Abstract
Mitochondria are essential organelles known to serve broad functions, including in cellular metabolism, calcium buffering, signaling pathways and the regulation of apoptotic cell death. Maintaining the integrity of the outer (OMM) and inner mitochondrial membranes (IMM) is vital for mitochondrial health. Cardiolipin (CL), a unique dimeric glycerophospholipid, is the signature lipid of energy-converting membranes. It plays a significant role in maintaining mitochondrial architecture and function, stabilizing protein complexes and facilitating efficient oxidative phosphorylation (OXPHOS) whilst regulating cytochrome c release from mitochondria. CL is especially enriched in the IMM and at sites of contact between the OMM and IMM. Disorders of protein misfolding, such as Alzheimer's and Parkinson's diseases, involve amyloidogenic peptides like amyloid-β, tau and α-synuclein, which form metastable toxic oligomeric species that interact with biological membranes. Electrophysiological studies have shown that these oligomers form ion-conducting nanopores in membranes mimicking the IMM's phospholipid composition. Poration of mitochondrial membranes disrupts the ionic balance, causing osmotic swelling, loss of the voltage potential across the IMM, release of pro-apoptogenic factors, and leads to cell death. The interaction between CL and amyloid oligomers appears to favour their membrane insertion and pore formation, directly implicating CL in amyloid toxicity. Additionally, pore formation in mitochondrial membranes is not limited to amyloid proteins and peptides; other biological peptides, as diverse as the pro-apoptotic Bcl-2 family members, gasdermin proteins, cobra venom cardiotoxins and bacterial pathogenic toxins, have all been described to punch holes in mitochondria, contributing to cell death processes. Collectively, these findings underscore the vulnerability of mitochondria and the involvement of CL in various pathogenic mechanisms, emphasizing the need for further research on targeting CL-amyloid interactions to mitigate mitochondrial dysfunction.
Collapse
Affiliation(s)
- Neville Vassallo
- Department of Physiology and Biochemistry, Faculty of Medicine and Surgery, University of Malta, Tal-Qroqq, Malta
- Centre for Molecular Medicine and Biobanking, University of Malta, Tal-Qroqq, Malta
| |
Collapse
|
14
|
Wisniewski BT, Casler JC, Lackner LL. Significantly reduced, but balanced, rates of mitochondrial fission and fusion are sufficient to maintain the integrity of yeast mitochondrial DNA. Mol Biol Cell 2024; 35:br25. [PMID: 39535883 PMCID: PMC11656474 DOI: 10.1091/mbc.e24-07-0306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 11/04/2024] [Accepted: 11/04/2024] [Indexed: 11/16/2024] Open
Abstract
Mitochondria exist as dynamic tubular networks and the morphology of these networks impacts organelle function and cell health. Mitochondrial morphology is maintained in part by the opposing activities of mitochondrial fission and fusion. Mitochondrial fission and fusion are also required to maintain mitochondrial DNA (mtDNA) integrity. In Saccharomyces cerevisiae, the simultaneous inhibition of mitochondrial fission and fusion results in increased mtDNA mutation and the consequent loss of respiratory competence. The mechanism by which fission and fusion maintain mtDNA integrity is not fully understood. Previous work demonstrates that mtDNA is spatially linked to mitochondrial fission sites. Here, we extend this finding using live-cell imaging to localize mtDNA to mitochondrial fusion sites. While mtDNA is present at sites of mitochondrial fission and fusion, mitochondrial fission and fusion rates are not altered in cells lacking mtDNA. Using alleles that alter mitochondrial fission and fusion rates, we find that mtDNA integrity can be maintained in cells with significantly reduced, but balanced, rates of fission and fusion. In addition, we find that increasing mtDNA copy number reduces the loss of respiratory competence in double mitochondrial fission-fusion mutants. Our findings add novel insights into the relationship between mitochondrial dynamics and mtDNA integrity.
Collapse
Affiliation(s)
- Brett T. Wisniewski
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208
| | - Jason C. Casler
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208
| | - Laura L. Lackner
- Department of Molecular Biosciences, Northwestern University, Evanston, IL 60208
| |
Collapse
|
15
|
Lee RG, Rudler DL, Rackham O, Filipovska A. Interorganelle phospholipid communication, a house not so divided. Trends Endocrinol Metab 2024; 35:872-883. [PMID: 38972781 DOI: 10.1016/j.tem.2024.06.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 06/12/2024] [Accepted: 06/13/2024] [Indexed: 07/09/2024]
Abstract
The presence of membrane-bound organelles with specific functions is one of the main hallmarks of eukaryotic cells. Organelle membranes are composed of specific lipids that govern their function and interorganelle communication. Discoveries in cell biology using imaging and omic technologies have revealed the mechanisms that drive membrane remodeling, organelle contact sites, and metabolite exchange. The interplay between multiple organelles and their interdependence is emerging as the next frontier for discovery using 3D reconstruction of volume electron microscopy (vEM) datasets. We discuss recent findings on the links between organelles that underlie common functions and cellular pathways. Specifically, we focus on the metabolism of ether glycerophospholipids that mediate organelle dynamics and their communication with each other, and the new imaging techniques that are powering these discoveries.
Collapse
Affiliation(s)
- Richard G Lee
- Australian Research Council (ARC) Centre of Excellence in Synthetic Biology, Queen Elizabeth II Medical Centre (QEIIMC), Nedlands, WA, Australia; Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, Nedlands, WA, Australia
| | - Danielle L Rudler
- Australian Research Council (ARC) Centre of Excellence in Synthetic Biology, Queen Elizabeth II Medical Centre (QEIIMC), Nedlands, WA, Australia; Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, Nedlands, WA, Australia
| | - Oliver Rackham
- Australian Research Council (ARC) Centre of Excellence in Synthetic Biology, Queen Elizabeth II Medical Centre (QEIIMC), Nedlands, WA, Australia; Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, Nedlands, WA, Australia; Curtin Medical School, Curtin University, Bentley, WA, Australia; Curtin Health Innovation Research Institute, Curtin University, Bentley, WA, Australia
| | - Aleksandra Filipovska
- Australian Research Council (ARC) Centre of Excellence in Synthetic Biology, Queen Elizabeth II Medical Centre (QEIIMC), Nedlands, WA, Australia; Telethon Kids Institute, Northern Entrance, Perth Children's Hospital, Nedlands, WA, Australia; The University of Western Australia Centre for Child Health Research, Northern Entrance, Perth Children's Hospital, Nedlands, WA, Australia.
| |
Collapse
|
16
|
Dumbali SP, Horton PD, Moore TI, Wenzel PL. Mitochondrial permeability transition dictates mitochondrial maturation upon switch in cellular identity of hematopoietic precursors. Commun Biol 2024; 7:967. [PMID: 39122870 PMCID: PMC11316084 DOI: 10.1038/s42003-024-06671-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: 10/22/2023] [Accepted: 08/02/2024] [Indexed: 08/12/2024] Open
Abstract
The mitochondrial permeability transition pore (mPTP) is a supramolecular channel that regulates exchange of solutes across cristae membranes, with executive roles in mitochondrial function and cell death. The contribution of the mPTP to normal physiology remains debated, although evidence implicates the mPTP in mitochondrial inner membrane remodeling in differentiating progenitor cells. Here, we demonstrate that strict control over mPTP conductance shapes metabolic machinery as cells transit toward hematopoietic identity. Cells undergoing the endothelial-to-hematopoietic transition (EHT) tightly control chief regulatory elements of the mPTP. During EHT, maturing arterial endothelium restricts mPTP activity just prior to hematopoietic commitment. After transition in cellular identity, mPTP conductance is restored. In utero treatment with NIM811, a molecule that blocks sensitization of the mPTP to opening by Cyclophilin D (CypD), amplifies oxidative phosphorylation (OXPHOS) in hematopoietic precursors and increases hematopoiesis in the embryo. Additionally, differentiating pluripotent stem cells (PSCs) acquire greater organization of mitochondrial cristae and hematopoietic activity following knockdown of the CypD gene, Ppif. Conversely, knockdown of Opa1, a GTPase critical for proper cristae architecture, induces cristae irregularity and impairs hematopoiesis. These data elucidate a mechanism that regulates mitochondrial maturation in hematopoietic precursors and underscore a role for the mPTP in the acquisition of hematopoietic fate.
Collapse
Affiliation(s)
- Sandeep P Dumbali
- Department of Integrative Biology & Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Paulina D Horton
- Department of Integrative Biology & Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Immunology Program, The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Travis I Moore
- Department of Integrative Biology & Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA
- Molecular & Translational Biology Program, The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA
| | - Pamela L Wenzel
- Department of Integrative Biology & Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, Houston, TX, USA.
- Center for Stem Cell and Regenerative Medicine, Brown Foundation Institute of Molecular Medicine, The University of Texas Health Science Center at Houston, Houston, TX, USA.
- Immunology Program, The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
- Molecular & Translational Biology Program, The University of Texas MD Anderson UTHealth Graduate School of Biomedical Sciences, Houston, TX, USA.
| |
Collapse
|
17
|
Yang TH, Kang EYC, Lin PH, Yu BBC, Wang JHH, Chen V, Wang NK. Mitochondria in Retinal Ganglion Cells: Unraveling the Metabolic Nexus and Oxidative Stress. Int J Mol Sci 2024; 25:8626. [PMID: 39201313 PMCID: PMC11354650 DOI: 10.3390/ijms25168626] [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: 05/31/2024] [Revised: 07/29/2024] [Accepted: 07/29/2024] [Indexed: 09/02/2024] Open
Abstract
This review explored the role of mitochondria in retinal ganglion cells (RGCs), which are essential for visual processing. Mitochondrial dysfunction is a key factor in the pathogenesis of various vision-related disorders, including glaucoma, hereditary optic neuropathy, and age-related macular degeneration. This review highlighted the critical role of mitochondria in RGCs, which provide metabolic support, regulate cellular health, and respond to cellular stress while also producing reactive oxygen species (ROS) that can damage cellular components. Maintaining mitochondrial function is essential for meeting RGCs' high metabolic demands and ensuring redox homeostasis, which is crucial for their proper function and visual health. Oxidative stress, exacerbated by factors like elevated intraocular pressure and environmental factors, contributes to diseases such as glaucoma and age-related vision loss by triggering cellular damage pathways. Strategies targeting mitochondrial function or bolstering antioxidant defenses include mitochondrial-based therapies, gene therapies, and mitochondrial transplantation. These advances can offer potential strategies for addressing mitochondrial dysfunction in the retina, with implications that extend beyond ocular diseases.
Collapse
Affiliation(s)
- Tsai-Hsuan Yang
- Department of Education, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan 33305, Taiwan;
- College of Medicine, National Yang Ming Chiao Tung University, Taipei 11217, Taiwan
| | - Eugene Yu-Chuan Kang
- Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan 33305, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Graduate Institute of Clinical Medical Sciences, College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; (P.-H.L.); (J.H.-H.W.); (V.C.)
| | - Pei-Hsuan Lin
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; (P.-H.L.); (J.H.-H.W.); (V.C.)
- National Taiwan University Hospital, Yunlin 640203, Taiwan
| | - Benjamin Ben-Chi Yu
- Fu Foundation School of Engineering & Applied Science, Columbia University, New York, NY 10027, USA;
| | - Jason Hung-Hsuan Wang
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; (P.-H.L.); (J.H.-H.W.); (V.C.)
- Columbian College of Arts and Sciences, George Washington University, Washington, DC 20052, USA
| | - Vincent Chen
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA; (P.-H.L.); (J.H.-H.W.); (V.C.)
- Faculty of Health Sciences, Queen’s University, Kingston, ON K7L 3N9, Canada
| | - Nan-Kai Wang
- Department of Ophthalmology, Chang Gung Memorial Hospital, Linkou Medical Center, Taoyuan 33305, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 33302, Taiwan
- Department of Ophthalmology, Edward S. Harkness Eye Institute, Vagelos College of Physicians and Surgeons, Columbia University Irving Medical Center, Columbia University, New York, NY 10032, USA
| |
Collapse
|
18
|
Shao B, Killion M, Oliver A, Vang C, Zeleke F, Neikirk K, Vue Z, Garza-Lopez E, Shao JQ, Mungai M, Lam J, Williams Q, Altamura CT, Whiteside A, Kabugi K, McKenzie J, Ezedimma M, Le H, Koh A, Scudese E, Vang L, Marshall AG, Crabtree A, Tanghal JI, Stephens D, Koh HJ, Jenkins BC, Murray SA, Cooper AT, Williams C, Damo SM, McReynolds MR, Gaddy JA, Wanjalla CN, Beasley HK, Hinton A. Ablation of Sam50 is associated with fragmentation and alterations in metabolism in murine and human myotubes. J Cell Physiol 2024; 239:e31293. [PMID: 38770789 PMCID: PMC11324413 DOI: 10.1002/jcp.31293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2023] [Revised: 03/30/2024] [Accepted: 04/26/2024] [Indexed: 05/22/2024]
Abstract
The sorting and assembly machinery (SAM) Complex is responsible for assembling β-barrel proteins in the mitochondrial membrane. Comprising three subunits, Sam35, Sam37, and Sam50, the SAM complex connects the inner and outer mitochondrial membranes by interacting with the mitochondrial contact site and cristae organizing system complex. Sam50, in particular, stabilizes the mitochondrial intermembrane space bridging (MIB) complex, which is crucial for protein transport, respiratory chain complex assembly, and regulation of cristae integrity. While the role of Sam50 in mitochondrial structure and metabolism in skeletal muscle remains unclear, this study aims to investigate its impact. Serial block-face-scanning electron microscopy and computer-assisted 3D renderings were employed to compare mitochondrial structure and networking in Sam50-deficient myotubes from mice and humans with wild-type (WT) myotubes. Furthermore, autophagosome 3D structure was assessed in human myotubes. Mitochondrial metabolic phenotypes were assessed using Gas Chromatography-Mass Spectrometry-based metabolomics to explore differential changes in WT and Sam50-deficient myotubes. The results revealed increased mitochondrial fragmentation and autophagosome formation in Sam50-deficient myotubes compared to controls. Metabolomic analysis indicated elevated metabolism of propanoate and several amino acids, including ß-Alanine, phenylalanine, and tyrosine, along with increased amino acid and fatty acid metabolism in Sam50-deficient myotubes. Furthermore, impairment of oxidative capacity was observed upon Sam50 ablation in both murine and human myotubes, as measured with the XF24 Seahorse Analyzer. Collectively, these findings support the critical role of Sam50 in establishing and maintaining mitochondrial integrity, cristae structure, and mitochondrial metabolism. By elucidating the impact of Sam50-deficiency, this study enhances our understanding of mitochondrial function in skeletal muscle.
