1
|
Balasco N, Modjtahedi N, Monti A, Ruvo M, Vitagliano L, Doti N. CHCHD4 Oxidoreductase Activity: A Comprehensive Analysis of the Molecular, Functional, and Structural Properties of Its Redox-Regulated Substrates. Molecules 2025; 30:2117. [PMID: 40430290 PMCID: PMC12114033 DOI: 10.3390/molecules30102117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/24/2025] [Accepted: 05/06/2025] [Indexed: 05/29/2025] Open
Abstract
The human CHCHD4 protein, which is a prototypical family member, carries a coiled-coil-helix-coiled-coil-helix motif that is stabilized by two disulfide bonds. Using its CPC sequence motif, CHCHD4 plays a key role in mitochondrial metabolism, cell survival, and response to stress conditions, controlling the mitochondrial import of diversified protein substrates that are specifically recognized through an interplay between covalent and non-covalent interactions. In the present review, we provide an updated and comprehensive analysis of CHCHD4 substrates controlled by its redox activities. A particular emphasis has been placed on the molecular and structural aspects of these partnerships. The literature survey has been integrated with the mining of structural databases reporting either experimental structures (Protein Data Bank) or structures predicted by AlphaFold, which provide protein three-dimensional models using machine learning-based approaches. In providing an updated view of the thirty-four CHCHD4 substrates that have been experimentally validated, our analyses highlight the notion that this protein can operate on a variety of structurally diversified substrates. Although in most cases, CHCHD4 plays a crucial role in the formation of disulfide bridges that stabilize helix-coil-helix motifs of its substrates, significant variations on this common theme are observed, especially for substrates that have been more recently identified.
Collapse
Affiliation(s)
- Nicole Balasco
- Institute of Molecular Biology and Pathology, National Research Council (CNR), Department of Chemistry, University of Rome Sapienza, Piazzale Aldo Moro 5, 00185 Rome, Italy;
| | - Nazanine Modjtahedi
- Unité Physiopathologie et Génétique du Neurone et du Muscle, UMR CNRS 5261, Inserm U1315, Université Claude Bernard Lyon 1, 69008 Lyon, France;
| | - Alessandra Monti
- Institute of Biostructures and Bioimaging, National Research Council (CNR), Via P. Castellino 111, 80131 Naples, Italy; (A.M.); (M.R.)
| | - Menotti Ruvo
- Institute of Biostructures and Bioimaging, National Research Council (CNR), Via P. Castellino 111, 80131 Naples, Italy; (A.M.); (M.R.)
| | - Luigi Vitagliano
- Institute of Biostructures and Bioimaging, National Research Council (CNR), Via P. Castellino 111, 80131 Naples, Italy; (A.M.); (M.R.)
| | - Nunzianna Doti
- Institute of Biostructures and Bioimaging, National Research Council (CNR), Via P. Castellino 111, 80131 Naples, Italy; (A.M.); (M.R.)
| |
Collapse
|
2
|
Ge A, Zhou H, Yang X, Zhao C, Xin C, Song Z. The Antifungal Effects of Equol Against Candida albicans Involve Mitochondrial Dysfunction. J Fungi (Basel) 2025; 11:339. [PMID: 40422673 DOI: 10.3390/jof11050339] [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: 03/17/2025] [Revised: 04/25/2025] [Accepted: 04/25/2025] [Indexed: 05/28/2025] Open
Abstract
Novel antifungal agents are urgently needed because of the increasing number of drug-resistant Candida strains encountered in clinical practice and the limited variety of available antifungal drugs. Equol, a metabolite of soy isoflavone glycosides, exhibits antifungal activities. In this study, Equol had good inhibitory activity against Candida species. The lowest inhibitory concentration of 125-500 μg/mL was confirmed by the gradient dilution method. In addition, transmission electron microscopy and the relative content assay showed that Equol altered the cell wall and membrane of Candida albicans. Further studies found that Equol treatment increased the intracellular levels of reactive oxygen species and Ca2+. Subsequent experiments suggested that Equol treatment depolarized the membrane potential of C. albicans and up-regulated the expression of the apoptosis-inducing factor gene. These results confirmed that Equol damaged the cell wall and membrane, dysregulated the intracellular components, induced oxidative stress and Ca2+ accumulation, and ultimately resulted in mitochondrial dysfunction. Collectively, these findings demonstrated that Equol is a potential antifungal agent.
Collapse
Affiliation(s)
- Anni Ge
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Hao Zhou
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Xi Yang
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Chunling Zhao
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Caiyan Xin
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
| | - Zhangyong Song
- School of Basic Medical Sciences, Southwest Medical University, Luzhou 646000, China
- The Public Platform of Molecular Biotechnology, Public Center of Experimental Technology, Southwest Medical University, Luzhou 646000, China
- Hemodynamics and Medical Engineering Combination Key Laboratory of Luzhou, Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
3
|
Reinhardt T, El Harraoui Y, Rothemann A, Jauch AT, Müller‐Deubert S, Köllen MF, Risch T, Jacobs LJHC, Müller R, Traube FR, Docheva D, Zahler S, Riemer J, Bach NC, Sieber SA. Chemical Proteomics Reveals Human Off-Targets of Fluoroquinolone Induced Mitochondrial Toxicity. Angew Chem Int Ed Engl 2025; 64:e202421424. [PMID: 39964703 PMCID: PMC12036814 DOI: 10.1002/anie.202421424] [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: 11/04/2024] [Accepted: 02/03/2025] [Indexed: 04/03/2025]
Abstract
Fluoroquinolones (FQs) are an important class of potent broad-spectrum antibiotics. However, their general use is more and more limited by adverse side effects. While general mechanisms for the fluoroquinolone-associated disability (FQAD) have been identified, the underlying molecular targets of toxicity remain elusive. In this study, focusing on the most commonly prescribed FQs Ciprofloxacin and Levofloxacin, whole proteome analyses revealed prominent mitochondrial dysfunction in human cells, specifically of the complexes I and IV of the electron transport chain (ETC). Furthermore, global untargeted chemo-proteomic methodologies such as photo-affinity profiling with FQ-derived probes, as well as derivatization-free thermal proteome profiling, were applied to elucidate human protein off-targets of FQs in living cells. Accordingly, the interactions of FQs with mitochondrial AIFM1 and IDH2 have been identified and biochemically validated for their contribution to mitochondrial dysfunction. Of note, the FQ induced ETC dysfunction via AIFM1 activates the reverse carboxylation pathway of IDH2 for rescue, however, its simultaneous inhibition further enhances mitochondrial toxicity. This off-target discovery study provides unique insights into FQ toxicity enabling the utilization of identified molecular principles for the design of a safer FQ generation.
Collapse
Affiliation(s)
- Till Reinhardt
- Center for Functional Protein AssembliesDepartment of BioscienceTUM School of Natural SciencesTechnische Universität MünchenErnst-Otto-Fischer-Straße 885748Garching, Deutschland.
| | - Yassmine El Harraoui
- Center for Functional Protein AssembliesDepartment of BioscienceTUM School of Natural SciencesTechnische Universität MünchenErnst-Otto-Fischer-Straße 885748Garching, Deutschland.
| | - Alex Rothemann
- Institute for Biochemistry and CECADUniversity of CologneKöln, Deutschland.
| | - Adrian T. Jauch
- Department of PharmacyPharmaceutical BiologyLudwig-Maximilians-Universität MünchenButenandtstraße 5–1381377München, Deutschland.
| | - Sigrid Müller‐Deubert
- Department of Musculoskeletal Tissue RegenerationOrthopaedic Hospital König-Ludwig-HausUniversity of Würzburg97076Würzburg, Deutschland.
| | - Martin F. Köllen
- Center for Functional Protein AssembliesDepartment of BioscienceTUM School of Natural SciencesTechnische Universität MünchenErnst-Otto-Fischer-Straße 885748Garching, Deutschland.
| | - Timo Risch
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) and Saarland University Department of PharmacyCampus Building E8.166123Saarbrücken, Deutschland.
| | - Lianne JHC Jacobs
- Institute for Biochemistry and CECADUniversity of CologneKöln, Deutschland.
| | - Rolf Müller
- Helmholtz Institute for Pharmaceutical Research Saarland (HIPS)Helmholtz Centre for Infection Research (HZI) and Saarland University Department of PharmacyCampus Building E8.166123Saarbrücken, Deutschland.
| | - Franziska R. Traube
- Institut für Biochemie und Technische BiochemieUniversität Stuttgart70569Stuttgart.
| | - Denitsa Docheva
- Department of Musculoskeletal Tissue RegenerationOrthopaedic Hospital König-Ludwig-HausUniversity of Würzburg97076Würzburg, Deutschland.
| | - Stefan Zahler
- Department of PharmacyPharmaceutical BiologyLudwig-Maximilians-Universität MünchenButenandtstraße 5–1381377München, Deutschland.
| | - Jan Riemer
- Institute for Biochemistry and CECADUniversity of CologneKöln, Deutschland.
| | - Nina C. Bach
- Center for Functional Protein AssembliesDepartment of BioscienceTUM School of Natural SciencesTechnische Universität MünchenErnst-Otto-Fischer-Straße 885748Garching, Deutschland.
| | - Stephan A. Sieber
- Center for Functional Protein AssembliesDepartment of BioscienceTUM School of Natural SciencesTechnische Universität MünchenErnst-Otto-Fischer-Straße 885748Garching, Deutschland.
| |
Collapse
|
4
|
Wang L, Lin Y, Lin Z, Wu Q, Zhong G, Chen L. Causal relationship between mitochondrial proteins and risks of aortic aneurysms and aortic dissection: a Mendelian randomization study. J Cardiothorac Surg 2025; 20:181. [PMID: 40186305 PMCID: PMC11971758 DOI: 10.1186/s13019-025-03389-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2024] [Accepted: 03/09/2025] [Indexed: 04/07/2025] Open
Abstract
BACKGROUND Mitochondrial dysfunction may be linked to the development of aortic aneurysm (AA) and aortic dissection (AD). This study aimed to evaluate the potential associations between proteins related to mitochondrial function and the risks of AA/AD using Mendelian randomization (MR). METHODS Large-scale publicly available genome-wide association studies (GWAS) and FinnGen summary data were utilized for MR analysis. The causal relationship between mitochondrial proteins and AA/AD was assessed using inverse-variance weighted (IVW) as the primary method. Sensitivity analyses were conducted to detect heterogeneity and pleiotropy by Cochran's Q test, MR-Egger test, MR-PRESSO global test, and "leave-one-out" analysis. RESULTS There were potential causal relationships between several mitochondrial proteins and AA/AD. Specifically, the iron-sulfur cluster assembly enzyme ISCU (OR = 1.165, 95% CI: 1.051-1.291, P = 0.004) and NFU1 iron-sulfur cluster scaffold homolog (OR = 1.184, 95% CI: 1.056-1.329, P = 0.004) were identified as potential risk factors for AA; whereas the 39 S ribosomal protein L14 (OR = 0.868, 95% CI: 0.764-0.987, P = 0.031) was found to be a protective factor for AA. Furthermore, 39 S ribosomal protein L33 (OR = 1.134, 95% CI: 1.010-1.274, P = 0.033) and cytochrome C oxidase subunit 5B (OR = 1.330, 95% CI: 1.037-1.706, P = 0.025) were associated with increased risks of AD; whereas the 39 S ribosomal protein L52 (OR = 0.736, 95% CI: 0.550-0.984, P = 0.038) and mitochondrial ubiquitin ligase activator of NFKB 1 (OR = 0.806, 95% CI: 0.656-0.989, P = 0.039) were identified as potential protective factors for AD. Sensitivity analysis confirmed the stability of the results. CONCLUSIONS This study identified potential genetic associations between mitochondrial proteins and AA/AD. Targeting these mitochondrial proteins may help prevent the occurrence of AA/AD.
