1
|
Brosey CA, Shen R, Tainer JA. NADH-bound AIF activates the mitochondrial CHCHD4/MIA40 chaperone by a substrate-mimicry mechanism. EMBO J 2025:10.1038/s44318-024-00360-6. [PMID: 39806100 DOI: 10.1038/s44318-024-00360-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [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
|
2
|
Wischhof L, Mathew AJ, Bonaguro L, Beyer M, Ehninger D, Nicotera P, Bano D. Mitochondrial complex I inhibition enhances astrocyte responsiveness to pro-inflammatory stimuli. Sci Rep 2024; 14:27182. [PMID: 39516523 PMCID: PMC11549212 DOI: 10.1038/s41598-024-78434-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024] Open
Abstract
Inhibition of the mitochondrial oxidative phosphorylation (OXPHOS) system can lead to metabolic disorders and neurodegenerative diseases. In primary mitochondrial disorders, reactive astrocytes often accompany neuronal degeneration and may contribute to neurotoxic inflammatory cascades that elicit brain lesions. The influence of mitochondria to astrocyte reactivity as well as the underlying molecular mechanisms remain elusive. Here we report that mitochondrial Complex I dysfunction promotes neural progenitor cell differentiation into astrocytes that are more responsive to neuroinflammatory stimuli. We show that the SWItch/Sucrose Non-Fermentable (SWI/SNF/BAF) chromatin remodeling complex takes part in the epigenetic regulation of astrocyte responsiveness, since its pharmacological inhibition abrogates the expression of inflammatory genes. Furthermore, we demonstrate that Complex I deficient human iPSC-derived astrocytes negatively influence neuronal physiology upon cytokine stimulation. Together, our data describe the SWI/SNF/BAF complex as a sensor of altered mitochondrial OXPHOS and a downstream epigenetic regulator of astrocyte-mediated neuroinflammation.
Collapse
Affiliation(s)
- Lena Wischhof
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Gebäude 99, 53127, Bonn, Germany
| | - Amal John Mathew
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Gebäude 99, 53127, Bonn, Germany
| | - Lorenzo Bonaguro
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Gebäude 99, 53127, Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, DZNE and University of Bonn and West German Genome Center, Bonn, Germany
- Genomics and Immunoregulation, LIMES Institute, University of Bonn, Bonn, Germany
| | - Marc Beyer
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Gebäude 99, 53127, Bonn, Germany
- PRECISE Platform for Single Cell Genomics and Epigenomics, DZNE and University of Bonn and West German Genome Center, Bonn, Germany
| | - Dan Ehninger
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Gebäude 99, 53127, Bonn, Germany
| | - Pierluigi Nicotera
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Gebäude 99, 53127, Bonn, Germany
| | - Daniele Bano
- German Center for Neurodegenerative Diseases (DZNE), Venusberg-Campus 1, Gebäude 99, 53127, Bonn, Germany.
| |
Collapse
|
3
|
Nisco A, Tolomeo M, Scalise M, Zanier K, Barile M. Exploring the impact of flavin homeostasis on cancer cell metabolism. Biochim Biophys Acta Rev Cancer 2024; 1879:189149. [PMID: 38971209 DOI: 10.1016/j.bbcan.2024.189149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2024] [Revised: 06/25/2024] [Accepted: 07/01/2024] [Indexed: 07/08/2024]
Abstract
Flavins and their associated proteins have recently emerged as compelling players in the landscape of cancer biology. Flavins, encompassing flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD), serve as coenzymes in a multitude of cellular processes, such as metabolism, apoptosis, and cell proliferation. Their involvement in oxidative phosphorylation, redox homeostasis, and enzymatic reactions has long been recognized. However, recent research has unveiled an extended role for flavins in the context of cancer. In parallel, riboflavin transporters (RFVTs), FAD synthase (FADS), and riboflavin kinase (RFK) have gained prominence in cancer research. These proteins, responsible for riboflavin uptake, FAD biosynthesis, and FMN generation, are integral components of the cellular machinery that governs flavin homeostasis. Dysregulation in the expression/function of these proteins has been associated with various cancers, underscoring their potential as diagnostic markers, therapeutic targets, and key determinants of cancer cell behavior. This review embarks on a comprehensive exploration of the multifaceted role of flavins and of the flavoproteins involved in nucleus-mitochondria crosstalk in cancer. We journey through the influence of flavins on cancer cell energetics, the modulation of RFVTs in malignant transformation, the diagnostic and prognostic significance of FADS, and the implications of RFK in drug resistance and apoptosis. This review also underscores the potential of these molecules and processes as targets for novel diagnostic and therapeutic strategies, offering new avenues for the battle against this relentless disease.
