1
|
Langley A, Abeling-Wang S, Wagner E, Salogiannis J. Movement of the endoplasmic reticulum is driven by multiple classes of vesicles marked by Rab-GTPases. Mol Biol Cell 2025; 36:ar9. [PMID: 39630612 DOI: 10.1091/mbc.e24-04-0197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2024] Open
Abstract
Peripheral endoplasmic reticulum (ER) tubules move along microtubules to interact with various organelles through membrane contact sites. Traditionally, ER moves by either sliding along stable microtubules via molecular motors or attaching to the plus ends of dynamic microtubules through tip attachment complexes (TAC). A recently discovered third process, hitchhiking, involves motile vesicles pulling ER tubules along microtubules. Previous research showed that ER hitchhikes on Rab5- and Rab7-marked endosomes, but it is uncertain whether other Rab-vesicles can do the same. In U2OS cells, we screened Rabs for their ability to cotransport with ER tubules and found that ER hitchhikes on post-Golgi vesicles marked by Rab6 (isoforms a and b). Rab6-ER hitchhiking occurs independently of ER-endolysosome contacts and TAC-mediated ER movement. Depleting Rab6 and the motility of Rab6-vesicles reduces overall ER movement. Conversely, relocating these vesicles to the cell periphery causes peripheral ER accumulation, indicating that Rab6-vesicle motility is crucial for a subset of ER movements. Proximal post-Golgi vesicles marked by TGN46 are involved in Rab6-ER hitchhiking, while late Golgi vesicles (Rabs 8/10/11/13/14) are not essential for ER movement. Our further analysis finds that ER to Golgi vesicles marked by Rab1 are also capable of driving a subset of ER movements. Taken together, our findings suggest that ER hitchhiking on Rab-vesicles is a significant mode of ER movement.
Collapse
Affiliation(s)
- Allison Langley
- Cellular, Molecular, and Biomedical Sciences Graduate Program, University of Vermont, Burlington, VT 05405
- Department of Molecular Physiology and Biophysics, Larner College of Medicine at the University of Vermont, Burlington, VT 05405
| | - Sarah Abeling-Wang
- Department of Molecular Physiology and Biophysics, Larner College of Medicine at the University of Vermont, Burlington, VT 05405
| | - Erinn Wagner
- Department of Molecular Physiology and Biophysics, Larner College of Medicine at the University of Vermont, Burlington, VT 05405
| | - John Salogiannis
- Department of Molecular Physiology and Biophysics, Larner College of Medicine at the University of Vermont, Burlington, VT 05405
| |
Collapse
|
2
|
Domingues N, Pires J, Milosevic I, Raimundo N. Role of lipids in interorganelle communication. Trends Cell Biol 2025; 35:46-58. [PMID: 38866684 PMCID: PMC11632148 DOI: 10.1016/j.tcb.2024.04.008] [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: 02/06/2024] [Revised: 04/26/2024] [Accepted: 04/29/2024] [Indexed: 06/14/2024]
Abstract
Cell homeostasis and function rely on well-orchestrated communication between different organelles. This communication is ensured by signaling pathways and membrane contact sites between organelles. Many players involved in organelle crosstalk have been identified, predominantly proteins and ions. The role of lipids in interorganelle communication remains poorly understood. With the development and broader availability of methods to quantify lipids, as well as improved spatiotemporal resolution in detecting different lipid species, the contribution of lipids to organelle interactions starts to be evident. However, the specific roles of various lipid molecules in intracellular communication remain to be studied systematically. We summarize new insights in the interorganelle communication field from the perspective of organelles and discuss the roles played by lipids in these complex processes.
Collapse
Affiliation(s)
- Neuza Domingues
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
| | - Joana Pires
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal
| | - Ira Milosevic
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal; Centre for Human Genetics, Nuffield Department of Medicine, University of Oxford, Oxford, UK
| | - Nuno Raimundo
- Multidisciplinary Institute of Ageing, University of Coimbra, Coimbra, Portugal; Department of Cellular and Molecular Physiology, Penn State College of Medicine, Hershey, PA, USA; Penn State Cancer Institute, Hershey, PA, USA.
| |
Collapse
|
3
|
Wang F, Han H, Wang C, Wang J, Peng Y, Chen Y, He Y, Deng Z, Li F, Rong Y, Wang D, Liu W, Chen H, Zhang Z. SARS-CoV-2 membrane protein induces neurodegeneration via affecting Golgi-mitochondria interaction. Transl Neurodegener 2024; 13:68. [PMID: 39726060 DOI: 10.1186/s40035-024-00458-1] [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: 05/25/2024] [Accepted: 11/12/2024] [Indexed: 12/28/2024] Open
Abstract
BACKGROUND Neurological complications are a significant concern of Coronavirus Disease 2019 (COVID-19). However, the pathogenic mechanism of neurological symptoms associated with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is poorly understood. METHODS We used Drosophila as a model to systematically analyze SARS-CoV-2 genes encoding structural and accessory proteins and identified the membrane protein (M) that disrupted mitochondrial functions in vivo. The M protein was stereotaxically injected to further assess its effects in the brains of wild-type (WT) and 5 × FAD mice. Omics technologies, including RNA sequencing and interactome analysis, were performed to explore the mechanisms of the effects of M protein both in vitro and in vivo. RESULTS Systematic analysis of SARS-CoV-2 structural and accessory proteins in Drosophila identified that the M protein induces mitochondrial fragmentation and dysfunction, leading to reduced ATP production, ROS overproduction, and eventually cell death in the indirect flight muscles. In WT mice, M caused hippocampal atrophy, neural apoptosis, glial activation, and mitochondrial damage. These changes were further aggravated in 5 × FAD mice. M was localized to the Golgi apparatus and genetically interacted with four wheel drive (FWD, a Drosophila homolog of mammalian PI4KIIIβ) to regulate Golgi functions in flies. Fwd RNAi, but not PI4KIIIα RNAi, reversed the M-induced Golgi abnormality, mitochondrial fragmentation, and ATP reduction. Inhibition of PI4KIIIβ activity suppressed the M-induced neuronal cell death. Therefore, M induced mitochondrial fragmentation and apoptosis likely through disruption of Golgi-derived PI(4)P-containing vesicles. CONCLUSIONS M disturbs the distribution and function of Golgi, leading to mitochondrial abnormality and eventually neurodegeneration via a PI4KIIIβ-mediated mechanism. This study reveals a potential mechanism for COVID-19 neurological symptoms and opens a new avenue for development of therapeutic strategies targeting SARS-CoV-2 M or mitochondria.
Collapse
Affiliation(s)
- Fang Wang
- Department of Neurosciences, Hengyang Medical School, University of South China, Hengyang, 421009, China
- Institute of Molecular Precision Medicine and Hunan Provincial Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Medical Genetics, College of Biological Sciences, Central South University, Changsha, 410078, China
| | - Hailong Han
- Department of Neurosciences, Hengyang Medical School, University of South China, Hengyang, 421009, China
- Institute of Molecular Precision Medicine and Hunan Provincial Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Medical Genetics, College of Biological Sciences, Central South University, Changsha, 410078, China
| | - Caifang Wang
- Institute of Molecular Precision Medicine and Hunan Provincial Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Medical Genetics, College of Biological Sciences, Central South University, Changsha, 410078, China
| | - Jingfei Wang
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150001, China
| | - Yanni Peng
- Institute of Molecular Precision Medicine and Hunan Provincial Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Medical Genetics, College of Biological Sciences, Central South University, Changsha, 410078, China
| | - Ye Chen
- Institute of Molecular Precision Medicine and Hunan Provincial Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, 410078, China
- Hunan Provincial Key Laboratory of Medical Genetics, College of Biological Sciences, Central South University, Changsha, 410078, China
| | - Yaohui He
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361000, China
| | - Zhouyang Deng
- Hunan Provincial Key Laboratory of Medical Genetics, College of Biological Sciences, Central South University, Changsha, 410078, China
| | - Fang Li
- Hunan Provincial Key Laboratory of Medical Genetics, College of Biological Sciences, Central South University, Changsha, 410078, China
| | - Yikang Rong
- Department of Neurosciences, Hengyang Medical School, University of South China, Hengyang, 421009, China
| | - Danling Wang
- Department of Neurosciences, Hengyang Medical School, University of South China, Hengyang, 421009, China
| | - Wen Liu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research, School of Pharmaceutical Sciences, Xiamen University, Xiamen, 361000, China
| | - Hualan Chen
- State Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, 150001, China
| | - Zhuohua Zhang
- Department of Neurosciences, Hengyang Medical School, University of South China, Hengyang, 421009, China.
- Institute of Molecular Precision Medicine and Hunan Provincial Key Laboratory of Molecular Precision Medicine, Xiangya Hospital, Central South University, Changsha, 410078, China.
- Hunan Provincial Key Laboratory of Medical Genetics, College of Biological Sciences, Central South University, Changsha, 410078, China.
| |
Collapse
|
4
|
Zhang T, Fan J, Wen X, Duan X. ECSIT: Biological function and involvement in diseases. Int Immunopharmacol 2024; 143:113524. [PMID: 39488037 DOI: 10.1016/j.intimp.2024.113524] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/28/2024] [Accepted: 10/25/2024] [Indexed: 11/04/2024]
Abstract
Evolutionary conserved signaling intermediate in Toll pathways (ECSIT), a multi-functional protein, was first identified as a cytosolic adaptor protein in Toll-like receptors (TLRs) signaling-mediated innate immune responses. In the past two decades, studies have expanded the understanding of ECSIT. Nevertheless, there are still large knowledge gaps due to the inadequate number of studies regarding ECSIT, especially an overall review of ECSIT is lacking. Here, we first comprehensively summarize the biological functions of ECSIT with particular focus on innate immune responses and mitochondrial homeostasis. Cumulative studies have reinforced that ECSIT is involved in the regulation of innate immune responses through activating NF-κB signaling and potentiating the Retinoic acid-induced gene Ⅰ (RIG-Ⅰ)/ mitochondrial antiviral- signaling protein (MAVS) pathway-mediated innate antiviral immunity. In addition, ECSIT determines the mitochondrial morphology and function including mitochondrial complex Ⅰ (CⅠ) assembly, mitochondrial reactive oxygen species (mROS) production, mitochondrial membrane potential (MMP) maintenance and mitochondrial quality control. Owing to these distinct functions, ECSIT is involved in the etiology and pathology of human diseases including Alzheimer's disease (AD), cardiac hypertrophy, musculoskeletal disintegration, cancer, extranodal natural killer/T cell lymphoma (ENKTL) and ischemic stroke. Collectively, the roles and mechanisms of ECSIT under physiological and pathological conditions are critically discussed to provide a clearer view of the therapeutic potential of ECSIT.
Collapse
Affiliation(s)
- Tan Zhang
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, PR China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai 200438, PR China.
| | - Jingcheng Fan
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, PR China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai 200438, PR China
| | - Xin Wen
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, PR China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai 200438, PR China
| | - Xuemei Duan
- School of Exercise and Health, Shanghai University of Sport, Shanghai 200438, PR China; Shanghai Frontiers Science Research Base of Exercise and Metabolic Health, Shanghai 200438, PR China
| |
Collapse
|
5
|
Li Y, Qi J, Guo L, Jiang X, He G. Organellar quality control crosstalk in aging-related disease: Innovation to pave the way. Aging Cell 2024:e14447. [PMID: 39668579 DOI: 10.1111/acel.14447] [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: 09/15/2024] [Revised: 11/04/2024] [Accepted: 12/02/2024] [Indexed: 12/14/2024] Open
Abstract
Organellar homeostasis and crosstalks within a cell have emerged as essential regulatory and determining factors for the survival and functions of cells. In response to various stimuli, cells can activate the organellar quality control systems (QCS) to maintain homeostasis. Numerous studies have demonstrated that dysfunction of QCS can lead to various aging-related diseases such as neurodegenerative, pulmonary, cardiometabolic diseases and cancers. However, the interplay between QCS and their potential role in these diseases are poorly understood. In this review, we present an overview of the current findings of QCS and their crosstalk, encompassing mitochondria, endoplasmic reticulum, Golgi apparatus, ribosomes, peroxisomes, lipid droplets, and lysosomes as well as the aberrant interplays among these organelles that contributes to the onset and progression of aging-related disorders. Furthermore, potential therapeutic approaches based on these quality control interactions are discussed. Our perspectives can enhance insights into the regulatory networks underlying QCS and the pathology of aging and aging-related diseases, which may pave the way for the development of novel therapeutic targets.
Collapse
Affiliation(s)
- Yu Li
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Jinxin Qi
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, China
| | - Linhong Guo
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Xian Jiang
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| | - Gu He
- Department of Dermatology & Venerology, West China Hospital, Sichuan University, Chengdu, China
- Laboratory of Dermatology, Clinical Institute of Inflammation and Immunology, Frontiers Science Center for Disease-Related Molecular Network, State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu, China
| |
Collapse
|
6
|
Ravindran S, Rau CD. The multifaceted role of mitochondria in cardiac function: insights and approaches. Cell Commun Signal 2024; 22:525. [PMID: 39472951 PMCID: PMC11523909 DOI: 10.1186/s12964-024-01899-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 10/19/2024] [Indexed: 11/02/2024] Open
Abstract
Cardiovascular disease (CVD) remains a global economic burden even in the 21st century with 85% of deaths resulting from heart attacks. Despite efforts in reducing the risk factors, and enhancing pharmacotherapeutic strategies, challenges persist in early identification of disease progression and functional recovery of damaged hearts. Targeting mitochondrial dysfunction, a key player in the pathogenesis of CVD has been less successful due to its role in other coexisting diseases. Additionally, it is the only organelle with an agathokakological function that is a remedy and a poison for the cell. In this review, we describe the origins of cardiac mitochondria and the role of heteroplasmy and mitochondrial subpopulations namely the interfibrillar, subsarcolemmal, perinuclear, and intranuclear mitochondria in maintaining cardiac function and in disease-associated remodeling. The cumulative evidence of mitochondrial retrograde communication with the nucleus is addressed, highlighting the need to study the genotype-phenotype relationships of specific organelle functions with CVD by using approaches like genome-wide association study (GWAS). Finally, we discuss the practicality of computational methods combined with single-cell sequencing technologies to address the challenges of genetic screening in the identification of heteroplasmy and contributory genes towards CVD.