Collapse
Affiliation(s)
- Lei Wang
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, Fujian province, China
- Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fujian Province University, Fuzhou, 350000, Fujian province, China
| | - Yuzuo Lin
- Union College of Clinical Medicine, Fujian Medical University Union Hospital, Fuzhou, 350000, Fujian province, China
| | - Ziyan Lin
- Union College of Clinical Medicine, Fujian Medical University Union Hospital, Fuzhou, 350000, Fujian province, China
| | - Qingtong Wu
- Union College of Clinical Medicine, Fujian Medical University Union Hospital, Fuzhou, 350000, Fujian province, China
| | - Guodong Zhong
- Department of Pathology, Fujian Province Second People's Hospital, The Second Affiliated Hospital of Fujian University of Traditional Chinese Medicine, Fuzhou, 350000, Fujian province, China.
| | - Liangwan Chen
- Department of Cardiovascular Surgery, Fujian Medical University Union Hospital, Fuzhou, 350000, Fujian province, China.
- Key Laboratory of Cardio-Thoracic Surgery, Fujian Medical University, Fujian Province University, Fuzhou, 350000, Fujian province, China.
- Engineering Research Center of Tissue and Organ Regeneration, Fujian Province University, Fuzhou, 350000, Fujian province, China.
| |
Collapse
|
5
|
Brosey CA, Shen R, Tainer JA. NADH-bound AIF activates the mitochondrial CHCHD4/MIA40 chaperone by a substrate-mimicry mechanism. EMBO J 2025; 44:1220-1248. [PMID: 39806100 PMCID: PMC11832770 DOI: 10.1038/s44318-024-00360-6] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 12/19/2024] [Accepted: 12/20/2024] [Indexed: 01/16/2025] Open
Abstract
Mitochondrial metabolism requires the chaperoned import of disulfide-stabilized proteins via CHCHD4/MIA40 and its enigmatic interaction with oxidoreductase Apoptosis-inducing factor (AIF). By crystallizing human CHCHD4's AIF-interaction domain with an activated AIF dimer, we uncover how NADH allosterically configures AIF to anchor CHCHD4's β-hairpin and histidine-helix motifs to the inner mitochondrial membrane. The structure further reveals a similarity between the AIF-interaction domain and recognition sequences of CHCHD4 substrates. NMR and X-ray scattering (SAXS) solution measurements, mutational analyses, and biochemistry show that the substrate-mimicking AIF-interaction domain shields CHCHD4's redox-sensitive active site. Disrupting this shield critically activates CHCHD4 substrate affinity and chaperone activity. Regulatory-domain sequestration by NADH-activated AIF directly stimulates chaperone binding and folding, revealing how AIF mediates CHCHD4 mitochondrial import. These results establish AIF as an integral component of the metazoan disulfide relay and point to NADH-activated dimeric AIF as an organizational import center for CHCHD4 and its substrates. Importantly, AIF regulation of CHCHD4 directly links AIF's cellular NAD(H) sensing to CHCHD4 chaperone function, suggesting a mechanism to balance tissue-specific oxidative phosphorylation (OXPHOS) capacity with NADH availability.
Collapse
Affiliation(s)
- Chris A Brosey
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - Runze Shen
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA
| | - John A Tainer
- Department of Molecular and Cellular Oncology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA.
- Department of Cancer Biology, The University of Texas M. D. Anderson Cancer Center, Houston, TX, 77030, USA.
- MBIB Division, Lawrence Berkeley National Laboratory, Berkeley, CA, 94720, USA.
| |
Collapse
|
6
|
Cwerman-Thibault H, Malko-Baverel V, Le Guilloux G, Torres-Cuevas I, Ratcliffe E, Mouri D, Mignon V, Saubaméa B, Boespflug-Tanguy O, Gressens P, Corral-Debrinski M. Harlequin mice exhibit cognitive impairment, severe loss of Purkinje cells and a compromised bioenergetic status due to the absence of Apoptosis Inducing Factor. Biochim Biophys Acta Mol Basis Dis 2024; 1870:167272. [PMID: 38897257 DOI: 10.1016/j.bbadis.2024.167272] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Revised: 05/15/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024]
Abstract
The functional integrity of the central nervous system relies on complex mechanisms in which the mitochondria are crucial actors because of their involvement in a multitude of bioenergetics and biosynthetic pathways. Mitochondrial diseases are among the most prevalent groups of inherited neurological disorders, affecting up to 1 in 5000 adults and despite considerable efforts around the world there is still limited curative treatments. Harlequin mice correspond to a relevant model of recessive X-linked mitochondrial disease due to a proviral insertion in the first intron of the Apoptosis-inducing factor gene, resulting in an almost complete depletion of the corresponding protein. These mice exhibit progressive degeneration of the retina, optic nerve, cerebellum, and cortical regions leading to irremediable blindness and ataxia, reminiscent of what is observed in patients suffering from mitochondrial diseases. We evaluated the progression of cerebellar degeneration in Harlequin mice, especially for Purkinje cells and its relationship with bioenergetics failure and behavioral damage. For the first time to our knowledge, we demonstrated that Harlequin mice display cognitive and emotional impairments at early stage of the disease with further deteriorations as ataxia aggravates. These functions, corresponding to higher-order cognitive processing, have been assigned to a complex network of reciprocal connections between the cerebellum and many cortical areas which could be dysfunctional in these mice. Consequently, Harlequin mice become a suitable experimental model to test innovative therapeutics, via the targeting of mitochondria which can become available to a large spectrum of neurological diseases.
Collapse
Affiliation(s)
| | | | | | - Isabel Torres-Cuevas
- Université Paris Cité, NeuroDiderot, Inserm, F-75019 Paris, France; Department of Physiology, University of Valencia, Vicent Andrés Estellés s/n, 46100 12 Burjassot, Spain
| | - Edward Ratcliffe
- Université Paris Cité, NeuroDiderot, Inserm, F-75019 Paris, France
| | - Djmila Mouri
- Université Paris Cité, NeuroDiderot, Inserm, F-75019 Paris, France
| | - Virginie Mignon
- Université de Paris, UMR-S 1144 Inserm, 75006 Paris, France; Université Paris Cité, Platform of Cellular and Molecular Imaging, US25 Inserm, UAR3612 CNRS, 75006 Paris, France
| | - Bruno Saubaméa
- Université de Paris, UMR-S 1144 Inserm, 75006 Paris, France
| | - Odile Boespflug-Tanguy
- Université Paris Cité, NeuroDiderot, Inserm, F-75019 Paris, France; Service de Neurologie et Maladies métaboliques, CHU Paris - Hôpital Robert Debré, F-75019 Paris, France
| | - Pierre Gressens
- Université Paris Cité, NeuroDiderot, Inserm, F-75019 Paris, France
| | | |
Collapse
|
7
|
Cwerman-Thibault H, Malko-Baverel V, Le Guilloux G, Ratcliffe E, Mouri D, Torres-Cuevas I, Millán I, Saubaméa B, Mignon V, Boespflug-Tanguy O, Gressens P, Corral-Debrinski M. Neuroglobin overexpression in cerebellar neurons of Harlequin mice improves mitochondrial homeostasis and reduces ataxic behavior. Mol Ther 2024; 32:2150-2175. [PMID: 38796706 PMCID: PMC11286817 DOI: 10.1016/j.ymthe.2024.05.030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Revised: 03/22/2024] [Accepted: 05/23/2024] [Indexed: 05/28/2024] Open
Abstract
Neuroglobin, a member of the globin superfamily, is abundant in the brain, retina, and cerebellum of mammals and localizes to mitochondria. The protein exhibits neuroprotective capacities by participating in electron transfer, oxygen supply, and protecting against oxidative stress. Our objective was to determine whether neuroglobin overexpression can be used to treat neurological disorders. We chose Harlequin mice, which harbor a retroviral insertion in the first intron of the apoptosis-inducing factor gene resulting in the depletion of the corresponding protein essential for mitochondrial biogenesis. Consequently, Harlequin mice display degeneration of the cerebellum and suffer from progressive blindness and ataxia. Cerebellar ataxia begins in Harlequin mice at the age of 4 months and is characterized by neuronal cell disappearance, bioenergetics failure, and motor and cognitive impairments, which aggravated with aging. Mice aged 2 months received adeno-associated viral vectors harboring the coding sequence of neuroglobin or apoptosis-inducing factor in both cerebellar hemispheres. Six months later, Harlequin mice exhibited substantial improvements in motor and cognitive skills; probably linked to the preservation of respiratory chain function, Purkinje cell numbers and connectivity. Thus, without sharing functional properties with apoptosis-inducing factor, neuroglobin was efficient in reducing ataxia in Harlequin mice.
Collapse
Affiliation(s)
- Hélène Cwerman-Thibault
- Université Paris Cité, Inserm, Maladies neurodéveloppementales et neurovasculaires, F-75019 Paris, France
| | - Vassilissa Malko-Baverel
- Université Paris Cité, Inserm, Maladies neurodéveloppementales et neurovasculaires, F-75019 Paris, France
| | - Gwendoline Le Guilloux
- Université Paris Cité, Inserm, Maladies neurodéveloppementales et neurovasculaires, F-75019 Paris, France
| | - Edward Ratcliffe
- Université Paris Cité, Inserm, Maladies neurodéveloppementales et neurovasculaires, F-75019 Paris, France
| | - Djmila Mouri
- Université Paris Cité, Inserm, Maladies neurodéveloppementales et neurovasculaires, F-75019 Paris, France
| | - Isabel Torres-Cuevas
- Université Paris Cité, Inserm, Maladies neurodéveloppementales et neurovasculaires, F-75019 Paris, France; Neonatal Research Group, Health Research Institute La Fe, 46026 Valencia, Spain
| | - Ivan Millán
- Université Paris Cité, Inserm, Maladies neurodéveloppementales et neurovasculaires, F-75019 Paris, France; Neonatal Research Group, Health Research Institute La Fe, 46026 Valencia, Spain; Laboratory of Comparative Neurobiology, Cavanilles Institute of Biodiversity and Evolutionary Biology, University of Valencia, Valencia, Spain
| | - Bruno Saubaméa
- Université Paris Cité, Platform of Cellular and Molecular Imaging (PICMO), US25 Inserm, UAR3612 CNRS, 75006 Paris, France; Université Paris Cité, Optimisation Thérapeutique en Neuropsychopharmacologie, UMR-S 1144 Inserm, 75006 Paris, France
| | - Virginie Mignon
- Université Paris Cité, Platform of Cellular and Molecular Imaging (PICMO), US25 Inserm, UAR3612 CNRS, 75006 Paris, France
| | - Odile Boespflug-Tanguy
- Université Paris Cité, Inserm, Maladies neurodéveloppementales et neurovasculaires, F-75019 Paris, France; Service de Neurologie et Maladies métaboliques, CHU Paris - Hôpital Robert Debré, F-75019 Paris, France
| | - Pierre Gressens
- Université Paris Cité, Inserm, Maladies neurodéveloppementales et neurovasculaires, F-75019 Paris, France
| | - Marisol Corral-Debrinski
- Université Paris Cité, Inserm, Maladies neurodéveloppementales et neurovasculaires, F-75019 Paris, France.
| |
Collapse
|
8
|
Huang D, Chen L, Ji Q, Xiang Y, Zhou Q, Chen K, Zhang X, Zou F, Zhang X, Zhao Z, Wang T, Zheng G, Meng X. Lead aggravates Alzheimer's disease pathology via mitochondrial copper accumulation regulated by COX17. Redox Biol 2024; 69:102990. [PMID: 38091880 PMCID: PMC10716782 DOI: 10.1016/j.redox.2023.102990] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 11/03/2023] [Accepted: 12/06/2023] [Indexed: 01/23/2024] Open
Abstract
Alzheimer's disease (AD) is a common neurodegenerative disease that is associated with multiple environmental risk factors, including heavy metals. Lead (Pb) is a heavy metal contaminant, which is closely related to the incidence of AD. However, the research on the role of microglia in Pb-induced AD-like pathology is limited. To determine the mechanism by which Pb exposure aggravates AD progression and the role of microglial activation, we exposed APP/PS1 mice and Aβ1-42-treated BV-2 cells to Pb. Our results suggested that chronic Pb exposure exacerbated learning and memory impairments in APP/PS1 mice. Pb exposure increased the activation of microglia in the hippocampus of APP/PS1 mice, which was associated with increased deposition of Aβ1-42, and induced hippocampal neuron damage. Pb exposure upregulated copper transporter 1 (CTR1) and downregulated copper P-type ATPase transporter (ATP7A) in the hippocampus of APP/PS1 mice and Aβ1-42-treated BV-2 cells. Moreover, Pb enhanced mitochondrial translocation of the mitochondrial copper transporter COX17, leading to an increase in mitochondrial copper concentration and mitochondrial damage. This could be reversed by copper-chelating agents or by inhibiting the mitochondrial translocation of COX17. The increased mitochondrial copper concentration caused by increased mitochondrial translocation of COX17 after Pb exposure may be related to the enhanced mitochondrial import pathway of AIF/CHCHD4. These results indicate that Pb induces the activation of microglia by increasing the concentration of copper in the mitochondria of microglia, and microglia release inflammatory factors to promote neuroinflammation, thus aggravating the pathology of AD. The present study provides new ideas for the prevention of Pb-induced AD.