Collapse
Affiliation(s)
- Alessia Nisco
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Italy
| | - Maria Tolomeo
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Italy; Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende, Italy
| | - Mariafrancesca Scalise
- Department of DiBEST (Biologia, Ecologia e Scienze della Terra), University of Calabria, Arcavacata di Rende, Italy
| | - Katia Zanier
- Biotechnology and Cell Signaling (CNRS/Université de Strasbourg, UMR 7242), Ecole Superieure de Biotechnologie de Strasbourg, Illkirch, France
| | - Maria Barile
- Department of Biosciences, Biotechnologies, and Environment, University of Bari Aldo Moro, Italy.
| |
Collapse
|
4
|
Wang L, Qi L, Huang X, Feng X, Gan J, Zhang J, Xi Y, Zhang S, Meng Q. RHBDF1 modulates cisplatin sensitivity of small cell lung cancer through YAP1/Smad2 signaling pathway. Heliyon 2024; 10:e33454. [PMID: 39027514 PMCID: PMC11254170 DOI: 10.1016/j.heliyon.2024.e33454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 06/20/2024] [Accepted: 06/21/2024] [Indexed: 07/20/2024] Open
Abstract
Small cell lung cancer (SCLC) is a fatal tumor type that is prone to drug resistance. In our previous study, we showed that human rhomboid-5 homolog-1 (RHBDF1) was differentially expressed in 5 intrinsic cisplatin-resistant SCLC tissues compared with 5 intrinsic cisplatin-sensitive SCLC tissues by RNA sequencing, which intrigued us. We performed gain- and loss-of-function experiments to investigate RHBDF1 function, bioinformatics analysis, qRT-PCR, western blotting, and immunoprecipitation to elucidate the molecular mechanisms as well as detect RHBDF1 expression in SCLC by immunohistochemistry. We found that RHBDF1 knockdown promoted cell proliferation and cisplatin chemoresistance and inhibited apoptosis in vitro and in vivo. These effects could be reversed by overexpressing RHBDF1 in vitro. Mechanistically, RHBDF1 interacted with YAP1, which increased the phosphorylation of Smad2 and transported Smad2 to the nucleus. Among clinical specimens, the RHBDF1 was a low expression in SCLC and was associated with clinicopathological features and prognosis. We are the first to reveal that RHBDF1 inhibited cell proliferation and promoted cisplatin sensitivity in SCLC and elucidate a novel mechanism through RHBDF1/YAP1/Smad2 signaling pathway which played a crucial role in cisplatin chemosensitivity. Targeting this pathway can be a promising therapeutic strategy for chemotherapy resistance in SCLC.
Collapse
Affiliation(s)
- Lei Wang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Lishuang Qi
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Xiaoyi Huang
- Biotherapy Center, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
- NHC Key Laboratory of Cell Transplantation, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Xiao Feng
- Department of Oncology, Jinan Central Hospital Affiliated to Shandong University, Jinan, Shandong, 250013, China
| | - Junqing Gan
- Department of Radiation Oncology, North China University of Science and Technology Affiliated Hospital, Tangshan, 063000, China
| | - Juxuan Zhang
- College of Bioinformatics Science and Technology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Yuhui Xi
- Department of Pathophysiology, Harbin Medical University, Harbin, Heilongjiang, 150081, China
| | - Shuai Zhang
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| | - Qingwei Meng
- Department of Medical Oncology, Harbin Medical University Cancer Hospital, Harbin, Heilongjiang, 150081, China
| |
Collapse
|
5
|
Shi T, Chen Z, Li J, Wang H, Wang Q. AIF translocation into nucleus caused by Aifm1 R450Q mutation: generation and characterization of a mouse model for AUNX1. Hum Mol Genet 2024; 33:905-918. [PMID: 38449065 PMCID: PMC11070138 DOI: 10.1093/hmg/ddae010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Revised: 10/30/2023] [Accepted: 01/05/2024] [Indexed: 03/08/2024] Open
Abstract
Mutations in AIFM1, encoding for apoptosis-inducing factor (AIF), cause AUNX1, an X-linked neurologic disorder with late-onset auditory neuropathy (AN) and peripheral neuropathy. Despite significant research on AIF, there are limited animal models with the disrupted AIFM1 representing the corresponding phenotype of human AUNX1, characterized by late-onset hearing loss and impaired auditory pathways. Here, we generated an Aifm1 p.R450Q knock-in mouse model (KI) based on the human AIFM1 p.R451Q mutation. Hemizygote KI male mice exhibited progressive hearing loss from P30 onward, with greater severity at P60 and stabilization until P210. Additionally, muscle atrophy was observed at P210. These phenotypic changes were accompanied by a gradual reduction in the number of spiral ganglion neuron cells (SGNs) at P30 and ribbons at P60, which coincided with the translocation of AIF into the nucleus starting from P21 and P30, respectively. The SGNs of KI mice at P210 displayed loss of cytomembrane integrity, abnormal nuclear morphology, and dendritic and axonal demyelination. Furthermore, the inner hair cells and myelin sheath displayed abnormal mitochondrial morphology, while fibroblasts from KI mice showed impaired mitochondrial function. In conclusion, we successfully generated a mouse model recapitulating AUNX1. Our findings indicate that disruption of Aifm1 induced the nuclear translocation of AIF, resulting in the impairment in the auditory pathway.