Collapse
Affiliation(s)
- Sriram Ravindran
- Computational Medicine Program, Department of Genetics, and McAllister Heart Institute, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC-27599, USA
| | - Christoph D Rau
- Computational Medicine Program, Department of Genetics, and McAllister Heart Institute, University of North Carolina at Chapel Hill, 116 Manning Drive, Chapel Hill, NC-27599, USA.
| |
Collapse
|
7
|
Pays E. Apolipoprotein-L1 (APOL1): From Sleeping Sickness to Kidney Disease. Cells 2024; 13:1738. [PMID: 39451256 PMCID: PMC11506758 DOI: 10.3390/cells13201738] [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: 09/30/2024] [Revised: 10/16/2024] [Accepted: 10/18/2024] [Indexed: 10/26/2024] Open
Abstract
Apolipoprotein-L1 (APOL1) is a membrane-interacting protein induced by inflammation, which confers human resistance to infection by African trypanosomes. APOL1 kills Trypanosoma brucei through induction of apoptotic-like parasite death, but two T. brucei clones acquired resistance to APOL1, allowing them to cause sleeping sickness. An APOL1 C-terminal sequence alteration, such as occurs in natural West African variants G1 and G2, restored human resistance to these clones. However, APOL1 unfolding induced by G1 or G2 mutations enhances protein hydrophobicity, resulting in kidney podocyte dysfunctions affecting renal filtration. The mechanism involved in these dysfunctions is debated. The ability of APOL1 to generate ion pores in trypanosome intracellular membranes or in synthetic membranes was provided as an explanation. However, transmembrane insertion of APOL1 strictly depends on acidic conditions, and podocyte cytopathology mainly results from secreted APOL1 activity on the plasma membrane, which occurs under non-acidic conditions. In this review, I argue that besides inactivation of APOL3 functions in membrane dynamics (fission and fusion), APOL1 variants induce inflammation-linked podocyte toxicity not through pore formation, but through plasma membrane disturbance resulting from increased interaction with cholesterol, which enhances cation channels activity. A natural mutation in the membrane-interacting domain (N264K) abrogates variant APOL1 toxicity at the expense of slightly increased sensitivity to trypanosomes, further illustrating the continuous mutual adaptation between host and parasite.
Collapse
Affiliation(s)
- Etienne Pays
- Laboratory of Molecular Parasitology, Institut de Biologie et de Médecine Moléculaires (IBMM), Université Libre de Bruxelles, 6041 Gosselies, Belgium
| |
Collapse
|
8
|
Tábara LC, Segawa M, Prudent J. Molecular mechanisms of mitochondrial dynamics. Nat Rev Mol Cell Biol 2024:10.1038/s41580-024-00785-1. [PMID: 39420231 DOI: 10.1038/s41580-024-00785-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2024] [Indexed: 10/19/2024]
Abstract
Mitochondria not only synthesize energy required for cellular functions but are also involved in numerous cellular pathways including apoptosis, calcium homoeostasis, inflammation and immunity. Mitochondria are dynamic organelles that undergo cycles of fission and fusion, and these transitions between fragmented and hyperfused networks ensure mitochondrial function, enabling adaptations to metabolic changes or cellular stress. Defects in mitochondrial morphology have been associated with numerous diseases, highlighting the importance of elucidating the molecular mechanisms regulating mitochondrial morphology. Here, we discuss recent structural insights into the assembly and mechanism of action of the core mitochondrial dynamics proteins, such as the dynamin-related protein 1 (DRP1) that controls division, and the mitofusins (MFN1 and MFN2) and optic atrophy 1 (OPA1) driving membrane fusion. Furthermore, we provide an updated view of the complex interplay between different proteins, lipids and organelles during the processes of mitochondrial membrane fusion and fission. Overall, we aim to present a valuable framework reflecting current perspectives on how mitochondrial membrane remodelling is regulated.
Collapse
Affiliation(s)
- Luis-Carlos Tábara
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Mayuko Segawa
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK
| | - Julien Prudent
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge, UK.
| |
Collapse
|
9
|
Tavoulari S, Lacabanne D, Pereira GC, Thangaratnarajah C, King MS, He J, Chowdhury SR, Tilokani L, Palmer SM, Prudent J, Walker JE, Kunji ERS. Distinct roles for the domains of the mitochondrial aspartate/glutamate carrier citrin in organellar localization and substrate transport. Mol Metab 2024; 90:102047. [PMID: 39419476 PMCID: PMC11539162 DOI: 10.1016/j.molmet.2024.102047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 10/19/2024] Open
Abstract
OBJECTIVE Citrin, the mitochondrial aspartate/glutamate carrier isoform 2 (AGC2), is structurally and mechanistically the most complex SLC25 family member, because it consists of three domains and forms a homo-dimer. Each protomer has an N-terminal calcium-binding domain with EF-hands, followed by a substrate-transporting carrier domain and a C-terminal domain with an amphipathic helix. The absence or dysfunction of citrin leads to citrin deficiency, a highly prevalent pan-ethnic mitochondrial disease. Here, we aim to understand the role of different citrin domains and how they contribute to pathogenic mechanisms in citrin deficiency. METHODS We have employed structural modeling and functional reconstitution of purified proteins in proteoliposomes to assess the transport activity and calcium regulation of wild-type citrin and pathogenic variants associated with citrin deficiency. We have also developed a double knockout of citrin and aralar (AGC1), the two paralogs of the mitochondrial aspartate/glutamate carrier, in HAP1 cells to perform mitochondrial imaging and to investigate mitochondrial localisation. RESULTS Using 33 pathogenic variants of citrin we clarify determinants of subcellular localization and transport mechanism. We identify crucial elements of the carrier domain that are required for transport, including those involved in substrate binding, network formation and dynamics. We show that the N-terminal domain is not involved in calcium regulation of transport, as previously thought, but when mutated causes a mitochondrial import defect. CONCLUSIONS Our work introduces a new role for the N-terminal domain of citrin and demonstrates that dysfunction of the different domains contributes to distinct pathogenic mechanisms in citrin deficiency.
Collapse
Affiliation(s)
- Sotiria Tavoulari
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY United Kingdom.
| | - Denis Lacabanne
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY United Kingdom
| | - Gonçalo C Pereira
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY United Kingdom
| | - Chancievan Thangaratnarajah
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY United Kingdom
| | - Martin S King
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY United Kingdom
| | - Jiuya He
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY United Kingdom
| | - Suvagata R Chowdhury
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY United Kingdom
| | - Lisa Tilokani
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY United Kingdom
| | - Shane M Palmer
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY United Kingdom
| | - Julien Prudent
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY United Kingdom
| | - John E Walker
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY United Kingdom
| | - Edmund R S Kunji
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Keith Peters Building, Cambridge Biomedical Campus, Hills Road, Cambridge, CB2 0XY United Kingdom.
| |
Collapse
|
10
|
Khatun J, Gelles JD, Chipuk JE. Dynamic death decisions: How mitochondrial dynamics shape cellular commitment to apoptosis and ferroptosis. Dev Cell 2024; 59:2549-2565. [PMID: 39378840 PMCID: PMC11469553 DOI: 10.1016/j.devcel.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Revised: 08/15/2024] [Accepted: 09/03/2024] [Indexed: 10/10/2024]
Abstract
The incorporation of mitochondria into early eukaryotes established organelle-based biochemistry and enabled metazoan development. Diverse mitochondrial biochemistry is essential for life, and its homeostatic control via mitochondrial dynamics supports organelle quality and function. Mitochondrial crosstalk with numerous regulated cell death (RCD) pathways controls the decision to die. In this review, we will focus on apoptosis and ferroptosis, two distinct forms of RCD that utilize divergent signaling to kill a targeted cell. We will highlight how proteins and processes involved in mitochondrial dynamics maintain biochemically diverse subcellular compartments to support apoptosis and ferroptosis machinery, as well as unite disparate RCD pathways through dual control of organelle biochemistry and the decision to die.
Collapse
Affiliation(s)
- Jesminara Khatun
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Jesse D Gelles
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA
| | - Jerry Edward Chipuk
- Department of Oncological Sciences, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Tisch Cancer Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; Department of Dermatology, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA; The Diabetes, Obesity, and Metabolism Institute, Icahn School of Medicine at Mount Sinai, One Gustave L. Levy Place, New York, NY 10029, USA.
| |
Collapse
|
11
|
Landoni JC, Kleele T, Winter J, Stepp W, Manley S. Mitochondrial Structure, Dynamics, and Physiology: Light Microscopy to Disentangle the Network. Annu Rev Cell Dev Biol 2024; 40:219-240. [PMID: 38976811 DOI: 10.1146/annurev-cellbio-111822-114733] [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: 07/10/2024]
Abstract
Mitochondria serve as energetic and signaling hubs of the cell: This function results from the complex interplay between their structure, function, dynamics, interactions, and molecular organization. The ability to observe and quantify these properties often represents the puzzle piece critical for deciphering the mechanisms behind mitochondrial function and dysfunction. Fluorescence microscopy addresses this critical need and has become increasingly powerful with the advent of superresolution methods and context-sensitive fluorescent probes. In this review, we delve into advanced light microscopy methods and analyses for studying mitochondrial ultrastructure, dynamics, and physiology, and highlight notable discoveries they enabled.
Collapse
Affiliation(s)
- Juan C Landoni
- Institute of Physics, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland;
| | - Tatjana Kleele
- Institute of Biochemistry, Swiss Federal Institute of Technology Zürich (ETH), Zürich, Switzerland;
- Institute of Physics, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland;
| | - Julius Winter
- Institute of Physics, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland;
| | - Willi Stepp
- Institute of Physics, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland;
| | - Suliana Manley
- Institute of Physics, Swiss Federal Institute of Technology Lausanne (EPFL), Lausanne, Switzerland;
| |
Collapse
|
12
|
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
|
13
|
Yao H, Xie Y, Li C, Liu W, Yi G. Mitochondria-Associated Organelle Crosstalk in Myocardial Ischemia/Reperfusion Injury. J Cardiovasc Transl Res 2024; 17:1106-1118. [PMID: 38807004 DOI: 10.1007/s12265-024-10523-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/09/2024] [Accepted: 05/10/2024] [Indexed: 05/30/2024]
Abstract
Organelle damage is a significant contributor to myocardial ischemia/reperfusion (I/R) injury. This damage often leads to disruption of endoplasmic reticulum protein regulatory programs and dysfunction of mitochondrial energy metabolism. Mitochondria and endoplasmic reticulum are seamlessly connected through the mitochondrial-associated endoplasmic reticulum membrane (MAM), which serves as a crucial site for the exchange of organelles and metabolites. However, there is a lack of reports regarding the communication of information and metabolites between mitochondria and related organelles, which is a crucial factor in triggering myocardial I/R damage. To address this research gap, this review described the role of crosstalk between mitochondria and the correlative organelles such as endoplasmic reticulum, lysosomal and nuclei involved in reperfusion injury of the heart. In summary, this review aims to provide a comprehensive understanding of the crosstalk between organelles in myocardial I/R injury, with the ultimate goal of facilitating the development of targeted therapies based on this knowledge.
Collapse
Affiliation(s)
- Hui Yao
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, 28 Chang Sheng West Road, Hunan, 421001, China
| | - Yuxin Xie
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, 28 Chang Sheng West Road, Hunan, 421001, China
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, Hunan, China
| | - Chaoquan Li
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, 28 Chang Sheng West Road, Hunan, 421001, China
| | - Wanting Liu
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, 28 Chang Sheng West Road, Hunan, 421001, China
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, Hunan, China
| | - Guanghui Yi
- Institute of Cardiovascular Disease, Key Laboratory for Arteriosclerology of Hunan Province, Hunan International Scientific and Technological Cooperation Base of Arteriosclerotic Disease, Hengyang Medical College, University of South China, 28 Chang Sheng West Road, Hunan, 421001, China.
- Institute of Pharmacy and Pharmacology, Hunan Province Cooperative Innovation Center for Molecular Target New Drug Study, University of South China, Hengyang, 421001, Hunan, China.
| |
Collapse
|
14
|
Wai T. Is mitochondrial morphology important for cellular physiology? Trends Endocrinol Metab 2024; 35:854-871. [PMID: 38866638 DOI: 10.1016/j.tem.2024.05.005] [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: 03/28/2024] [Revised: 05/07/2024] [Accepted: 05/13/2024] [Indexed: 06/14/2024]
Abstract
Mitochondria are double membrane-bound organelles the network morphology of which in cells is shaped by opposing events of fusion and fission executed by dynamin-like GTPases. Mutations in these genes can perturb the form and functions of mitochondria in cell and animal models of mitochondrial diseases. An expanding array of chemical, mechanical, and genetic stressors can converge on mitochondrial-shaping proteins and disrupt mitochondrial morphology. In recent years, studies aimed at disentangling the multiple roles of mitochondrial-shaping proteins beyond fission or fusion have provided insights into the homeostatic relevance of mitochondrial morphology. Here, I review the pleiotropy of mitochondrial fusion and fission proteins with the aim of understanding whether mitochondrial morphology is important for cell and tissue physiology.
Collapse
Affiliation(s)
- Timothy Wai
- Institut Pasteur, Mitochondrial Biology, CNRS UMR 3691, Université Paris Cité, Paris, France.
| |
Collapse
|
15
|
Posor Y, Haucke V. A three-way organelle junction controls PI(4)P metabolism and mitochondrial division. J Cell Biol 2024; 223:e202407125. [PMID: 39083022 PMCID: PMC11291957 DOI: 10.1083/jcb.202407125] [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] [Indexed: 09/13/2024] Open
Abstract
Membrane contact sites (MCS) facilitate communication between organelles. Casler et al. (https://doi.org/10.1083/jcb.202308144) show that tripartite MCS between mitochondria, the endoplasmic reticulum (ER), and the plasma membrane (PM) regulate mitochondrial division and the distribution of phosphatidylinositol 4-phosphate [PI(4)P] on the PM.
Collapse
Affiliation(s)
- York Posor
- Leibniz Forschungsinstitut für Molekulare Pharmakologie (FMP) , Berlin, Germany
| | - Volker Haucke
- Leibniz Forschungsinstitut für Molekulare Pharmakologie (FMP) , Berlin, Germany
- Faculty of Biology, Chemistry, Pharmacy, Freie Universität Berlin, Berlin, Germany
| |
Collapse
|
16
|
Casler JC, Harper CS, White AJ, Anderson HL, Lackner LL. Mitochondria-ER-PM contacts regulate mitochondrial division and PI(4)P distribution. J Cell Biol 2024; 223:e202308144. [PMID: 38781029 PMCID: PMC11116812 DOI: 10.1083/jcb.202308144] [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: 08/18/2023] [Revised: 12/13/2023] [Accepted: 05/07/2024] [Indexed: 05/25/2024] Open
Abstract
The mitochondria-ER-cortex anchor (MECA) forms a tripartite membrane contact site between mitochondria, the endoplasmic reticulum (ER), and the plasma membrane (PM). The core component of MECA, Num1, interacts with the PM and mitochondria via two distinct lipid-binding domains; however, the molecular mechanism by which Num1 interacts with the ER is unclear. Here, we demonstrate that Num1 contains a FFAT motif in its C-terminus that interacts with the integral ER membrane protein Scs2. While dispensable for Num1's functions in mitochondrial tethering and dynein anchoring, the FFAT motif is required for Num1's role in promoting mitochondrial division. Unexpectedly, we also reveal a novel function of MECA in regulating the distribution of phosphatidylinositol-4-phosphate (PI(4)P). Breaking Num1 association with any of the three membranes it tethers results in an accumulation of PI(4)P on the PM, likely via disrupting Sac1-mediated PI(4)P turnover. This work establishes MECA as an important regulatory hub that spatially organizes mitochondria, ER, and PM to coordinate crucial cellular functions.