Collapse
Affiliation(s)
- Bryanna Shao
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Mason Killion
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Ashton Oliver
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Chia Vang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Faben Zeleke
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Edgar Garza-Lopez
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Jian-Qiang Shao
- Central Microscopy Research Facility, University of Iowa, Iowa City, Iowa, USA
| | - Margaret Mungai
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Jacob Lam
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, USA
| | - Qiana Williams
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Christopher T Altamura
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Aaron Whiteside
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, Ohio, USA
| | - Kinuthia Kabugi
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Jessica McKenzie
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Maria Ezedimma
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Han Le
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Alice Koh
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Estevão Scudese
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Larry Vang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Andrea G Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Amber Crabtree
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | | | - Dominique Stephens
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Ho-Jin Koh
- Department of Biological Sciences, Tennessee State University, Nashville, Tennessee, USA
| | - Brenita C Jenkins
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, USA
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Sandra A Murray
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Anthonya T Cooper
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA
| | - Clintoria Williams
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, Ohio, USA
| | - Steven M Damo
- Department of Life and Physical Sciences, Fisk University, Nashville, Tennessee, USA
| | - Melanie R McReynolds
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, Pennsylvania, USA
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Jennifer A Gaddy
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
- US Department of Veterans Affairs, Tennessee Valley Healthcare Systems, Nashville, Tennessee, USA
| | - Celestine N Wanjalla
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, USA
| | - Heather K Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, USA
| |
Collapse
|
19
|
Benaroya H. Mitochondria and MICOS - function and modeling. Rev Neurosci 2024; 35:503-531. [PMID: 38369708 DOI: 10.1515/revneuro-2024-0004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 01/14/2024] [Indexed: 02/20/2024]
Abstract
An extensive review is presented on mitochondrial structure and function, mitochondrial proteins, the outer and inner membranes, cristae, the role of F1FO-ATP synthase, the mitochondrial contact site and cristae organizing system (MICOS), the sorting and assembly machinery morphology and function, and phospholipids, in particular cardiolipin. Aspects of mitochondrial regulation under physiological and pathological conditions are outlined, in particular the role of dysregulated MICOS protein subunit Mic60 in Parkinson's disease, the relations between mitochondrial quality control and proteins, and mitochondria as signaling organelles. A mathematical modeling approach of cristae and MICOS using mechanical beam theory is introduced and outlined. The proposed modeling is based on the premise that an optimization framework can be used for a better understanding of critical mitochondrial function and also to better map certain experiments and clinical interventions.
Collapse
Affiliation(s)
- Haym Benaroya
- Department of Mechanical and Aerospace Engineering, Rutgers University, 98 Brett Road, Piscataway, NJ 08854, USA
| |
Collapse
|
20
|
Wisniewski BT, Lackner LL. Significantly reduced, but balanced, rates of mitochondrial fission and fusion are sufficient to maintain the integrity of yeast mitochondrial DNA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.18.604121. [PMID: 39071310 PMCID: PMC11275889 DOI: 10.1101/2024.07.18.604121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/30/2024]
Abstract
Mitochondria exist as dynamic tubular networks and the morphology of these networks impacts organelle function and cell health. Mitochondrial morphology is maintained in part by the opposing activities of mitochondrial fission and fusion. Mitochondrial fission and fusion are also required to maintain mitochondrial DNA (mtDNA) integrity. In Saccharomyces cerevisiae , the simultaneous inhibition of mitochondrial fission and fusion results in increased mtDNA mutation and the consequent loss of respiratory competence. The mechanism by which fission and fusion maintain mtDNA integrity is not fully understood. Previous work demonstrates that mtDNA is spatially linked to mitochondrial fission sites. Here, we extend this finding using live-cell imaging to localize mtDNA to mitochondrial fusion sites. While mtDNA is present at sites of mitochondrial fission and fusion, mitochondrial fission and fusion rates are not altered in cells lacking mtDNA. Using alleles that alter mitochondrial fission and fusion rates, we find that mtDNA integrity can be maintained in cells with significantly reduced, but balanced, rates of fission and fusion. In addition, we find that increasing mtDNA copy number reduces the loss of respiratory competence in double mitochondrial fission-fusion mutants. Our findings add novel insights into the relationship between mitochondrial dynamics and mtDNA integrity.
Collapse
|
21
|
Marada A, Walter C, Suhm T, Shankar S, Nandy A, Brummer T, Dhaouadi I, Vögtle FN, Meisinger C. DYRK1A signalling synchronizes the mitochondrial import pathways for metabolic rewiring. Nat Commun 2024; 15:5265. [PMID: 38902238 PMCID: PMC11189921 DOI: 10.1038/s41467-024-49611-4] [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: 09/19/2023] [Accepted: 06/12/2024] [Indexed: 06/22/2024] Open
Abstract
Mitochondria require an extensive proteome to maintain a variety of metabolic reactions, and changes in cellular demand depend on rapid adaptation of the mitochondrial protein composition. The TOM complex, the organellar entry gate for mitochondrial precursors in the outer membrane, is a target for cytosolic kinases to modulate protein influx. DYRK1A phosphorylation of the carrier import receptor TOM70 at Ser91 enables its efficient docking and thus transfer of precursor proteins to the TOM complex. Here, we probe TOM70 phosphorylation in molecular detail and find that TOM70 is not a CK2 target nor import receptor for MIC19 as previously suggested. Instead, we identify TOM20 as a MIC19 import receptor and show off-target inhibition of the DYRK1A-TOM70 axis with the clinically used CK2 inhibitor CX4945 which activates TOM20-dependent import pathways. Taken together, modulation of DYRK1A signalling adapts the central mitochondrial protein entry gate via synchronization of TOM70- and TOM20-dependent import pathways for metabolic rewiring. Thus, DYRK1A emerges as a cytosolic surveillance kinase to regulate and fine-tune mitochondrial protein biogenesis.
Collapse
Affiliation(s)
- Adinarayana Marada
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Corvin Walter
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
| | - Tamara Suhm
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Sahana Shankar
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Arpita Nandy
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
- Faculty of Biology, University of Freiburg, 79104, Freiburg, Germany
- Spemann Graduate School of Biology and Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - Tilman Brummer
- Institute of Molecular Medicine, ZBMZ, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany
- German Cancer Consortium DKTK Partner Site Freiburg, German Cancer Research Center (DKFZ), Heidelberg, Germany
| | - Ines Dhaouadi
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany
| | - F-Nora Vögtle
- Center for Molecular Biology of Heidelberg University (ZMBH), DKFZ-ZMBH Alliance, 69120, Heidelberg, Germany.
- Network Aging Research, Heidelberg University, 69120, Heidelberg, Germany.
- CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany.
| | - Chris Meisinger
- Institute of Biochemistry and Molecular Biology, ZBMZ, Faculty of Medicine, University of Freiburg, 79104, Freiburg, Germany.
- BIOSS Centre for Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany.
- CIBSS - Centre for Integrative Biological Signalling Studies, University of Freiburg, 79104, Freiburg, Germany.
| |
Collapse
|
22
|
Kumar A, Gok MO, Nguyen KN, Connor OM, Reese ML, Wideman JG, Muñoz-Gómez SA, Friedman JR. A dynamin superfamily-like pseudoenzyme coordinates with MICOS to promote cristae architecture. Curr Biol 2024; 34:2606-2622.e9. [PMID: 38692277 PMCID: PMC11187654 DOI: 10.1016/j.cub.2024.04.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 03/19/2024] [Accepted: 04/10/2024] [Indexed: 05/03/2024]
Abstract
Mitochondrial cristae architecture is crucial for optimal respiratory function of the organelle. Cristae shape is maintained in part by the mitochondrial contact site and cristae organizing system (MICOS) complex. While MICOS is required for normal cristae morphology, the precise mechanistic role of each of the seven human MICOS subunits, and how the complex coordinates with other cristae-shaping factors, has not been fully determined. Here, we examine the MICOS complex in Schizosaccharomyces pombe, a minimal model whose genome only encodes for four core subunits. Using an unbiased proteomics approach, we identify a poorly characterized inner mitochondrial membrane protein that interacts with MICOS and is required to maintain cristae morphology, which we name Mmc1. We demonstrate that Mmc1 works in concert with MICOS to promote normal mitochondrial morphology and respiratory function. Mmc1 is a distant relative of the dynamin superfamily of proteins (DSPs), GTPases, which are well established to shape and remodel membranes. Similar to DSPs, Mmc1 self-associates and forms high-molecular-weight assemblies. Interestingly, however, Mmc1 is a pseudoenzyme that lacks key residues required for GTP binding and hydrolysis, suggesting that it does not dynamically remodel membranes. These data are consistent with the model that Mmc1 stabilizes cristae architecture by acting as a scaffold to support cristae ultrastructure on the matrix side of the inner membrane. Our study reveals a new class of proteins that evolved early in fungal phylogeny and is required for the maintenance of cristae architecture. This highlights the possibility that functionally analogous proteins work with MICOS to establish cristae morphology in metazoans.
Collapse
Affiliation(s)
- Abhishek Kumar
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Mehmet Oguz Gok
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Kailey N Nguyen
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Olivia M Connor
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Michael L Reese
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jeremy G Wideman
- Center for Mechanisms of Evolution, Biodesign Institute, School of Life Sciences, Arizona State University, Tempe, AZ 85281, USA
| | - Sergio A Muñoz-Gómez
- Department of Biological Sciences, Purdue University, West Lafayette, IN 47907, USA
| | - Jonathan R Friedman
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
23
|
Vue Z, Prasad P, Le H, Neikirk K, Harris C, Garza-Lopez E, Wang E, Murphy A, Jenkins B, Vang L, Scudese E, Shao B, Kadam A, Shao J, Marshall AG, Crabtree A, Kirk B, Koh A, Wilson G, Oliver A, Rodman T, Kabugi K, Koh HJ, Smith Q, Zaganjor E, Wanjalla CN, Dash C, Evans C, Phillips MA, Hubert D, Ajijola O, Whiteside A, Do Koo Y, Kinder A, Demirci M, Albritton CF, Wandira N, Jamison S, Ahmed T, Saleem M, Tomar D, Williams CR, Sweetwyne MT, Murray SA, Cooper A, Kirabo A, Jadiya P, Quintana A, Katti P, Fu Dai D, McReynolds MR, Hinton A. The MICOS Complex Regulates Mitochondrial Structure and Oxidative Stress During Age-Dependent Structural Deficits in the Kidney. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.09.598108. [PMID: 38915644 PMCID: PMC11195114 DOI: 10.1101/2024.06.09.598108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/26/2024]
Abstract
The kidney filters nutrient waste and bodily fluids from the bloodstream, in addition to secondary functions of metabolism and hormone secretion, requiring an astonishing amount of energy to maintain its functions. In kidney cells, mitochondria produce adenosine triphosphate (ATP) and help maintain kidney function. Due to aging, the efficiency of kidney functions begins to decrease. Dysfunction in mitochondria and cristae, the inner folds of mitochondria, is a hallmark of aging. Therefore, age-related kidney function decline could be due to changes in mitochondrial ultrastructure, increased reactive oxygen species (ROS), and subsequent alterations in metabolism and lipid composition. We sought to understand if there is altered mitochondrial ultrastructure, as marked by 3D morphological changes, across time in tubular kidney cells. Serial block facing-scanning electron microscope (SBF-SEM) and manual segmentation using the Amira software were used to visualize murine kidney samples during the aging process at 3 months (young) and 2 years (old). We found that 2-year mitochondria are more fragmented, compared to the 3-month, with many uniquely shaped mitochondria observed across aging, concomitant with shifts in ROS, metabolomics, and lipid homeostasis. Furthermore, we show that the mitochondrial contact site and cristae organizing system (MICOS) complex is impaired in the kidney due to aging. Disruption of the MICOS complex shows altered mitochondrial calcium uptake and calcium retention capacity, as well as generation of oxidative stress. We found significant, detrimental structural changes to aged kidney tubule mitochondria suggesting a potential mechanism underlying why kidney diseases occur more readily with age. We hypothesize that disruption in the MICOS complex further exacerbates mitochondrial dysfunction, creating a vicious cycle of mitochondrial degradation and oxidative stress, thus impacting kidney health.