Collapse
Affiliation(s)
- Tao Shi
- Senior Department of Otolaryngology-Head and Neck Surgery, the Sixth Medical Center of PLA General Hospital, Medical School of Chinese PLA, 6 Fucheng Road, Beijing 100048, P. R. China
- National Clinical Research Center for Otolaryngologic Diseases, Chinese PLA General Hospital, 6 Fucheng Road, Beijing 100048, P. R. China
| | - Ziyi Chen
- Senior Department of Otolaryngology-Head and Neck Surgery, the Sixth Medical Center of PLA General Hospital, Medical School of Chinese PLA, 6 Fucheng Road, Beijing 100048, P. R. China
- National Clinical Research Center for Otolaryngologic Diseases, Chinese PLA General Hospital, 6 Fucheng Road, Beijing 100048, P. R. China
| | - Jin Li
- Senior Department of Otolaryngology-Head and Neck Surgery, the Sixth Medical Center of PLA General Hospital, Medical School of Chinese PLA, 6 Fucheng Road, Beijing 100048, P. R. China
- National Clinical Research Center for Otolaryngologic Diseases, Chinese PLA General Hospital, 6 Fucheng Road, Beijing 100048, P. R. China
| | - Hongyang Wang
- Senior Department of Otolaryngology-Head and Neck Surgery, the Sixth Medical Center of PLA General Hospital, Medical School of Chinese PLA, 6 Fucheng Road, Beijing 100048, P. R. China
- National Clinical Research Center for Otolaryngologic Diseases, Chinese PLA General Hospital, 6 Fucheng Road, Beijing 100048, P. R. China
| | - Qiuju Wang
- Senior Department of Otolaryngology-Head and Neck Surgery, the Sixth Medical Center of PLA General Hospital, Medical School of Chinese PLA, 6 Fucheng Road, Beijing 100048, P. R. China
- National Clinical Research Center for Otolaryngologic Diseases, Chinese PLA General Hospital, 6 Fucheng Road, Beijing 100048, P. R. China
| |
Collapse
|
6
|
Matveeva A, Watters O, Rukhadze A, Khemka N, Gentile D, Perez IF, Llorente-Folch I, Farrell C, Lo Cacciato E, Jackson J, Piazzesi A, Wischhof L, Woods I, Halang L, Hogg M, Muñoz AG, Dillon ET, Matallanas D, Arijs I, Lambrechts D, Bano D, Connolly NMC, Prehn JHM. Integrated analysis of transcriptomic and proteomic alterations in mouse models of ALS/FTD identify early metabolic adaptions with similarities to mitochondrial dysfunction disorders. Amyotroph Lateral Scler Frontotemporal Degener 2024; 25:135-149. [PMID: 37779364 DOI: 10.1080/21678421.2023.2261979] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Accepted: 09/10/2023] [Indexed: 10/03/2023]
Abstract
OBJECTIVE Sporadic and familial amyotrophic lateral sclerosis (ALS) is a fatal progressive neurodegenerative disease that results in loss of motor neurons and, in some patients, associates with frontotemporal dementia (FTD). Apart from the accumulation of proteinaceous deposits, emerging literature indicates that aberrant mitochondrial bioenergetics may contribute to the onset and progression of ALS/FTD. Here we sought to investigate the pathophysiological signatures of mitochondrial dysfunction associated with ALS/FTD. METHODS By means of label-free mass spectrometry (MS) and mRNA sequencing (mRNA-seq), we report pre-symptomatic changes in the cortices of TDP-43 and FUS mutant mouse models. Using tissues from transgenic mouse models of mitochondrial diseases as a reference, we performed comparative analyses and extracted unique and common mitochondrial signatures that revealed neuroprotective compensatory mechanisms in response to early damage. RESULTS In this regard, upregulation of both Acyl-CoA Synthetase Long-Chain Family Member 3 (ACSL3) and mitochondrial tyrosyl-tRNA synthetase 2 (YARS2) were the most representative change in pre-symptomatic ALS/FTD tissues, suggesting that fatty acid beta-oxidation and mitochondrial protein translation are mechanisms of adaptation in response to ALS/FTD pathology. CONCLUSIONS Together, our unbiased integrative analyses unveil novel molecular components that may influence mitochondrial homeostasis in the earliest phase of ALS.