Collapse
Affiliation(s)
- Jason C. Casler
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Clare S. Harper
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Antoineen J. White
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Heidi L. Anderson
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Laura L. Lackner
- Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| |
Collapse
|
17
|
Brar KK, Hughes DT, Morris JL, Subramanian K, Krishna S, Gao F, Rieder LS, Uhrig S, Freeman J, Smith HL, Jukes-Jones R, Avezov E, Nunnari J, Prudent J, Butcher AJ, Mallucci GR. PERK-ATAD3A interaction provides a subcellular safe haven for protein synthesis during ER stress. Science 2024; 385:eadp7114. [PMID: 39116259 DOI: 10.1126/science.adp7114] [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: 04/09/2024] [Accepted: 07/10/2024] [Indexed: 08/10/2024]
Abstract
Endoplasmic reticulum (ER) stress induces the repression of protein synthesis throughout the cell. Attempts to understand how localized stress leads to widespread repression have been limited by difficulties in resolving translation rates at the subcellular level. Here, using live-cell imaging of reporter mRNA translation, we unexpectedly found that during ER stress, active translation at mitochondria was significantly protected. The mitochondrial protein ATPase family AAA domain-containing protein 3A (ATAD3A) interacted with protein kinase RNA-like endoplasmic reticulum kinase (PERK) and mediated this effect on localized translation by competing for binding with PERK's target, eukaryotic initiation factor 2 (eIF2). PERK-ATAD3A interactions increased during ER stress, forming mitochondria-ER contact sites. Furthermore, ATAD3A binding attenuated local PERK signaling and rescued the expression of some mitochondrial proteins. Thus, PERK-ATAD3A interactions can control translational repression at a subcellular level, mitigating the impact of ER stress on the cell.
Collapse
Affiliation(s)
- Karinder K Brar
- Altos Labs, Cambridge Institute of Science, Granta Park, Cambridge CB21 6GP, UK
- UK Dementia Research Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 OAH, UK
| | - Daniel T Hughes
- UK Dementia Research Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 OAH, UK
| | - Jordan L Morris
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | - Kelly Subramanian
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA
| | - Shivaani Krishna
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA
- Altos Labs, Bay Area Institute of Science, Redwood Shores, CA 94065, USA
| | - Fei Gao
- UK Dementia Research Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 OAH, UK
| | - Lara-Sophie Rieder
- Altos Labs, Cambridge Institute of Science, Granta Park, Cambridge CB21 6GP, UK
- UK Dementia Research Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 OAH, UK
| | - Sebastian Uhrig
- Altos Labs, Cambridge Institute of Science, Granta Park, Cambridge CB21 6GP, UK
| | - Joshua Freeman
- Altos Labs, Cambridge Institute of Science, Granta Park, Cambridge CB21 6GP, UK
- UK Dementia Research Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 OAH, UK
| | - Heather L Smith
- Altos Labs, Cambridge Institute of Science, Granta Park, Cambridge CB21 6GP, UK
- UK Dementia Research Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 OAH, UK
| | | | - Edward Avezov
- UK Dementia Research Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 OAH, UK
| | - Jodi Nunnari
- Department of Molecular and Cellular Biology, University of California, Davis, Davis, CA 95616, USA
- Altos Labs, Bay Area Institute of Science, Redwood Shores, CA 94065, USA
| | - Julien Prudent
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | - Adrian J Butcher
- Altos Labs, Cambridge Institute of Science, Granta Park, Cambridge CB21 6GP, UK
- UK Dementia Research Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 OAH, UK
| | - Giovanna R Mallucci
- Altos Labs, Cambridge Institute of Science, Granta Park, Cambridge CB21 6GP, UK
- UK Dementia Research Institute and Department of Clinical Neurosciences, University of Cambridge, Cambridge Biomedical Campus, Cambridge CB2 OAH, UK
| |
Collapse
|
18
|
Li Z, Liang S, Ke L, Wang M, Gao K, Li D, Xu Z, Li N, Zhang P, Cheng W. Cell life-or-death events in osteoporosis: All roads lead to mitochondrial dynamics. Pharmacol Res 2024; 208:107383. [PMID: 39214266 DOI: 10.1016/j.phrs.2024.107383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 08/14/2024] [Accepted: 08/26/2024] [Indexed: 09/04/2024]
Abstract
Mitochondria exhibit heterogeneous shapes and networks within and among cell types and tissues, also in normal or osteoporotic bone tissues with complex cell types. This dynamic characteristic is determined by the high plasticity provided by mitochondrial dynamics and is stemmed from responding to the survival and functional requirements of various bone cells in a specific microenvironments. In contrast, mitochondrial dysfunction, induced by dysregulation of mitochondrial dynamics, may act as a trigger of cell death signals, including common apoptosis and other forms of programmed cell death (PCD). These PCD processes consisting of tightly structured cascade gene expression events, can further influence the bone remodeling by facilitating the death of various bone cells. Mitochondrial dynamics, therefore, drive the bone cells to stand at the crossroads of life and death by integrating external signals and altering metabolism, shape, and signal-response properties of mitochondria. This implies that targeting mitochondrial dynamics displays significant potential in treatment of osteoporosis. Considerable effort has been made in osteoporosis to emphasize the parallel roles of mitochondria in regulating energy metabolism, calcium signal transduction, oxidative stress, inflammation, and cell death. However, the emerging field of mitochondrial dynamics-related PCD is not well understood. Herein, to bridge the gap, we outline the latest knowledge on mitochondrial dynamics regulating bone cell life or death during normal bone remodeling and osteoporosis.
Collapse
Affiliation(s)
- Zhichao Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Songlin Liang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Liqing Ke
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Mengjie Wang
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Kuanhui Gao
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Dandan Li
- College of Integrated Traditional Chinese and Western Medicine, Hebei University of Chinese Medicine, Shijiazhuang, 050011, China
| | - Zhanwang Xu
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China
| | - Nianhu Li
- First College of Clinical Medicine, Shandong University of Traditional Chinese Medicine, Jinan, 250014, China; Department of Orthopedics, Affiliated Hospital of Shandong University of Traditional Chinese Medicine, Jinan, 250014, China.
| | - Peng Zhang
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Faculty of Biomedical Engineering, Shenzhen University of Advanced Technology, Shenzhen, 518000, China; Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518000, China; Shandong Zhongke Advanced Technology Co., Ltd., Jinan, 250300, China.
| | - Wenxiang Cheng
- Center for Translational Medicine Research and Development, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
19
|
Jankauskas SS, Varzideh F, Kansakar U, Al Tibi G, Densu Agyapong E, Gambardella J, Santulli G. Insights into molecular and cellular functions of the Golgi calcium/manganese-proton antiporter TMEM165. J Biol Chem 2024; 300:107567. [PMID: 39002685 PMCID: PMC11345563 DOI: 10.1016/j.jbc.2024.107567] [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: 04/30/2024] [Revised: 06/19/2024] [Accepted: 07/08/2024] [Indexed: 07/15/2024] Open
Abstract
The Golgi compartment performs a number of crucial roles in the cell. However, the exact molecular mechanisms underlying these actions are not fully defined. Pathogenic mutations in genes encoding Golgi proteins may serve as an important source for expanding our knowledge. For instance, mutations in the gene encoding Transmembrane protein 165 (TMEM165) were discovered as a cause of a new type of congenital disorder of glycosylation (CDG). Comprehensive studies of TMEM165 in different model systems, including mammals, yeast, and fish uncovered the new realm of Mn2+ homeostasis regulation. TMEM165 was shown to act as a Ca2+/Mn2+:H+ antiporter in the medial- and trans-Golgi network, pumping the metal ions into the Golgi lumen and protons outside. Disruption of TMEM165 antiporter activity results in defects in N- and O-glycosylation of proteins and glycosylation of lipids. Impaired glycosylation of TMEM165-CDG arises from a lack of Mn2+ within the Golgi. Nevertheless, Mn2+ insufficiency in the Golgi is compensated by the activity of the ATPase SERCA2. TMEM165 turnover has also been found to be regulated by Mn2+ cytosolic concentration. Besides causing CDG, recent investigations have demonstrated the functional involvement of TMEM165 in several other pathologies including cancer and mental health disorders. This systematic review summarizes the available information on TMEM165 molecular structure, cellular function, and its roles in health and disease.
Collapse
Affiliation(s)
- Stanislovas S Jankauskas
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York, USA
| | - Fahimeh Varzideh
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York, USA
| | - Urna Kansakar
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York, USA
| | - Ghaith Al Tibi
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York, USA
| | - Esther Densu Agyapong
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York, USA
| | - Jessica Gambardella
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York, USA; Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy
| | - Gaetano Santulli
- Department of Medicine, Wilf Family Cardiovascular Research Institute, Einstein-Mount Sinai Diabetes Research Center (ES-DRC), Albert Einstein College of Medicine, New York City, New York, USA; Department of Advanced Biomedical Sciences, "Federico II" University, Naples, Italy; International Translational Research and Medical Education (ITME) Consortium, Academic Research Unit, Naples, Italy; Department of Molecular Pharmacology, Einstein Institute for Aging Research, Fleischer Institute for Diabetes and Metabolism (FIDAM), Albert Einstein College of Medicine, New York City, New York, USA.
| |
Collapse
|
20
|
Kourkoulou A, Martzoukou O, Fischer R, Amillis S. A type II phosphatidylinositol-4-kinase coordinates sorting of cargo polarizing by endocytic recycling. Commun Biol 2024; 7:855. [PMID: 38997419 PMCID: PMC11245547 DOI: 10.1038/s42003-024-06553-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 07/04/2024] [Indexed: 07/14/2024] Open
Abstract
Depending on their phosphorylation status, derivatives of phosphatidylinositol play important roles in vesicle identity, recognition and intracellular trafficking processes. In eukaryotic cells, phosphatidylinositol-4 phosphate pools generated by specific kinases are key determinants of the conventional secretion pathways. Earlier work in yeast has classified phosphatidylinositol-4 kinases in two types, Stt4p and Pik1p belonging to type III and Lsb6p to type II, with distinct cellular localizations and functions. Eurotiomycetes appear to lack Pik1p homologues. In Aspergillus nidulans, unlike homologues in other fungi, AnLsb6 is associated to late Golgi membranes and when heterologously overexpressed, it compensates for the thermosensitive phenotype in a Saccharomyces cerevisiae pik1 mutant, whereas its depletion leads to disorganization of Golgi-associated PHOSBP-labelled membranes, that tend to aggregate dependent on functional Rab5 GTPases. Evidence provided herein, indicates that the single type II phosphatidylinositol-4 kinase AnLsb6 is the main contributor for decorating secretory vesicles with relevant phosphatidylinositol-phosphate species, which navigate essential cargoes following the route of apical polarization via endocytic recycling.
Collapse
Affiliation(s)
- Anezia Kourkoulou
- National and Kapodistrian University of Athens, Department of Biology, Athens, Hellas, Greece
| | - Olga Martzoukou
- National and Kapodistrian University of Athens, Department of Biology, Athens, Hellas, Greece
| | - Reinhard Fischer
- Karlsruhe Institute of Technology - South Campus, Institute for Applied Biosciences, Department of Microbiology, Karlsruhe, Germany
| | - Sotiris Amillis
- National and Kapodistrian University of Athens, Department of Biology, Athens, Hellas, Greece.
- Karlsruhe Institute of Technology - South Campus, Institute for Applied Biosciences, Department of Microbiology, Karlsruhe, Germany.
| |
Collapse
|
21
|
Tábara LC, Burr SP, Frison M, Chowdhury SR, Paupe V, Nie Y, Johnson M, Villar-Azpillaga J, Viegas F, Segawa M, Anand H, Petkevicius K, Chinnery PF, Prudent J. MTFP1 controls mitochondrial fusion to regulate inner membrane quality control and maintain mtDNA levels. Cell 2024; 187:3619-3637.e27. [PMID: 38851188 DOI: 10.1016/j.cell.2024.05.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 03/19/2024] [Accepted: 05/09/2024] [Indexed: 06/10/2024]
Abstract
Mitochondrial dynamics play a critical role in cell fate decisions and in controlling mtDNA levels and distribution. However, the molecular mechanisms linking mitochondrial membrane remodeling and quality control to mtDNA copy number (CN) regulation remain elusive. Here, we demonstrate that the inner mitochondrial membrane (IMM) protein mitochondrial fission process 1 (MTFP1) negatively regulates IMM fusion. Moreover, manipulation of mitochondrial fusion through the regulation of MTFP1 levels results in mtDNA CN modulation. Mechanistically, we found that MTFP1 inhibits mitochondrial fusion to isolate and exclude damaged IMM subdomains from the rest of the network. Subsequently, peripheral fission ensures their segregation into small MTFP1-enriched mitochondria (SMEM) that are targeted for degradation in an autophagic-dependent manner. Remarkably, MTFP1-dependent IMM quality control is essential for basal nucleoid recycling and therefore to maintain adequate mtDNA levels within the cell.
Collapse
Affiliation(s)
- Luis Carlos Tábara
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK.
| | - Stephen P Burr
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK; Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Michele Frison
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK; Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Suvagata R Chowdhury
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | - Vincent Paupe
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | - Yu Nie
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK; Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Mark Johnson
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | - Jara Villar-Azpillaga
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | - Filipa Viegas
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | - Mayuko Segawa
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | - Hanish Anand
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | - Kasparas Petkevicius
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK
| | - Patrick F Chinnery
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK; Department of Clinical Neurosciences, Cambridge Biomedical Campus, University of Cambridge, Cambridge CB2 0QQ, UK
| | - Julien Prudent
- Medical Research Council Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK.
| |
Collapse
|
22
|
Sun F, Fang M, Zhang H, Song Q, Li S, Li Y, Jiang S, Yang L. Drp1: Focus on Diseases Triggered by the Mitochondrial Pathway. Cell Biochem Biophys 2024; 82:435-455. [PMID: 38438751 DOI: 10.1007/s12013-024-01245-5] [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] [Accepted: 02/26/2024] [Indexed: 03/06/2024]
Abstract
Drp1 (Dynamin-Related Protein 1) is a cytoplasmic GTPase protein encoded by the DNM1L gene that influences mitochondrial dynamics by mediating mitochondrial fission processes. Drp1 has been demonstrated to play an important role in a variety of life activities such as cell survival, proliferation, migration, and death. Drp1 has been shown to play different physiological roles under different physiological conditions, such as normal and inflammation. Recently studies have revealed that Drp1 plays a critical role in the occurrence, development, and aggravation of a series of diseases, thereby it serves as a potential therapeutic target for them. In this paper, we review the structure and biological properties of Drp1, summarize the biological processes that occur in the inflammatory response to Drp1, discuss its role in various cancers triggered by the mitochondrial pathway and investigate effective methods for targeting Drp1 in cancer treatment. We also synthesized the phenomena of Drp1 involving in the triggering of other diseases. The results discussed herein contribute to our deeper understanding of mitochondrial kinetic pathway-induced diseases and their therapeutic applications. It is critical for advancing the understanding of the mechanisms of Drp1-induced mitochondrial diseases and preventive therapies.