Collapse
Affiliation(s)
- Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Praveena Prasad
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA 16801
| | - Han Le
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Chanel Harris
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Edgar Garza-Lopez
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Eric Wang
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA, 92697, USA
| | - Alexandria Murphy
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA 16801
| | - Brenita Jenkins
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA 16801
| | - Larry Vang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Estevão Scudese
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Bryanna Shao
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Ashlesha Kadam
- Department of Internal Medicine, Section of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157 USA
| | - Jianqiang Shao
- Central Microscopy Research Facility, University of Iowa, Iowa City, IA, 52242, USA
| | - Andrea G. Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Amber Crabtree
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Benjamin Kirk
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Alice Koh
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Genesis Wilson
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Ashton Oliver
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Taylor Rodman
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Kinuthia Kabugi
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Ho-Jin Koh
- Department of Biological Sciences, Tennessee State University, Nashville, TN 37209, USA
| | - Quinton Smith
- Department of Chemical and Biomolecular Engineering, University of California, Irvine, CA, 92697, USA
| | - Elma Zaganjor
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | | | - Chandravanu Dash
- Department of Biochemistry, Cancer Biology, Pharmacology and Neuroscience, Meharry Medical College, Nashville, TN, United States
| | - Chantell Evans
- Department of Cell Biology, Duke University School of Medicine, Durham, NC, 27708, USA
| | - Mark A. Phillips
- Department of Integrative Biology, Oregon State University, Corvallis, OR, 97331, USA
| | - David Hubert
- Department of Integrative Biology, Oregon State University, Corvallis, OR, 97331, USA
| | - Olujimi Ajijola
- UCLA Cardiac Arrhythmia Center, University of California, Los Angeles, CA, USA
| | - Aaron Whiteside
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH 45435 USA
| | - Young Do Koo
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
- Fraternal Order of Eagles Diabetes Research Center, Iowa City, Iowa, USA
| | - André Kinder
- Artur Sá Earp Neto University Center - UNIFASE-FMP, Petrópolis Medical School, Brazil
| | - Mert Demirci
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Claude F. Albritton
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
- Department of Biomedical Sciences, School of Graduate Studies, Meharry Medical College, Nashville, TN 37208-3501, USA
| | - Nelson Wandira
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Sydney Jamison
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Taseer Ahmed
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Mohammad Saleem
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
| | - Dhanendra Tomar
- Department of Internal Medicine, Section of Cardiovascular Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157 USA
| | - Clintoria R. Williams
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH 45435 USA
| | - Mariya T. Sweetwyne
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, 98195, USA
| | - Sandra A. Murray
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Anthonya Cooper
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Annet Kirabo
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN 37232
- Vanderbilt Center for Immunobiology, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University, Nashville, TN, 37232, USA
- Vanderbilt Institute for Global Health, Vanderbilt University, Nashville, TN, 37232, USA
| | - Pooja Jadiya
- Department of Internal Medicine, Section of Gerontology and Geriatric Medicine, Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest University School of Medicine, Winston-Salem, NC
| | - Anita Quintana
- Department of Biological Sciences, Border Biomedical Research Center, The University of Texas at El Paso, El Paso, Texas, USA
| | - Prasanna Katti
- National Heart, Lung and Blood Institute, National Institutes of Health, 9000 Rockville Pike, Bethesda, MD 20892, USA
- Department of Biology, Indian Institute of Science Education and Research (IISER) Tirupati, AP, 517619, India
| | - Dao Fu Dai
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Melanie R. McReynolds
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life Sciences, Pennsylvania State University, State College, PA 16801
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| |
Collapse
|
24
|
Suomalainen A, Nunnari J. Mitochondria at the crossroads of health and disease. Cell 2024; 187:2601-2627. [PMID: 38788685 DOI: 10.1016/j.cell.2024.04.037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2024] [Revised: 04/25/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024]
Abstract
Mitochondria reside at the crossroads of catabolic and anabolic metabolism-the essence of life. How their structure and function are dynamically tuned in response to tissue-specific needs for energy, growth repair, and renewal is being increasingly understood. Mitochondria respond to intrinsic and extrinsic stresses and can alter cell and organismal function by inducing metabolic signaling within cells and to distal cells and tissues. Here, we review how the centrality of mitochondrial functions manifests in health and a broad spectrum of diseases and aging.
Collapse
Affiliation(s)
- Anu Suomalainen
- University of Helsinki, Stem Cells and Metabolism Program, Faculty of Medicine, Helsinki, Finland; HiLife, University of Helsinki, Helsinki, Finland; HUS Diagnostics, Helsinki University Hospital, Helsinki, Finland.
| | - Jodi Nunnari
- Altos Labs, Bay Area Institute, Redwood Shores, CA, USA.
| |
Collapse
|
25
|
Leventoux N, Morimoto S, Ishikawa M, Nakamura S, Ozawa F, Kobayashi R, Watanabe H, Supakul S, Okamoto S, Zhou Z, Kobayashi H, Kato C, Hirokawa Y, Aiba I, Takahashi S, Shibata S, Takao M, Yoshida M, Endo F, Yamanaka K, Kokubo Y, Okano H. Aberrant CHCHD2-associated mitochondriopathy in Kii ALS/PDC astrocytes. Acta Neuropathol 2024; 147:84. [PMID: 38750212 DOI: 10.1007/s00401-024-02734-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 02/28/2024] [Accepted: 04/15/2024] [Indexed: 05/25/2024]
Abstract
Amyotrophic Lateral Sclerosis/Parkinsonism-Dementia Complex (ALS/PDC), a rare and complex neurological disorder, is predominantly observed in the Western Pacific islands, including regions of Japan, Guam, and Papua. This enigmatic condition continues to capture medical attention due to affected patients displaying symptoms that parallel those seen in either classical amyotrophic lateral sclerosis (ALS) or Parkinson's disease (PD). Distinctly, postmortem examinations of the brains of affected individuals have shown the presence of α-synuclein aggregates and TDP-43, which are hallmarks of PD and classical ALS, respectively. These observations are further complicated by the detection of phosphorylated tau, accentuating the multifaceted proteinopathic nature of ALS/PDC. The etiological foundations of this disease remain undetermined, and genetic investigations have yet to provide conclusive answers. However, emerging evidence has implicated the contribution of astrocytes, pivotal cells for maintaining brain health, to neurodegenerative onset, and likely to play a significant role in the pathogenesis of ALS/PDC. Leveraging advanced induced pluripotent stem cell technology, our team cultivated multiple astrocyte lines to further investigate the Japanese variant of ALS/PDC (Kii ALS/PDC). CHCHD2 emerged as a significantly dysregulated gene when disease astrocytes were compared to healthy controls. Our analyses also revealed imbalances in the activation of specific pathways: those associated with astrocytic cilium dysfunction, known to be involved in neurodegeneration, and those related to major neurological disorders, including classical ALS and PD. Further in-depth examinations revealed abnormalities in the mitochondrial morphology and metabolic processes of the affected astrocytes. A particularly striking observation was the reduced expression of CHCHD2 in the spinal cord, motor cortex, and oculomotor nuclei of patients with Kii ALS/PDC. In summary, our findings suggest a potential reduction in the support Kii ALS/PDC astrocytes provide to neurons, emphasizing the need to explore the role of CHCHD2 in maintaining mitochondrial health and its implications for the disease.
Collapse
Affiliation(s)
- Nicolas Leventoux
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Satoru Morimoto
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Keio Regenerative Medicine Research Centre, Keio University, Kanagawa, Japan
- Division of Neurodegenerative Disease Research, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
- Department of Oncologic Pathology, Mie University Graduate School of Medicine, Mie, Japan
| | - Mitsuru Ishikawa
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Shiho Nakamura
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Keio Regenerative Medicine Research Centre, Keio University, Kanagawa, Japan
- Division of Neurodegenerative Disease Research, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Fumiko Ozawa
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Keio Regenerative Medicine Research Centre, Keio University, Kanagawa, Japan
- Division of Neurodegenerative Disease Research, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Reona Kobayashi
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Hirotaka Watanabe
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Keio Regenerative Medicine Research Centre, Keio University, Kanagawa, Japan
| | - Sopak Supakul
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Satoshi Okamoto
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Zhi Zhou
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Hiroya Kobayashi
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Keio Regenerative Medicine Research Centre, Keio University, Kanagawa, Japan
- Division of Neurodegenerative Disease Research, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Chris Kato
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Keio Regenerative Medicine Research Centre, Keio University, Kanagawa, Japan
- Division of Neurodegenerative Disease Research, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan
| | - Yoshifumi Hirokawa
- Department of Oncologic Pathology, Mie University Graduate School of Medicine, Mie, Japan
| | - Ikuko Aiba
- Department of Neurology, NHO, Higashinagoya National Hospital, Aichi, Japan
| | - Shinichi Takahashi
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Keio Regenerative Medicine Research Centre, Keio University, Kanagawa, Japan
- Department of Neurology and Stroke, International Medical Centre, Saitama Medical University, Saitama, Japan
| | - Shinsuke Shibata
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
- Division of Microscopic Anatomy, Graduate School of Medical and Dental Sciences, Niigata University, Niigata, Japan
| | - Masaki Takao
- Department of Clinical Laboratory, National Centre of Neurology and Psychiatry (NCNP), Tokyo, Japan
| | - Mari Yoshida
- Department of Neuropathology, Institute for Medical Science of Aging, Aichi Medical University, Aichi, Japan
| | - Fumito Endo
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Aichi, Japan
| | - Koji Yamanaka
- Department of Neuroscience and Pathobiology, Research Institute of Environmental Medicine, Nagoya University, Aichi, Japan
| | - Yasumasa Kokubo
- Kii ALS/PDC Research Centre, Mie University Graduate School of Regional Innovation Studies, Mie, Japan.
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan.
- Keio Regenerative Medicine Research Centre, Keio University, Kanagawa, Japan.
- Division of Neurodegenerative Disease Research, Tokyo Metropolitan Institute for Geriatrics and Gerontology, Tokyo, Japan.
| |
Collapse
|
26
|
Li D, Gao X, Ma X, Wang M, Cheng C, Xue T, Gao F, Shen Y, Zhang J, Liu Q. Aging-induced tRNA Glu-derived fragment impairs glutamate biosynthesis by targeting mitochondrial translation-dependent cristae organization. Cell Metab 2024; 36:1059-1075.e9. [PMID: 38458203 DOI: 10.1016/j.cmet.2024.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 10/31/2023] [Accepted: 02/15/2024] [Indexed: 03/10/2024]
Abstract
Mitochondrial cristae, infoldings of the mitochondrial inner membrane, undergo aberrant changes in their architecture with age. However, the underlying molecular mechanisms and their contribution to brain aging are largely elusive. Here, we observe an age-dependent accumulation of Glu-5'tsRNA-CTC, a transfer-RNA-derived small RNA (tsRNA), derived from nuclear-encoded tRNAGlu in the mitochondria of glutaminergic neurons. Mitochondrial Glu-5'tsRNA-CTC disrupts the binding of mt-tRNALeu and leucyl-tRNA synthetase2 (LaRs2), impairing mt-tRNALeu aminoacylation and mitochondria-encoded protein translation. Mitochondrial translation defects disrupt cristae organization, leading to damaged glutaminase (GLS)-dependent glutamate formation and reduced synaptosomal glutamate levels. Moreover, reduction of Glu-5'tsRNA-CTC protects aged brains from age-related defects in mitochondrial cristae organization, glutamate metabolism, synaptic structures, and memory. Thus, beyond illustrating a physiological role for normal mitochondrial cristae ultrastructure in maintaining glutamate levels, our study defines a pathological role for tsRNAs in brain aging and age-related memory decline.