Collapse
Affiliation(s)
- Anna Matveeva
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Orla Watters
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- SFI FutureNeuro Research Centre, Dublin 2, Ireland
| | - Ani Rukhadze
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Niraj Khemka
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Debora Gentile
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Ivan Fernandez Perez
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Irene Llorente-Folch
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Cliona Farrell
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | | | - Joshua Jackson
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Antonia Piazzesi
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Lena Wischhof
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Ina Woods
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Luise Halang
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Marion Hogg
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- SFI FutureNeuro Research Centre, Dublin 2, Ireland
- Department of Biosciences, Nottingham Trent University, Clifton Campus, Nottingham, UK
| | - Amaya Garcia Muñoz
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin 4, Belfield, Ireland
| | - Eugène T Dillon
- Mass Spectrometry Resource, Conway Institute of Biomolecular & Biomedical Research, University College Dublin, Dublin 4, Ireland
| | - David Matallanas
- Systems Biology Ireland, School of Medicine, University College Dublin, Dublin 4, Belfield, Ireland
| | - Ingrid Arijs
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium, and
- VIB Center for Cancer Biology, Leuven, Belgium
| | - Diether Lambrechts
- Laboratory for Translational Genetics, Department of Human Genetics, KU Leuven, Leuven, Belgium, and
- VIB Center for Cancer Biology, Leuven, Belgium
| | - Daniele Bano
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Niamh M C Connolly
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
| | - Jochen H M Prehn
- Centre for Systems Medicine, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- Department of Physiology and Medical Physics, Royal College of Surgeons in Ireland, Dublin 2, Ireland
- SFI FutureNeuro Research Centre, Dublin 2, Ireland
| |
Collapse
|
7
|
Hanaford AR, Khanna A, Truong V, James K, Chen Y, Mulholland M, Kayser B, Liao RW, Sedensky M, Morgan P, Andrew Baertsch N, Kalia V, Sarkar S, Johnson SC. Peripheral macrophages drive CNS disease in the Ndufs4(-/-) model of Leigh syndrome. Brain Pathol 2023; 33:e13192. [PMID: 37552802 PMCID: PMC10580015 DOI: 10.1111/bpa.13192] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Accepted: 07/07/2023] [Indexed: 08/10/2023] Open
Abstract
Subacute necrotizing encephalopathy, or Leigh syndrome (LS), is the most common pediatric presentation of genetic mitochondrial disease. LS is a multi-system disorder with severe neurologic, metabolic, and musculoskeletal symptoms. The presence of progressive, symmetric, and necrotizing lesions in the brainstem are a defining feature of the disease, and the major cause of morbidity and mortality, but the mechanisms underlying their pathogenesis have been elusive. Recently, we demonstrated that high-dose pexidartinib, a CSF1R inhibitor, prevents LS CNS lesions and systemic disease in the Ndufs4(-/-) mouse model of LS. While the dose-response in this study implicated peripheral immune cells, the immune populations involved have not yet been elucidated. Here, we used a targeted genetic tool, deletion of the colony-stimulating Factor 1 receptor (CSF1R) macrophage super-enhancer FIRE (Csf1rΔFIRE), to specifically deplete microglia and define the role of microglia in the pathogenesis of LS. Homozygosity for the Csf1rΔFIRE allele ablates microglia in both control and Ndufs4(-/-) animals, but onset of CNS lesions and sequalae in the Ndufs4(-/-), including mortality, are only marginally impacted by microglia depletion. The overall development of necrotizing CNS lesions is not altered, though microglia remain absent. Finally, histologic analysis of brainstem lesions provides direct evidence of a causal role for peripheral macrophages in the characteristic CNS lesions. These data demonstrate that peripheral macrophages play a key role in the pathogenesis of disease in the Ndufs4(-/-) model.