Collapse
Affiliation(s)
- Dingbang Huang
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Lixuan Chen
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Qiuyi Ji
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Yang Xiang
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Qin Zhou
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Kaiju Chen
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Xiaoshun Zhang
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Fei Zou
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China
| | - Xingmei Zhang
- Department of Neurobiology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, 510515, China
| | - Zaihua Zhao
- Department of Occupational and Environmental Health and the Ministry of Education's Key Laboratory of Hazard Assessment and Control in Special Operational Environment, School of Military Preventive Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Tao Wang
- Department of Occupational and Environmental Health and the Ministry of Education's Key Laboratory of Hazard Assessment and Control in Special Operational Environment, School of Military Preventive Medicine, Fourth Military Medical University, Xi'an, 710032, China
| | - Gang Zheng
- Department of Occupational and Environmental Health and the Ministry of Education's Key Laboratory of Hazard Assessment and Control in Special Operational Environment, School of Military Preventive Medicine, Fourth Military Medical University, Xi'an, 710032, China.
| | - Xiaojing Meng
- Department of Occupational Health and Occupational Medicine, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, China.
| |
Collapse
|
9
|
González-Arzola K, Díaz-Quintana A. Mitochondrial Factors in the Cell Nucleus. Int J Mol Sci 2023; 24:13656. [PMID: 37686461 PMCID: PMC10563088 DOI: 10.3390/ijms241713656] [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: 07/18/2023] [Revised: 08/31/2023] [Accepted: 08/31/2023] [Indexed: 09/10/2023] Open
Abstract
The origin of eukaryotic organisms involved the integration of mitochondria into the ancestor cell, with a massive gene transfer from the original proteobacterium to the host nucleus. Thus, mitochondrial performance relies on a mosaic of nuclear gene products from a variety of genomes. The concerted regulation of their synthesis is necessary for metabolic housekeeping and stress response. This governance involves crosstalk between mitochondrial, cytoplasmic, and nuclear factors. While anterograde and retrograde regulation preserve mitochondrial homeostasis, the mitochondria can modulate a wide set of nuclear genes in response to an extensive variety of conditions, whose response mechanisms often merge. In this review, we summarise how mitochondrial metabolites and proteins-encoded either in the nucleus or in the organelle-target the cell nucleus and exert different actions modulating gene expression and the chromatin state, or even causing DNA fragmentation in response to common stress conditions, such as hypoxia, oxidative stress, unfolded protein stress, and DNA damage.
Collapse
Affiliation(s)
- Katiuska González-Arzola
- Centro Andaluz de Biología Molecular y Medicina Regenerativa—CABIMER, Consejo Superior de Investigaciones Científicas—Universidad de Sevilla—Universidad Pablo de Olavide, 41092 Seville, Spain
- Departamento de Bioquímica Vegetal y Biología Molecular, Universidad de Sevilla, 41012 Seville, Spain
| | - Antonio Díaz-Quintana
- Departamento de Bioquímica Vegetal y Biología Molecular, Universidad de Sevilla, 41012 Seville, Spain
- Instituto de Investigaciones Químicas—cicCartuja, Universidad de Sevilla—C.S.I.C, 41092 Seville, Spain
| |
Collapse
|
10
|
Peker E, Weiss K, Song J, Zarges C, Gerlich S, Boehm V, Trifunovic A, Langer T, Gehring NH, Becker T, Riemer J. A two-step mitochondrial import pathway couples the disulfide relay with matrix complex I biogenesis. J Cell Biol 2023; 222:e202210019. [PMID: 37159021 PMCID: PMC10174193 DOI: 10.1083/jcb.202210019] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2022] [Revised: 03/03/2023] [Accepted: 04/06/2023] [Indexed: 05/10/2023] Open
Abstract
Mitochondria critically rely on protein import and its tight regulation. Here, we found that the complex I assembly factor NDUFAF8 follows a two-step import pathway linking IMS and matrix import systems. A weak targeting sequence drives TIM23-dependent NDUFAF8 matrix import, and en route, allows exposure to the IMS disulfide relay, which oxidizes NDUFAF8. Import is closely surveyed by proteases: YME1L prevents accumulation of excess NDUFAF8 in the IMS, while CLPP degrades reduced NDUFAF8 in the matrix. Therefore, NDUFAF8 can only fulfil its function in complex I biogenesis if both oxidation in the IMS and subsequent matrix import work efficiently. We propose that the two-step import pathway for NDUFAF8 allows integration of the activity of matrix complex I biogenesis pathways with the activity of the mitochondrial disulfide relay system in the IMS. Such coordination might not be limited to NDUFAF8 as we identified further proteins that can follow such a two-step import pathway.
Collapse
Affiliation(s)
- Esra Peker
- Institute for Biochemistry, University of Cologne, Cologne, Germany
| | - Konstantin Weiss
- Institute for Biochemistry, University of Cologne, Cologne, Germany
| | - Jiyao Song
- Institute of Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Christine Zarges
- Institute for Biochemistry, University of Cologne, Cologne, Germany
| | - Sarah Gerlich
- Institute for Biochemistry, University of Cologne, Cologne, Germany
| | - Volker Boehm
- Institute for Genetics, University of Cologne, Cologne, Germany
| | - Aleksandra Trifunovic
- Institute for Mitochondrial Diseases and Aging, Medical Faculty, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Thomas Langer
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
- Department of Mitochondrial Proteostasis, Max Planck Institute for Biology of Ageing, Cologne, Germany
| | - Niels H. Gehring
- Institute for Genetics, University of Cologne, Cologne, Germany
- Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Thomas Becker
- Institute of Biochemistry and Molecular Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Jan Riemer
- Institute for Biochemistry, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| |
Collapse
|
11
|
Fernández de la Torre M, Fiuza-Luces C, Laine-Menéndez S, Delmiro A, Arenas J, Martín MÁ, Lucia A, Morán M. Pathophysiology of Cerebellar Degeneration in Mitochondrial Disorders: Insights from the Harlequin Mouse. Int J Mol Sci 2023; 24:10973. [PMID: 37446148 DOI: 10.3390/ijms241310973] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2023] [Revised: 06/22/2023] [Accepted: 06/28/2023] [Indexed: 07/15/2023] Open
Abstract
By means of a proteomic approach, we assessed the pathways involved in cerebellar neurodegeneration in a mouse model (Harlequin, Hq) of mitochondrial disorder. A differential proteomic profile study (iTRAQ) was performed in cerebellum homogenates of male Hq and wild-type (WT) mice 8 weeks after the onset of clear symptoms of ataxia in the Hq mice (aged 5.2 ± 0.2 and 5.3 ± 0.1 months for WT and Hq, respectively), followed by a biochemical validation of the most relevant changes. Additional groups of 2-, 3- and 6-month-old WT and Hq mice were analyzed to assess the disease progression on the proteins altered in the proteomic study. The proteomic analysis showed that beyond the expected deregulation of oxidative phosphorylation, the cerebellum of Hq mice showed a marked astroglial activation together with alterations in Ca2+ homeostasis and neurotransmission, with an up- and downregulation of GABAergic and glutamatergic neurotransmission, respectively, and the downregulation of cerebellar "long-term depression", a synaptic plasticity phenomenon that is a major player in the error-driven learning that occurs in the cerebellar cortex. Our study provides novel insights into the mechanisms associated with cerebellar degeneration in the Hq mouse model, including a complex deregulation of neuroinflammation, oxidative phosphorylation and glutamate, GABA and amino acids' metabolism.
Collapse
Affiliation(s)
- Miguel Fernández de la Torre
- Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital '12 de Octubre' ('imas12'), 28041 Madrid, Spain
| | - Carmen Fiuza-Luces
- Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital '12 de Octubre' ('imas12'), 28041 Madrid, Spain
| | - Sara Laine-Menéndez
- Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital '12 de Octubre' ('imas12'), 28041 Madrid, Spain
| | - Aitor Delmiro
- Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital '12 de Octubre' ('imas12'), 28041 Madrid, Spain
- Spanish Network for Biomedical Research in Rare Diseases (CIBERER), U723, 28029 Madrid, Spain
- Servicio de Bioquímica Clínica, Hospital Universitario "12 de Octubre", 28041 Madrid, Spain
| | - Joaquín Arenas
- Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital '12 de Octubre' ('imas12'), 28041 Madrid, Spain
- Spanish Network for Biomedical Research in Rare Diseases (CIBERER), U723, 28029 Madrid, Spain
| | - Miguel Ángel Martín
- Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital '12 de Octubre' ('imas12'), 28041 Madrid, Spain
- Spanish Network for Biomedical Research in Rare Diseases (CIBERER), U723, 28029 Madrid, Spain
- Servicio de Genética, Hospital Universitario "12 de Octubre", 28041 Madrid, Spain
| | - Alejandro Lucia
- Faculty of Sports Sciences, European University of Madrid, 28670 Madrid, Spain
- Spanish Network for Biomedical Research in Fragility and Healthy Aging (CIBERFES), 28029 Madrid, Spain
| | - María Morán
- Mitochondrial and Neuromuscular Diseases Laboratory, Instituto de Investigación Sanitaria Hospital '12 de Octubre' ('imas12'), 28041 Madrid, Spain
- Spanish Network for Biomedical Research in Rare Diseases (CIBERER), U723, 28029 Madrid, Spain
| |
Collapse
|
12
|
Qiu Y, Wang H, Fan M, Pan H, Guan J, Jiang Y, Jia Z, Wu K, Zhou H, Zhuang Q, Lei Z, Ding X, Cai H, Dong Y, Yan L, Lin A, Fu Y, Zhang D, Yan Q, Wang Q. Impaired AIF-CHCHD4 interaction and mitochondrial calcium overload contribute to auditory neuropathy spectrum disorder in patient-iPSC-derived neurons with AIFM1 variant. Cell Death Dis 2023; 14:375. [PMID: 37365177 PMCID: PMC10293272 DOI: 10.1038/s41419-023-05899-6] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Revised: 06/08/2023] [Accepted: 06/16/2023] [Indexed: 06/28/2023]
Abstract
Auditory neuropathy spectrum disorder (ANSD) is a hearing impairment caused by dysfunction of inner hair cells, ribbon synapses, spiral ganglion neurons and/or the auditory nerve itself. Approximately 1/7000 newborns have abnormal auditory nerve function, accounting for 10%-14% of cases of permanent hearing loss in children. Although we previously identified the AIFM1 c.1265 G > A variant to be associated with ANSD, the mechanism by which ANSD is associated with AIFM1 is poorly understood. We generated induced pluripotent stem cells (iPSCs) from peripheral blood mononuclear cells (PBMCs) via nucleofection with episomal plasmids. The patient-specific iPSCs were edited via CRISPR/Cas9 technology to generate gene-corrected isogenic iPSCs. These iPSCs were further differentiated into neurons via neural stem cells (NSCs). The pathogenic mechanism was explored in these neurons. In patient cells (PBMCs, iPSCs, and neurons), the AIFM1 c.1265 G > A variant caused a novel splicing variant (c.1267-1305del), resulting in AIF p.R422Q and p.423-435del proteins, which impaired AIF dimerization. Such impaired AIF dimerization then weakened the interaction between AIF and coiled-coil-helix-coiled-coil-helix domain-containing protein 4 (CHCHD4). On the one hand, the mitochondrial import of ETC complex subunits was inhibited, subsequently leading to an increased ADP/ATP ratio and elevated ROS levels. On the other hand, MICU1-MICU2 heterodimerization was impaired, leading to mCa2+ overload. Calpain was activated by mCa2+ and subsequently cleaved AIF for its translocation into the nucleus, ultimately resulting in caspase-independent apoptosis. Interestingly, correction of the AIFM1 variant significantly restored the structure and function of AIF, further improving the physiological state of patient-specific iPSC-derived neurons. This study demonstrates that the AIFM1 variant is one of the molecular bases of ANSD. Mitochondrial dysfunction, especially mCa2+ overload, plays a prominent role in ANSD associated with AIFM1. Our findings help elucidate the mechanism of ANSD and may lead to the provision of novel therapies.