Collapse
Affiliation(s)
- Dingfeng Li
- Department of Neurology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China; Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Xinyi Gao
- Department of Neurology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China; Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Xiaolin Ma
- Department of Neurology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China; Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Ming Wang
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Chuandong Cheng
- Department of Neurosurgery, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Tian Xue
- Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China
| | - Feng Gao
- Department of Neurology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China
| | - Yong Shen
- Department of Neurology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China; Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Neurodegenerative Disorder Research Center, Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei 230026, China
| | - Juan Zhang
- Department of Neurology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China; Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China.
| | - Qiang Liu
- Department of Neurology, The First Affiliated Hospital of USTC, Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, Hefei National Research Center for Physical Sciences at the Microscale, University of Science and Technology of China, Hefei 230001, China; Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; CAS Key Laboratory of Brain Function and Diseases, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230001, China; Neurodegenerative Disorder Research Center, Anhui Province Key Laboratory of Biomedical Aging Research, University of Science and Technology of China, Hefei 230026, China; Key Laboratory of Immune Response and Immunotherapy, University of Science and Technology of China, Hefei 230001, China.
| |
Collapse
|
27
|
Fuentes JM, Morcillo P. The Role of Cardiolipin in Mitochondrial Function and Neurodegenerative Diseases. Cells 2024; 13:609. [PMID: 38607048 PMCID: PMC11012098 DOI: 10.3390/cells13070609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 03/28/2024] [Accepted: 03/29/2024] [Indexed: 04/13/2024] Open
Abstract
Cardiolipin (CL) is a mitochondria-exclusive phospholipid synthesized in the inner mitochondrial membrane. CL plays a key role in mitochondrial membranes, impacting a plethora of functions this organelle performs. Consequently, it is conceivable that abnormalities in the CL content, composition, and level of oxidation may negatively impact mitochondrial function and dynamics, with important implications in a variety of diseases. This review concentrates on papers published in recent years, combined with basic and underexplored research in CL. We capture new findings on its biological functions in the mitochondria, as well as its association with neurodegenerative diseases such as Alzheimer's disease or Parkinson's disease. Lastly, we explore the potential applications of CL as a biomarker and pharmacological target to mitigate mitochondrial dysfunction.
Collapse
Affiliation(s)
- José M. Fuentes
- Departamento de Bioquímica y Biología Molecular y Genética, Facultad de Enfermería y Terapia Ocupacional, Universidad de Extremadura, 10003 Cáceres, Spain
- Centro de Investigación Biomédica en Red en Enfermedades Neurodegenerativas, Instituto de Salud Carlos III (CIBER-CIBERNED-ISCIII), 28029 Madrid, Spain
- Instituto Universitario de Investigación Biosanitaria de Extremadura (INUBE), 10003 Cáceres, Spain
| | - Patricia Morcillo
- Departmentof Neurology, Columbia University, New York, NY 10032, USA
| |
Collapse
|
28
|
Kumar M, Sharma S, Kumar J, Barik S, Mazumder S. Mitochondrial electron transport chain in macrophage reprogramming: Potential role in antibacterial immune response. CURRENT RESEARCH IN IMMUNOLOGY 2024; 5:100077. [PMID: 38572399 PMCID: PMC10987323 DOI: 10.1016/j.crimmu.2024.100077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/21/2024] [Accepted: 03/22/2024] [Indexed: 04/05/2024] Open
Abstract
Macrophages restrain microbial infection and reinstate tissue homeostasis. The mitochondria govern macrophage metabolism and serve as pivot in innate immunity, thus acting as immunometabolic regulon. Metabolic pathways produce electron flows that end up in mitochondrial electron transport chain (mtETC), made of super-complexes regulating multitude of molecular and biochemical processes. Cell-intrinsic and extrinsic factors influence mtETC structure and function, impacting several aspects of macrophage immunity. These factors provide the macrophages with alternate fuel sources and metabolites, critical to gain functional competence and overcoming pathogenic stress. Mitochondrial reactive oxygen species (mtROS) and oxidative phosphorylation (OXPHOS) generated through the mtETC are important innate immune attributes, which help macrophages in mounting antibacterial responses. Recent studies have demonstrated the role of mtETC in governing mitochondrial dynamics and macrophage polarization (M1/M2). M1 macrophages are important for containing bacterial pathogens and M2 macrophages promote tissue repair and wound healing. Thus, mitochondrial bioenergetics and metabolism are intimately coupled with innate immunity. In this review, we have addressed mtETC function as innate rheostats that regulate macrophage reprogramming and innate immune responses. Advancement in this field encourages further exploration and provides potential novel macrophage-based therapeutic targets to control unsolicited inflammation.
Collapse
Affiliation(s)
- Manmohan Kumar
- Department of Laboratory Medicine, University of California, San Francisco, San Francisco, CA 94143, USA
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Shagun Sharma
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
- Department of Zoology, Gargi College, University of Delhi, Delhi, India
| | - Jai Kumar
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
| | - Sailen Barik
- EonBio, 3780 Pelham Drive, Mobile, AL 36619, USA
| | - Shibnath Mazumder
- Immunobiology Laboratory, Department of Zoology, University of Delhi, Delhi, India
- Faculty of Life Sciences and Biotechnology, South Asian University, Delhi, India
| |
Collapse
|
29
|
Mise K, Long J, Galvan DL, Ye Z, Fan G, Sharma R, Serysheva II, Moore TI, Jeter CR, Anna Zal M, Araki M, Wada J, Schumacker PT, Chang BH, Danesh FR. NDUFS4 regulates cristae remodeling in diabetic kidney disease. Nat Commun 2024; 15:1965. [PMID: 38438382 PMCID: PMC10912198 DOI: 10.1038/s41467-024-46366-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Accepted: 02/22/2024] [Indexed: 03/06/2024] Open
Abstract
The mitochondrial electron transport chain (ETC) is a highly adaptive process to meet metabolic demands of the cell, and its dysregulation has been associated with diverse clinical pathologies. However, the role and nature of impaired ETC in kidney diseases remains poorly understood. Here, we generate diabetic mice with podocyte-specific overexpression of Ndufs4, an accessory subunit of mitochondrial complex I, as a model investigate the role of ETC integrity in diabetic kidney disease (DKD). We find that conditional male mice with genetic overexpression of Ndufs4 exhibit significant improvements in cristae morphology, mitochondrial dynamics, and albuminuria. By coupling proximity labeling with super-resolution imaging, we also identify the role of cristae shaping protein STOML2 in linking NDUFS4 with improved cristae morphology. Together, we provide the evidence on the central role of NDUFS4 as a regulator of cristae remodeling and mitochondrial function in kidney podocytes. We propose that targeting NDUFS4 represents a promising approach to slow the progression of DKD.
Collapse
Affiliation(s)
- Koki Mise
- Section of Nephrology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
- Department of Nephrology, Rheumatology, Endocrinology & Metabolism, Okayama University Graduate School of Medicine, Dentistry & Pharmaceutical Sciences, Okayama, Japan
| | - Jianyin Long
- Section of Nephrology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Daniel L Galvan
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Zengchun Ye
- Division of Nephrology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Guizhen Fan
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Rajesh Sharma
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Irina I Serysheva
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Travis I Moore
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Collene R Jeter
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - M Anna Zal
- Department of Epigenetics and Molecular Carcinogenesis, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Motoo Araki
- Department of Urology, Okayama University Graduate School of Medicine, Dentistry & Pharmaceutical Sciences, Okayama, Japan
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology & Metabolism, Okayama University Graduate School of Medicine, Dentistry & Pharmaceutical Sciences, Okayama, Japan
| | - Paul T Schumacker
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Benny H Chang
- Section of Nephrology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Farhad R Danesh
- Section of Nephrology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
- Department of Biochemistry and Molecular Pharmacology, Baylor College of Medicine, Houston, TX, USA.
| |
Collapse
|
30
|
Ren X, Shi P, Su J, Wei T, Li J, Hu Y, Wu C. Loss of Myo19 increases metastasis by enhancing microenvironmental ROS gradient and chemotaxis. EMBO Rep 2024; 25:971-990. [PMID: 38279020 PMCID: PMC10933354 DOI: 10.1038/s44319-023-00052-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: 06/10/2023] [Revised: 12/10/2023] [Accepted: 12/19/2023] [Indexed: 01/28/2024] Open
Abstract
Tumor metastasis involves cells migrating directionally in response to external chemical signals. Reactive oxygen species (ROS) in the form of H2O2 has been demonstrated as a chemoattractant for neutrophils but its spatial characteristics in tumor microenvironment and potential role in tumor cell dissemination remain unknown. Here we investigate the spatial ROS distribution in 3D tumor spheroids and identify a ROS concentration gradient in spheroid periphery, which projects into a H2O2 gradient in tumor microenvironment. We further reveal the role of H2O2 gradient to induce chemotaxis of tumor cells by activating Src and subsequently inhibiting RhoA. Finally, we observe that the absence of mitochondria cristae remodeling proteins including the mitochondria-localized actin motor Myosin 19 (Myo19) enhances ROS gradient and promotes tumor dissemination. Myo19 downregulation is seen in many tumors, and Myo19 expression is negatively associated with tumor metastasis in vivo. Together, our study reveals the chemoattractant role of tumor microenvironmental ROS and implies the potential impact of mitochondria cristae disorganization on tumor invasion and metastasis.
Collapse
Affiliation(s)
- Xiaoyu Ren
- Institute of Systems Biomedicine, Peking University Health Science Center, Key Laboratory of Tumor Systems Biology, Beijing, 100191, China
| | - Peng Shi
- Institute of Systems Biomedicine, Peking University Health Science Center, Key Laboratory of Tumor Systems Biology, Beijing, 100191, China.
- International Cancer Institute, Peking University, Beijing, 100191, China.
| | - Jing Su
- Department of Pathology, School of Basic Medical Sciences, Peking University Third Hospital, Peking University Health Science Center, Beijing, 100191, China
| | - Tonghua Wei
- Institute of Systems Biomedicine, Peking University Health Science Center, Key Laboratory of Tumor Systems Biology, Beijing, 100191, China
| | - Jiayi Li
- Institute of Systems Biomedicine, Peking University Health Science Center, Key Laboratory of Tumor Systems Biology, Beijing, 100191, China
| | - Yiping Hu
- Institute of Systems Biomedicine, Peking University Health Science Center, Key Laboratory of Tumor Systems Biology, Beijing, 100191, China
| | - Congying Wu
- Institute of Systems Biomedicine, Peking University Health Science Center, Key Laboratory of Tumor Systems Biology, Beijing, 100191, China.
- International Cancer Institute, Peking University, Beijing, 100191, China.
| |
Collapse
|
31
|
Murphy MP, O'Neill LAJ. A break in mitochondrial endosymbiosis as a basis for inflammatory diseases. Nature 2024; 626:271-279. [PMID: 38326590 DOI: 10.1038/s41586-023-06866-z] [Citation(s) in RCA: 39] [Impact Index Per Article: 39.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2023] [Accepted: 11/14/2023] [Indexed: 02/09/2024]
Abstract
Mitochondria retain bacterial traits due to their endosymbiotic origin, but host cells do not recognize them as foreign because the organelles are sequestered. However, the regulated release of mitochondrial factors into the cytosol can trigger cell death, innate immunity and inflammation. This selective breakdown in the 2-billion-year-old endosymbiotic relationship enables mitochondria to act as intracellular signalling hubs. Mitochondrial signals include proteins, nucleic acids, phospholipids, metabolites and reactive oxygen species, which have many modes of release from mitochondria, and of decoding in the cytosol and nucleus. Because these mitochondrial signals probably contribute to the homeostatic role of inflammation, dysregulation of these processes may lead to autoimmune and inflammatory diseases. A potential reason for the increased incidence of these diseases may be changes in mitochondrial function and signalling in response to such recent phenomena as obesity, dietary changes and other environmental factors. Focusing on the mixed heritage of mitochondria therefore leads to predictions for future insights, research paths and therapeutic opportunities. Thus, whereas mitochondria can be considered 'the enemy within' the cell, evolution has used this strained relationship in intriguing ways, with increasing evidence pointing to the recent failure of endosymbiosis being critical for the pathogenesis of inflammatory diseases.
Collapse
Affiliation(s)
- Michael P Murphy
- MRC Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK.
- Department of Medicine, University of Cambridge, Cambridge, UK.
| | - Luke A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
32
|
Vue Z, Garza‐Lopez E, Neikirk K, Katti P, Vang L, Beasley H, Shao J, Marshall AG, Crabtree A, Murphy AC, Jenkins BC, Prasad P, Evans C, Taylor B, Mungai M, Killion M, Stephens D, Christensen TA, Lam J, Rodriguez B, Phillips MA, Daneshgar N, Koh H, Koh A, Davis J, Devine N, Saleem M, Scudese E, Arnold KR, Vanessa Chavarin V, Daniel Robinson R, Chakraborty M, Gaddy JA, Sweetwyne MT, Wilson G, Zaganjor E, Kezos J, Dondi C, Reddy AK, Glancy B, Kirabo A, Quintana AM, Dai D, Ocorr K, Murray SA, Damo SM, Exil V, Riggs B, Mobley BC, Gomez JA, McReynolds MR, Hinton A. 3D reconstruction of murine mitochondria reveals changes in structure during aging linked to the MICOS complex. Aging Cell 2023; 22:e14009. [PMID: 37960952 PMCID: PMC10726809 DOI: 10.1111/acel.14009] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Revised: 09/01/2023] [Accepted: 09/19/2023] [Indexed: 11/15/2023] Open
Abstract
During aging, muscle gradually undergoes sarcopenia, the loss of function associated with loss of mass, strength, endurance, and oxidative capacity. However, the 3D structural alterations of mitochondria associated with aging in skeletal muscle and cardiac tissues are not well described. Although mitochondrial aging is associated with decreased mitochondrial capacity, the genes responsible for the morphological changes in mitochondria during aging are poorly characterized. We measured changes in mitochondrial morphology in aged murine gastrocnemius, soleus, and cardiac tissues using serial block-face scanning electron microscopy and 3D reconstructions. We also used reverse transcriptase-quantitative PCR, transmission electron microscopy quantification, Seahorse analysis, and metabolomics and lipidomics to measure changes in mitochondrial morphology and function after loss of mitochondria contact site and cristae organizing system (MICOS) complex genes, Chchd3, Chchd6, and Mitofilin. We identified significant changes in mitochondrial size in aged murine gastrocnemius, soleus, and cardiac tissues. We found that both age-related loss of the MICOS complex and knockouts of MICOS genes in mice altered mitochondrial morphology. Given the critical role of mitochondria in maintaining cellular metabolism, we characterized the metabolomes and lipidomes of young and aged mouse tissues, which showed profound alterations consistent with changes in membrane integrity, supporting our observations of age-related changes in muscle tissues. We found a relationship between changes in the MICOS complex and aging. Thus, it is important to understand the mechanisms that underlie the tissue-dependent 3D mitochondrial phenotypic changes that occur in aging and the evolutionary conservation of these mechanisms between Drosophila and mammals.