Collapse
Affiliation(s)
- Fulin Sun
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China
- Health Science Center, Qingdao University, Qingdao, China
| | - Min Fang
- Department of Gynaecology, Qingdao Women and Children's Hospital, Qingdao, 266021, Shandong, China
| | - Huhu Zhang
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China
| | - Qinghang Song
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China
- Health Science Center, Qingdao University, Qingdao, China
| | - Shuang Li
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China
| | - Ya Li
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China
| | - Shuyao Jiang
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China
- Health Science Center, Qingdao University, Qingdao, China
| | - Lina Yang
- Department of Genetics and Cell Biology, Basic Medical College, Qingdao University, Qingdao, China.
| |
Collapse
|
23
|
Liu M, Duan Y, Dong J, Zhang K, Jin X, Gao M, Jia H, Chen J, Liu M, Wei M, Zhong X. Early signs of neurodegenerative diseases: Possible mechanisms and targets for Golgi stress. Biomed Pharmacother 2024; 175:116646. [PMID: 38692058 DOI: 10.1016/j.biopha.2024.116646] [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: 02/28/2024] [Revised: 04/17/2024] [Accepted: 04/24/2024] [Indexed: 05/03/2024] Open
Abstract
The Golgi apparatus plays a crucial role in mediating the modification, transport, and sorting of intracellular proteins and lipids. The morphological changes occurring in the Golgi apparatus are exceptionally important for maintaining its function. When exposed to external pressure or environmental stimulation, the Golgi apparatus undergoes adaptive changes in both structure and function, which are known as Golgi stress. Although certain signal pathway responses or post-translational modifications have been observed following Golgi stress, further research is needed to comprehensively summarize and understand the related mechanisms. Currently, there is evidence linking Golgi stress to neurodegenerative diseases; however, the role of Golgi stress in the progression of neurodegenerative diseases such as Alzheimer's disease remains largely unexplored. This review focuses on the structural and functional alterations of the Golgi apparatus during stress, elucidating potential mechanisms underlying the involvement of Golgi stress in regulating immunity, autophagy, and metabolic processes. Additionally, it highlights the pivotal role of Golgi stress as an early signaling event implicated in the pathogenesis and progression of neurodegenerative diseases. Furthermore, this study summarizes prospective targets that can be therapeutically exploited to mitigate neurodegenerative diseases by targeting Golgi stress. These findings provide a theoretical foundation for identifying novel breakthroughs in preventing and treating neurodegenerative diseases.
Collapse
Affiliation(s)
- Mengyu Liu
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Ying Duan
- Liaoning Maternal and Child Health Hospital, Shayang, Liaoning 110005, China
| | - Jianru Dong
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Kaisong Zhang
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Xin Jin
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Menglin Gao
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Huachao Jia
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Ju Chen
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China
| | - Mingyan Liu
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China.
| | - Minjie Wei
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China; Liaoning Medical Diagnosis and Treatment Center, Shenyang, Liaoning 110167, China.
| | - Xin Zhong
- School of Pharmacy, China Medical University, Shenyang, Liaoning 110122, China.
| |
Collapse
|
24
|
Langley A, Abeling-Wang S, Wagner E, Salogiannis J. Movement of the endoplasmic reticulum is driven by multiple classes of vesicles marked by Rab-GTPases. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.14.592021. [PMID: 38798686 PMCID: PMC11118391 DOI: 10.1101/2024.05.14.592021] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2024]
Abstract
Peripheral endoplasmic reticulum (ER) tubules move along microtubules to interact with various organelles through membrane contact sites (MCS). Traditionally, ER moves by either sliding along stable microtubules via molecular motors or attaching to the plus ends of dynamic microtubules through tip attachment complexes (TAC). A recently discovered third process, hitchhiking, involves motile vesicles pulling ER tubules along microtubules. Previous research showed that ER hitchhikes on Rab5- and Rab7-marked endosomes, but it is uncertain if other Rab-vesicles can do the same. In U2OS cells, we screened Rabs for their ability to cotransport with ER tubules and found that ER hitchhikes on post-Golgi vesicles marked by Rab6 (isoforms a and b). Rab6-ER hitchhiking occurs independently of ER-endolysosome contacts and TAC-mediated ER movement. Disrupting either Rab6 or the motility of Rab6-vesicles reduces overall ER movement. Conversely, relocating these vesicles to the cell periphery causes peripheral ER accumulation, indicating that Rab6-vesicle motility is crucial for a subset of ER movements. Proximal post-Golgi vesicles marked by TGN46 are involved in Rab6-ER hitchhiking, while other post-Golgi vesicles (Rabs 8/10/11/13/14) are not essential for ER movement. Our further analysis finds that ER to Golgi vesicles marked by Rab1 are also capable of driving a subset of ER movements. Taken together, our findings suggest that ER hitchhiking on Rab-vesicles is a significant mode of ER movement. SIGNIFICANCE STATEMENT Peripheral endoplasmic reticulum tubules move on microtubules by either attaching to motors (cargo adaptor-mediated), dynamic microtubule-plus ends (tip attachment complexes) or motile vesicles (hitchhiking) but the prevalence of each mode is not clearPost-Golgi vesicles marked by Rab6/TGN46 and ER to Golgi vesicles marked by Rab1 drive ER movementsER hitchhiking on multiple classes of vesicles (endolysosomal, post-Golgi and ER to Golgi) marked by Rabs plays a prominent role in ER movement.
Collapse
|
25
|
Wu Z, Xiao C, Li F, Huang W, You F, Li X. Mitochondrial fusion-fission dynamics and its involvement in colorectal cancer. Mol Oncol 2024; 18:1058-1075. [PMID: 38158734 PMCID: PMC11076987 DOI: 10.1002/1878-0261.13578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Revised: 10/21/2023] [Accepted: 12/28/2023] [Indexed: 01/03/2024] Open
Abstract
The incidence and mortality rates of colorectal cancer have elevated its status as a significant public health concern. Recent research has elucidated the crucial role of mitochondrial fusion-fission dynamics in the initiation and progression of colorectal cancer. Elevated mitochondrial fission or fusion activity can contribute to the metabolic reprogramming of tumor cells, thereby activating oncogenic pathways that drive cell proliferation, invasion, migration, and drug resistance. Nevertheless, excessive mitochondrial fission can induce apoptosis, whereas moderate mitochondrial fusion can protect cells from oxidative stress. This imbalance in mitochondrial dynamics can exert dual roles as both promoters and inhibitors of colorectal cancer progression. This review provides an in-depth analysis of the fusion-fission dynamics and the underlying pathological mechanisms in colorectal cancer cells. Additionally, it offers partial insights into the mitochondrial kinetics in colorectal cancer-associated cells, such as immune and endothelial cells. This review is aimed at identifying key molecular events involved in colorectal cancer progression and highlighting the potential of mitochondrial dynamic proteins as emerging targets for pharmacological intervention.
Collapse
Affiliation(s)
- Zihong Wu
- Hospital of Chengdu University of Traditional Chinese MedicineChina
| | - Chong Xiao
- Hospital of Chengdu University of Traditional Chinese MedicineChina
- Oncology Teaching and Research DepartmentChengdu University of Traditional Chinese MedicineChina
| | - Fang Li
- Hospital of Chengdu University of Traditional Chinese MedicineChina
| | - Wenbo Huang
- Hospital of Chengdu University of Traditional Chinese MedicineChina
| | - Fengming You
- Hospital of Chengdu University of Traditional Chinese MedicineChina
- Institute of OncologyChengdu University of Traditional Chinese MedicineChina
| | - Xueke Li
- Hospital of Chengdu University of Traditional Chinese MedicineChina
- Oncology Teaching and Research DepartmentChengdu University of Traditional Chinese MedicineChina
| |
Collapse
|
26
|
Saukko-Paavola AJ, Klemm RW. Remodelling of mitochondrial function by import of specific lipids at multiple membrane-contact sites. FEBS Lett 2024; 598:1274-1291. [PMID: 38311340 DOI: 10.1002/1873-3468.14813] [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: 11/29/2023] [Revised: 12/14/2023] [Accepted: 12/28/2023] [Indexed: 02/08/2024]
Abstract
Organelles form physical and functional contact between each other to exchange information, metabolic intermediates, and signaling molecules. Tethering factors and contact site complexes bring partnering organelles into close spatial proximity to establish membrane contact sites (MCSs), which specialize in unique functions like lipid transport or Ca2+ signaling. Here, we discuss how MCSs form dynamic platforms that are important for lipid metabolism. We provide a perspective on how import of specific lipids from the ER and other organelles may contribute to remodeling of mitochondria during nutrient starvation. We speculate that mitochondrial adaptation is achieved by connecting several compartments into a highly dynamic organelle network. The lipid droplet appears to be a central hub in coordinating the function of these organelle neighborhoods.
Collapse
Affiliation(s)
| | - Robin W Klemm
- Department of Physiology, Anatomy and Genetics, University of Oxford, UK
| |
Collapse
|
27
|
Monteiro-Cardoso VF, Giordano F. Emerging functions of the mitochondria-ER-lipid droplet three-way junction in coordinating lipid transfer, metabolism, and storage in cells. FEBS Lett 2024; 598:1252-1273. [PMID: 38774950 DOI: 10.1002/1873-3468.14893] [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: 02/05/2024] [Revised: 03/18/2024] [Accepted: 04/05/2024] [Indexed: 05/25/2024]
Abstract
Over the past two decades, we have witnessed a growing appreciation for the importance of membrane contact sites (CS) in facilitating direct communication between organelles. CS are tiny regions where the membranes of two organelles meet but do not fuse and allow the transfer of metabolites between organelles, playing crucial roles in the coordination of cellular metabolic activities. The significant advancements in imaging techniques and molecular and cell biology research have revealed that CS are more complex than what originally thought, and as they are extremely dynamic, they can remodel their shape, composition, and functions in accordance with metabolic and environmental changes and can occur between more than two organelles. Here, we describe how recent studies led to the identification of a three-way mitochondria-ER-lipid droplet CS and discuss the emerging functions of these contacts in maintaining lipid storage, homeostasis, and balance. We also summarize the properties and functions of key protein components localized at the mitochondria-ER-lipid droplet interface, with a special focus on lipid transfer proteins. Understanding tripartite CS is essential for unraveling the complexities of inter-organelle communication and cooperation within cells.
Collapse
Affiliation(s)
- Vera Filipa Monteiro-Cardoso
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, Gif-sur-Yvette cedex, France
- Inserm U1280, Gif-sur-Yvette cedex, France
| | - Francesca Giordano
- Institute for Integrative Biology of the Cell (I2BC), CEA, CNRS, Université Paris-Saclay, Gif-sur-Yvette cedex, France
- Inserm U1280, Gif-sur-Yvette cedex, France
| |
Collapse
|
28
|
Kalvelage J, Wöhlbrand L, Senkler J, Schumacher J, Ditz N, Bischof K, Winklhofer M, Klingl A, Braun HP, Rabus R. Conspicuous chloroplast with light harvesting-photosystem I/II megacomplex in marine Prorocentrum cordatum. PLANT PHYSIOLOGY 2024; 195:306-325. [PMID: 38330164 PMCID: PMC11181951 DOI: 10.1093/plphys/kiae052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 01/05/2024] [Accepted: 01/06/2024] [Indexed: 02/10/2024]
Abstract
Marine photosynthetic (micro)organisms drive multiple biogeochemical cycles and display a large diversity. Among them, the bloom-forming, free-living dinoflagellate Prorocentrum cordatum CCMP 1329 (formerly P. minimum) stands out with its distinct cell biological features. Here, we obtained insights into the structural properties of the chloroplast and the photosynthetic machinery of P. cordatum using microscopic and proteogenomic approaches. High-resolution FIB/SEM analysis revealed a single large chloroplast (∼40% of total cell volume) with a continuous barrel-like structure, completely lining the inner face of the cell envelope and enclosing a single reticular mitochondrium, the Golgi apparatus, as well as diverse storage inclusions. Enriched thylakoid membrane fractions of P. cordatum were comparatively analyzed with those of the well-studied model-species Arabidopsis (Arabidopsis thaliana) using 2D BN DIGE. Strikingly, P. cordatum possessed a large photosystem-light harvesting megacomplex (>1.5 MDa), which is dominated by photosystems I and II (PSI, PSII), chloroplast complex I, and chlorophyll a-b binding light harvesting complex proteins. This finding parallels the absence of grana in its chloroplast and distinguishes from the predominant separation of PSI and PSII complexes in A. thaliana, indicating a different mode of flux balancing. Except for the core elements of the ATP synthase and the cytb6f-complex, the composition of the other complexes (PSI, PSII, and pigment-binding proteins, PBPs) of P. cordatum differed markedly from those of A. thaliana. Furthermore, a high number of PBPs was detected, accounting for a large share of the total proteomic data (∼65%) and potentially providing P. cordatum with flexible adaptation to changing light regimes.
Collapse
Affiliation(s)
- Jana Kalvelage
- School of Mathematics and Science, Institute for Chemistry and Biology of the Marine Environment (ICBM), Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany
| | - Lars Wöhlbrand
- School of Mathematics and Science, Institute for Chemistry and Biology of the Marine Environment (ICBM), Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany
| | - Jennifer Senkler
- Faculty of Natural Sciences, Institute of Plant Genetics, Leibniz Universität Hannover, 30419 Hannover, Germany
| | - Julian Schumacher
- School of Mathematics and Science, Institute for Chemistry and Biology of the Marine Environment (ICBM), Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany
| | - Noah Ditz
- Faculty of Natural Sciences, Institute of Plant Genetics, Leibniz Universität Hannover, 30419 Hannover, Germany
| | - Kai Bischof
- Faculty Biology/Chemistry, University of Bremen & MARUM, 28359 Bremen, Germany
| | - Michael Winklhofer
- School of Mathematics and Science, Institute of Biology and Environmental Sciences (IBU), Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany
- Research Center Neurosensory Science, School of Mathematics and Science, Carl von Ossietzky University of Oldenburg, 26129 Oldenburg, Germany
| | - Andreas Klingl
- Faculty of Biology, Botany, Ludwig-Maximilians-Universität LMU München, 82152 Planegg-Martinsried, Germany
| | - Hans-Peter Braun
- Faculty of Natural Sciences, Institute of Plant Genetics, Leibniz Universität Hannover, 30419 Hannover, Germany
| | - Ralf Rabus
- School of Mathematics and Science, Institute for Chemistry and Biology of the Marine Environment (ICBM), Carl von Ossietzky Universität Oldenburg, 26129 Oldenburg, Germany
| |
Collapse
|
29
|
Lei Y, Gan M, Qiu Y, Chen Q, Wang X, Liao T, Zhao M, Chen L, Zhang S, Zhao Y, Niu L, Wang Y, Zhu L, Shen L. The role of mitochondrial dynamics and mitophagy in skeletal muscle atrophy: from molecular mechanisms to therapeutic insights. Cell Mol Biol Lett 2024; 29:59. [PMID: 38654156 PMCID: PMC11036639 DOI: 10.1186/s11658-024-00572-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: 01/09/2024] [Accepted: 04/04/2024] [Indexed: 04/25/2024] Open
Abstract
Skeletal muscle is the largest metabolic organ of the human body. Maintaining the best quality control and functional integrity of mitochondria is essential for the health of skeletal muscle. However, mitochondrial dysfunction characterized by mitochondrial dynamic imbalance and mitophagy disruption can lead to varying degrees of muscle atrophy, but the underlying mechanism of action is still unclear. Although mitochondrial dynamics and mitophagy are two different mitochondrial quality control mechanisms, a large amount of evidence has indicated that they are interrelated and mutually regulated. The former maintains the balance of the mitochondrial network, eliminates damaged or aged mitochondria, and enables cells to survive normally. The latter degrades damaged or aged mitochondria through the lysosomal pathway, ensuring cellular functional health and metabolic homeostasis. Skeletal muscle atrophy is considered an urgent global health issue. Understanding and gaining knowledge about muscle atrophy caused by mitochondrial dysfunction, particularly focusing on mitochondrial dynamics and mitochondrial autophagy, can greatly contribute to the prevention and treatment of muscle atrophy. In this review, we critically summarize the recent research progress on mitochondrial dynamics and mitophagy in skeletal muscle atrophy, and expound on the intrinsic molecular mechanism of skeletal muscle atrophy caused by mitochondrial dynamics and mitophagy. Importantly, we emphasize the potential of targeting mitochondrial dynamics and mitophagy as therapeutic strategies for the prevention and treatment of muscle atrophy, including pharmacological treatment and exercise therapy, and summarize effective methods for the treatment of skeletal muscle atrophy.