Collapse
Affiliation(s)
- Yue Qiu
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Hongyang Wang
- Senior Department of Otolaryngology, Head and Neck Surgery, Chinese PLA Institute of Otolaryngology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Mingjie Fan
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- Department of Pediatrics, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China
| | - Huaye Pan
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Jing Guan
- Senior Department of Otolaryngology, Head and Neck Surgery, Chinese PLA Institute of Otolaryngology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Yangwei Jiang
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Zexiao Jia
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Kaiwen Wu
- Senior Department of Otolaryngology, Head and Neck Surgery, Chinese PLA Institute of Otolaryngology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Hui Zhou
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Qianqian Zhuang
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Zhaoying Lei
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Xue Ding
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Huajian Cai
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Yufei Dong
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Lei Yan
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Aifu Lin
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Yong Fu
- The Children's Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310052, China
| | - Dong Zhang
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| | - Qingfeng Yan
- College of Life Sciences, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
- Department of Pediatrics, The First Affiliated Hospital of Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310003, China.
- Key Laboratory for Cell and Gene Engineering of Zhejiang Province, Hangzhou, Zhejiang, 310058, China.
| | - Qiuju Wang
- Senior Department of Otolaryngology, Head and Neck Surgery, Chinese PLA Institute of Otolaryngology, Chinese PLA General Hospital, Beijing, 100853, China.
| |
Collapse
|
13
|
Shi JF, Liu Y, Wang Y, Gao R, Wang Y, Liu J. Targeting ferroptosis, a novel programmed cell death, for the potential of alcohol-related liver disease therapy. Front Pharmacol 2023; 14:1194343. [PMID: 37214434 PMCID: PMC10196366 DOI: 10.3389/fphar.2023.1194343] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Accepted: 04/27/2023] [Indexed: 05/24/2023] Open
Abstract
Ferroptosis is a new iron-dependent cell death mode, which is different from the other types of programmed cell death, such as apoptosis, necrosis, and autophagy. Ferroptosis is characterized by a process in which fatal lipids from lipid peroxidation accumulate in cells and eventually lead to cell death. Alcohol-related liver disease (ALD) is a type of liver injury caused by excessive alcohol intake. Alcohol-related liver disease is a broad-spectrum disease category, which includes fatty liver, steatohepatitis, hepatitis, cirrhosis, and hepatocellular tumors. Recent studies have found that ferroptosis is involved in the pathological development of non-viral liver diseases. Therefore, ferroptosis may be an ideal target for the treatment of non-viral liver diseases. In this review article, we will elaborate the molecular mechanism and regulatory mechanism of ferroptosis, explore the key role of ferroptosis in the Alcohol-related liver disease process, and summarize the existing targeted ferroptosis drugs and their feasibility for the treatment of Alcohol-related liver disease.
Collapse
Affiliation(s)
- Jing-Fen Shi
- Institute for Health Policy and Hospital Management, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
- Wenjiang District People’s Hospital of Chengdu, Chengdu, China
| | - Yu’e Liu
- Tongji University Cancer Center, Shanghai Tenth People’s Hospital of Tongji University, School of Medicine, Tongji University, Shanghai, China
| | - Yan Wang
- Wenjiang District People’s Hospital of Chengdu, Chengdu, China
| | - Ru Gao
- Wenjiang District People’s Hospital of Chengdu, Chengdu, China
| | - Yi Wang
- Department of Critical Care Medicine, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| | - Jun Liu
- Wenjiang District People’s Hospital of Chengdu, Chengdu, China
- Department of Ultrasound Medicine, Sichuan Academy of Medical Science and Sichuan Provincial People’s Hospital, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
14
|
Shoshan-Barmatz V, Arif T, Shteinfer-Kuzmine A. Apoptotic proteins with non-apoptotic activity: expression and function in cancer. Apoptosis 2023; 28:730-753. [PMID: 37014578 PMCID: PMC10071271 DOI: 10.1007/s10495-023-01835-3] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/17/2023] [Indexed: 04/05/2023]
Abstract
Apoptosis is a process of programmed cell death in which a cell commits suicide while maintaining the integrity and architecture of the tissue as a whole. Apoptosis involves activation of one of two major pathways: the extrinsic pathway, where extracellular pro-apoptotic signals, transduced through plasma membrane death receptors, activate a caspase cascade leading to apoptosis. The second, the intrinsic apoptotic pathway, where damaged DNA, oxidative stress, or chemicals, induce the release of pro-apoptotic proteins from the mitochondria, leading to the activation of caspase-dependent and independent apoptosis. However, it has recently become apparent that proteins involved in apoptosis also exhibit non-cell death-related physiological functions that are related to the cell cycle, differentiation, metabolism, inflammation or immunity. Such non-conventional activities were predominantly reported in non-cancer cells although, recently, such a dual function for pro-apoptotic proteins has also been reported in cancers where they are overexpressed. Interestingly, some apoptotic proteins translocate to the nucleus in order to perform a non-apoptotic function. In this review, we summarize the unconventional roles of the apoptotic proteins from a functional perspective, while focusing on two mitochondrial proteins: VDAC1 and SMAC/Diablo. Despite having pro-apoptotic functions, these proteins are overexpressed in cancers and this apparent paradox and the associated pathophysiological implications will be discussed. We will also present possible mechanisms underlying the switch from apoptotic to non-apoptotic activities although a deeper investigation into the process awaits further study.
Collapse
Affiliation(s)
- Varda Shoshan-Barmatz
- Department of Life Sciences, Ben-Gurion University of the Negev, 84105, Beer Sheva, Israel.
- National Institute for Biotechnology in the Negev, Beer Sheva, Israel.
| | - Tasleem Arif
- Department of Cell, Developmental, and Regenerative Biology, Icahn School of Medicine at Mount Sinai, New York, NY, 10029, USA
| | | |
Collapse
|
15
|
Mao J, Wang D, Wang D, Wu Q, Shang Q, Gao C, Wang H, Wang H, Du M, Peng P, Jia H, Xu X, Wang J, Yang L, Luo Z. SIRT5-related desuccinylation modification of AIFM1 protects against compression-induced intervertebral disc degeneration by regulating mitochondrial homeostasis. Exp Mol Med 2023; 55:253-268. [PMID: 36653443 PMCID: PMC9898264 DOI: 10.1038/s12276-023-00928-y] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2022] [Revised: 11/17/2022] [Accepted: 11/22/2022] [Indexed: 01/19/2023] Open
Abstract
Mitochondrial dysfunction plays a major role in the development of intervertebral disc degeneration (IDD). Sirtuin 5 (SIRT5) participates in the maintenance of mitochondrial homeostasis through its desuccinylase activity. However, it is still unclear whether succinylation or SIRT5 is involved in the impairment of mitochondria and development of IDD induced by excessive mechanical stress. Our 4D label-free quantitative proteomic results showed decreased expression of the desuccinylase SIRT5 in rat nucleus pulposus (NP) tissues under mechanical loading. Overexpression of Sirt5 effectively alleviated, whereas knockdown of Sirt5 aggravated, the apoptosis and dysfunction of NP cells under mechanical stress, consistent with the more severe IDD phenotype of Sirt5 KO mice than wild-type mice that underwent lumbar spine instability (LSI) surgery. Moreover, immunoprecipitation-coupled mass spectrometry (IP-MS) results suggested that AIFM1 was a downstream target of SIRT5, which was verified by a Co-IP assay. We further demonstrated that reduced SIRT5 expression resulted in the increased succinylation of AIFM1, which in turn abolished the interaction between AIFM1 and CHCHD4 and thus led to the reduced electron transfer chain (ETC) complex subunits in NP cells. Reduced ETC complex subunits resulted in mitochondrial dysfunction and the subsequent occurrence of IDD under mechanical stress. Finally, we validated the efficacy of treatments targeting disrupted mitochondrial protein importation by upregulating SIRT5 expression or methylene blue (MB) administration in the compression-induced rat IDD model. In conclusion, our study provides new insights into the occurrence and development of IDD and offers promising therapeutic approaches for IDD.
Collapse
Affiliation(s)
- Jianxin Mao
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Di Wang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Dong Wang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Qi Wu
- Intensive Care Unit, Heze Municipal Hospital, Heze, 274031, People's Republic of China
| | - Qiliang Shang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Chu Gao
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Huanbo Wang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Han Wang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Mu Du
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Pandi Peng
- Medical Research Institute, Northwestern Polytechnical University, Xi'an, 710068, People's Republic of China
| | - Haoruo Jia
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Xiaolong Xu
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Jie Wang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Liu Yang
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China.
- Medical Research Institute, Northwestern Polytechnical University, Xi'an, 710068, People's Republic of China.
| | - Zhuojing Luo
- Institute of Orthopedic Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China.
- Medical Research Institute, Northwestern Polytechnical University, Xi'an, 710068, People's Republic of China.
| |
Collapse
|
16
|
Zhou Y, Zhao Y, Ma W, Zhang L, Jiang Y, Dong W. USF1-CHCHD4 axis promotes lung adenocarcinoma progression partially via activating the MYC pathway. Discov Oncol 2022; 13:136. [PMID: 36482116 PMCID: PMC9732179 DOI: 10.1007/s12672-022-00600-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2022] [Accepted: 12/05/2022] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND This study aimed to identify genes related to lung adenocarcinoma (LUAD) and investigate the effects and molecular mechanisms of coiled-coil-helix-coiled-coil-helix domain containing 4 (CHCHD4) in the progression of LUAD. METHODS The GEPIA database was used to evaluate the differential expression of CHCHD4 and the survival data of LUAD patients compared to controls. TCGA-LUAD database, JASPAR website, and GSEA were used to analyse the relationship between CHCHD4 and the upstream stimulating factor 1 (USF1) or MYC pathways. The proliferation, apoptosis, migration, and invasion of LUAD cells were evaluated using cell counting kit-8, 5-ethynyl-2'-deoxyuridine, colony formation, flow cytometry, wound healing, and transwell assays. qRT-PCR, western blotting, and immunohistochemistry were used to detect the mRNA and protein expression, respectively. Furthermore, xenograft tumours from nude mice were used to verify the effect of CHCHD4 on LUAD in vivo. RESULTS CHCHD4 overexpression was found in LUAD tumor tissues and cells, and high CHCHD4 was associated with a poor prognosis. Interestingly, CHCHD4 knockdown suppressed the malignant phenotype of the LUAD cells. Moreover, we found that USF1 upregulated CHCHD4 and promoted LUAD progression. CHCHD4 knockdown also inhibited the progression of LUAD. In addition, CHCHD4 knockdown suppressed xenograft tumour growth. CONCLUSION USF1-CHCHD4 axis can promote LUAD progress, which may be through activating MYC pathway.