Collapse
Affiliation(s)
- Zer Vue
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | | | - Kit Neikirk
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - Prasanna Katti
- National Heart, Lung and Blood Institute, National Institutes of HealthMarylandBethesdaUSA
| | - Larry Vang
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - Heather Beasley
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - Jianqiang Shao
- Central Microscopy Research FacilityUniversity of IowaIowaIowa CityUSA
| | - Andrea G. Marshall
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - Amber Crabtree
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - Alexandria C. Murphy
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life SciencesPennsylvania State UniversityPennsylvaniaState CollegeUSA
| | - Brenita C. Jenkins
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life SciencesPennsylvania State UniversityPennsylvaniaState CollegeUSA
| | - Praveena Prasad
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life SciencesPennsylvania State UniversityPennsylvaniaState CollegeUSA
| | - Chantell Evans
- Department of Cell BiologyDuke University School of MedicineNorth CarolinaDurhamUSA
| | - Brittany Taylor
- J. Crayton Pruitt Family Department of Biomedical EngineeringUniversity of FloridaFloridaGainesvilleUSA
| | - Margaret Mungai
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - Mason Killion
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - Dominique Stephens
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | | | - Jacob Lam
- Department of Internal MedicineUniversity of IowaIowaIowa CityUSA
| | | | - Mark A. Phillips
- Department of Integrative BiologyOregon State UniversityOregonCorvallisUSA
| | - Nastaran Daneshgar
- Department of Integrative BiologyOregon State UniversityOregonCorvallisUSA
| | - Ho‐Jin Koh
- Department of Biological SciencesTennessee State UniversityTennesseeNashvilleUSA
| | - Alice Koh
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
- Department of MedicineVanderbilt University Medical CenterTennesseeNashvilleUSA
| | - Jamaine Davis
- Department of Biochemistry, Cancer Biology, Neuroscience, and PharmacologyMeharry Medical CollegeTennesseeNashvilleUSA
| | - Nina Devine
- Department of Integrative BiologyOregon State UniversityOregonCorvallisUSA
| | - Mohammad Saleem
- Department of MedicineVanderbilt University Medical CenterTennesseeNashvilleUSA
| | - Estevão Scudese
- Laboratory of Biosciences of Human Motricity (LABIMH) of the Federal University of State of Rio de Janeiro (UNIRIO)Rio de JaneiroBrazil
- Sport Sciences and Exercise Laboratory (LaCEE)Catholic University of Petrópolis (UCP)PetrópolisState of Rio de JaneiroBrazil
| | - Kenneth Ryan Arnold
- Department of Ecology and Evolutionary BiologyUniversity of California at IrvineCaliforniaIrvineUSA
| | - Valeria Vanessa Chavarin
- Department of Ecology and Evolutionary BiologyUniversity of California at IrvineCaliforniaIrvineUSA
| | - Ryan Daniel Robinson
- Department of Ecology and Evolutionary BiologyUniversity of California at IrvineCaliforniaIrvineUSA
| | | | - Jennifer A. Gaddy
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
- Department of MedicineVanderbilt University Medical CenterTennesseeNashvilleUSA
- Department of Medicine Health and SocietyVanderbilt UniversityTennesseeNashvilleUSA
- Department of Pathology, Microbiology and ImmunologyVanderbilt University Medical CenterTennesseeNashvilleUSA
- Department of Veterans AffairsTennessee Valley Healthcare SystemsTennesseeNashvilleUSA
| | - Mariya T. Sweetwyne
- Department of Laboratory Medicine and PathologyUniversity of WashingtonWashingtonSeattleUSA
| | - Genesis Wilson
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - Elma Zaganjor
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| | - James Kezos
- Sanford Burnham Prebys Medical Discovery InstituteCaliforniaLa JollaUSA
| | - Cristiana Dondi
- Sanford Burnham Prebys Medical Discovery InstituteCaliforniaLa JollaUSA
| | | | - Brian Glancy
- National Heart, Lung and Blood Institute, National Institutes of HealthMarylandBethesdaUSA
- National Institute of Arthritis and Musculoskeletal and Skin Diseases, National Institutes of HealthMarylandBethesdaUSA
| | - Annet Kirabo
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
- Department of MedicineVanderbilt University Medical CenterTennesseeNashvilleUSA
| | - Anita M. Quintana
- Department of Biological Sciences, Border Biomedical Research CenterUniversity of Texas at El PasoTexasEl PasoUSA
| | - Dao‐Fu Dai
- Department of PathologyUniversity of Johns Hopkins School of MedicineMarylandBaltimoreUSA
| | - Karen Ocorr
- Sanford Burnham Prebys Medical Discovery InstituteCaliforniaLa JollaUSA
| | - Sandra A. Murray
- Department of Cell Biology, School of MedicineUniversity of PittsburghPennsylvaniaPittsburghUSA
| | - Steven M. Damo
- Department of Life and Physical SciencesFisk UniversityTennesseeNashvilleUSA
- Center for Structural BiologyVanderbilt UniversityTennesseeNashvilleUSA
| | - Vernat Exil
- Department of Pediatrics, Carver College of MedicineUniversity of IowaIowaIowa CityUSA
- Department of Pediatrics, Division of CardiologySt. Louis University School of MedicineMissouriSt. LouisUSA
| | - Blake Riggs
- Department of BiologySan Francisco State UniversityCaliforniaSan FranciscoUSA
| | - Bret C. Mobley
- Department of PathologyVanderbilt University Medical CenterTennesseeNashvilleUSA
| | - Jose A. Gomez
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
- Department of MedicineVanderbilt University Medical CenterTennesseeNashvilleUSA
| | - Melanie R. McReynolds
- Department of Biochemistry and Molecular Biology, The Huck Institute of the Life SciencesPennsylvania State UniversityPennsylvaniaState CollegeUSA
| | - Antentor Hinton
- Department of Molecular Physiology and BiophysicsVanderbilt UniversityTennesseeNashvilleUSA
| |
Collapse
|
33
|
Vue Z, Neikirk K, Vang L, Garza-Lopez E, Christensen TA, Shao J, Lam J, Beasley HK, Marshall AG, Crabtree A, Anudokem J, Rodriguez B, Kirk B, Bacevac S, Barongan T, Shao B, Stephens DC, Kabugi K, Koh HJ, Koh A, Evans CS, Taylor B, Reddy AK, Miller-Fleming T, Actkins KV, Zaganjor E, Daneshgar N, Murray SA, Mobley BC, Damo SM, Gaddy JA, Riggs B, Wanjalla C, Kirabo A, McReynolds M, Gomez JA, Phillips MA, Exil V, Dai DF, Hinton A. Three-dimensional mitochondria reconstructions of murine cardiac muscle changes in size across aging. Am J Physiol Heart Circ Physiol 2023; 325:H965-H982. [PMID: 37624101 PMCID: PMC10977873 DOI: 10.1152/ajpheart.00202.2023] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 07/26/2023] [Accepted: 08/12/2023] [Indexed: 08/26/2023]
Abstract
With sparse treatment options, cardiac disease remains a significant cause of death among humans. As a person ages, mitochondria breakdown and the heart becomes less efficient. Heart failure is linked to many mitochondria-associated processes, including endoplasmic reticulum stress, mitochondrial bioenergetics, insulin signaling, autophagy, and oxidative stress. The roles of key mitochondrial complexes that dictate the ultrastructure, such as the mitochondrial contact site and cristae organizing system (MICOS), in aging cardiac muscle are poorly understood. To better understand the cause of age-related alteration in mitochondrial structure in cardiac muscle, we used transmission electron microscopy (TEM) and serial block facing-scanning electron microscopy (SBF-SEM) to quantitatively analyze the three-dimensional (3-D) networks in cardiac muscle samples of male mice at aging intervals of 3 mo, 1 yr, and 2 yr. Here, we present the loss of cristae morphology, the inner folds of the mitochondria, across age. In conjunction with this, the three-dimensional (3-D) volume of mitochondria decreased. These findings mimicked observed phenotypes in murine cardiac fibroblasts with CRISPR/Cas9 knockout of Mitofilin, Chchd3, Chchd6 (some members of the MICOS complex), and Opa1, which showed poorer oxidative consumption rate and mitochondria with decreased mitochondrial length and volume. In combination, these data show the need to explore if loss of the MICOS complex in the heart may be involved in age-associated mitochondrial and cristae structural changes.NEW & NOTEWORTHY This article shows how mitochondria in murine cardiac changes, importantly elucidating age-related changes. It also is the first to show that the MICOS complex may play a role in outer membrane mitochondrial structure.
Collapse
Affiliation(s)
- Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Larry Vang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Edgar Garza-Lopez
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Trace A Christensen
- Microscopy and Cell Analysis Core Facility, Mayo Clinic, Rochester, Minnesota, United States
| | - Jianqiang Shao
- Central Microscopy Research Facility, University of Iowa, Iowa City, Iowa, United States
| | - Jacob Lam
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Heather K Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Andrea G Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Amber Crabtree
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Josephs Anudokem
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Benjamin Rodriguez
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Benjamin Kirk
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Serif Bacevac
- Department of Internal Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Taylor Barongan
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Bryanna Shao
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Dominique C Stephens
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
- Department of Life and Physical Sciences, Fisk University, Nashville, Tennessee, United States
| | - Kinuthia Kabugi
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Ho-Jin Koh
- Department of Biological Sciences, Tennessee State University, Nashville, Tennessee, United States
| | - Alice Koh
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Chantell S Evans
- Department of Cell Biology, Duke University School of Medicine, Durham, North Carolina, United States
| | - Brittany Taylor
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida, United States
| | - Anilkumar K Reddy
- Department of Medicine, Baylor College of Medicine, Houston, Texas, United States
| | - Tyne Miller-Fleming
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Ky'Era V Actkins
- Division of Genetic Medicine, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Elma Zaganjor
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| | - Nastaran Daneshgar
- Department of Pathology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Sandra A Murray
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, United States
| | - Bret C Mobley
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Steven M Damo
- Department of Life and Physical Sciences, Fisk University, Nashville, Tennessee, United States
| | - Jennifer A Gaddy
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
- Tennessee Valley Healthcare Systems, United States Department of Veterans Affairs, Nashville, Tennessee, United States
| | - Blake Riggs
- Department of Biology at San Francisco State University, San Francisco, California, United States
| | - Celestine Wanjalla
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Annet Kirabo
- Division of Clinical Pharmacology, Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Melanie McReynolds
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, State College, Pennsylvania, United States
| | - Jose A Gomez
- Department of Medicine, Vanderbilt University Medical Center, Nashville, Tennessee, United States
| | - Mark A Phillips
- Department of Integrative Biology, Oregon State University, Corvallis, Oregon, United States
| | - Vernat Exil
- Division of Cardiology, Department of Pediatrics, St. Louis University School of Medicine, St. Louis, Missouri, United States
- Department of Pediatrics, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States
| | - Dao-Fu Dai
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, Tennessee, United States
| |
Collapse
|
34
|
Su H, Guo H, Qiu X, Lin TY, Qin C, Celio G, Yong P, Senders M, Han X, Bernlohr DA, Chen X. Lipocalin 2 regulates mitochondrial phospholipidome remodeling, dynamics, and function in brown adipose tissue in male mice. Nat Commun 2023; 14:6729. [PMID: 37872178 PMCID: PMC10593768 DOI: 10.1038/s41467-023-42473-2] [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: 11/17/2022] [Accepted: 10/11/2023] [Indexed: 10/25/2023] Open
Abstract
Mitochondrial function is vital for energy metabolism in thermogenic adipocytes. Impaired mitochondrial bioenergetics in brown adipocytes are linked to disrupted thermogenesis and energy balance in obesity and aging. Phospholipid cardiolipin (CL) and phosphatidic acid (PA) jointly regulate mitochondrial membrane architecture and dynamics, with mitochondria-associated endoplasmic reticulum membranes (MAMs) serving as the platform for phospholipid biosynthesis and metabolism. However, little is known about the regulators of MAM phospholipid metabolism and their connection to mitochondrial function. We discover that LCN2 is a PA binding protein recruited to the MAM during inflammation and metabolic stimulation. Lcn2 deficiency disrupts mitochondrial fusion-fission balance and alters the acyl-chain composition of mitochondrial phospholipids in brown adipose tissue (BAT) of male mice. Lcn2 KO male mice exhibit an increase in the levels of CLs containing long-chain polyunsaturated fatty acids (LC-PUFA), a decrease in CLs containing monounsaturated fatty acids, resulting in mitochondrial dysfunction. This dysfunction triggers compensatory activation of peroxisomal function and the biosynthesis of LC-PUFA-containing plasmalogens in BAT. Additionally, Lcn2 deficiency alters PA production, correlating with changes in PA-regulated phospholipid-metabolizing enzymes and the mTOR signaling pathway. In conclusion, LCN2 plays a critical role in the acyl-chain remodeling of phospholipids and mitochondrial bioenergetics by regulating PA production and its function in activating signaling pathways.