Collapse
Affiliation(s)
- Yuhang Lei
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Mailin Gan
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yanhao Qiu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Qiuyang Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Xingyu Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Tianci Liao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Mengying Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Lei Chen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Shunhua Zhang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Ye Zhao
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Lili Niu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Yan Wang
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China
| | - Li Zhu
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China.
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
| | - Linyuan Shen
- Farm Animal Genetic Resources Exploration and Innovation Key Laboratory of Sichuan Province, Sichuan Agricultural University, Chengdu, 611130, China.
- Key Laboratory of Livestock and Poultry Multi-Omics, Ministry of Agriculture and Rural Affairs, College of Animal and Technology, Sichuan Agricultural University, Chengdu, 611130, China.
| |
Collapse
|
30
|
Pays E. The Janus-faced functions of Apolipoproteins L in membrane dynamics. Cell Mol Life Sci 2024; 81:134. [PMID: 38478101 PMCID: PMC10937811 DOI: 10.1007/s00018-024-05180-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2023] [Revised: 02/06/2024] [Accepted: 02/18/2024] [Indexed: 03/17/2024]
Abstract
The functions of human Apolipoproteins L (APOLs) are poorly understood, but involve diverse activities like lysis of bloodstream trypanosomes and intracellular bacteria, modulation of viral infection and induction of apoptosis, autophagy, and chronic kidney disease. Based on recent work, I propose that the basic function of APOLs is the control of membrane dynamics, at least in the Golgi and mitochondrion. Together with neuronal calcium sensor-1 (NCS1) and calneuron-1 (CALN1), APOL3 controls the activity of phosphatidylinositol-4-kinase-IIIB (PI4KB), involved in both Golgi and mitochondrion membrane fission. Whereas secreted APOL1 induces African trypanosome lysis through membrane permeabilization of the parasite mitochondrion, intracellular APOL1 conditions non-muscular myosin-2A (NM2A)-mediated transfer of PI4KB and APOL3 from the Golgi to the mitochondrion under conditions interfering with PI4KB-APOL3 interaction, such as APOL1 C-terminal variant expression or virus-induced inflammatory signalling. APOL3 controls mitophagy through complementary interactions with the membrane fission factor PI4KB and the membrane fusion factor vesicle-associated membrane protein-8 (VAMP8). In mice, the basic APOL1 and APOL3 activities could be exerted by mAPOL9 and mAPOL8, respectively. Perspectives regarding the mechanism and treatment of APOL1-related kidney disease are discussed, as well as speculations on additional APOLs functions, such as APOL6 involvement in adipocyte membrane dynamics through interaction with myosin-10 (MYH10).
Collapse
Affiliation(s)
- Etienne Pays
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041, Gosselies, Belgium.
| |
Collapse
|
31
|
Wilson EL, Yu Y, Leal NS, Woodward JA, Patikas N, Morris JL, Field SF, Plumbly W, Paupe V, Chowdhury SR, Antrobus R, Lindop GE, Adia YM, Loh SHY, Prudent J, Martins LM, Metzakopian E. Genome-wide CRISPR/Cas9 screen shows that loss of GET4 increases mitochondria-endoplasmic reticulum contact sites and is neuroprotective. Cell Death Dis 2024; 15:203. [PMID: 38467609 PMCID: PMC10928201 DOI: 10.1038/s41419-024-06568-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: 06/26/2023] [Revised: 02/13/2024] [Accepted: 02/19/2024] [Indexed: 03/13/2024]
Abstract
Organelles form membrane contact sites between each other, allowing for the transfer of molecules and signals. Mitochondria-endoplasmic reticulum (ER) contact sites (MERCS) are cellular subdomains characterized by close apposition of mitochondria and ER membranes. They have been implicated in many diseases, including neurodegenerative, metabolic, and cardiac diseases. Although MERCS have been extensively studied, much remains to be explored. To uncover novel regulators of MERCS, we conducted a genome-wide, flow cytometry-based screen using an engineered MERCS reporter cell line. We found 410 genes whose downregulation promotes MERCS and 230 genes whose downregulation decreases MERCS. From these, 29 genes were selected from each population for arrayed screening and 25 were validated from the high population and 13 from the low population. GET4 and BAG6 were highlighted as the top 2 genes that upon suppression increased MERCS from both the pooled and arrayed screens, and these were subjected to further investigation. Multiple microscopy analyses confirmed that loss of GET4 or BAG6 increased MERCS. GET4 and BAG6 were also observed to interact with the known MERCS proteins, inositol 1,4,5-trisphosphate receptors (IP3R) and glucose-regulated protein 75 (GRP75). In addition, we found that loss of GET4 increased mitochondrial calcium uptake upon ER-Ca2+ release and mitochondrial respiration. Finally, we show that loss of GET4 rescues motor ability, improves lifespan and prevents neurodegeneration in a Drosophila model of Alzheimer's disease (Aβ42Arc). Together, these results suggest that GET4 is involved in decreasing MERCS and that its loss is neuroprotective.
Collapse
Affiliation(s)
- Emma L Wilson
- UK Dementia Research Institute, University of Cambridge, Clifford Albutt building, Cambridge biomedical campus, Cambridge, CB2 0AH, UK
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters building, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK
| | - Yizhou Yu
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK
| | - Nuno S Leal
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK
| | - James A Woodward
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK
| | - Nikolaos Patikas
- UK Dementia Research Institute, University of Cambridge, Clifford Albutt building, Cambridge biomedical campus, Cambridge, CB2 0AH, UK
| | - Jordan L Morris
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters building, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK
| | - Sarah F Field
- UK Dementia Research Institute, University of Cambridge, Clifford Albutt building, Cambridge biomedical campus, Cambridge, CB2 0AH, UK
| | - William Plumbly
- UK Dementia Research Institute, University of Cambridge, Clifford Albutt building, Cambridge biomedical campus, Cambridge, CB2 0AH, UK
| | - Vincent Paupe
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters building, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK
| | - Suvagata R Chowdhury
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters building, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK
| | - Robin Antrobus
- Cambridge Institute for Medical Research, University of Cambridge, The Keith Peters Building, Cambridge Biomedical Campus, Hills Rd, Cambridge, CB2 0XY, UK
| | - Georgina E Lindop
- Cambridge Advanced Imaging Centre, University of Cambridge, Anatomy Building, Downing Site, Cambridge, CB2 3DY, UK
| | - Yusuf M Adia
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK
| | - Samantha H Y Loh
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK
| | - Julien Prudent
- MRC Mitochondrial Biology Unit, University of Cambridge, The Keith Peters building, Cambridge Biomedical Campus, Cambridge, CB2 0XY, UK.
| | - L Miguel Martins
- MRC Toxicology Unit, University of Cambridge, Gleeson Building, Tennis Court Road, Cambridge, CB2 1QR, UK.
| | - Emmanouil Metzakopian
- UK Dementia Research Institute, University of Cambridge, Clifford Albutt building, Cambridge biomedical campus, Cambridge, CB2 0AH, UK.
- bit bio, The Dorothy Hodgkin Building, Babraham Research Campus, Cambridge, CB22 3FH, UK.
| |
Collapse
|
32
|
He J, Liu K, Fu C. Recent insights into the control of mitochondrial fission. Biochem Soc Trans 2024; 52:99-110. [PMID: 38288744 DOI: 10.1042/bst20230220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 01/07/2024] [Accepted: 01/09/2024] [Indexed: 02/29/2024]
Abstract
Mitochondria are the powerhouse of the cell. They undergo fission and fusion to maintain cellular homeostasis. In this review, we explore the intricate regulation of mitochondrial fission at various levels, including the protein level, the post-translational modification level, and the organelle level. Malfunctions in mitochondrial fission can have detrimental effects on cells. Therefore, we also examine the association between mitochondrial fission with diseases such as breast cancer and cardiovascular disorders. We anticipate that a comprehensive investigation into the control of mitochondrial fission will pave the way for the development of innovative therapeutic strategies.
Collapse
Affiliation(s)
- Jiajia He
- MOE Key Laboratory for Cellular Dynamics and Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
- Anhui Key Laboratory of Cellular Dynamics and Chemical Biology and Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Ke Liu
- MOE Key Laboratory for Cellular Dynamics and Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
- Anhui Key Laboratory of Cellular Dynamics and Chemical Biology and Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| | - Chuanhai Fu
- MOE Key Laboratory for Cellular Dynamics and Center for Advanced Interdisciplinary Science and Biomedicine of IHM, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei 230027, China
- Anhui Key Laboratory of Cellular Dynamics and Chemical Biology and Hefei National Research Center for Interdisciplinary Sciences at the Microscale, School of Life Sciences, University of Science and Technology of China, Hefei 230027, China
| |
Collapse
|
33
|
Giamogante F, Barazzuol L, Maiorca F, Poggio E, Esposito A, Masato A, Napolitano G, Vagnoni A, Calì T, Brini M. A SPLICS reporter reveals [Formula: see text]-synuclein regulation of lysosome-mitochondria contacts which affects TFEB nuclear translocation. Nat Commun 2024; 15:1516. [PMID: 38374070 PMCID: PMC10876553 DOI: 10.1038/s41467-024-46007-2] [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: 06/19/2023] [Accepted: 02/07/2024] [Indexed: 02/21/2024] Open
Abstract
Mitochondrial and lysosomal activities are crucial to maintain cellular homeostasis: optimal coordination is achieved at their membrane contact sites where distinct protein machineries regulate organelle network dynamics, ions and metabolites exchange. Here we describe a genetically encoded SPLICS reporter for short- and long- juxtapositions between mitochondria and lysosomes. We report the existence of narrow and wide lysosome-mitochondria contacts differently modulated by mitophagy, autophagy and genetic manipulation of tethering factors. The overexpression of α-synuclein (α-syn) reduces the apposition of mitochondria/lysosomes membranes and affects their privileged Ca2+ transfer, impinging on TFEB nuclear translocation. We observe enhanced TFEB nuclear translocation in α-syn-overexpressing cells. We propose that α-syn, by interfering with mitochondria/lysosomes tethering impacts on local Ca2+ regulated pathways, among which TFEB mediated signaling, and in turn mitochondrial and lysosomal function. Defects in mitochondria and lysosome represent a common hallmark of neurodegenerative diseases: targeting their communication could open therapeutic avenues.
Collapse
Affiliation(s)
- Flavia Giamogante
- Department of Biomedical Sciences (DSB), University of Padova, Padova, Italy
| | - Lucia Barazzuol
- Department of Biomedical Sciences (DSB), University of Padova, Padova, Italy
| | | | - Elena Poggio
- Department of Biology (DIBIO), University of Padova, Padova, Italy
| | - Alessandra Esposito
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Department of Medical and Translational Science, Federico II University, Naples, Italy
| | - Anna Masato
- Department of Biology (DIBIO), University of Padova, Padova, Italy
- UK-Dementia Research Institute at UCL, University College London, London, UK
| | - Gennaro Napolitano
- Telethon Institute of Genetics and Medicine (TIGEM), Naples, Italy
- Department of Medical and Translational Science, Federico II University, Naples, Italy
| | - Alessio Vagnoni
- Department of Basic and Clinical Neurosciences, Maurice Wohl Clinical Neuroscience Institute, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
| | - Tito Calì
- Department of Biomedical Sciences (DSB), University of Padova, Padova, Italy.
- Padova Neuroscience Center (PNC), University of Padova, Padova, Italy.
- Study Center for Neurodegeneration (CESNE), University of Padova, Padova, Italy.
| | - Marisa Brini
- Department of Biology (DIBIO), University of Padova, Padova, Italy.
- Study Center for Neurodegeneration (CESNE), University of Padova, Padova, Italy.
- Department of Pharmaceutical and Pharmacological Sciences (DSF), University of Padova, Padova, Italy.
| |
Collapse
|
34
|
Polanco CM, Cavieres VA, Galarza AJ, Jara C, Torres AK, Cancino J, Varas-Godoy M, Burgos PV, Tapia-Rojas C, Mardones GA. GOLPH3 Participates in Mitochondrial Fission and Is Necessary to Sustain Bioenergetic Function in MDA-MB-231 Breast Cancer Cells. Cells 2024; 13:316. [PMID: 38391929 PMCID: PMC10887169 DOI: 10.3390/cells13040316] [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: 12/17/2023] [Revised: 02/03/2024] [Accepted: 02/05/2024] [Indexed: 02/24/2024] Open
Abstract
In this study, we investigated the inter-organelle communication between the Golgi apparatus (GA) and mitochondria. Previous observations suggest that GA-derived vesicles containing phosphatidylinositol 4-phosphate (PI(4)P) play a role in mitochondrial fission, colocalizing with DRP1, a key protein in this process. However, the functions of these vesicles and potentially associated proteins remain unknown. GOLPH3, a PI(4)P-interacting GA protein, is elevated in various types of solid tumors, including breast cancer, yet its precise role is unclear. Interestingly, GOLPH3 levels influence mitochondrial mass by affecting cardiolipin synthesis, an exclusive mitochondrial lipid. However, the mechanism by which GOLPH3 influences mitochondria is not fully understood. Our live-cell imaging analysis showed GFP-GOLPH3 associating with PI(4)P vesicles colocalizing with YFP-DRP1 at mitochondrial fission sites. We tested the functional significance of these observations with GOLPH3 knockout in MDA-MB-231 cells of breast cancer, resulting in a fragmented mitochondrial network and reduced bioenergetic function, including decreased mitochondrial ATP production, mitochondrial membrane potential, and oxygen consumption. Our findings suggest a potential negative regulatory role for GOLPH3 in mitochondrial fission, impacting mitochondrial function and providing insights into GA-mitochondria communication.
Collapse
Affiliation(s)
- Catalina M. Polanco
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia, Santiago 7510156, Chile; (C.M.P.); (V.A.C.); (C.J.); (A.K.T.); (J.C.); (M.V.-G.); (P.V.B.)
| | - Viviana A. Cavieres
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia, Santiago 7510156, Chile; (C.M.P.); (V.A.C.); (C.J.); (A.K.T.); (J.C.); (M.V.-G.); (P.V.B.)
- Departamento de Ciencias Biológicas y Químicas, Facultad de Medicina y Ciencia, Universidad San Sebastián, Campus Los Leones, Providencia, Santiago 7510156, Chile
| | - Abigail J. Galarza
- Escuela de Medicina, Facultad de Medicina y Ciencia, Universidad San Sebastián, Valdivia 5110693, Chile;
| | - Claudia Jara
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia, Santiago 7510156, Chile; (C.M.P.); (V.A.C.); (C.J.); (A.K.T.); (J.C.); (M.V.-G.); (P.V.B.)
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba, Santiago 8580702, Chile
| | - Angie K. Torres
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia, Santiago 7510156, Chile; (C.M.P.); (V.A.C.); (C.J.); (A.K.T.); (J.C.); (M.V.-G.); (P.V.B.)