Collapse
Affiliation(s)
- Yuhui Zhou
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, 250021, People's Republic of China
| | - Yunxia Zhao
- Department of Neurology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, People's Republic of China
| | - Wei Ma
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, 250021, People's Republic of China
| | - Lin Zhang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, 250021, People's Republic of China
| | - Yuanzhu Jiang
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, 250021, People's Republic of China
| | - Wei Dong
- Department of Thoracic Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 324 Jingwu Road, Jinan, 250021, People's Republic of China.
| |
Collapse
|
17
|
Nedara K, Reinhardt C, Lebraud E, Arena G, Gracia C, Buard V, Pioche-Durieu C, Castelli F, Colsch B, Bénit P, Rustin P, Albaud B, Gestraud P, Baulande S, Servant N, Deutsch E, Verbavatz JM, Brenner C, Milliat F, Modjtahedi N. Relevance of the TRIAP1/p53 axis in colon cancer cell proliferation and adaptation to glutamine deprivation. Front Oncol 2022; 12:958155. [DOI: 10.3389/fonc.2022.958155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 10/10/2022] [Indexed: 11/13/2022] Open
Abstract
Human TRIAP1 (TP53-regulated inhibitor of apoptosis 1; also known as p53CSV for p53-inducible cell survival factor) is the homolog of yeast Mdm35, a well-known chaperone that interacts with the Ups/PRELI family proteins and participates in the intramitochondrial transfer of lipids for the synthesis of cardiolipin (CL) and phosphatidylethanolamine. Although recent reports indicate that TRIAP1 is a prosurvival factor abnormally overexpressed in various types of cancer, knowledge about its molecular and metabolic function in human cells is still elusive. It is therefore critical to understand the metabolic and proliferative advantages that TRIAP1 expression provides to cancer cells. Here, in a colorectal cancer cell model, we report that the expression of TRIAP1 supports cancer cell proliferation and tumorigenesis. Depletion of TRIAP1 perturbed the mitochondrial ultrastructure, without a major impact on CL levels and mitochondrial activity. TRIAP1 depletion caused extramitochondrial perturbations resulting in changes in the endoplasmic reticulum-dependent lipid homeostasis and induction of a p53-mediated stress response. Furthermore, we observed that TRIAP1 depletion conferred a robust p53-mediated resistance to the metabolic stress caused by glutamine deprivation. These findings highlight the importance of TRIAP1 in tumorigenesis and indicate that the loss of TRIAP1 has extramitochondrial consequences that could impact on the metabolic plasticity of cancer cells and their response to conditions of nutrient deprivation.
Collapse
|
18
|
Dong Y, Zhang Y, Feng Y, An W. The protective roles of augmenter of liver regeneration in hepatocytes in the non-alcoholic fatty liver disease. Front Pharmacol 2022; 13:928606. [PMID: 36304168 PMCID: PMC9592723 DOI: 10.3389/fphar.2022.928606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2022] [Accepted: 09/20/2022] [Indexed: 11/23/2022] Open
Abstract
Non-alcoholic fatty liver disease (NAFLD) occurs in 25% of the global population and manifests as lipid deposition, hepatocyte injury, activation of Kupffer and stellate cells, and steatohepatitis. Predominantly expressed in hepatocytes, the augmenter of liver regeneration (ALR) is a key factor in liver regulation that can alleviate fatty liver disease and protect the liver from abnormal liver lipid metabolism. ALR has three isoforms (15-, 21-, and 23-kDa), amongst which 23-kDa ALR is the most extensively studied. The 23-kDa ALR isoform is a sulfhydryl oxidase that resides primarily in the mitochondrial intermembrane space (IMS), whereby it protects the liver against various types of injury. In this review, we describe the role of ALR in regulating hepatocytes in the context of NAFLD. We also discuss questions about ALR that remain to be explored in the future. In conclusion, ALR appears to be a promising therapeutic target for treating NAFLD.
Collapse
Affiliation(s)
- Yuan Dong
- Department of Science and Technology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yuejie Zhang
- Department of Science and Technology, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yingmei Feng
- Department of Science and Technology, Beijing Youan Hospital, Capital Medical University, Beijing, China
- *Correspondence: Yingmei Feng, ; Wei An,
| | - Wei An
- Department of Cell Biology, Capital Medical University and the Municipal Key Laboratory for Liver Protection and Regulation of Regeneration, Beijing, China
- *Correspondence: Yingmei Feng, ; Wei An,
| |
Collapse
|
19
|
Wischhof L, Scifo E, Ehninger D, Bano D. AIFM1 beyond cell death: An overview of this OXPHOS-inducing factor in mitochondrial diseases. EBioMedicine 2022; 83:104231. [PMID: 35994922 PMCID: PMC9420475 DOI: 10.1016/j.ebiom.2022.104231] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2022] [Revised: 08/01/2022] [Accepted: 08/02/2022] [Indexed: 11/17/2022] Open
Abstract
Apoptosis-inducing factor (AIF) is a mitochondrial intermembrane space flavoprotein with diverse functions in cellular physiology. In this regard, a large number of studies have elucidated AIF's participation to chromatin condensation during cell death in development, cancer, cardiovascular and brain disorders. However, the discovery of rare AIFM1 mutations in patients has shifted the interest of biomedical researchers towards AIF's contribution to pathogenic mechanisms underlying inherited AIFM1-linked metabolic diseases. The functional characterization of AIF binding partners has rapidly advanced our understanding of AIF biology within the mitochondria and beyond its widely reported role in cell death. At the present time, it is reasonable to assume that AIF contributes to cell survival by promoting biogenesis and maintenance of the mitochondrial oxidative phosphorylation (OXPHOS) system. With this review, we aim to outline the current knowledge around the vital role of AIF by primarily focusing on currently reported human diseases that have been linked to AIFM1 deficiency.
Collapse
Affiliation(s)
- Lena Wischhof
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Enzo Scifo
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Dan Ehninger
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Daniele Bano
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
| |
Collapse
|
20
|
Chiappetta G, Gamberi T, Faienza F, Limaj X, Rizza S, Messori L, Filomeni G, Modesti A, Vinh J. Redox proteome analysis of auranofin exposed ovarian cancer cells (A2780). Redox Biol 2022; 52:102294. [PMID: 35358852 PMCID: PMC8966199 DOI: 10.1016/j.redox.2022.102294] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2022] [Accepted: 03/16/2022] [Indexed: 01/03/2023] Open
Abstract
The effects of Auranofin (AF) on protein expression and protein oxidation in A2780 cancer cells were investigated through a strategy based on simultaneous expression proteomics and redox proteomics determinations. Bioinformatics analysis of the proteomics data supports the view that the most critical cellular changes elicited by AF treatment consist of thioredoxin reductase inhibition, alteration of the cell redox state, impairment of the mitochondrial functions, metabolic changes associated with conversion to a glycolytic phenotype, induction of ER stress. The occurrence of the above cellular changes was extensively validated by performing direct biochemical assays. Our data are consistent with the concept that AF produces its effects through a multitarget mechanism that mainly affects the redox metabolism and the mitochondrial functions and results into severe ER stress. Results are discussed in the context of the current mechanistic knowledge existing on AF. Redox proteomics allows to underline cell adaptation mechanisms in response to Auranofin treatment in ovarian cancer cells. BRCA1 is one of the major candidates of the ovarian cancer cell adaptation to Auranofin treatment. Auranofin alters the oxidative phosphorylation and mitochondrial protein import machinery. TRAP1 C501 modulates Auranofin toxicity. Auranofin induces severe stress of the endoplasmic reticulum.
Collapse
Affiliation(s)
- Giovanni Chiappetta
- Biological Mass Spectrometry and Proteomics Group, SMBP, PDC CNRS UMR, 8249, ESPCI Paris, Université PSL, 10 rue Vauquelin, 75005, Paris, France.
| | - Tania Gamberi
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134, Florence, Italy.
| | - Fiorella Faienza
- Department of Biology, University of Rome Tor Vergata, Rome, Italy
| | - Xhesika Limaj
- Biological Mass Spectrometry and Proteomics Group, SMBP, PDC CNRS UMR, 8249, ESPCI Paris, Université PSL, 10 rue Vauquelin, 75005, Paris, France
| | - Salvatore Rizza
- Redox Signaling and Oxidative Stress Group, Danish Cancer Society Research Center, Copenhagen, Denmark
| | - Luigi Messori
- Metmed Lab, Department of Chemistry, University of Florence, via della lastruccia 3, 50019, Sesto Fiorentino, Italy
| | - Giuseppe Filomeni
- Department of Biology, University of Rome Tor Vergata, Rome, Italy; Redox Signaling and Oxidative Stress Group, Danish Cancer Society Research Center, Copenhagen, Denmark; Center for Healthy Aging, University of Copenhagen, Denmark
| | - Alessandra Modesti
- Department of Experimental and Clinical Biomedical Sciences, University of Florence, Viale G.B. Morgagni 50, 50134, Florence, Italy
| | - Joelle Vinh
- Biological Mass Spectrometry and Proteomics Group, SMBP, PDC CNRS UMR, 8249, ESPCI Paris, Université PSL, 10 rue Vauquelin, 75005, Paris, France
| |
Collapse
|
21
|
Ma S, Adzavon YM, Wen X, Zhao P, Xie F, Liu M, Ma X. Novel Insights in the Regulatory Mechanisms of Ferroptosis in Hepatocellular Carcinoma. Front Cell Dev Biol 2022; 10:873029. [PMID: 35663406 PMCID: PMC9160826 DOI: 10.3389/fcell.2022.873029] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 05/05/2022] [Indexed: 12/12/2022] Open
Abstract
Ferroptosis is a newly defined programmed cell death, which by its mechanism differs from other programmed cell death processes such as apoptosis, necrosis, and autophagy. It has a unique morphology and biological properties that antioxidants and iron-chelating agents can regulate. Ferroptosis has the characteristics of iron ion deposition and dependence on lipid peroxidation. It can affect the progression of many cancers, including liver cancer, by inducing an intracellular iron-dependent accumulation of reactive oxygen species, providing new possibilities for cancer treatment. At present, great progress has been made in exploring the molecular mechanism of ferroptosis. In this review, we summarize the characteristics, mechanisms, and regulatory factors of ferroptosis in detail, discuss the progress of ferroptosis research in liver cancer, and provide directions and new ideas for the treatment of hepatocellular carcinoma.
Collapse
Affiliation(s)
- Shiwen Ma
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
- Beijing Molecular Hydrogen Research Center, Beijing, China
| | - Yao Mawulikplimi Adzavon
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
- Beijing Molecular Hydrogen Research Center, Beijing, China
- *Correspondence: Yao Mawulikplimi Adzavon,
| | - Xiaohu Wen
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
- Beijing Molecular Hydrogen Research Center, Beijing, China
| | - Pengxiang Zhao
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
- Beijing Molecular Hydrogen Research Center, Beijing, China
| | - Fei Xie
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
- Beijing Molecular Hydrogen Research Center, Beijing, China
| | - Mengyu Liu
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
- Beijing Molecular Hydrogen Research Center, Beijing, China
| | - Xuemei Ma
- Faculty of Environment and Life, Beijing University of Technology, Beijing, China
- Beijing Molecular Hydrogen Research Center, Beijing, China
| |
Collapse
|
22
|
Stairs CW, Táborský P, Salomaki ED, Kolisko M, Pánek T, Eme L, Hradilová M, Vlček Č, Jerlström-Hultqvist J, Roger AJ, Čepička I. Anaeramoebae are a divergent lineage of eukaryotes that shed light on the transition from anaerobic mitochondria to hydrogenosomes. Curr Biol 2021; 31:5605-5612.e5. [PMID: 34710348 DOI: 10.1016/j.cub.2021.10.010] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2021] [Revised: 07/07/2021] [Accepted: 10/05/2021] [Indexed: 01/02/2023]
Abstract
Discoveries of diverse microbial eukaryotes and their inclusion in comprehensive phylogenomic analyses have crucially re-shaped the eukaryotic tree of life in the 21st century.1 At the deepest level, eukaryotic diversity comprises 9-10 "supergroups." One of these supergroups, the Metamonada, is particularly important to our understanding of the evolutionary dynamics of eukaryotic cells, including the remodeling of mitochondrial function. All metamonads thrive in low-oxygen environments and lack classical aerobic mitochondria, instead possessing mitochondrion-related organelles (MROs) with metabolisms that are adapted to low-oxygen conditions. These MROs lack an organellar genome, do not participate in the Krebs cycle and oxidative phosphorylation,2 and often synthesize ATP by substrate-level phosphorylation coupled to hydrogen production.3,4 The events that occurred during the transition from an oxygen-respiring mitochondrion to a functionally streamlined MRO early in metamonad evolution remain largely unknown. Here, we report transcriptomes of two recently described, enigmatic, anaerobic protists from the genus Anaeramoeba.5 Using phylogenomic analysis, we show that these species represent a divergent, phylum-level lineage in the tree of metamonads, emerging as a sister group of the Parabasalia and reordering the deep branching order of the metamonad tree. Metabolic reconstructions of the Anaeramoeba MROs reveal many "classical" mitochondrial features previously not seen in metamonads, including a disulfide relay import system, propionate production, and amino acid metabolism. Our findings suggest that the cenancestor of Metamonada likely had MROs with more classical mitochondrial features than previously anticipated and demonstrate how discoveries of novel lineages of high taxonomic rank continue to transform our understanding of early eukaryote evolution.