Collapse
Affiliation(s)
- Hongming Su
- Department of Food Science and Nutrition, University of Minnesota-Twin Cities, St. Paul, MN, 55108, USA
| | - Hong Guo
- Department of Food Science and Nutrition, University of Minnesota-Twin Cities, St. Paul, MN, 55108, USA
| | - Xiaoxue Qiu
- Department of Food Science and Nutrition, University of Minnesota-Twin Cities, St. Paul, MN, 55108, USA
| | - Te-Yueh Lin
- Department of Food Science and Nutrition, University of Minnesota-Twin Cities, St. Paul, MN, 55108, USA
| | - Chao Qin
- Barshop Institute for Longevity and Aging Studies, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229-3900, USA
| | - Gail Celio
- University Imaging Centers, University of Minnesota-Twin Cities, Minneapolis, MN, 55455, USA
| | - Peter Yong
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN, 55455, USA
| | - Mark Senders
- University Imaging Centers, University of Minnesota-Twin Cities, Minneapolis, MN, 55455, USA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, Department of Medicine, University of Texas Health Science Center at San Antonio, San Antonio, TX, 78229-3900, USA
| | - David A Bernlohr
- Department of Biochemistry, Molecular Biology and Biophysics, University of Minnesota-Twin Cities, Minneapolis, MN, 55455, USA
| | - Xiaoli Chen
- Department of Food Science and Nutrition, University of Minnesota-Twin Cities, St. Paul, MN, 55108, USA.
| |
Collapse
|
35
|
Shao B, Killion M, Oliver A, Vang C, Zeleke F, Neikirk K, Vue Z, Garza-Lopez E, Shao JQ, Mungai M, Lam J, Williams Q, Altamura CT, Whiteside A, Kabugi K, McKenzie J, Koh A, Scudese E, Vang L, Marshall AG, Crabtree A, Tanghal JI, Stephens D, Koh HJ, Jenkins BC, Murray SA, Cooper AT, Williams C, Damo SM, McReynolds MR, Gaddy JA, Wanjalla CN, Beasley HK, Hinton A. Ablation of Sam50 is associated with fragmentation and alterations in metabolism in murine and human myotubes. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.05.20.541602. [PMID: 37292887 PMCID: PMC10245823 DOI: 10.1101/2023.05.20.541602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/10/2023]
Abstract
The Sorting and Assembly Machinery (SAM) Complex is responsible for assembling β-barrel proteins in the mitochondrial membrane. Comprising three subunits, Sam35, Sam37, and Sam50, the SAM complex connects the inner and outer mitochondrial membranes by interacting with the mitochondrial contact site and cristae organizing system (MICOS) complex. Sam50, in particular, stabilizes the mitochondrial intermembrane space bridging (MIB) complex, which is crucial for protein transport, respiratory chain complex assembly, and regulation of cristae integrity. While the role of Sam50 in mitochondrial structure and metabolism in skeletal muscle remains unclear, this study aims to investigate its impact. Serial block-face-scanning electron microscopy (SBF-SEM) and computer-assisted 3D renderings were employed to compare mitochondrial structure and networking in Sam50-deficient myotubes from mice and humans with wild-type (WT) myotubes. Furthermore, autophagosome 3D structure was assessed in human myotubes. Mitochondrial metabolic phenotypes were assessed using Gas Chromatography-Mass Spectrometry-based metabolomics to explore differential changes in WT and Sam50-deficient myotubes. The results revealed increased mitochondrial fragmentation and autophagosome formation in Sam50-deficient myotubes compared to controls. Metabolomic analysis indicated elevated metabolism of propanoate and several amino acids, including ß-Alanine, phenylalanine, and tyrosine, along with increased amino acid and fatty acid metabolism in Sam50-deficient myotubes. Furthermore, impairment of oxidative capacity was observed upon Sam50 ablation in both murine and human myotubes, as measured with the XF24 Seahorse Analyzer. Collectively, these findings support the critical role of Sam50 in establishing and maintaining mitochondrial integrity, cristae structure, and mitochondrial metabolism. By elucidating the impact of Sam50-deficiency, this study enhances our understanding of mitochondrial function in skeletal muscle.
Collapse
Affiliation(s)
- Bryanna Shao
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Mason Killion
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Ashton Oliver
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Chia Vang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Faben Zeleke
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Kit Neikirk
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Zer Vue
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Edgar Garza-Lopez
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Jian-Qiang Shao
- Central Microscopy Research Facility, University of Iowa, Iowa City, IA, 52242, USA
| | - Margaret Mungai
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Jacob Lam
- Department of Internal Medicine, University of Iowa, Iowa City, IA, 52242, USA
| | - Qiana Williams
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Christopher T Altamura
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Aaron Whiteside
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH 45435 USA
| | - Kinuthia Kabugi
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Jessica McKenzie
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Alice Koh
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Estevão Scudese
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Larry Vang
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Andrea G Marshall
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Amber Crabtree
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | | | - Dominique Stephens
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Ho-Jin Koh
- Department of Biological Sciences, Tennessee State University, Nashville, TN 37209
| | - Brenita C Jenkins
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA
| | - Sandra A Murray
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Anthonya T Cooper
- Department of Cell Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA, 15260, USA
| | - Clintoria Williams
- Department of Neuroscience, Cell Biology and Physiology, Wright State University, Dayton, OH 45435 USA
| | - Steven M Damo
- Department of Life and Physical Sciences, Fisk University, Nashville, TN, 37208, USA
| | - Melanie R McReynolds
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, PA
- Huck Institutes of the Life Sciences, Pennsylvania State University, University Park, PA
| | - Jennifer A Gaddy
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
- Tennessee Valley Healthcare Systems, U.S. Department of Veterans Affairs, Nashville, TN, 37212, USA
| | - Celestine N Wanjalla
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, 37232, USA
| | - Heather K Beasley
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| | - Antentor Hinton
- Department of Molecular Physiology and Biophysics, Vanderbilt University, Nashville, TN, 37232, USA
| |
Collapse
|
36
|
Kumar A, Gok MO, Nguyen KN, Reese ML, Wideman JG, Muñoz-Gómez SA, Friedman JR. A DRP-like pseudoenzyme coordinates with MICOS to promote cristae architecture. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.10.03.560745. [PMID: 37873150 PMCID: PMC10592917 DOI: 10.1101/2023.10.03.560745] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/25/2023]
Abstract
Mitochondrial cristae architecture is crucial for optimal respiratory function of the organelle. Cristae shape is maintained in part by the mitochondrial inner membrane-localized MICOS complex. While MICOS is required for normal cristae morphology, the precise mechanistic role of each of the seven human MICOS subunits, and how the complex coordinates with other cristae shaping factors, has not been fully determined. Here, we examine the MICOS complex in Schizosaccharomyces pombe, a minimal model whose genome only encodes for four core subunits. Using an unbiased proteomics approach, we identify a poorly characterized inner mitochondrial membrane protein that interacts with MICOS and is required to maintain cristae morphology, which we name Mmc1. We demonstrate that Mmc1 works in concert with MICOS complexes to promote normal mitochondrial morphology and respiratory function. Bioinformatic analyses reveal that Mmc1 is a distant relative of the Dynamin-Related Protein (DRP) family of GTPases, which are well established to shape and remodel membranes. We find that, like DRPs, Mmc1 self-associates and forms high molecular weight assemblies. Interestingly, however, Mmc1 is a pseudoenzyme that lacks key residues required for GTP binding and hydrolysis, suggesting it does not dynamically remodel membranes. These data are consistent with a model in which Mmc1 stabilizes cristae architecture by acting as a scaffold to support cristae ultrastructure on the matrix side of the inner membrane. Our study reveals a new class of proteins that evolved early in fungal phylogeny and is required for the maintenance of cristae architecture. This highlights the possibility that functionally analogous proteins work with MICOS to establish cristae morphology in metazoans.
Collapse
Affiliation(s)
- Abhishek Kumar
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Mehmet Oguz Gok
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Kailey N. Nguyen
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX
| | - Michael L. Reese
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX
| | - Jeremy G. Wideman
- Center for Mechanisms of Evolution, Biodesign Institute, School of Life Sciences, Arizona State University, Tempe, AZ
| | | | - Jonathan R. Friedman
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX
| |
Collapse
|
37
|
Connor OM, Matta SK, Friedman JR. Completion of mitochondrial division requires the intermembrane space protein Mdi1/Atg44. J Cell Biol 2023; 222:e202303147. [PMID: 37540145 PMCID: PMC10403340 DOI: 10.1083/jcb.202303147] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 07/05/2023] [Accepted: 07/17/2023] [Indexed: 08/05/2023] Open
Abstract
Mitochondria are highly dynamic double membrane-bound organelles that maintain their shape in part through fission and fusion. Mitochondrial fission is performed by a dynamin-related protein, Dnm1 (Drp1 in humans), that constricts and divides the mitochondria in a GTP hydrolysis-dependent manner. However, it is unclear whether factors inside mitochondria help coordinate the process and if Dnm1/Drp1 activity is sufficient to complete the fission of both mitochondrial membranes. Here, we identify an intermembrane space protein required for mitochondrial fission in yeast, which we propose to name Mdi1 (also named Atg44). Loss of Mdi1 causes mitochondrial hyperfusion due to defects in fission, but not the lack of Dnm1 recruitment to mitochondria. Mdi1 is conserved in fungal species, and its homologs contain an amphipathic α-helix, mutations of which disrupt mitochondrial morphology. One model is that Mdi1 distorts mitochondrial membranes to enable Dnm1 to robustly complete fission. Our work reveals that Dnm1 cannot efficiently divide mitochondria without the coordinated function of Mdi1 inside mitochondria.
Collapse
Affiliation(s)
- Olivia M. Connor
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Srujan K. Matta
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jonathan R. Friedman
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| |
Collapse
|
38
|
Sohn JH, Mutlu B, Latorre-Muro P, Liang J, Bennett CF, Sharabi K, Kantorovich N, Jedrychowski M, Gygi SP, Banks AS, Puigserver P. Liver mitochondrial cristae organizing protein MIC19 promotes energy expenditure and pedestrian locomotion by altering nucleotide metabolism. Cell Metab 2023; 35:1356-1372.e5. [PMID: 37473754 PMCID: PMC10528355 DOI: 10.1016/j.cmet.2023.06.015] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 03/24/2023] [Accepted: 06/23/2023] [Indexed: 07/22/2023]
Abstract
Liver mitochondria undergo architectural remodeling that maintains energy homeostasis in response to feeding and fasting. However, the specific components and molecular mechanisms driving these changes and their impact on energy metabolism remain unclear. Through comparative mouse proteomics, we found that fasting induces strain-specific mitochondrial cristae formation in the liver by upregulating MIC19, a subunit of the MICOS complex. Enforced MIC19 expression in the liver promotes cristae formation, mitochondrial respiration, and fatty acid oxidation while suppressing gluconeogenesis. Mice overexpressing hepatic MIC19 show resistance to diet-induced obesity and improved glucose homeostasis. Interestingly, MIC19 overexpressing mice exhibit elevated energy expenditure and increased pedestrian locomotion. Metabolite profiling revealed that uracil accumulates in the livers of these mice due to increased uridine phosphorylase UPP2 activity. Furthermore, uracil-supplemented diet increases locomotion in wild-type mice. Thus, MIC19-induced mitochondrial cristae formation in the liver increases uracil as a signal to promote locomotion, with protective effects against diet-induced obesity.