- Centro de Excelencia en Biomedicina de Magallanes (CEBIMA), Universidad de Magallanes, Punta Arenas 6210427, Chile
| | - Jorge Cancino
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia, Santiago 7510156, Chile; (C.M.P.); (V.A.C.); (C.J.); (A.K.T.); (J.C.); (M.V.-G.); (P.V.B.)
- Escuela de Medicina, Facultad de Medicina y Ciencia, Universidad San Sebastián, Valdivia 5110693, Chile;
| | - Manuel Varas-Godoy
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia, Santiago 7510156, Chile; (C.M.P.); (V.A.C.); (C.J.); (A.K.T.); (J.C.); (M.V.-G.); (P.V.B.)
- Escuela de Medicina, Facultad de Medicina y Ciencia, Universidad San Sebastián, Valdivia 5110693, Chile;
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba, Santiago 8580702, Chile
| | - Patricia V. Burgos
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia, Santiago 7510156, Chile; (C.M.P.); (V.A.C.); (C.J.); (A.K.T.); (J.C.); (M.V.-G.); (P.V.B.)
- Escuela de Medicina, Facultad de Medicina y Ciencia, Universidad San Sebastián, Valdivia 5110693, Chile;
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba, Santiago 8580702, Chile
| | - Cheril Tapia-Rojas
- Centro de Biología Celular y Biomedicina (CEBICEM), Facultad de Medicina y Ciencia, Universidad San Sebastián, Providencia, Santiago 7510156, Chile; (C.M.P.); (V.A.C.); (C.J.); (A.K.T.); (J.C.); (M.V.-G.); (P.V.B.)
- Centro Científico y Tecnológico de Excelencia Ciencia & Vida, Fundación Ciencia & Vida, Huechuraba, Santiago 8580702, Chile
| | - Gonzalo A. Mardones
- Escuela de Medicina, Facultad de Medicina y Ciencia, Universidad San Sebastián, Valdivia 5110693, Chile;
| |
Collapse
|
35
|
Wit M, Belykh A, Sumara G. Protein kinase D (PKD) on the crossroad of lipid absorption, synthesis and utilization. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119653. [PMID: 38104800 DOI: 10.1016/j.bbamcr.2023.119653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 10/19/2023] [Accepted: 11/30/2023] [Indexed: 12/19/2023]
Abstract
Inappropriate lipid levels in the blood, as well as its content and composition in different organs, underlie multiple metabolic disorders including obesity, non-alcoholic fatty liver disease, type 2 diabetes, and atherosclerosis. Multiple processes contribute to the complex metabolism of triglycerides (TGs), fatty acids (FAs), and other lipid species. These consist of digestion and absorption of dietary lipids, de novo FAs synthesis (lipogenesis), uptake of TGs and FAs by peripheral tissues, TGs storage in the intracellular depots as well as lipid utilization for β-oxidation and their conversion to lipid-derivatives. A majority of the enzymatic reactions linked to lipogenesis, TGs synthesis, lipid absorption, and transport are happening at the endoplasmic reticulum, while β-oxidation takes place in mitochondria and peroxisomes. The Golgi apparatus is a central sorting, protein- and lipid-modifying organelle and hence is involved in lipid metabolism as well. However, the impact of the processes taking part in the Golgi apparatus are often overseen. The protein kinase D (PKD) family (composed of three members, PKD1, 2, and 3) is the master regulator of Golgi dynamics. PKDs are also a sensor of different lipid species in distinct cellular compartments. In this review, we discuss the roles of PKD family members in the regulation of lipid metabolism including the processes executed by PKDs at the Golgi apparatus. We also discuss the role of PKDs-dependent signaling in different cellular compartments and organs in the context of the development of metabolic disorders.
Collapse
Affiliation(s)
- Magdalena Wit
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warszawa, Poland
| | - Andrei Belykh
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warszawa, Poland
| | - Grzegorz Sumara
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Street, 02-093 Warszawa, Poland.
| |
Collapse
|
36
|
Wu Z, Xiao C, Long J, Huang W, You F, Li X. Mitochondrial dynamics and colorectal cancer biology: mechanisms and potential targets. Cell Commun Signal 2024; 22:91. [PMID: 38302953 PMCID: PMC10835948 DOI: 10.1186/s12964-024-01490-4] [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: 08/22/2023] [Accepted: 01/11/2024] [Indexed: 02/03/2024] Open
Abstract
Colorectal cancer (CRC) is a significant public health concern, and its development is associated with mitochondrial dysfunction. Mitochondria can adapt to the high metabolic demands of cancer cells owing to their plasticity and dynamic nature. The fusion-fission dynamics of mitochondria play a crucial role in signal transduction and metabolic functions of CRC cells. Enhanced mitochondrial fission promotes the metabolic reprogramming of CRC cells, leading to cell proliferation, metastasis, and chemoresistance. Excessive fission can also trigger mitochondria-mediated apoptosis. In contrast, excessive mitochondrial fusion leads to adenosine triphosphate (ATP) overproduction and abnormal tumor proliferation, whereas moderate fusion protects intestinal epithelial cells from oxidative stress-induced mitochondrial damage, thus preventing colitis-associated cancer (CAC). Therefore, an imbalance in mitochondrial dynamics can either promote or inhibit CRC progression. This review provides an overview of the mechanism underlying mitochondrial fusion-fission dynamics and their impact on CRC biology. This revealed the dual role of mitochondrial fusion-fission dynamics in CRC development and identified potential drug targets. Additionally, this study partially explored mitochondrial dynamics in immune and vascular endothelial cells in the tumor microenvironment, suggesting promising prospects for targeting key fusion/fission effector proteins against CRC.
Collapse
Affiliation(s)
- Zihong Wu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Chong Xiao
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
- Oncology Teaching and Research Department of Chengdu, University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Jing Long
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Wenbo Huang
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Fengming You
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
- Institute of Oncology, Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| | - Xueke Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
- Oncology Teaching and Research Department of Chengdu, University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
37
|
Voeltz GK, Sawyer EM, Hajnóczky G, Prinz WA. Making the connection: How membrane contact sites have changed our view of organelle biology. Cell 2024; 187:257-270. [PMID: 38242082 DOI: 10.1016/j.cell.2023.11.040] [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: 09/19/2023] [Revised: 11/13/2023] [Accepted: 11/29/2023] [Indexed: 01/21/2024]
Abstract
The view of organelles and how they operate together has changed dramatically over the last two decades. The textbook view of organelles was that they operated largely independently and were connected by vesicular trafficking and the diffusion of signals through the cytoplasm. We now know that all organelles make functional close contacts with one another, often called membrane contact sites. The study of these sites has moved to center stage in cell biology as it has become clear that they play critical roles in healthy and developing cells and during cell stress and disease states. Contact sites have important roles in intracellular signaling, lipid metabolism, motor-protein-mediated membrane dynamics, organelle division, and organelle biogenesis. Here, we summarize the major conceptual changes that have occurred in cell biology as we have come to appreciate how contact sites integrate the activities of organelles.
Collapse
Affiliation(s)
- G K Voeltz
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Boulder, CO 80309, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA.
| | - E M Sawyer
- Department of Molecular, Cellular, and Developmental Biology, University of Colorado, Boulder, Boulder, CO 80309, USA; Howard Hughes Medical Institute, Chevy Chase, MD 20815, USA
| | - G Hajnóczky
- MitoCare Center, Department of Pathology, Anatomy and Cell Biology, Thomas Jefferson University, Philadelphia, PA 19107, USA
| | - W A Prinz
- Department of Cell Biology, UT Southwestern Medical Center, Dallas, TX 75390, USA.
| |
Collapse
|
38
|
Lourdes SR, Gurung R, Giri S, Mitchell CA, McGrath MJ. A new role for phosphoinositides in regulating mitochondrial dynamics. Adv Biol Regul 2024; 91:101001. [PMID: 38057188 DOI: 10.1016/j.jbior.2023.101001] [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: 11/17/2023] [Accepted: 11/27/2023] [Indexed: 12/08/2023]
Abstract
Phosphoinositides are a minor group of membrane-associated phospholipids that are transiently generated on the cytoplasmic leaflet of many organelle membranes and the plasma membrane. There are seven functionally distinct phosphoinositides, each derived via the reversible phosphorylation of phosphatidylinositol in various combinations on the inositol ring. Their generation and termination is tightly regulated by phosphatidylinositol-kinases and -phosphatases. These enzymes can function together in an integrated and coordinated manner, whereby the phosphoinositide product of one enzyme may subsequently serve as a substrate for another to generate a different phosphoinositide species. This regulatory mechanism not only enables the transient generation of phosphoinositides on membranes, but also more complex sequential or bidirectional conversion pathways, and phosphoinositides can also be transferred between organelles via membrane contacts. It is this capacity to fine-tune phosphoinositide signals that makes them ideal regulators of membrane organization and dynamics, through their recruitment of signalling, membrane altering and lipid transfer proteins. Research spanning several decades has provided extensive evidence that phosphoinositides are major gatekeepers of membrane organization, with roles in endocytosis, exocytosis, autophagy, lysosome dynamics, vesicular transport and secretion, cilia, inter-organelle membrane contact, endosome maturation and nuclear function. By contrast, there has been remarkably little known about the role of phosphoinositides at mitochondria - an enigmatic and major knowledge gap, with challenges in reliably detecting phosphoinositides at this site. Here we review recent significant breakthroughs in understanding the role of phosphoinositides in regulating mitochondrial dynamics and metabolic function.
Collapse
Affiliation(s)
- Sonia Raveena Lourdes
- Cancer Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Rajendra Gurung
- Cancer Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Saveen Giri
- Cancer Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Christina A Mitchell
- Cancer Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia.
| | - Meagan J McGrath
- Cancer Program, Monash Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia; Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| |
Collapse
|
39
|
Area-Gomez E, Schon EA. Towards a Unitary Hypothesis of Alzheimer's Disease Pathogenesis. J Alzheimers Dis 2024; 98:1243-1275. [PMID: 38578892 DOI: 10.3233/jad-231318] [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: 04/07/2024]
Abstract
The "amyloid cascade" hypothesis of Alzheimer's disease (AD) pathogenesis invokes the accumulation in the brain of plaques (containing the amyloid-β protein precursor [AβPP] cleavage product amyloid-β [Aβ]) and tangles (containing hyperphosphorylated tau) as drivers of pathogenesis. However, the poor track record of clinical trials based on this hypothesis suggests that the accumulation of these peptides is not the only cause of AD. Here, an alternative hypothesis is proposed in which the AβPP cleavage product C99, not Aβ, is the main culprit, via its role as a regulator of cholesterol metabolism. C99, which is a cholesterol sensor, promotes the formation of mitochondria-associated endoplasmic reticulum (ER) membranes (MAM), a cholesterol-rich lipid raft-like subdomain of the ER that communicates, both physically and biochemically, with mitochondria. We propose that in early-onset AD (EOAD), MAM-localized C99 is elevated above normal levels, resulting in increased transport of cholesterol from the plasma membrane to membranes of intracellular organelles, such as ER/endosomes, thereby upregulating MAM function and driving pathology. By the same token, late-onset AD (LOAD) is triggered by any genetic variant that increases the accumulation of intracellular cholesterol that, in turn, boosts the levels of C99 and again upregulates MAM function. Thus, the functional cause of AD is upregulated MAM function that, in turn, causes the hallmark disease phenotypes, including the plaques and tangles. Accordingly, the MAM hypothesis invokes two key interrelated elements, C99 and cholesterol, that converge at the MAM to drive AD pathogenesis. From this perspective, AD is, at bottom, a lipid disorder.
Collapse
Affiliation(s)
- Estela Area-Gomez
- Department of Neurology, Columbia University, New York, NY, USA
- Centro de Investigaciones Biológicas "Margarita Salas", Spanish National Research Council, Madrid, Spain
| | - Eric A Schon
- Department of Neurology, Columbia University, New York, NY, USA
- Department of Genetics and Development>, Columbia University, New York, NY, USA
| |
Collapse
|
40
|
Zhang R, Lan J, Chen Q, Liu Y, Hu L, Cao J, Zhao H, Shen Y. Hesperidin Alleviates Acute Necrotizing Pancreatitis by Activating SIRT1 - Molecular Docking, Molecular Dynamics Simulation, and Experimental Validation. Comb Chem High Throughput Screen 2024; 27:1745-1757. [PMID: 37534793 DOI: 10.2174/1386207326666230803140408] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 06/11/2023] [Accepted: 06/21/2023] [Indexed: 08/04/2023]
Abstract
BACKGROUND Acute necrotizing pancreatitis is a serious pancreatic injury with limited effective treatments. This study aims to investigate the therapeutic effects of hesperidin on Larginine- induced acute pancreatitis and its potential targets. METHODS The authors induced acute pancreatitis in mice by administering two hourly intraperitoneal injections of L-arginine-HCl, and evaluated the impact of hesperidin on pancreatic and lung tissues, plasma amylase activity, and myeloperoxidase content. Additionally, necrosis and mitochondrial function was tested in primary pancreatic acinar cells. The interactions between hesperidin and proteins involved in necrosis and mitochondrial dysfunction were further invested using in silico molecular docking and molecular dynamic simulations. RESULTS Hesperidin effectively ameliorated the severity of acute necrotizing pancreatitis by reducing plasma amylase, pancreatic MPO, serum IL-6 levels, pancreatic edema, inflammation, and pancreatic necrosis. Hesperidin also protected against acute pancreatitis-associated lung injury and prevented acinar cell necrosis, mitochondrial membrane potential loss, and ATP depletion. In addition, hesperidin exhibited a high binding affinity with SIRT1 and increased the protein levels of SIRT1. The SIRT1 inhibitor EX527 abolished the protective effect of hesperidin against necrosis in acinar cells. CONCLUSION These findings indicate that hesperidin alleviates the severity of acute necrotizing pancreatitis by activating SIRT1, which may provide insight into the mechanisms of natural compounds in treating AP. Hesperidin has potential as a therapeutic agent for acute necrotizing pancreatitis and provides a new approach for novel therapeutic strategies.