Collapse
Affiliation(s)
- Courtney W Stairs
- Department of Biology, Lund University, Sölvegatan 35, 223 62 Lund, Sweden.
| | - Petr Táborský
- Department of Zoology, Faculty of Science, Charles University, Viničná 7, 128 44 Prague, Czech Republic
| | - Eric D Salomaki
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, 370 05 České Budějovice, Czech Republic
| | - Martin Kolisko
- Institute of Parasitology, Biology Centre, Czech Academy of Sciences, 370 05 České Budějovice, Czech Republic
| | - Tomáš Pánek
- Department of Zoology, Faculty of Science, Charles University, Viničná 7, 128 44 Prague, Czech Republic
| | - Laura Eme
- Université Paris-Saclay, CNRS, AgroParisTech, Ecologie Systématique Evolution, 91400 Orsay, France
| | - Miluše Hradilová
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Čestmír Vlček
- Institute of Molecular Genetics, Academy of Sciences of the Czech Republic, Vídeňská 1083, 142 20 Prague, Czech Republic
| | - Jon Jerlström-Hultqvist
- Department of Biochemistry and Molecular Biology, Dalhousie University, 5850 College St. Halifax, NS B3H 4R2, Canada
| | - Andrew J Roger
- Department of Biochemistry and Molecular Biology, Dalhousie University, 5850 College St. Halifax, NS B3H 4R2, Canada
| | - Ivan Čepička
- Department of Zoology, Faculty of Science, Charles University, Viničná 7, 128 44 Prague, Czech Republic.
| |
Collapse
|
23
|
Geldon S, Fernández-Vizarra E, Tokatlidis K. Redox-Mediated Regulation of Mitochondrial Biogenesis, Dynamics, and Respiratory Chain Assembly in Yeast and Human Cells. Front Cell Dev Biol 2021; 9:720656. [PMID: 34557489 PMCID: PMC8452992 DOI: 10.3389/fcell.2021.720656] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2021] [Accepted: 08/04/2021] [Indexed: 12/24/2022] Open
Abstract
Mitochondria are double-membrane organelles that contain their own genome, the mitochondrial DNA (mtDNA), and reminiscent of its endosymbiotic origin. Mitochondria are responsible for cellular respiration via the function of the electron oxidative phosphorylation system (OXPHOS), located in the mitochondrial inner membrane and composed of the four electron transport chain (ETC) enzymes (complexes I-IV), and the ATP synthase (complex V). Even though the mtDNA encodes essential OXPHOS components, the large majority of the structural subunits and additional biogenetical factors (more than seventy proteins) are encoded in the nucleus and translated in the cytoplasm. To incorporate these proteins and the rest of the mitochondrial proteome, mitochondria have evolved varied, and sophisticated import machineries that specifically target proteins to the different compartments defined by the two membranes. The intermembrane space (IMS) contains a high number of cysteine-rich proteins, which are mostly imported via the MIA40 oxidative folding system, dependent on the reduction, and oxidation of key Cys residues. Several of these proteins are structural components or assembly factors necessary for the correct maturation and function of the ETC complexes. Interestingly, many of these proteins are involved in the metalation of the active redox centers of complex IV, the terminal oxidase of the mitochondrial ETC. Due to their function in oxygen reduction, mitochondria are the main generators of reactive oxygen species (ROS), on both sides of the inner membrane, i.e., in the matrix and the IMS. ROS generation is important due to their role as signaling molecules, but an excessive production is detrimental due to unwanted oxidation reactions that impact on the function of different types of biomolecules contained in mitochondria. Therefore, the maintenance of the redox balance in the IMS is essential for mitochondrial function. In this review, we will discuss the role that redox regulation plays in the maintenance of IMS homeostasis as well as how mitochondrial ROS generation may be a key regulatory factor for ETC biogenesis, especially for complex IV.
Collapse
Affiliation(s)
| | - Erika Fernández-Vizarra
- Institute of Molecular Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Kostas Tokatlidis
- Institute of Molecular Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
24
|
Goyal S, Tiwari S, Seth B, Tandon A, Shankar J, Sinha M, Singh SJ, Priya S, Chaturvedi RK. Bisphenol-A inhibits mitochondrial biogenesis via impairment of GFER mediated mitochondrial protein import in the rat brain hippocampus. Neurotoxicology 2021; 85:18-32. [PMID: 33878312 DOI: 10.1016/j.neuro.2021.04.001] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 04/05/2021] [Accepted: 04/06/2021] [Indexed: 12/22/2022]
Abstract
Mitochondrial biogenesis relies on different protein import machinery, as mitochondrial proteins are imported from the cytosol. The mitochondrial intermembrane space assembly (MIA) pathway consists of GFER/ALR and CHCHD4/Mia40, responsible for importing proteins and their oxidative folding inside the mitochondria. The MIA pathway plays an essential role in complex IV (COX IV) biogenesis via importing copper chaperone COX17, associated with the respiratory chain. BPA, an environmental toxicant, found in consumable plastics, causes neurotoxicity via impairment in mitochondrial dynamics, neurogenesis, and cognitive functions. We studied the levels of key regulatory proteins of mitochondrial import pathways and mitochondrial biogenesis after BPA exposure in the rat hippocampus. BPA caused a significant reduction in the levels of mitochondrial biogenesis proteins (PGC1α, and TFAM) and mitochondrial import protein (GFER). Immunohistochemical analysis showed reduced co-localization of NeuN with GFER, PGC-1α, and TFAM suggesting impaired mitochondrial biogenesis and protein import. BPA exposure resulted in damaged mitochondria with distorted cristae in neurons and caused a significant reduction in GFER localization inside IMS as depicted by immunogold electron microscopy. The reduced levels of GFER resulted in defective COX17 import. The translocation of cytochrome c into the cytosol and increased cleaved caspase-3 levels triggered apoptosis due to BPA toxicity. Overall, our study implicates GFER as a potential target for impaired mitochondrial protein machinery, biogenesis, and apoptosis against BPA neurotoxicity in the rat hippocampus.
Collapse
Affiliation(s)
- Shweta Goyal
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Saurabh Tiwari
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Brashket Seth
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Ankit Tandon
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India; Department of Biochemistry, School of Dental Sciences, Babu Banarasi Das University, BBD City, Faizabad Road, Lucknow, U.P, 226 028, India
| | - Jai Shankar
- Advanced Imaging Facility, CSIR-IITR, Lucknow, India
| | - Meetali Sinha
- Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India; Computational Toxicology Facility, CSIR-IITR, Lucknow, India
| | - Sangh Jyoti Singh
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Smriti Priya
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India
| | - Rajnish Kumar Chaturvedi
- Developmental Toxicology Laboratory, Systems Toxicology and Health Risk Assessment Group, CSIR-Indian Institute of Toxicology Research (CSIR-IITR), Vishvigyan Bhavan, 31, Mahatma Gandhi Marg, Lucknow, Uttar Pradesh, 226001, India; Academy of Scientific and Innovative Research (AcSIR), Ghaziabad, 201002, India.
| |
Collapse
|
25
|
Kit O, Frantsiyants E, Neskubina I, Cheryarina N, Shikhlyarova A, Przhedetskiy Y, Pozdnyakova V, Surikova E, Kaplieva I, Bandovkina V. Influence of standard and stimulated growth of B16/F10 melanoma on AIF levels in mitochondria in cells of the heart and other somatic organs in female mice. CARDIOMETRY 2021. [DOI: 10.18137/cardiometry.2021.18.113120] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
studying AIF levels in mitochondria in cells of the heart and various organs in female mice with the growth of experimental melanoma B16 / F10 and comorbid pathology
Collapse
|
26
|
Needs HI, Protasoni M, Henley JM, Prudent J, Collinson I, Pereira GC. Interplay between Mitochondrial Protein Import and Respiratory Complexes Assembly in Neuronal Health and Degeneration. Life (Basel) 2021; 11:432. [PMID: 34064758 PMCID: PMC8151517 DOI: 10.3390/life11050432] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/27/2021] [Accepted: 05/02/2021] [Indexed: 12/14/2022] Open
Abstract
The fact that >99% of mitochondrial proteins are encoded by the nuclear genome and synthesised in the cytosol renders the process of mitochondrial protein import fundamental for normal organelle physiology. In addition to this, the nuclear genome comprises most of the proteins required for respiratory complex assembly and function. This means that without fully functional protein import, mitochondrial respiration will be defective, and the major cellular ATP source depleted. When mitochondrial protein import is impaired, a number of stress response pathways are activated in order to overcome the dysfunction and restore mitochondrial and cellular proteostasis. However, prolonged impaired mitochondrial protein import and subsequent defective respiratory chain function contributes to a number of diseases including primary mitochondrial diseases and neurodegeneration. This review focuses on how the processes of mitochondrial protein translocation and respiratory complex assembly and function are interlinked, how they are regulated, and their importance in health and disease.
Collapse
Affiliation(s)
- Hope I. Needs
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK; (H.I.N.); (J.M.H.)
| | - Margherita Protasoni
- Medical Research Council-Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK; (M.P.); (J.P.)
| | - Jeremy M. Henley
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK; (H.I.N.); (J.M.H.)
- Centre for Neuroscience and Regenerative Medicine, Faculty of Science, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Julien Prudent
- Medical Research Council-Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK; (M.P.); (J.P.)
| | - Ian Collinson
- School of Biochemistry, University of Bristol, Bristol BS8 1TD, UK; (H.I.N.); (J.M.H.)
| | - Gonçalo C. Pereira
- Medical Research Council-Mitochondrial Biology Unit, University of Cambridge, Cambridge CB2 0XY, UK; (M.P.); (J.P.)
| |
Collapse
|
27
|
Erv1 and Cytochrome c Mediate Rapid Electron Transfer via A Collision-Type Interaction. J Mol Biol 2021; 433:167045. [PMID: 33971209 DOI: 10.1016/j.jmb.2021.167045] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/01/2021] [Accepted: 04/29/2021] [Indexed: 12/11/2022]
Abstract
Being essential for oxidative protein folding in the mitochondrial intermembrane space, the mitochondrial disulfide relay relies on the electron transfer (ET) from the sulfhydryl oxidase Erv1 to cytochrome c (Cc). Using solution NMR spectroscopy, we demonstrate that while the yeast Cc-Erv1 system is functionally active, no observable binding of the protein partners takes place. The transient interaction between Erv1 and Cc can be rationalized by molecular modeling, suggesting that a large surface area of Erv1 can sustain a fast ET to Cc via a collision-type mechanism, without the need for a canonical protein complex formation. We suggest that, by preventing the direct ET to molecular oxygen (O2), the collision-type Cc-Erv1 interaction plays a role in protecting the organism against reactive oxygen species.