Collapse
Affiliation(s)
- Allison R. Hanaford
- Center for Integrative Brain ResearchSeattle Children's Research InstituteSeattleWashingtonUSA
| | - Asheema Khanna
- Ben Towne Center for Childhood Cancer ResearchSeattle Children's Research InstituteSeattleWashingtonUSA
| | - Vivian Truong
- Center for Integrative Brain ResearchSeattle Children's Research InstituteSeattleWashingtonUSA
| | - Katerina James
- Center for Integrative Brain ResearchSeattle Children's Research InstituteSeattleWashingtonUSA
| | - Yihan Chen
- Center for Integrative Brain ResearchSeattle Children's Research InstituteSeattleWashingtonUSA
| | - Michael Mulholland
- Center for Integrative Brain ResearchSeattle Children's Research InstituteSeattleWashingtonUSA
| | - Bernhard Kayser
- Center for Integrative Brain ResearchSeattle Children's Research InstituteSeattleWashingtonUSA
| | - Ryan W. Liao
- Center for Integrative Brain ResearchSeattle Children's Research InstituteSeattleWashingtonUSA
| | - Margaret Sedensky
- Center for Integrative Brain ResearchSeattle Children's Research InstituteSeattleWashingtonUSA
- Department of Anesthesiology and Pain MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Phil Morgan
- Center for Integrative Brain ResearchSeattle Children's Research InstituteSeattleWashingtonUSA
- Department of Anesthesiology and Pain MedicineUniversity of WashingtonSeattleWashingtonUSA
| | - Nathan Andrew Baertsch
- Center for Integrative Brain ResearchSeattle Children's Research InstituteSeattleWashingtonUSA
- Department of PediatricsUniversity of Washington School of MedicineSeattleWashingtonUSA
| | - Vandana Kalia
- Ben Towne Center for Childhood Cancer ResearchSeattle Children's Research InstituteSeattleWashingtonUSA
- Department of PediatricsUniversity of Washington School of MedicineSeattleWashingtonUSA
| | - Surojit Sarkar
- Ben Towne Center for Childhood Cancer ResearchSeattle Children's Research InstituteSeattleWashingtonUSA
- Department of PediatricsUniversity of Washington School of MedicineSeattleWashingtonUSA
| | - Simon C. Johnson
- Center for Integrative Brain ResearchSeattle Children's Research InstituteSeattleWashingtonUSA
- Department of Anesthesiology and Pain MedicineUniversity of WashingtonSeattleWashingtonUSA
- Department of Laboratory Medicine and PathologyUniversity of WashingtonSeattleWashingtonUSA
- Department of NeurologyUniversity of WashingtonSeattleWashingtonUSA
- Department of Applied Sciences, Translational BioscienceNorthumbria UniversityNewcastle Upon TyneUK
| |
Collapse
|
8
|
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: 0] [Impact Index Per Article: 0] [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
|
9
|
Unveiling Human Proteome Signatures of Heart Failure with Preserved Ejection Fraction. Biomedicines 2022; 10:biomedicines10112943. [PMID: 36428511 PMCID: PMC9687619 DOI: 10.3390/biomedicines10112943] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 11/08/2022] [Accepted: 11/12/2022] [Indexed: 11/18/2022] Open
Abstract
Heart failure with preserved ejection fraction (HFpEF) is a highly prevalent but still poorly understood clinical entity. Its current pathophysiological understanding supports a critical role of comorbidities and their chronic effect on cardiac function and structure. Importantly, despite the replication of some HFpEF phenotypic features, to this day, experimental models have failed to bring new effective therapies to the clinical setting. Thus, the direct investigation of HFpEF human myocardial samples may unveil key, and possibly human-specific, pathophysiological mechanisms. This study employed quantitative proteomic analysis by advanced mass spectrometry (SWATH-MS) to investigate signaling pathways and pathophysiological mechanisms in HFpEF. Protein-expression profiles were analyzed in human left ventricular myocardial samples of HFpEF patients and compared with a mixed control group. Functional analysis revealed several proteins that correlate with HFpEF, including those associated with mitochondrial dysfunction, oxidative stress, and inflammation. Despite the known disease heterogeneity, proteomic profiles could indicate a reduced mitochondrial oxidative phosphorylation and fatty-acid oxidation capacity in HFpEF patients with diabetes. The proteomic characterization described in this work provides new insights. Furthermore, it fosters further questions related to HFpEF cellular pathophysiology, paving the way for additional studies focused on developing novel therapies and diagnosis strategies for HFpEF patients.