Collapse
Affiliation(s)
- Rui Zhang
- Department of Pharmacy, Guizhou Provincial People's Hospital, 550002 Guiyang, China
| | - Junjie Lan
- Department of Pharmacy, Guizhou Provincial People's Hospital, 550002 Guiyang, China
| | - Qi Chen
- Department of Pharmacy, Guizhou Provincial People's Hospital, 550002 Guiyang, China
| | - Yang Liu
- Department of Hepatobiliary Surgery II, Guizhou Provincial People's Hospital, 550002 Guiyang, China
| | - Linfang Hu
- Department of Pharmacy, Guizhou Provincial People's Hospital, 550002 Guiyang, China
| | - Jinyong Cao
- Department of Endoscopy, Guizhou Provincial People's Hospital, 550002 Guiyang, China
| | - Huaye Zhao
- Department of Pharmacy, Guizhou Provincial People's Hospital, 550002 Guiyang, China
| | - Yan Shen
- School of Pharmacy and Bioengineering, Chongqing University of Technology, 405400 Chongqing, China
| |
Collapse
|
41
|
Lecordier L, Heo P, Graversen JH, Hennig D, Skytthe MK, Cornet d'Elzius A, Pincet F, Pérez-Morga D, Pays E. Apolipoproteins L1 and L3 control mitochondrial membrane dynamics. Cell Rep 2023; 42:113528. [PMID: 38041817 PMCID: PMC10765320 DOI: 10.1016/j.celrep.2023.113528] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Revised: 11/08/2023] [Accepted: 11/17/2023] [Indexed: 12/04/2023] Open
Abstract
Apolipoproteins L1 and L3 (APOLs) are associated at the Golgi with the membrane fission factors phosphatidylinositol 4-kinase-IIIB (PI4KB) and non-muscular myosin 2A. Either APOL1 C-terminal truncation (APOL1Δ) or APOL3 deletion (APOL3-KO [knockout]) reduces PI4KB activity and triggers actomyosin reorganization. We report that APOL3, but not APOL1, controls PI4KB activity through interaction with PI4KB and neuronal calcium sensor-1 or calneuron-1. Both APOLs are present in Golgi-derived autophagy-related protein 9A vesicles, which are involved in PI4KB trafficking. Like APOL3-KO, APOL1Δ induces PI4KB dissociation from APOL3, linked to reduction of mitophagy flux and production of mitochondrial reactive oxygen species. APOL1 and APOL3, respectively, can interact with the mitophagy receptor prohibitin-2 and the mitophagosome membrane fusion factor vesicle-associated membrane protein-8 (VAMP8). While APOL1 conditions PI4KB and APOL3 involvement in mitochondrion fission and mitophagy, APOL3-VAMP8 interaction promotes fusion between mitophagosomal and endolysosomal membranes. We propose that APOL3 controls mitochondrial membrane dynamics through interactions with the fission factor PI4KB and the fusion factor VAMP8.
Collapse
Affiliation(s)
- Laurence Lecordier
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041 Gosselies, Belgium
| | - Paul Heo
- Laboratoire de Physique de l'Ecole Normale Supérieure, Ecole Normale Supérieure (ENS), Université Paris Sciences et Lettres (PSL), CNRS, Sorbonne Université, Université Paris-Cité, 75005 Paris, France; Institute of Psychiatry and Neuroscience of Paris, INSERM U1266, 75014 Paris, France
| | - Jonas H Graversen
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Dorle Hennig
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, 5000 Odense C, Denmark
| | - Maria Kløjgaard Skytthe
- Department of Molecular Medicine, Cancer and Inflammation Research, University of Southern Denmark, 5000 Odense C, Denmark
| | | | - Frédéric Pincet
- Laboratoire de Physique de l'Ecole Normale Supérieure, Ecole Normale Supérieure (ENS), Université Paris Sciences et Lettres (PSL), CNRS, Sorbonne Université, Université Paris-Cité, 75005 Paris, France
| | - David Pérez-Morga
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041 Gosselies, Belgium; Center for Microscopy and Molecular Imaging (CMMI), Université Libre de Bruxelles, 6041 Gosselies, Belgium
| | - Etienne Pays
- Laboratory of Molecular Parasitology, IBMM, Université Libre de Bruxelles, 6041 Gosselies, Belgium.
| |
Collapse
|
42
|
Lurette O, Martín-Jiménez R, Khan M, Sheta R, Jean S, Schofield M, Teixeira M, Rodriguez-Aller R, Perron I, Oueslati A, Hebert-Chatelain E. Aggregation of alpha-synuclein disrupts mitochondrial metabolism and induce mitophagy via cardiolipin externalization. Cell Death Dis 2023; 14:729. [PMID: 37949858 PMCID: PMC10638290 DOI: 10.1038/s41419-023-06251-8] [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: 01/03/2023] [Revised: 10/16/2023] [Accepted: 10/27/2023] [Indexed: 11/12/2023]
Abstract
Accumulation of α-synuclein aggregates in the substantia nigra pars compacta is central in the pathophysiology of Parkinson's disease, leading to the degeneration of dopaminergic neurons and the manifestation of motor symptoms. Although several PD models mimic the pathological accumulation of α-synuclein after overexpression, they do not allow for controlling and monitoring its aggregation. We recently generated a new optogenetic tool by which we can spatiotemporally control the aggregation of α-synuclein using a light-induced protein aggregation system. Using this innovative tool, we aimed to characterize the impact of α-synuclein clustering on mitochondria, whose activity is crucial to maintain neuronal survival. We observed that aggregates of α-synuclein transiently and dynamically interact with mitochondria, leading to mitochondrial depolarization, lower ATP production, mitochondrial fragmentation and degradation via cardiolipin externalization-dependent mitophagy. Aggregation of α-synuclein also leads to lower mitochondrial content in human dopaminergic neurons and in mouse midbrain. Interestingly, overexpression of α-synuclein alone did not induce mitochondrial degradation. This work is among the first to clearly discriminate between the impact of α-synuclein overexpression and aggregation on mitochondria. This study thus represents a new framework to characterize the role of mitochondria in PD.
Collapse
Affiliation(s)
- Olivier Lurette
- Canada Research Chair in Mitochondrial Signaling and Physiopathology, Moncton, NB, Canada
- Department of Biology, University of Moncton, Moncton, NB, Canada
| | - Rebeca Martín-Jiménez
- Canada Research Chair in Mitochondrial Signaling and Physiopathology, Moncton, NB, Canada
- Department of Biology, University of Moncton, Moncton, NB, Canada
| | - Mehtab Khan
- Canada Research Chair in Mitochondrial Signaling and Physiopathology, Moncton, NB, Canada
- Department of Biology, University of Moncton, Moncton, NB, Canada
| | - Razan Sheta
- CHU de Québec Research Center, Axe Neurosciences, Quebec City, QC, Canada
- Department of Molecular Medecine, Université Laval, Quebec City, QC, Canada
| | - Stéphanie Jean
- Canada Research Chair in Mitochondrial Signaling and Physiopathology, Moncton, NB, Canada
- Department of Biology, University of Moncton, Moncton, NB, Canada
| | - Mia Schofield
- Canada Research Chair in Mitochondrial Signaling and Physiopathology, Moncton, NB, Canada
- Department of Biology, University of Moncton, Moncton, NB, Canada
| | - Maxime Teixeira
- CHU de Québec Research Center, Axe Neurosciences, Quebec City, QC, Canada
- Department of Molecular Medecine, Université Laval, Quebec City, QC, Canada
| | - Raquel Rodriguez-Aller
- CHU de Québec Research Center, Axe Neurosciences, Quebec City, QC, Canada
- Department of Molecular Medecine, Université Laval, Quebec City, QC, Canada
| | - Isabelle Perron
- Canada Research Chair in Mitochondrial Signaling and Physiopathology, Moncton, NB, Canada
- Department of Biology, University of Moncton, Moncton, NB, Canada
| | - Abid Oueslati
- CHU de Québec Research Center, Axe Neurosciences, Quebec City, QC, Canada
- Department of Molecular Medecine, Université Laval, Quebec City, QC, Canada
| | - Etienne Hebert-Chatelain
- Canada Research Chair in Mitochondrial Signaling and Physiopathology, Moncton, NB, Canada.
- Department of Biology, University of Moncton, Moncton, NB, Canada.
| |
Collapse
|
43
|
Hirschenberger M, Lepelley A, Rupp U, Klute S, Hunszinger V, Koepke L, Merold V, Didry-Barca B, Wondany F, Bergner T, Moreau T, Rodero MP, Rösler R, Wiese S, Volpi S, Gattorno M, Papa R, Lynch SA, Haug MG, Houge G, Wigby KM, Sprague J, Lenberg J, Read C, Walther P, Michaelis J, Kirchhoff F, de Oliveira Mann CC, Crow YJ, Sparrer KMJ. ARF1 prevents aberrant type I interferon induction by regulating STING activation and recycling. Nat Commun 2023; 14:6770. [PMID: 37914730 PMCID: PMC10620153 DOI: 10.1038/s41467-023-42150-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2023] [Accepted: 09/29/2023] [Indexed: 11/03/2023] Open
Abstract
Type I interferon (IFN) signalling is tightly controlled. Upon recognition of DNA by cyclic GMP-AMP synthase (cGAS), stimulator of interferon genes (STING) translocates along the endoplasmic reticulum (ER)-Golgi axis to induce IFN signalling. Termination is achieved through autophagic degradation or recycling of STING by retrograde Golgi-to-ER transport. Here, we identify the GTPase ADP-ribosylation factor 1 (ARF1) as a crucial negative regulator of cGAS-STING signalling. Heterozygous ARF1 missense mutations cause a previously unrecognized type I interferonopathy associated with enhanced IFN-stimulated gene expression. Disease-associated, GTPase-defective ARF1 increases cGAS-STING dependent type I IFN signalling in cell lines and primary patient cells. Mechanistically, mutated ARF1 perturbs mitochondrial morphology, causing cGAS activation by aberrant mitochondrial DNA release, and leads to accumulation of active STING at the Golgi/ERGIC due to defective retrograde transport. Our data show an unexpected dual role of ARF1 in maintaining cGAS-STING homeostasis, through promotion of mitochondrial integrity and STING recycling.
Collapse
Affiliation(s)
| | - Alice Lepelley
- Université Paris Cité, Imagine Institute, Laboratory of Neurogenetics and Neuroinflammation, INSERM UMR1163, F-75015, Paris, France
| | - Ulrich Rupp
- Central Facility for Electron Microscopy, Ulm University, 89081, Ulm, Germany
| | - Susanne Klute
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Victoria Hunszinger
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Lennart Koepke
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | - Veronika Merold
- Institute of Virology, Technical University of Munich, 81675, Munich, Germany
| | - Blaise Didry-Barca
- Université Paris Cité, Imagine Institute, Laboratory of Neurogenetics and Neuroinflammation, INSERM UMR1163, F-75015, Paris, France
| | - Fanny Wondany
- Institute of Biophysics, Ulm University, 89081, Ulm, Germany
| | - Tim Bergner
- Central Facility for Electron Microscopy, Ulm University, 89081, Ulm, Germany
| | - Tatiana Moreau
- Université Paris Cité, Imagine Institute, Laboratory of Neurogenetics and Neuroinflammation, INSERM UMR1163, F-75015, Paris, France
| | - Mathieu P Rodero
- Université Paris Cité, Imagine Institute, Laboratory of Neurogenetics and Neuroinflammation, INSERM UMR1163, F-75015, Paris, France
| | - Reinhild Rösler
- Core Unit Mass Spectrometry and Proteomics, Ulm University, 89081, Ulm, Germany
| | - Sebastian Wiese
- Core Unit Mass Spectrometry and Proteomics, Ulm University, 89081, Ulm, Germany
| | - Stefano Volpi
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genoa, Italy
- Università degli Studi di Genova, Genoa, Italy
| | - Marco Gattorno
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Riccardo Papa
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto Giannina Gaslini, Genoa, Italy
| | - Sally-Ann Lynch
- Children's Health Ireland, Crumlin, Dublin, Eire
- University College Dublin, Dublin, Eire
| | - Marte G Haug
- Department of Medical Genetics, St. Olav's Hospital, Trondheim, Norway
| | - Gunnar Houge
- Department of Medical Genetics, Haukeland University Hospital, 5021, Bergen, Norway
| | - Kristen M Wigby
- Division of Genomic Medicine, Department of Pediatrics, University of California, Davis in Sacramento, CA, USA
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
| | - Jessica Sprague
- Division of Pediatric and Adolescent Dermatology, Rady Children's Hospital San Diego, San Diego, CA, USA
- Department of Dermatology, University of California San Diego School of Medicine, La Jolla, USA
| | - Jerica Lenberg
- Rady Children's Institute for Genomic Medicine, San Diego, CA, USA
| | - Clarissa Read
- Central Facility for Electron Microscopy, Ulm University, 89081, Ulm, Germany
| | - Paul Walther
- Central Facility for Electron Microscopy, Ulm University, 89081, Ulm, Germany
| | - Jens Michaelis
- Institute of Biophysics, Ulm University, 89081, Ulm, Germany
| | - Frank Kirchhoff
- Institute of Molecular Virology, Ulm University Medical Center, 89081, Ulm, Germany
| | | | - Yanick J Crow
- Université Paris Cité, Imagine Institute, Laboratory of Neurogenetics and Neuroinflammation, INSERM UMR1163, F-75015, Paris, France.
- MRC Human Genetics Unit, Institute of Genetics and Cancer, University of Edinburgh, Edinburgh, UK.
| | | |
Collapse
|
44
|
Davenport AM, Morris M, Sabti F, Sabti S, Shakya D, Hynds DL, Cheriyath V. G1P3/IFI6, an interferon stimulated protein, promotes the association of RAB5 + endosomes with mitochondria in breast cancer cells. Cell Biol Int 2023; 47:1868-1879. [PMID: 37598317 DOI: 10.1002/cbin.12079] [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: 02/07/2023] [Revised: 07/31/2023] [Accepted: 08/05/2023] [Indexed: 08/21/2023]
Abstract
G1P3/IFI6 is an interferon stimulated gene with antiapoptotic, prometastatic, and antiviral functions. Despite its pleiotropic functions, subcellular localization of G1P3 remains unclear. Using biochemical- and confocal microscopic approaches, this study identified the localization of G1P3 in organelles of the endomembrane system and in the mitochondria of breast cancer cells. In cell fractionation studies, both interferon-induced endogenous- and stably expressed G1P3 cofractionated with affinity-isolated mitochondria. Results of the protease protection assay have suggested that ~24% of mitochondrial G1P3 resides within the mitochondria. Conforming to this, confocal microscopy studies of cells stably expressing epitope-tagged G1P3 (MCF-7/G1P3-FLAG), identified its localization in mitochondria (~38%) as well as in ER, trans-Golgi network (TGN), lysosomes, and in RAB5 positive (RAB5+ ) endosomes. These results suggested the trafficking of G1P3 from TGN into endolysosomes. Both G1P3 and RAB5 were known to confer apoptosis resistance through mitochondrial stabilization. Therefore, the effects of G1P3 on the localization of RAB5 in mitochondria were tested. Compared to vector control, the co-occurrence of RAB5 with the mitochondria was increased by 1.5-fold in MCF-7/G1P3-FLAG expressing cells (p ≤ .005). Taken together, our results demonstrate a role for G1P3 to promote the association of RAB5+ endosomes with mitochondria and provide insight into yet another mechanism of G1P3-induced cancer cell survival.