Collapse
|
28
|
Liu S, Zhou M, Ruan Z, Wang Y, Chang C, Sasaki M, Rajaram V, Lemoff A, Nambiar K, Wang JE, Hatanpaa KJ, Luo W, Dawson TM, Dawson VL, Wang Y. AIF3 splicing switch triggers neurodegeneration. Mol Neurodegener 2021; 16:25. [PMID: 33853653 PMCID: PMC8048367 DOI: 10.1186/s13024-021-00442-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2020] [Accepted: 03/12/2021] [Indexed: 11/18/2022] Open
Abstract
BACKGROUND Apoptosis-inducing factor (AIF), as a mitochondrial flavoprotein, plays a fundamental role in mitochondrial bioenergetics that is critical for cell survival and also mediates caspase-independent cell death once it is released from mitochondria and translocated to the nucleus under ischemic stroke or neurodegenerative diseases. Although alternative splicing regulation of AIF has been implicated, it remains unknown which AIF splicing isoform will be induced under pathological conditions and how it impacts mitochondrial functions and neurodegeneration in adult brain. METHODS AIF splicing induction in brain was determined by multiple approaches including 5' RACE, Sanger sequencing, splicing-specific PCR assay and bottom-up proteomic analysis. The role of AIF splicing in mitochondria and neurodegeneration was determined by its biochemical properties, cell death analysis, morphological and functional alterations and animal behavior. Three animal models, including loss-of-function harlequin model, gain-of-function AIF3 knockin model and conditional inducible AIF splicing model established using either Cre-loxp recombination or CRISPR/Cas9 techniques, were applied to explore underlying mechanisms of AIF splicing-induced neurodegeneration. RESULTS We identified a nature splicing AIF isoform lacking exons 2 and 3 named as AIF3. AIF3 was undetectable under physiological conditions but its expression was increased in mouse and human postmortem brain after stroke. AIF3 splicing in mouse brain caused enlarged ventricles and severe neurodegeneration in the forebrain regions. These AIF3 splicing mice died 2-4 months after birth. AIF3 splicing-triggered neurodegeneration involves both mitochondrial dysfunction and AIF3 nuclear translocation. We showed that AIF3 inhibited NADH oxidase activity, ATP production, oxygen consumption, and mitochondrial biogenesis. In addition, expression of AIF3 significantly increased chromatin condensation and nuclear shrinkage leading to neuronal cell death. However, loss-of-AIF alone in harlequin or gain-of-AIF3 alone in AIF3 knockin mice did not cause robust neurodegeneration as that observed in AIF3 splicing mice. CONCLUSIONS We identified AIF3 as a disease-inducible isoform and established AIF3 splicing mouse model. The molecular mechanism underlying AIF3 splicing-induced neurodegeneration involves mitochondrial dysfunction and AIF3 nuclear translocation resulting from the synergistic effect of loss-of-AIF and gain-of-AIF3. Our study provides a valuable tool to understand the role of AIF3 splicing in brain and a potential therapeutic target to prevent/delay the progress of neurodegenerative diseases.
Collapse
Affiliation(s)
- Shuiqiao Liu
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Mi Zhou
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Zhi Ruan
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Yanan Wang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Calvin Chang
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Masayuki Sasaki
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
- Departments of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Veena Rajaram
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Andrew Lemoff
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Kalyani Nambiar
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Jennifer E. Wang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Kimmo J. Hatanpaa
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Weibo Luo
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
| | - Ted M. Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
- Departments of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Valina L. Dawson
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
- Departments of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
- Solomon H. Snyder Department of Neuroscience, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
- Physiology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| | - Yingfei Wang
- Department of Pathology, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
- Department of Neurology, University of Texas Southwestern Medical Center, Dallas, TX 75390 USA
- Neuroregeneration and Stem Cell Programs, Institute for Cell Engineering, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
- Departments of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21205 USA
| |
Collapse
|
29
|
The Mia40/CHCHD4 Oxidative Folding System: Redox Regulation and Signaling in the Mitochondrial Intermembrane Space. Antioxidants (Basel) 2021; 10:antiox10040592. [PMID: 33921425 PMCID: PMC8069373 DOI: 10.3390/antiox10040592] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 04/06/2021] [Accepted: 04/09/2021] [Indexed: 12/20/2022] Open
Abstract
Mitochondria are critical for several cellular functions as they control metabolism, cell physiology, and cell death. The mitochondrial proteome consists of around 1500 proteins, the vast majority of which (about 99% of them) are encoded by nuclear genes, with only 13 polypeptides in human cells encoded by mitochondrial DNA. Therefore, it is critical for all the mitochondrial proteins that are nuclear-encoded to be targeted precisely and sorted specifically to their site of action inside mitochondria. These processes of targeting and sorting are catalysed by protein translocases that operate in each one of the mitochondrial sub-compartments. The main protein import pathway for the intermembrane space (IMS) recognises proteins that are cysteine-rich, and it is the only import pathway that chemically modifies the imported precursors by introducing disulphide bonds to them. In this manner, the precursors are trapped in the IMS in a folded state. The key component of this pathway is Mia40 (called CHCHD4 in human cells), which itself contains cysteine motifs and is subject to redox regulation. In this review, we detail the basic components of the MIA pathway and the disulphide relay mechanism that underpins the electron transfer reaction along the oxidative folding mechanism. Then, we discuss the key protein modulators of this pathway and how they are interlinked to the small redox-active molecules that critically affect the redox state in the IMS. We present also evidence that the mitochondrial redox processes that are linked to iron–sulfur clusters biogenesis and calcium homeostasis coalesce in the IMS at the MIA machinery. The fact that the MIA machinery and several of its interactors and substrates are linked to a variety of common human diseases connected to mitochondrial dysfunction highlight the potential of redox processes in the IMS as a promising new target for developing new treatments for some of the most complex and devastating human diseases.
Collapse
|
30
|
Jiang X, Stockwell BR, Conrad M. Ferroptosis: mechanisms, biology and role in disease. Nat Rev Mol Cell Biol 2021; 22:266-282. [PMID: 33495651 PMCID: PMC8142022 DOI: 10.1038/s41580-020-00324-8] [Citation(s) in RCA: 3975] [Impact Index Per Article: 993.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/14/2020] [Indexed: 02/06/2023]
Abstract
The research field of ferroptosis has seen exponential growth over the past few years, since the term was coined in 2012. This unique modality of cell death, driven by iron-dependent phospholipid peroxidation, is regulated by multiple cellular metabolic pathways, including redox homeostasis, iron handling, mitochondrial activity and metabolism of amino acids, lipids and sugars, in addition to various signalling pathways relevant to disease. Numerous organ injuries and degenerative pathologies are driven by ferroptosis. Intriguingly, therapy-resistant cancer cells, particularly those in the mesenchymal state and prone to metastasis, are exquisitely vulnerable to ferroptosis. As such, pharmacological modulation of ferroptosis, via both its induction and its inhibition, holds great potential for the treatment of drug-resistant cancers, ischaemic organ injuries and other degenerative diseases linked to extensive lipid peroxidation. In this Review, we provide a critical analysis of the current molecular mechanisms and regulatory networks of ferroptosis, the potential physiological functions of ferroptosis in tumour suppression and immune surveillance, and its pathological roles, together with a potential for therapeutic targeting. Importantly, as in all rapidly evolving research areas, challenges exist due to misconceptions and inappropriate experimental methods. This Review also aims to address these issues and to provide practical guidelines for enhancing reproducibility and reliability in studies of ferroptosis. Finally, we discuss important concepts and pressing questions that should be the focus of future ferroptosis research.
Collapse
Affiliation(s)
- Xuejun Jiang
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Brent R Stockwell
- Department of Biological Sciences, Columbia University, New York, NY, USA.
- Department of Chemistry, Columbia University, New York, NY, USA.
| | - Marcus Conrad
- Institute of Metabolism and Cell Death, Helmholtz Zentrum München, Neuherberg, Germany.
- Laboratory of Experimental Oncology, Pirogov Russian National Research Medical University, Moscow, Russia.
| |
Collapse
|
31
|
Edwards R, Eaglesfield R, Tokatlidis K. The mitochondrial intermembrane space: the most constricted mitochondrial sub-compartment with the largest variety of protein import pathways. Open Biol 2021; 11:210002. [PMID: 33715390 PMCID: PMC8061763 DOI: 10.1098/rsob.210002] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The mitochondrial intermembrane space (IMS) is the most constricted sub-mitochondrial compartment, housing only about 5% of the mitochondrial proteome, and yet is endowed with the largest variability of protein import mechanisms. In this review, we summarize our current knowledge of the major IMS import pathway based on the oxidative protein folding pathway and discuss the stunning variability of other IMS protein import pathways. As IMS-localized proteins only have to cross the outer mitochondrial membrane, they do not require energy sources like ATP hydrolysis in the mitochondrial matrix or the inner membrane electrochemical potential which are critical for import into the matrix or insertion into the inner membrane. We also explore several atypical IMS import pathways that are still not very well understood and are guided by poorly defined or completely unknown targeting peptides. Importantly, many of the IMS proteins are linked to several human diseases, and it is therefore crucial to understand how they reach their normal site of function in the IMS. In the final part of this review, we discuss current understanding of how such IMS protein underpin a large spectrum of human disorders.
Collapse
Affiliation(s)
- Ruairidh Edwards
- Institute of Molecular Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, University Avenue, Glasgow G12 8QQ, UK
| | - Ross Eaglesfield
- Institute of Molecular Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, University Avenue, Glasgow G12 8QQ, UK
| | - Kostas Tokatlidis
- Institute of Molecular Cell and Systems Biology, College of Medical, Veterinary and Life Sciences, University of Glasgow, University Avenue, Glasgow G12 8QQ, UK
| |
Collapse
|
32
|
CHCHD4 (MIA40) and the mitochondrial disulfide relay system. Biochem Soc Trans 2021; 49:17-27. [PMID: 33599699 PMCID: PMC7925007 DOI: 10.1042/bst20190232] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2020] [Revised: 12/19/2020] [Accepted: 01/06/2021] [Indexed: 12/13/2022]
Abstract
Mitochondria are pivotal for normal cellular physiology, as they perform a crucial role in diverse cellular functions and processes, including respiration and the regulation of bioenergetic and biosynthetic pathways, as well as regulating cellular signalling and transcriptional networks. In this way, mitochondria are central to the cell's homeostatic machinery, and as such mitochondrial dysfunction underlies the pathology of a diverse range of diseases including mitochondrial disease and cancer. Mitochondrial import pathways and targeting mechanisms provide the means to transport into mitochondria the hundreds of nuclear-encoded mitochondrial proteins that are critical for the organelle's many functions. One such import pathway is the highly evolutionarily conserved disulfide relay system (DRS) within the mitochondrial intermembrane space (IMS), whereby proteins undergo a form of oxidation-dependent protein import. A central component of the DRS is the oxidoreductase coiled-coil-helix-coiled-coil-helix (CHCH) domain-containing protein 4 (CHCHD4, also known as MIA40), the human homologue of yeast Mia40. Here, we summarise the recent advances made to our understanding of the role of CHCHD4 and the DRS in physiology and disease, with a specific focus on the emerging importance of CHCHD4 in regulating the cellular response to low oxygen (hypoxia) and metabolism in cancer.
Collapse
|
33
|
Arena G, Modjtahedi N, Kruger R. Exploring the contribution of the mitochondrial disulfide relay system to Parkinson's disease: the PINK1/CHCHD4 interplay. Neural Regen Res 2021; 16:2222-2224. [PMID: 33818502 PMCID: PMC8354137 DOI: 10.4103/1673-5374.310679] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Affiliation(s)
- Giuseppe Arena
- Translational Neuroscience group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg, Esch-sur-Alzette, Luxembourg
| | - Nazanine Modjtahedi
- Université Paris-Saclay, Gustave Roussy Institute, CNRS, Metabolic and systemic aspects of oncogenesis for new therapeutic approaches, Villejuif, France
| | - Rejko Kruger
- Translational Neuroscience group, Luxembourg Centre for Systems Biomedicine (LCSB), University of Luxembourg; Parkinson Research Clinic, Centre Hospitalier du Luxembourg (CHL); Transversal Translational Medicine, Luxembourg Institute of Health (LIH), Esch-sur-Alzette, Luxembourg
| |
Collapse
|
34
|
"Oncometabolism: The switchboard of cancer - An editorial". Biochim Biophys Acta Mol Basis Dis 2020; 1867:166031. [PMID: 33310398 DOI: 10.1016/j.bbadis.2020.166031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
35
|
Zhao T, Goedhart C, Pfeffer G, Greenway SC, Lines M, Khan A, Innes AM, Shutt TE. Skeletal Phenotypes Due to Abnormalities in Mitochondrial Protein Homeostasis and Import. Int J Mol Sci 2020; 21:8327. [PMID: 33171986 PMCID: PMC7664180 DOI: 10.3390/ijms21218327] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 10/28/2020] [Accepted: 11/03/2020] [Indexed: 12/19/2022] Open
Abstract
Mitochondrial disease represents a collection of rare genetic disorders caused by mitochondrial dysfunction. These disorders can be quite complex and heterogeneous, and it is recognized that mitochondrial disease can affect any tissue at any age. The reasons for this variability are not well understood. In this review, we develop and expand a subset of mitochondrial diseases including predominantly skeletal phenotypes. Understanding how impairment ofdiverse mitochondrial functions leads to a skeletal phenotype will help diagnose and treat patients with mitochondrial disease and provide additional insight into the growing list of human pathologies associated with mitochondrial dysfunction. The underlying disease genes encode factors involved in various aspects of mitochondrial protein homeostasis, including proteases and chaperones, mitochondrial protein import machinery, mediators of inner mitochondrial membrane lipid homeostasis, and aminoacylation of mitochondrial tRNAs required for translation. We further discuss a complex of frequently associated phenotypes (short stature, cataracts, and cardiomyopathy) potentially explained by alterations to steroidogenesis, a process regulated by mitochondria. Together, these observations provide novel insight into the consequences of impaired mitochondrial protein homeostasis.