Collapse
Affiliation(s)
- Jee Hyung Sohn
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Beste Mutlu
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Pedro Latorre-Muro
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Jiaxin Liang
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Christopher F Bennett
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Kfir Sharabi
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Noa Kantorovich
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Mark Jedrychowski
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Steven P Gygi
- Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA
| | - Alexander S Banks
- Division of Endocrinology, Diabetes and Metabolism, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, USA
| | - Pere Puigserver
- Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Blavatnik Institute, Harvard Medical School, Boston, MA 02215, USA.
| |
Collapse
|
39
|
Jang S, Javadov S. Unraveling the mechanisms of cardiolipin function: The role of oxidative polymerization of unsaturated acyl chains. Redox Biol 2023; 64:102774. [PMID: 37300954 PMCID: PMC10363451 DOI: 10.1016/j.redox.2023.102774] [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: 04/20/2023] [Revised: 05/26/2023] [Accepted: 06/03/2023] [Indexed: 06/12/2023] Open
Abstract
Cardiolipin is a unique phospholipid of the inner mitochondrial membrane (IMM) as well as in bacteria. It performs several vital functions such as resisting osmotic rupture and stabilizing the supramolecular structure of large membrane proteins, like ATP synthases and respirasomes. The process of cardiolipin biosynthesis results in the production of immature cardiolipin. A subsequent step is required for its maturation when its acyl groups are replaced with unsaturated acyl chains, primarily linoleic acid. Linoleic acid is the major fatty acid of cardiolipin across all organs and tissues, except for the brain. Linoleic acid is not synthesized by mammalian cells. It has the unique ability to undergo oxidative polymerization at a moderately accelerated rate compared to other unsaturated fatty acids. This property can enable cardiolipin to form covalently bonded net-like structures essential for maintaining the complex geometry of the IMM and gluing the quaternary structure of large IMM protein complexes. Unlike triglycerides, phospholipids possess only two covalently linked acyl chains, which constrain their capacity to develop robust and complicated structures through oxidative polymerization of unsaturated acyl chains. Cardiolipin, on the other hand, has four fatty acids at its disposal to form covalently bonded polymer structures. Despite its significance, the oxidative polymerization of cardiolipin has been overlooked due to the negative perception surrounding biological oxidation and methodological difficulties. Here, we discuss an intriguing hypothesis that oxidative polymerization of cardiolipin is essential for the structure and function of cardiolipin in the IMM in physiological conditions. In addition, we highlight current challenges associated with the identification and characterization of oxidative polymerization of cardiolipin in vivo. Altogether, the study provides a better understanding of the structural and functional role of cardiolipin in mitochondria.
Collapse
Affiliation(s)
- Sehwan Jang
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR 00936-5067, USA
| | - Sabzali Javadov
- Department of Physiology, University of Puerto Rico School of Medicine, San Juan, PR 00936-5067, USA.
| |
Collapse
|
40
|
Adams RA, Liu Z, Hsieh C, Marko M, Lederer WJ, Jafri MS, Mannella C. Structural Analysis of Mitochondria in Cardiomyocytes: Insights into Bioenergetics and Membrane Remodeling. Curr Issues Mol Biol 2023; 45:6097-6115. [PMID: 37504301 PMCID: PMC10378267 DOI: 10.3390/cimb45070385] [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: 06/28/2023] [Revised: 07/15/2023] [Accepted: 07/18/2023] [Indexed: 07/29/2023] Open
Abstract
Mitochondria in mammalian cardiomyocytes display considerable structural heterogeneity, the significance of which is not currently understood. We use electron microscopic tomography to analyze a dataset of 68 mitochondrial subvolumes to look for correlations among mitochondrial size and shape, crista morphology and membrane density, and organelle location within rat cardiac myocytes. A tomographic analysis guided the definition of four classes of crista morphology: lamellar, tubular, mixed and transitional, the last associated with remodeling between lamellar and tubular cristae. Correlations include an apparent bias for mitochondria with lamellar cristae to be located in the regions between myofibrils and a two-fold larger crista membrane density in mitochondria with lamellar cristae relative to mitochondria with tubular cristae. The examination of individual cristae inside mitochondria reveals local variations in crista topology, such as extent of branching, alignment of fenestrations and progressive changes in membrane morphology and packing density. The findings suggest both a rationale for the interfibrillar location of lamellar mitochondria and a pathway for crista remodeling from lamellar to tubular morphology.
Collapse
Affiliation(s)
- Raquel A. Adams
- Krasnow Institute for Advanced Study and School of Systems Biology, George Mason University, Fairfax, VA 22030, USA;
| | - Zheng Liu
- Wadsworth Center, New York State Department of Health, Albany, NY 12201, USA (M.M.)
| | - Chongere Hsieh
- Wadsworth Center, New York State Department of Health, Albany, NY 12201, USA (M.M.)
| | - Michael Marko
- Wadsworth Center, New York State Department of Health, Albany, NY 12201, USA (M.M.)
| | - W. Jonathan Lederer
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA;
- Center for Biomedical Engineering and Technology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - M. Saleet Jafri
- Krasnow Institute for Advanced Study and School of Systems Biology, George Mason University, Fairfax, VA 22030, USA;
- Center for Biomedical Engineering and Technology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| | - Carmen Mannella
- Department of Physiology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA;
- Center for Biomedical Engineering and Technology, School of Medicine, University of Maryland, Baltimore, MD 21201, USA
| |
Collapse
|
41
|
Yang S, Yin X, Wang J, Li H, Shen H, Sun Q, Li X. MIC19 Exerts Neuroprotective Role via Maintaining the Mitochondrial Structure in a Rat Model of Intracerebral Hemorrhage. Int J Mol Sci 2023; 24:11553. [PMID: 37511310 PMCID: PMC10380515 DOI: 10.3390/ijms241411553] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 07/07/2023] [Accepted: 07/09/2023] [Indexed: 07/30/2023] Open
Abstract
As an essential constituent of the mitochondrial contact site and cristae organization system (MICOS), MIC19 plays a crucial role in maintaining the stability of mitochondrial function and microstructure. However, the mechanisms and functions of MIC19 in intracerebral hemorrhage (ICH) remain unknown and need to be investigated. Sprague Dawley (SD) rats injected with autologous blood obtained from the caudal artery, and cultured neurons exposed to oxygen hemoglobin (OxyHb) were used to establish and emulate the ICH model in vivo and in vitro. Lentiviral vector encoding MIC19 or MIC19 short hairpin ribonucleic acid (shRNA) was constructed and administered to rats by intracerebroventricular injection to overexpress or knock down MIC19, respectively. First, MIC19 protein levels were increased after ICH modeling. After virus transfection and subsequent ICH modeling, we observed that overexpression of MIC19 could mitigate cell apoptosis and neuronal death, as well as abnormalities in mitochondrial structure and function, oxidative stress within mitochondria, and neurobehavioral deficits in rats following ICH. Conversely, knockdown of MIC19 had the opposite effect. Moreover, we found that the connection between MIC19 and SAM50 was disrupted after ICH, which may be a reason for the impairment of the mitochondrial structure after ICH. In conclusion, MIC19 exerts a protective role in the subsequent injury induced by ICH. The investigation of MIC19 may offer clinicians novel therapeutic insights for patients afflicted with ICH.
Collapse
Affiliation(s)
- Siyuan Yang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Xulong Yin
- Institute of Stroke Research, Soochow University, Suzhou 215006, China
- Department of Neurology, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
| | - Jiahe Wang
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Haiying Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Haitao Shen
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Qing Sun
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Institute of Stroke Research, Soochow University, Suzhou 215006, China
| | - Xiang Li
- Department of Neurosurgery & Brain and Nerve Research Laboratory, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Institute of Stroke Research, Soochow University, Suzhou 215006, China
| |
Collapse
|
42
|
Mise K, Long J, Galvan DL, Ye Z, Fan G, Serysheva II, Moore TI, Wada J, Schumacker PT, Chang BH, Danesh FR. NDUFS4 Regulates Cristae Remodeling in Diabetic Kidney Disease. RESEARCH SQUARE 2023:rs.3.rs-3070079. [PMID: 37461606 PMCID: PMC10350115 DOI: 10.21203/rs.3.rs-3070079/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/24/2023]
Abstract
The mitochondrial electron transport chain (ETC) is a highly adaptive process to meet metabolic demands of the cell, and its dysregulation has been associated with diverse clinical pathologies. However, the role and nature of impaired ETC in kidney diseases remains poorly understood. Here, we generated diabetic mice with podocyte-specific overexpression of Ndufs4, an accessory subunit of mitochondrial complex I, as a model to investigate the role of ETC integrity in diabetic kidney disease (DKD). We find that these conditional mice exhibit significant improvements in cristae morphology, mitochondrial dynamics, and albuminuria. By coupling proximity labeling with super-resolution imaging, we also identify the role of cristae shaping proteins in linking NDUFS4 with improved cristae morphology. Taken together, we discover the central role of NDUFS4 as a powerful regulator of cristae remodeling, respiratory supercomplexes assembly, and mitochondrial ultrastructure in vitro and in vivo. We propose that targeting NDUFS4 represents a promising approach to slow the progression of DKD.
Collapse
Affiliation(s)
- Koki Mise
- Section of Nephrology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Nephrology, Rheumatology, Endocrinology & Metabolism, Okayama University Graduate School of Medicine, Dentistry & Pharmaceutical Sciences, Okayama, Japan
| | - Jianyin Long
- Section of Nephrology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Daniel L. Galvan
- Department of Hematopathology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Zengchun Ye
- Division of Nephrology, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, China
| | - Guizhen Fan
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Irina I. Serysheva
- Department of Biochemistry and Molecular Biology, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Travis I. Moore
- Department of Integrative Biology and Pharmacology, The University of Texas Health Science Center at Houston, Houston, Texas
| | - Jun Wada
- Department of Nephrology, Rheumatology, Endocrinology & Metabolism, Okayama University Graduate School of Medicine, Dentistry & Pharmaceutical Sciences, Okayama, Japan
| | - Paul T. Schumacker
- Department of Pediatrics, Feinberg School of Medicine, Northwestern University, Chicago, IL
| | - Benny H. Chang
- Section of Nephrology, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Farhad R. Danesh
- Section of Nephrology, The University of Texas MD Anderson Cancer Center, Houston, Texas
- Department of Pharmacology and Chemical Biology, Baylor College of Medicine, Houston, TX
| |
Collapse
|
43
|
Wang G, Wang Y, Wang K, Zhao H, Liu M, Liang W, Li D. Perillaldehyde Functions as a Potential Antifungal Agent by Triggering Metacaspase-Independent Apoptosis in Botrytis cinerea. Microbiol Spectr 2023; 11:e0052623. [PMID: 37191530 PMCID: PMC10269628 DOI: 10.1128/spectrum.00526-23] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2023] [Accepted: 04/20/2023] [Indexed: 05/17/2023] Open
Abstract
Botrytis cinerea, the causal agent of gray mold, is an important plant pathogen causing preharvest and postharvest diseases. Due to the extensive use of commercial fungicides, fungicide-resistant strains have emerged. Natural compounds with antifungal properties are widely present in various kinds of organisms. Perillaldehyde (PA), derived from the plant species Perilla frutescens, is generally recognized as a potent antimicrobial substance and to be safe to humans and the environment. In this study, we demonstrated that PA could significantly inhibit the mycelial growth of B. cinerea and reduced its pathogenicity on tomato leaves. We also found that PA had a significant protective effect on tomato, grape, and strawberry. The antifungal mechanism of PA was investigated by measuring the reactive oxygen species (ROS) accumulation, the intracellular Ca2+ level, the mitochondrial membrane potential, DNA fragmentation, and phosphatidylserine exposure. Further analyses revealed that PA promoted protein ubiquitination and induced autophagic activities and then triggered protein degradation. When the two metacaspase genes, BcMca1 and BcMca2, were knocked out from B. cinerea, all mutants did not exhibit reduced sensitivity to PA. These findings demonstrated that PA could induce metacaspase-independent apoptosis in B. cinerea. Based on our results, we proposed that PA could be used as an effective control agent for gray mold management. IMPORTANCE Botrytis cinerea causes gray mold disease, is considered one of the most important dangerous pathogens worldwide, and leads to severe economic losses worldwide. Due to the lack of resistant varieties of B. cinerea, gray mold control has mainly relied on application of synthetic fungicides. However, long-term and extensive use of synthetic fungicides has increased fungicide resistance in B. cinerea and is harmful to humans and the environment. In this study, we found that perillaldehyde has a significant protective effect on tomato, grape, and strawberry. We further characterized the antifungal mechanism of PA on B. cinerea. Our results indicated that PA induced apoptosis that was independent of metacaspase function.
Collapse
Affiliation(s)
- Guanbo Wang
- Shandong Engineering Research Center for Environment-Friendly Agricultural Pest Management, College of Plant Health and Medicine, Qingdao Agricultural University, Qingdao, China
| | - Yadi Wang
- Shandong Engineering Research Center for Environment-Friendly Agricultural Pest Management, College of Plant Health and Medicine, Qingdao Agricultural University, Qingdao, China
| | - Kunchun Wang
- The Linzi Center for Agricultural and Rural Development, Zibo, China
| | - Haonan Zhao
- Shandong Engineering Research Center for Environment-Friendly Agricultural Pest Management, College of Plant Health and Medicine, Qingdao Agricultural University, Qingdao, China
| | - Mengjie Liu
- Shandong Engineering Research Center for Environment-Friendly Agricultural Pest Management, College of Plant Health and Medicine, Qingdao Agricultural University, Qingdao, China
| | - Wenxing Liang
- Shandong Engineering Research Center for Environment-Friendly Agricultural Pest Management, College of Plant Health and Medicine, Qingdao Agricultural University, Qingdao, China
| | - Delong Li
- Shandong Engineering Research Center for Environment-Friendly Agricultural Pest Management, College of Plant Health and Medicine, Qingdao Agricultural University, Qingdao, China
| |
Collapse
|
44
|
Zheng Y, Gibb AA, Xu H, Liu S, Hill BG. The metabolic state of the heart regulates mitochondrial supercomplex abundance in mice. Redox Biol 2023; 63:102740. [PMID: 37210780 DOI: 10.1016/j.redox.2023.102740] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 05/10/2023] [Indexed: 05/23/2023] Open
Abstract
Mitochondrial supercomplexes are observed in mammalian tissues with high energy demand and may influence metabolism and redox signaling. Nevertheless, the mechanisms that regulate supercomplex abundance remain unclear. In this study, we examined the composition of supercomplexes derived from murine cardiac mitochondria and determined how their abundance changes with substrate provision or by genetically induced changes to the cardiac glucose-fatty acid cycle. Protein complexes from digitonin-solubilized cardiac mitochondria were resolved by blue-native polyacrylamide gel electrophoresis and were identified by mass spectrometry and immunoblotting to contain constituents of Complexes I, III, IV, and V as well as accessory proteins involved in supercomplex assembly and stability, cristae architecture, carbohydrate and fat oxidation, and oxidant detoxification. Respiratory analysis of high molecular mass supercomplexes confirmed the presence of intact respirasomes, capable of transferring electrons from NADH to O2. Provision of respiratory substrates to isolated mitochondria augmented supercomplex abundance, with fatty acyl substrate (octanoylcarnitine) promoting higher supercomplex abundance than carbohydrate-derived substrate (pyruvate). Mitochondria isolated from transgenic hearts that express kinase-deficient 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase (GlycoLo), which decreases glucose utilization and increases reliance on fatty acid oxidation for energy, had higher mitochondrial supercomplex abundance and activity compared with mitochondria from wild-type or phosphatase-deficient 6-phosphofructo-2-kinase/fructose-2,6-bisphosphatase-expressing hearts (GlycoHi), the latter of which encourages reliance on glucose catabolism for energy. These findings indicate that high energetic reliance on fatty acid catabolism bolsters levels of mitochondrial supercomplexes, supporting the idea that the energetic state of the heart is regulatory factor in supercomplex assembly or stability.