Collapse
|
10
|
Hanaford A, Johnson SC. The immune system as a driver of mitochondrial disease pathogenesis: a review of evidence. Orphanet J Rare Dis 2022; 17:335. [PMID: 36056365 PMCID: PMC9438277 DOI: 10.1186/s13023-022-02495-3] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Accepted: 08/15/2022] [Indexed: 12/04/2022] Open
Abstract
BACKGROUND Genetic mitochondrial diseases represent a significant challenge to human health. These diseases are extraordinarily heterogeneous in clinical presentation and genetic origin, and often involve multi-system disease with severe progressive symptoms. Mitochondrial diseases represent the most common cause of inherited metabolic disorders and one of the most common causes of inherited neurologic diseases, yet no proven therapeutic strategies yet exist. The basic cell and molecular mechanisms underlying the pathogenesis of mitochondrial diseases have not been resolved, hampering efforts to develop therapeutic agents. MAIN BODY In recent pre-clinical work, we have shown that pharmacologic agents targeting the immune system can prevent disease in the Ndufs4(KO) model of Leigh syndrome, indicating that the immune system plays a causal role in the pathogenesis of at least this form of mitochondrial disease. Intriguingly, a number of case reports have indicated that immune-targeting therapeutics may be beneficial in the setting of genetic mitochondrial disease. Here, we summarize clinical and pre-clinical evidence suggesting a key role for the immune system in mediating the pathogenesis of at least some forms of genetic mitochondrial disease. CONCLUSIONS Significant clinical and pre-clinical evidence indicates a key role for the immune system as a significant in the pathogenesis of at least some forms of genetic mitochondrial disease.
Collapse
Affiliation(s)
- Allison Hanaford
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 9th Ave., JMB-925, Seattle, WA, 98101, USA
| | - Simon C Johnson
- Center for Integrative Brain Research, Seattle Children's Research Institute, 1900 9th Ave., JMB-925, Seattle, WA, 98101, USA.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
- Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA.
- Department of Neurology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
11
|
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: 15] [Impact Index Per Article: 5.0] [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
|
12
|
Salscheider SL, Gerlich S, Cabrera-Orefice A, Peker E, Rothemann RA, Murschall LM, Finger Y, Szczepanowska K, Ahmadi ZA, Guerrero-Castillo S, Erdogan A, Becker M, Ali M, Habich M, Petrungaro C, Burdina N, Schwarz G, Klußmann M, Neundorf I, Stroud DA, Ryan MT, Trifunovic A, Brandt U, Riemer J. AIFM1 is a component of the mitochondrial disulfide relay that drives complex I assembly through efficient import of NDUFS5. EMBO J 2022; 41:e110784. [PMID: 35859387 PMCID: PMC9434101 DOI: 10.15252/embj.2022110784] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 06/26/2022] [Accepted: 06/30/2022] [Indexed: 12/12/2022] Open
Abstract
The mitochondrial intermembrane space protein AIFM1 has been reported to mediate the import of MIA40/CHCHD4, which forms the import receptor in the mitochondrial disulfide relay. Here, we demonstrate that AIFM1 and MIA40/CHCHD4 cooperate beyond this MIA40/CHCHD4 import. We show that AIFM1 and MIA40/CHCHD4 form a stable long‐lived complex in vitro, in different cell lines, and in tissues. In HEK293 cells lacking AIFM1, levels of MIA40 are unchanged, but the protein is present in the monomeric form. Monomeric MIA40 neither efficiently interacts with nor mediates the import of specific substrates. The import defect is especially severe for NDUFS5, a subunit of complex I of the respiratory chain. As a consequence, NDUFS5 accumulates in the cytosol and undergoes rapid proteasomal degradation. Lack of mitochondrial NDUFS5 in turn results in stalling of complex I assembly. Collectively, we demonstrate that AIFM1 serves two overlapping functions: importing MIA40/CHCHD4 and constituting an integral part of the disulfide relay that ensures efficient interaction of MIA40/CHCHD4 with specific substrates.