Collapse
Affiliation(s)
- Tian Zhao
- Departments of Medical Genetics and Biochemistry & Molecular Biology, Cumming School of Medicine, Alberta Children’s Hospital Research Institute, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada;
| | - Caitlin Goedhart
- Departments of Pediatrics and Medical Genetics, Cumming School of Medicine, Alberta Children’s Hospital Research Institute, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada; (C.G.); (M.L.); (A.M.I)
| | - Gerald Pfeffer
- Departments of Clinical Neurosciences and Medical Genetics, Cumming School of Medicine, Hotchkiss Brain Institute, Alberta Child Health Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada;
| | - Steven C Greenway
- Departments of Pediatrics, Cardiac Sciences and Biochemistry & Molecular Biology, Cumming School of Medicine, Alberta Children’s Hospital Research Institute and Libin Cardiovascular Institute, University of Calgary, Calgary, AB T2N 4N1, Canada;
| | - Matthew Lines
- Departments of Pediatrics and Medical Genetics, Cumming School of Medicine, Alberta Children’s Hospital Research Institute, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada; (C.G.); (M.L.); (A.M.I)
| | - Aneal Khan
- Departments of Pediatrics and Medical Genetics, Cumming School of Medicine, Alberta Children’s Hospital Research Institute, University of Calgary, Calgary, AB T3B 6A8, Canada;
| | - A Micheil Innes
- Departments of Pediatrics and Medical Genetics, Cumming School of Medicine, Alberta Children’s Hospital Research Institute, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada; (C.G.); (M.L.); (A.M.I)
| | - Timothy E Shutt
- Departments of Medical Genetics and Biochemistry & Molecular Biology, Cumming School of Medicine, Alberta Children’s Hospital Research Institute, Hotchkiss Brain Institute, University of Calgary, Calgary, AB T2N 4N1, Canada;
| |
Collapse
|
36
|
Thiriveedi VR, Mattam U, Pattabhi P, Bisoyi V, Talari NK, Krishnamoorthy T, Sepuri NBV. Glutathionylated and Fe-S cluster containing hMIA40 (CHCHD4) regulates ROS and mitochondrial complex III and IV activities of the electron transport chain. Redox Biol 2020; 37:101725. [PMID: 32971361 PMCID: PMC7511737 DOI: 10.1016/j.redox.2020.101725] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2020] [Revised: 09/02/2020] [Accepted: 09/11/2020] [Indexed: 12/14/2022] Open
Abstract
Human MIA40, an intermembrane space (IMS) import receptor of mitochondria harbors twin CX9C motifs for stability while its CPC motif is known to facilitate the import of IMS bound proteins. Site-directed mutagenesis complemented by MALDI on in vivo hMIA40 protein shows that a portion of MIA40 undergoes reversible S-glutathionylation at three cysteines in the twin CX9C motifs and the lone cysteine 4 residue. We find that HEK293T cells expressing hMIA40 mutant defective for glutathionylation are compromised in the activities of complexes III and IV of the Electron Transport Chain (ETC) and enhance Reactive Oxygen Species (ROS) levels. Immunocapture studies show MIA40 interacting with complex III. Interestingly, glutathionylated MIA40 can transfer electrons to cytochrome C directly. However, Fe–S clusters associated with the CPC motif are essential to facilitate the two-electron to one-electron transfer for reducing cytochrome C. These results suggest that hMIA40 undergoes glutathionylation to maintain ROS levels and for optimum function of complexes III and IV of ETC. Our studies shed light on a novel post-translational modification of hMIA40 and its ability to act as a redox switch to regulate the ETC and cellular redox homeostasis.
Collapse
Affiliation(s)
| | - Ushodaya Mattam
- Department of Biochemistry, University of Hyderabad, Gachibowli, Hyderabad, TS, 500046, India
| | - Prasad Pattabhi
- Department of Biochemistry, University of Hyderabad, Gachibowli, Hyderabad, TS, 500046, India
| | - Vandana Bisoyi
- Department of Biochemistry, University of Hyderabad, Gachibowli, Hyderabad, TS, 500046, India
| | - Noble Kumar Talari
- Department of Biochemistry, University of Hyderabad, Gachibowli, Hyderabad, TS, 500046, India
| | - Thanuja Krishnamoorthy
- Vectrogen Biologicals Pvt.Ltd., BioNEST, School of Life Sciences, University of Hyderabad, Gachibowli, Hyderabad, TS, 500046, India
| | - Naresh Babu V Sepuri
- Department of Biochemistry, University of Hyderabad, Gachibowli, Hyderabad, TS, 500046, India.
| |
Collapse
|
37
|
Antonicka H, Lin ZY, Janer A, Aaltonen MJ, Weraarpachai W, Gingras AC, Shoubridge EA. A High-Density Human Mitochondrial Proximity Interaction Network. Cell Metab 2020; 32:479-497.e9. [PMID: 32877691 DOI: 10.1016/j.cmet.2020.07.017] [Citation(s) in RCA: 121] [Impact Index Per Article: 24.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2020] [Revised: 06/24/2020] [Accepted: 07/28/2020] [Indexed: 12/17/2022]
Abstract
We used BioID, a proximity-dependent biotinylation assay with 100 mitochondrial baits from all mitochondrial sub-compartments, to create a high-resolution human mitochondrial proximity interaction network. We identified 1,465 proteins, producing 15,626 unique high-confidence proximity interactions. Of these, 528 proteins were previously annotated as mitochondrial, nearly half of the mitochondrial proteome defined by Mitocarta 2.0. Bait-bait analysis showed a clear separation of mitochondrial compartments, and correlation analysis among preys across all baits allowed us to identify functional clusters involved in diverse mitochondrial functions and to assign uncharacterized proteins to specific modules. We demonstrate that this analysis can assign isoforms of the same mitochondrial protein to different mitochondrial sub-compartments and show that some proteins may have multiple cellular locations. Outer membrane baits showed specific proximity interactions with cytosolic proteins and proteins in other organellar membranes, suggesting specialization of proteins responsible for contact site formation between mitochondria and individual organelles.
Collapse
Affiliation(s)
- Hana Antonicka
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada; Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Zhen-Yuan Lin
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada
| | - Alexandre Janer
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada; Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Mari J Aaltonen
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada; Department of Human Genetics, McGill University, Montreal, QC, Canada
| | - Woranontee Weraarpachai
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada; Department of Biochemistry, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Anne-Claude Gingras
- Lunenfeld-Tanenbaum Research Institute, Toronto, ON, Canada; Department of Molecular Genetics, University of Toronto, Toronto, ON, Canada.
| | - Eric A Shoubridge
- Montreal Neurological Institute, McGill University, Montreal, QC, Canada; Department of Human Genetics, McGill University, Montreal, QC, Canada.
| |
Collapse
|
38
|
High Frequency of AIFM1 Variants and Phenotype Progression of Auditory Neuropathy in a Chinese Population. Neural Plast 2020; 2020:5625768. [PMID: 32684920 PMCID: PMC7350177 DOI: 10.1155/2020/5625768] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 05/14/2020] [Accepted: 06/02/2020] [Indexed: 12/12/2022] Open
Abstract
To decipher the genotype-phenotype correlation of auditory neuropathy (AN) caused by AIFM1 variations, as well as the phenotype progression of these patients, exploring the potential molecular pathogenic mechanism of AN. A total of 36 families of individuals with AN (50 cases) with AIFM1 variations were recruited and identified by Sanger sequencing or next-generation sequencing; the participants included 30 patients from 16 reported families and 20 new cases. We found that AIFM1-positive cases accounted for 18.6% of late-onset AN cases. Of the 50 AN patients with AIFM1 variants, 45 were male and 5 were female. The hotspot variation of this gene was p.Leu344Phe, accounting for 36.1%. A total of 19 AIFM1 variants were reported in this study, including 7 novel ones. A follow-up study was performed on 30 previously reported AIFM1-positive subjects, 16 follow-up cases (53.3%) were included in this study, and follow-up periods were recorded from 1 to 23 years with average 9.75 ± 9.89 years. There was no hearing threshold increase during the short-term follow-up period (1-10 years), but the low-frequency and high-frequency hearing thresholds showed a significant increase with the prolongation of follow-up time. The speech discrimination score progressed gradually and significantly along with the course of the disease and showed a more serious decline, which was disproportionately worse than the pure tone threshold. In addition to the X-linked recessive inheritance pattern, the X-linked dominant inheritance pattern is also observed in AIFM1-related AN and affects females. In conclusion, we confirmed that AIFM1 is the primary related gene among late-onset AN cases, and the most common recurrent variant is p.Leu344Phe. Except for the X-linked recessive inheritance pattern, the X-linked dominant inheritance pattern is another probability of AIFM1-related AN, with females affected. Phenotypical features of AIFM1-related AN suggested that auditory dyssynchrony progressively worsened over time.
Collapse
|
39
|
Reina S, Pittalà MGG, Guarino F, Messina A, De Pinto V, Foti S, Saletti R. Cysteine Oxidations in Mitochondrial Membrane Proteins: The Case of VDAC Isoforms in Mammals. Front Cell Dev Biol 2020; 8:397. [PMID: 32582695 PMCID: PMC7287182 DOI: 10.3389/fcell.2020.00397] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2020] [Accepted: 04/29/2020] [Indexed: 12/16/2022] Open
Abstract
Cysteine residues are reactive amino acids that can undergo several modifications driven by redox reagents. Mitochondria are the source of an abundant production of radical species, and it is surprising that such a large availability of highly reactive chemicals is compatible with viable and active organelles, needed for the cell functions. In this work, we review the results highlighting the modifications of cysteines in the most abundant proteins of the outer mitochondrial membrane (OMM), that is, the voltage-dependent anion selective channel (VDAC) isoforms. This interesting protein family carries several cysteines exposed to the oxidative intermembrane space (IMS). Through mass spectrometry (MS) analysis, cysteine posttranslational modifications (PTMs) were precisely determined, and it was discovered that such cysteines can be subject to several oxidization degrees, ranging from the disulfide bridge to the most oxidized, the sulfonic acid, one. The large spectra of VDAC cysteine oxidations, which is unique for OMM proteins, indicate that they have both a regulative function and a buffering capacity able to counteract excess of mitochondrial reactive oxygen species (ROS) load. The consequence of these peculiar cysteine PTMs is discussed.
Collapse
Affiliation(s)
- Simona Reina
- Section of Molecular Biology, Department of Biological, Geological and Environmental Sciences, University of Catania, Catania, Italy
| | - Maria Gaetana Giovanna Pittalà
- Section of Biology and Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Francesca Guarino
- Section of Biology and Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Angela Messina
- Section of Molecular Biology, Department of Biological, Geological and Environmental Sciences, University of Catania, Catania, Italy
| | - Vito De Pinto
- Section of Biology and Genetics, Department of Biomedical and Biotechnological Sciences, University of Catania, Catania, Italy
| | - Salvatore Foti
- Organic Mass Spectrometry Laboratory, Department of Chemical Sciences, University of Catania, Catania, Italy
| | - Rosaria Saletti
- Organic Mass Spectrometry Laboratory, Department of Chemical Sciences, University of Catania, Catania, Italy
| |
Collapse
|