Collapse
Affiliation(s)
- Yuting Zheng
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Department of Medicine, University of Louisville, Louisville, KY, USA
| | - Andrew A Gibb
- Cardiovascular Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine, Temple University, Philadelphia, PA, 19140, USA
| | - Hongkai Xu
- Center of Proteomic Analysis, Beijing Genomics Institute (BGI-Shenzhen), Shenzhen, 518000, China
| | - Siqi Liu
- Center of Proteomic Analysis, Beijing Genomics Institute (BGI-Shenzhen), Shenzhen, 518000, China
| | - Bradford G Hill
- Center for Cardiometabolic Science, Christina Lee Brown Envirome Institute, Department of Medicine, University of Louisville, Louisville, KY, USA.
| |
Collapse
|
45
|
Xin Y, Zhao L, Peng R. HIF-1 signaling: an emerging mechanism for mitochondrial dynamics. J Physiol Biochem 2023:10.1007/s13105-023-00966-0. [PMID: 37178248 DOI: 10.1007/s13105-023-00966-0] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Accepted: 05/02/2023] [Indexed: 05/15/2023]
Abstract
A growing emphasis has been paid to the function of mitochondria in tumors, neurodegenerative disorders (NDs), and cardiovascular diseases. Mitochondria are oxygen-sensitive organelles whose function depends on their structural basis. Mitochondrial dynamics are critical in regulating the structure. Mitochondrial dynamics include fission, fusion, motility, cristae remodeling, and mitophagy. These processes could alter mitochondrial morphology, number, as well as distribution, to regulate complicated cellular signaling processes like metabolism. Meanwhile, they also could modulate cell proliferation and apoptosis. The initiation and progression of several diseases, such as tumors, NDs, cardiovascular disease, were all interrelated with mitochondrial dynamics. HIF-1 is a nuclear protein presented as heterodimers, and its transcriptional activity is triggered by hypoxia. It plays an important role in numerous physiological processes including the development of cardiovascular system, immune system, and cartilage. Additionally, it could evoke compensatory responses in cells during hypoxia through upstream and downstream signaling networks. Moreover, the alteration of oxygen level is a pivotal factor to promote mitochondrial dynamics and HIF-1 activation. HIF-1α might be a promising target for modulating mitochondrial dynamics to develop therapeutic approaches for NDs, immunological diseases, and other related diseases. Here, we reviewed the research progress of mitochondrial dynamics and the potential regulatory mechanism of HIF-1 in mitochondrial dynamics.
Collapse
Affiliation(s)
- Yu Xin
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China
| | - Li Zhao
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China.
| | - Ruiyun Peng
- Beijing Institute of Radiation Medicine, Beijing, 100850, People's Republic of China.
| |
Collapse
|
46
|
Tomar D, Thomas M, Garbincius JF, Kolmetzky DW, Salik O, Jadiya P, Joseph SK, Carpenter AC, Hajnóczky G, Elrod JW. MICU1 regulates mitochondrial cristae structure and function independently of the mitochondrial Ca 2+ uniporter channel. Sci Signal 2023; 16:eabi8948. [PMID: 37098122 PMCID: PMC10388395 DOI: 10.1126/scisignal.abi8948] [Citation(s) in RCA: 29] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Accepted: 03/30/2023] [Indexed: 04/27/2023]
Abstract
MICU1 is a calcium (Ca2+)-binding protein that regulates the mitochondrial Ca2+ uniporter channel complex (mtCU) and mitochondrial Ca2+ uptake. MICU1 knockout mice display disorganized mitochondrial architecture, a phenotype that is distinct from that of mice with deficiencies in other mtCU subunits and, thus, is likely not explained by changes in mitochondrial matrix Ca2+ content. Using proteomic and cellular imaging techniques, we found that MICU1 localized to the mitochondrial contact site and cristae organizing system (MICOS) and directly interacted with the MICOS components MIC60 and CHCHD2 independently of the mtCU. We demonstrated that MICU1 was essential for MICOS complex formation and that MICU1 ablation resulted in altered cristae organization, mitochondrial ultrastructure, mitochondrial membrane dynamics, and cell death signaling. Together, our results suggest that MICU1 is an intermembrane space Ca2+ sensor that modulates mitochondrial membrane dynamics independently of matrix Ca2+ uptake. This system enables distinct Ca2+ signaling in the mitochondrial matrix and at the intermembrane space to modulate cellular energetics and cell death in a concerted manner.
Collapse
Affiliation(s)
- Dhanendra Tomar
- Cardiovascular Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140
| | - Manfred Thomas
- Cardiovascular Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140
| | - Joanne F. Garbincius
- Cardiovascular Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140
| | - Devin W. Kolmetzky
- Cardiovascular Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140
| | - Oniel Salik
- Cardiovascular Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140
- Health and Exercise Physiology, Ursinus College, Collegeville, PA 19426, USA
| | - Pooja Jadiya
- Cardiovascular Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140
| | - Suresh K. Joseph
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - April C. Carpenter
- Health and Exercise Physiology, Ursinus College, Collegeville, PA 19426, USA
| | - György Hajnóczky
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA, USA
| | - John W. Elrod
- Cardiovascular Research Center, Department of Cardiovascular Sciences, Lewis Katz School of Medicine at Temple University, Philadelphia, PA 19140
| |
Collapse
|
47
|
Altieri DC. Mitochondria in cancer: clean windmills or stressed tinkerers? Trends Cell Biol 2023; 33:293-299. [PMID: 36055942 PMCID: PMC9938083 DOI: 10.1016/j.tcb.2022.08.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 07/29/2022] [Accepted: 08/01/2022] [Indexed: 11/23/2022]
Abstract
There is now a consensus that mitochondria are important tumor drivers, sophisticated biological machines that can engender a panoply of key disease traits. How this happens, however, is still mostly elusive. The opinion presented here is that what cancer exploits are not the normal mitochondria of oxygenated and nutrient-replete tissues, but the unfit, damaged, and dysfunctional organelles generated by the hostile environment of tumor growth. These 'ghost' mitochondria survive quality control and thwart cell death to relay multiple comprehensive 'danger signals' of metabolic starvation, cellular stress, and reprogrammed gene expression. The result is a new, treacherous cellular phenotype, proliferatively quiescent but highly motile, that enables tumor cell escape from a threatening environment and colonization of distant, more favorable sites (metastasis).
Collapse
Affiliation(s)
- Dario C Altieri
- Immunology, Microenvironment and Metastasis Program, The Wistar Institute, Philadelphia, PA 19104, USA.
| |
Collapse
|
48
|
Sokolova IM. Ectotherm mitochondrial economy and responses to global warming. Acta Physiol (Oxf) 2023; 237:e13950. [PMID: 36790303 DOI: 10.1111/apha.13950] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/24/2023] [Accepted: 02/10/2023] [Indexed: 02/16/2023]
Abstract
Temperature is a key abiotic factor affecting ecology, biogeography, and evolution of species. Alterations of energy metabolism play an important role in adaptations and plastic responses to temperature shifts on different time scales. Mitochondrial metabolism affects cellular bioenergetics and redox balance making these organelles an important determinant of organismal performances such as growth, locomotion, or development. Here I analyze the impacts of environmental temperature on the mitochondrial functions (including oxidative phosphorylation, proton leak, production of reactive oxygen species(ROS), and ATP synthesis) of ectotherms and discuss the mechanisms underlying negative shifts in the mitochondrial energy economy caused by supraoptimal temperatures. Owing to the differences in the thermal sensitivity of different mitochondrial processes, elevated temperatures (beyond the species- and population-specific optimal range) cause reallocation of the electron flux and the protonmotive force (Δp) in a way that decreases ATP synthesis efficiency, elevates the relative cost of the mitochondrial maintenance, causes excessive production of ROS and raises energy cost for antioxidant defense. These shifts in the mitochondrial energy economy might have negative consequences for the organismal fitness traits such as the thermal tolerance or growth. Correlation between the thermal sensitivity indices of the mitochondria and the whole organism indicate that these traits experience similar selective pressures but further investigations are needed to establish whether there is a cause-effect relationship between the mitochondrial failure and loss of organismal performance during temperature change.
Collapse
Affiliation(s)
- Inna M Sokolova
- Department of Marine Biology, Institute of Biological Sciences, University of Rostock, Rostock, Germany
- Department of Maritime Systems, Interdisciplinary Faculty, University of Rostock, Rostock, Germany
| |
Collapse
|
49
|
Mahley RW. Apolipoprotein E4 targets mitochondria and the mitochondria-associated membrane complex in neuropathology, including Alzheimer's disease. Curr Opin Neurobiol 2023; 79:102684. [PMID: 36753858 DOI: 10.1016/j.conb.2023.102684] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2023] [Accepted: 01/06/2023] [Indexed: 02/08/2023]
Abstract
Apolipoprotein (apo) E4 sets the stage for neuropathology in Alzheimer's disease (AD) by causing mitochondrial dysfunction and altering mitochondria-associated membranes. Contact and apposition of mitochondrial-endoplasmic reticulum membranes are enhanced in brain cells in AD and associated with increases in tethering and spacing proteins that modulate many cellular processes. Contact site protein levels are higher in apoE4 cells. In apoE4 neurons, the NAD+/NADH ratio is lowered, reactive oxygen species are increased, and NAD/NADH pathway components and redox proteins are decreased. Oxidative phosphorylation is impaired and reserve ATP generation capacity is lacking. ApoE4 neurons have ∼50% fewer respiratory complex subunits (e.g., ATP synthase) and may increase translocase levels of the outer and inner mitochondrial membranes to facilitate delivery of nucleus-encoded complex subunits. Respiratory complex assembly relies on mitochondrial cristae organizing system subunits that are altered in apoE4 cells, and apoE4 increases mitochondrial proteases that control respiratory subunit composition for complex assembly.
Collapse
Affiliation(s)
- Robert W Mahley
- Gladstone Institute of Neurological Disease, 1650 Owens Street, San Francisco, CA 94158, USA; Departments of Pathology and Medicine, University of California, San Francisco, CA 94143, USA.
| |
Collapse
|
50
|
Connor OM, Matta SK, Friedman JR. An intermembrane space protein facilitates completion of mitochondrial division in yeast. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.03.31.535139. [PMID: 37034761 PMCID: PMC10081322 DOI: 10.1101/2023.03.31.535139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 06/19/2023]
Abstract
Mitochondria are highly dynamic double membrane-bound organelles that maintain their shape in part through fission and fusion. Mitochondrial fission is performed by the dynamin-related protein Dnm1 (Drp1 in humans), a large GTPase that constricts and divides the mitochondria in a GTP hydrolysis-dependent manner. However, it is unclear whether factors inside mitochondria help coordinate the process and if Dnm1/Drp1 activity alone is sufficient to complete fission of both mitochondrial membranes. Here, we identify an intermembrane space protein required for mitochondrial fission in yeast, which we propose to name Mdi1. Loss of Mdi1 leads to hyper-fused mitochondria networks due to defects in mitochondrial fission, but not lack of Dnm1 recruitment to mitochondria. Mdi1 plays a conserved role in fungal species and its homologs contain a putative amphipathic α-helix, mutations in which disrupt mitochondrial morphology. One model to explain these findings is that Mdi1 associates with and distorts the mitochondrial inner membrane to enable Dnm1 to robustly complete fission. Our work reveals that Dnm1 cannot efficiently divide mitochondria without the coordinated function of a protein that resides inside mitochondria.
Collapse
Affiliation(s)
- Olivia M. Connor
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Srujan K. Matta
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| | - Jonathan R. Friedman
- Department of Cell Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390
| |
Collapse
|