Collapse
Affiliation(s)
| | - Sarah Gerlich
- Institute for Biochemistry, University of Cologne, Cologne, Germany
| | - Alfredo Cabrera-Orefice
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Esra Peker
- Institute for Biochemistry, University of Cologne, Cologne, Germany
| | | | | | - Yannik Finger
- Institute for Biochemistry, University of Cologne, Cologne, Germany
| | - Karolina Szczepanowska
- Medical Faculty, Institute for Mitochondrial Diseases and Aging, University of Cologne, Cologne, Germany.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Zeinab Alsadat Ahmadi
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Sergio Guerrero-Castillo
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Alican Erdogan
- Institute for Biochemistry, University of Cologne, Cologne, Germany
| | - Mark Becker
- Institute for Biochemistry, University of Cologne, Cologne, Germany
| | - Muna Ali
- Institute for Biochemistry, University of Cologne, Cologne, Germany
| | - Markus Habich
- Institute for Biochemistry, University of Cologne, Cologne, Germany
| | | | - Nele Burdina
- Institute for Biochemistry, University of Cologne, Cologne, Germany
| | - Guenter Schwarz
- 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.,Center for Molecular Medicine, University of Cologne, Cologne, Germany
| | - Merlin Klußmann
- Institute for Biochemistry, University of Cologne, Cologne, Germany
| | - Ines Neundorf
- Institute for Biochemistry, University of Cologne, Cologne, Germany
| | - David A Stroud
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Vic., Australia
| | - Michael T Ryan
- Department of Biochemistry and Molecular Biology, Monash Biomedicine Discovery Institute, Monash University, Melbourne, Vic., Australia
| | - Aleksandra Trifunovic
- Medical Faculty, Institute for Mitochondrial Diseases and Aging, 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
| | - Ulrich Brandt
- Radboud Institute for Molecular Life Sciences, Radboud University Medical Center, Nijmegen, The Netherlands.,Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, 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
|
13
|
Piazzesi A, Wang Y, Jackson J, Wischhof L, Zeisler-Diehl V, Scifo E, Oganezova I, Hoffmann T, Gómez Martín P, Bertan F, Wrobel CJJ, Schroeder FC, Ehninger D, Händler K, Schultze JL, Schreiber L, van Echten-Deckert G, Nicotera P, Bano D. CEST-2.2 overexpression alters lipid metabolism and extends longevity of mitochondrial mutants. EMBO Rep 2022; 23:e52606. [PMID: 35297148 PMCID: PMC9066074 DOI: 10.15252/embr.202152606] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 02/14/2022] [Accepted: 02/22/2022] [Indexed: 12/17/2022] Open
Abstract
Mitochondrial dysfunction can either extend or decrease Caenorhabditis elegans lifespan, depending on whether transcriptionally regulated responses can elicit durable stress adaptation to otherwise detrimental lesions. Here, we test the hypothesis that enhanced metabolic flexibility is sufficient to circumvent bioenergetic abnormalities associated with the phenotypic threshold effect, thereby transforming short‐lived mitochondrial mutants into long‐lived ones. We find that CEST‐2.2, a carboxylesterase mainly localizes in the intestine, may stimulate the survival of mitochondrial deficient animals. We report that genetic manipulation of cest‐2.2 expression has a minor lifespan impact on wild‐type nematodes, whereas its overexpression markedly extends the lifespan of complex I‐deficient gas‐1(fc21) mutants. We profile the transcriptome and lipidome of cest‐2.2 overexpressing animals and show that CEST‐2.2 stimulates lipid metabolism and fatty acid beta‐oxidation, thereby enhancing mitochondrial respiratory capacity through complex II and LET‐721/ETFDH, despite the inherited genetic lesion of complex I. Together, our findings unveil a metabolic pathway that, through the tissue‐specific mobilization of lipid deposits, may influence the longevity of mitochondrial mutant C. elegans.