Collapse
Affiliation(s)
- Anne M Davenport
- Department of Biological and Environmental Sciences, Texas A&M University-Commerce, Commerce, Texas, USA
- Department of Biology, Texas Woman's University, Denton, Texas, USA
| | - Madeleine Morris
- Department of Biological and Environmental Sciences, Texas A&M University-Commerce, Commerce, Texas, USA
| | - Fatima Sabti
- Department of Biological and Environmental Sciences, Texas A&M University-Commerce, Commerce, Texas, USA
| | - Sarah Sabti
- Department of Biological and Environmental Sciences, Texas A&M University-Commerce, Commerce, Texas, USA
| | - Diksha Shakya
- Department of Biological and Environmental Sciences, Texas A&M University-Commerce, Commerce, Texas, USA
| | - DiAnna L Hynds
- Department of Biology, Texas Woman's University, Denton, Texas, USA
| | - Venugopalan Cheriyath
- Department of Biological and Environmental Sciences, Texas A&M University-Commerce, Commerce, Texas, USA
| |
Collapse
|
45
|
Hao S, Huang H, Ma RY, Zeng X, Duan CY. Multifaceted functions of Drp1 in hypoxia/ischemia-induced mitochondrial quality imbalance: from regulatory mechanism to targeted therapeutic strategy. Mil Med Res 2023; 10:46. [PMID: 37833768 PMCID: PMC10571487 DOI: 10.1186/s40779-023-00482-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2023] [Accepted: 09/21/2023] [Indexed: 10/15/2023] Open
Abstract
Hypoxic-ischemic injury is a common pathological dysfunction in clinical settings. Mitochondria are sensitive organelles that are readily damaged following ischemia and hypoxia. Dynamin-related protein 1 (Drp1) regulates mitochondrial quality and cellular functions via its oligomeric changes and multiple modifications, which plays a role in mediating the induction of multiple organ damage during hypoxic-ischemic injury. However, there is active controversy and gaps in knowledge regarding the modification, protein interaction, and functions of Drp1, which both hinder and promote development of Drp1 as a novel therapeutic target. Here, we summarize recent findings on the oligomeric changes, modification types, and protein interactions of Drp1 in various hypoxic-ischemic diseases, as well as the Drp1-mediated regulation of mitochondrial quality and cell functions following ischemia and hypoxia. Additionally, potential clinical translation prospects for targeting Drp1 are discussed. This review provides new ideas and targets for proactive interventions on multiple organ damage induced by various hypoxic-ischemic diseases.
Collapse
Affiliation(s)
- Shuai Hao
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China
- Research Institute of General Surgery, Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210002 China
| | - He Huang
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China
| | - Rui-Yan Ma
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China
- Department of Cardiovascular Surgery, Xinqiao Hospital, Army Medical University, Chongqing, 400037 China
| | - Xue Zeng
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China
- Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, 400010 China
| | - Chen-Yang Duan
- Department of Anesthesiology, the Second Affiliated Hospital of Chongqing Medical University, Chongqing, 400010 China
| |
Collapse
|
46
|
Chen Y, Li P, Chen X, Yan R, Zhang Y, Wang M, Qin X, Li S, Zheng C, You F, Li T, Liu Y. Endoplasmic reticulum-mitochondrial calcium transport contributes to soft extracellular matrix-triggered mitochondrial dynamics and mitophagy in breast carcinoma cells. Acta Biomater 2023; 169:192-208. [PMID: 37541606 DOI: 10.1016/j.actbio.2023.07.060] [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: 04/13/2023] [Revised: 07/03/2023] [Accepted: 07/28/2023] [Indexed: 08/06/2023]
Abstract
Although mitochondrial morphology and function are considered to be closely related to matrix stiffness-driven tumor progression, it remains poorly understood how extracellular matrix (ECM) stiffness affects mitochondrial dynamics and mitophagy. Here, we found that soft substrate triggered calcium transport by increasing endoplasmic reticulum (ER) calcium release and mitochondrial (MITO) calcium uptake. ER-MITO calcium transport promoted the recruitment of dynamin-related protein 1 (Drp1) to mitochondria and phosphorylation at the serine 616 site, which induced mitochondrial fragmentation and Parkin/PINK1-mediated mitophagy. Furthermore, in vivo experiments demonstrated that soft ECM enhanced calcium levels in tumor tissue, Drp1 activity was required for soft ECM-induced mitochondrial dynamics impairment, and inhibition of Drp1 activity enhanced soft ECM-induced tumor necrosis. In conclusion, we revealed a new mechanism whereby ER-MITO calcium transport regulated mitochondrial dynamics and mitophagy through Drp1 translocation in response to soft substrates. These findings provide valuable insights into ECM stiffness as a potential target for antitumor therapy. STATEMENT OF SIGNIFICANCE: Here, we examined the relationship between substrate stiffness and mitochondrial dynamics by using polyacrylamide (PAA) substrates to simulate the stages of breast cancer or BAPN to reduce tumor tissue stiffness. The results elucidated that soft substrate triggered the recruitment of DRP1 and subsequent mitochondrial fission and mitophagy by ER-MITO calcium transport. Furthermore, mitophagy partly attenuated soft ECM-mediated tumor tissue necrosis and contributed to tumor survival in vivo. Our discoveries revealed the molecular mechanisms by which mechanical stimulation regulates mitochondrial dynamics, providing valuable insights into ECM stiffness as a target for anti-tumor approaches, which could be beneficial for both biomechanics research and clinical applications.
Collapse
Affiliation(s)
- Yu Chen
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Ping Li
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Xiangyan Chen
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Ran Yan
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Yixi Zhang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Meng Wang
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Xiang Qin
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Shun Li
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China
| | - Chuan Zheng
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan, PR China
| | - Fengming You
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan, PR China
| | - Tingting Li
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China.
| | - Yiyao Liu
- Department of Pharmacy, Personalized Drug Therapy Key Laboratory of Sichuan Province, Sichuan Provincial People's Hospital, and School of Life Science and Technology, University of Electronic Science and Technology of China, Chengdu, PR China; TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, No. 39 Shi-er-qiao Road, Chengdu 610072, Sichuan, PR China.
| |
Collapse
|
47
|
Huang D, Chen S, Xiong D, Wang H, Zhu L, Wei Y, Li Y, Zou S. Mitochondrial Dynamics: Working with the Cytoskeleton and Intracellular Organelles to Mediate Mechanotransduction. Aging Dis 2023; 14:1511-1532. [PMID: 37196113 PMCID: PMC10529762 DOI: 10.14336/ad.2023.0201] [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: 10/21/2022] [Accepted: 02/01/2023] [Indexed: 05/19/2023] Open
Abstract
Cells are constantly exposed to various mechanical environments; therefore, it is important that they are able to sense and adapt to changes. It is known that the cytoskeleton plays a critical role in mediating and generating extra- and intracellular forces and that mitochondrial dynamics are crucial for maintaining energy homeostasis. Nevertheless, the mechanisms by which cells integrate mechanosensing, mechanotransduction, and metabolic reprogramming remain poorly understood. In this review, we first discuss the interaction between mitochondrial dynamics and cytoskeletal components, followed by the annotation of membranous organelles intimately related to mitochondrial dynamic events. Finally, we discuss the evidence supporting the participation of mitochondria in mechanotransduction and corresponding alterations in cellular energy conditions. Notable advances in bioenergetics and biomechanics suggest that the mechanotransduction system composed of mitochondria, the cytoskeletal system, and membranous organelles is regulated through mitochondrial dynamics, which may be a promising target for further investigation and precision therapies.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Yuyu Li
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| | - Shujuan Zou
- State Key Laboratory of Oral Diseases, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, China
| |
Collapse
|
48
|
Oleinik N, Albayram O, Kassir MF, Atilgan FC, Walton C, Karakaya E, Kurtz J, Alekseyenko A, Alsudani H, Sheridan M, Szulc ZM, Ogretmen B. Alterations of lipid-mediated mitophagy result in aging-dependent sensorimotor defects. Aging Cell 2023; 22:e13954. [PMID: 37614052 PMCID: PMC10577547 DOI: 10.1111/acel.13954] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Revised: 07/20/2023] [Accepted: 07/24/2023] [Indexed: 08/25/2023] Open
Abstract
The metabolic consequences of mitophagy alterations due to age-related stress in healthy aging brains versus neurodegeneration remain unknown. Here, we demonstrate that ceramide synthase 1 (CerS1) is transported to the outer mitochondrial membrane by the p17/PERMIT transporter that recognizes mislocalized mitochondrial ribosomes (mitoribosomes) via 39-FLRN-42 residues, inducing ceramide-mediated mitophagy. P17/PERMIT-CerS1-mediated mitophagy attenuated the argininosuccinate/fumarate/malate axis and induced d-glucose and fructose accumulation in neurons in culture and brain tissues (primarily in the cerebellum) of wild-type mice in vivo. These metabolic changes in response to sodium-selenite were nullified in the cerebellum of CerS1to/to (catalytically inactive for C18-ceramide production CerS1 mutant), PARKIN-/- or p17/PERMIT-/- mice that have dysfunctional mitophagy. Whereas sodium selenite induced mitophagy in the cerebellum and improved motor-neuron deficits in aged wild-type mice, exogenous fumarate or malate prevented mitophagy. Attenuating ceramide-mediated mitophagy enhanced damaged mitochondria accumulation and age-dependent sensorimotor abnormalities in p17/PERMIT-/- mice. Reinstituting mitophagy using a ceramide analog drug with selenium conjugate, LCL768, restored mitophagy and reduced malate/fumarate metabolism, improving sensorimotor deficits in old p17/PERMIT-/- mice. Thus, these data describe the metabolic consequences of alterations to p17/PERMIT/ceramide-mediated mitophagy associated with the loss of mitochondrial quality control in neurons and provide therapeutic options to overcome age-dependent sensorimotor deficits and related disorders like amyotrophic lateral sclerosis (ALS).
Collapse
Affiliation(s)
- Natalia Oleinik
- Department of Biochemistry and Molecular BiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
- Hollings Cancer CenterMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Onder Albayram
- Departments of Pathology and Laboratory MedicineMedical University of South CarolinaCharlestonSouth CarolinaUSA
- Department of NeuroscienceMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Mohamed Faisal Kassir
- Department of Biochemistry and Molecular BiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
- Hollings Cancer CenterMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - F. Cansu Atilgan
- Department of Biochemistry and Molecular BiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
- Hollings Cancer CenterMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Chase Walton
- Department of Biochemistry and Molecular BiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
- Hollings Cancer CenterMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Eda Karakaya
- Departments of Pathology and Laboratory MedicineMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - John Kurtz
- Departments of Pathology and Laboratory MedicineMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Alexander Alekseyenko
- Hollings Cancer CenterMedical University of South CarolinaCharlestonSouth CarolinaUSA
- Public HealthMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Habeeb Alsudani
- Cancer CenterCold Spring Harbor LaboratoryCold Spring HarborNew YorkUSA
| | - Megan Sheridan
- Department of Biochemistry and Molecular BiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
- Hollings Cancer CenterMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Zdzislaw M. Szulc
- Department of Biochemistry and Molecular BiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
- Hollings Cancer CenterMedical University of South CarolinaCharlestonSouth CarolinaUSA
| | - Besim Ogretmen
- Department of Biochemistry and Molecular BiologyMedical University of South CarolinaCharlestonSouth CarolinaUSA
- Hollings Cancer CenterMedical University of South CarolinaCharlestonSouth CarolinaUSA
| |
Collapse
|
49
|
Hu D, Tan M, Lu D, Kleiboeker B, Liu X, Park H, Kravitz AV, Shoghi KI, Tseng YH, Razani B, Ikeda A, Lodhi IJ. TMEM135 links peroxisomes to the regulation of brown fat mitochondrial fission and energy homeostasis. Nat Commun 2023; 14:6099. [PMID: 37773161 PMCID: PMC10541902 DOI: 10.1038/s41467-023-41849-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Accepted: 09/20/2023] [Indexed: 10/01/2023] Open
Abstract
Mitochondrial morphology, which is controlled by mitochondrial fission and fusion, is an important regulator of the thermogenic capacity of brown adipocytes. Adipose-specific peroxisome deficiency impairs thermogenesis by inhibiting cold-induced mitochondrial fission due to decreased mitochondrial membrane content of the peroxisome-derived lipids called plasmalogens. Here, we identify TMEM135 as a critical mediator of the peroxisomal regulation of mitochondrial fission and thermogenesis. Adipose-specific TMEM135 knockout in mice blocks mitochondrial fission, impairs thermogenesis, and increases diet-induced obesity and insulin resistance. Conversely, TMEM135 overexpression promotes mitochondrial division, counteracts obesity and insulin resistance, and rescues thermogenesis in peroxisome-deficient mice. Mechanistically, thermogenic stimuli promote association between peroxisomes and mitochondria and plasmalogen-dependent localization of TMEM135 in mitochondria, where it mediates PKA-dependent phosphorylation and mitochondrial retention of the fission factor Drp1. Together, these results reveal a previously unrecognized inter-organelle communication regulating mitochondrial fission and energy homeostasis and identify TMEM135 as a potential target for therapeutic activation of BAT.
Collapse
Affiliation(s)
- Donghua Hu
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Min Tan
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Dongliang Lu
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Brian Kleiboeker
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Xuejing Liu
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Hongsuk Park
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Alexxai V Kravitz
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO, USA
| | - Kooresh I Shoghi
- Mallinckrodt Institute of Radiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Yu-Hua Tseng
- Section on Integrative Physiology and Metabolism, Research Division, Joslin Diabetes Center, Harvard Medical School, Boston, MA, USA
| | - Babak Razani
- Cardiovascular Division, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
- University of Pittsburgh School of Medicine and UPMC, Pittsburgh, PA, USA
| | - Akihiro Ikeda
- Department of Medical Genetics, University of Wisconsin-Madison, Madison, WI, USA
| | - Irfan J Lodhi
- Division of Endocrinology, Metabolism & Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
50
|
Boutry M, DiGiovanni LF, Demers N, Fountain A, Mamand S, Botelho RJ, Kim PK. Arf1-PI4KIIIβ positive vesicles regulate PI(3)P signaling to facilitate lysosomal tubule fission. J Cell Biol 2023; 222:e202205128. [PMID: 37289133 PMCID: PMC10250553 DOI: 10.1083/jcb.202205128] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 04/03/2023] [Accepted: 05/22/2023] [Indexed: 06/09/2023] Open
Abstract
Formation and fission of tubules from autolysosomes, endolysosomes, or phagolysosomes are required for lysosome reformation. However, the mechanisms governing these processes in these different lysosomal organelles are poorly understood. Thus, the role of phosphatidylinositol-4-phosphate (PI(4)P) is unclear as it was shown to promote the formation of tubules from phagolysosomes but was proposed to inhibit tubule formation on autolysosomes because the loss of PI4KIIIβ causes extensive lysosomal tubulation. Using super-resolution live-cell imaging, we show that Arf1-PI4KIIIβ positive vesicles are recruited to tubule fission sites from autolysosomes, endolysosomes, and phagolysosomes. Moreover, we show that PI(4)P is required to form autolysosomal tubules and that increased lysosomal tubulation caused by loss of PI4KIIIβ represents impaired tubule fission. At the site of fission, we propose that Arf1-PI4KIIIβ positive vesicles mediate a PI(3)P signal on lysosomes in a process requiring the lipid transfer protein SEC14L2. Our findings indicate that Arf1-PI4KIIIβ positive vesicles and their regulation of PI(3)P are critical components of the lysosomal tubule fission machinery.
Collapse
Affiliation(s)
- Maxime Boutry
- Cell Biology Program, Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, Ontario, Canada
| | - Laura F. DiGiovanni
- Cell Biology Program, Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Nicholas Demers
- Cell Biology Program, Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Aaron Fountain
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
- Graduate Program in Molecular Science, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Sami Mamand
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
- Graduate Program in Molecular Science, Toronto Metropolitan University, Toronto, Ontario, Canada
- Polytechnic Research Center, Erbil Polytechnic University, Erbil, Kurdistan, Iraq
| | - Roberto J. Botelho
- Department of Chemistry and Biology, Toronto Metropolitan University, Toronto, Ontario, Canada
- Graduate Program in Molecular Science, Toronto Metropolitan University, Toronto, Ontario, Canada
| | - Peter K. Kim
- Cell Biology Program, Hospital for Sick Children, Peter Gilgan Centre for Research and Learning, Toronto, Ontario, Canada
- Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
- Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, South Korea
| |
Collapse
|