Collapse
Affiliation(s)
- Antonia Piazzesi
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Yiru Wang
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Joshua Jackson
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Lena Wischhof
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | - Enzo Scifo
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Ina Oganezova
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Thorben Hoffmann
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | - Fabio Bertan
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Chester J J Wrobel
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, USA
| | - Frank C Schroeder
- Boyce Thompson Institute and Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY, USA
| | - Dan Ehninger
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Kristian Händler
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE), University of Bonn, Bonn, Germany
| | - Joachim L Schultze
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.,PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE), University of Bonn, Bonn, Germany.,Department for Genomics and Immunoregulation, LIMES Institute, University of Bonn, Bonn, Germany
| | - Lukas Schreiber
- Institute of Cellular and Molecular Botany (IZMB), University of Bonn, Bonn, Germany
| | | | | | - Daniele Bano
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| |
Collapse
|
14
|
Jackson J, Wischhof L, Scifo E, Pellizzer A, Wang Y, Piazzesi A, Gentile D, Siddig S, Stork M, Hopkins CE, Händler K, Weis J, Roos A, Schultze JL, Nicotera P, Ehninger D, Bano D. SGPL1 stimulates VPS39 recruitment to the mitochondria in MICU1 deficient cells. Mol Metab 2022; 61:101503. [PMID: 35452878 PMCID: PMC9170783 DOI: 10.1016/j.molmet.2022.101503] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Revised: 04/08/2022] [Accepted: 04/15/2022] [Indexed: 01/21/2023] Open
Abstract
Objective Mitochondrial “retrograde” signaling may stimulate organelle biogenesis as a compensatory adaptation to aberrant activity of the oxidative phosphorylation (OXPHOS) system. To maintain energy-consuming processes in OXPHOS deficient cells, alternative metabolic pathways are functionally coupled to the degradation, recycling and redistribution of biomolecules across distinct intracellular compartments. While transcriptional regulation of mitochondrial network expansion has been the focus of many studies, the molecular mechanisms promoting mitochondrial maintenance in energy-deprived cells remain poorly investigated. Methods We performed transcriptomics, quantitative proteomics and lifespan assays to identify pathways that are mechanistically linked to mitochondrial network expansion and homeostasis in Caenorhabditis elegans lacking the mitochondrial calcium uptake protein 1 (MICU-1/MICU1). To support our findings, we carried out biochemical and image analyses in mammalian cells and mouse-derived tissues. Results We report that micu-1(null) mutations impair the OXPHOS system and promote C. elegans longevity through a transcriptional program that is independent of the mitochondrial calcium uniporter MCU-1/MCU and the essential MCU regulator EMRE-1/EMRE. We identify sphingosine phosphate lyase SPL-1/SGPL1 and the ATFS-1-target HOPS complex subunit VPS-39/VPS39 as critical lifespan modulators of micu-1(null) mutant animals. Cross-species investigation indicates that SGPL1 upregulation stimulates VPS39 recruitment to the mitochondria, thereby enhancing mitochondria-lysosome contacts. Consistently, VPS39 downregulation compromises mitochondrial network maintenance and basal autophagic flux in MICU1 deficient cells. In mouse-derived muscles, we show that VPS39 recruitment to the mitochondria may represent a common signature associated with altered OXPHOS system. Conclusions Our findings reveal a previously unrecognized SGPL1/VPS39 axis that stimulates intracellular organelle interactions and sustains autophagy and mitochondrial homeostasis in OXPHOS deficient cells. micu-1(null) nematodes are long-lived mitochondrial mutants. MICU-1/MICU1 deficiency stimulates VPS-39/VPS39 and SPL-1/SGPL1 upregulation. VPS-39 sustains mitochondrial network expansion in micu-1(null) nematodes. VPS39 and SGPL1 expression influences mitochondria-lysosome contact sites in MICU1 deficient cells. VPS39/SGPL1 signaling may be a common signature of mitochondrial deficient cells.
Collapse
Affiliation(s)
- Joshua Jackson
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Lena Wischhof
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Enzo Scifo
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Anna Pellizzer
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Yiru Wang
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Antonia Piazzesi
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Debora Gentile
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Sana Siddig
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Miriam Stork
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | - Kristian Händler
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany; PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE), University of Bonn, Bonn, Germany
| | - Joachim Weis
- Institute of Neuropathology, RWTH Aachen University Hospital, Aachen, Germany
| | - Andreas Roos
- Universitätsklinikum Essen and Universität Duisburg-Essen, Essen, Germany
| | - Joachim L Schultze
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany; PRECISE Platform for Single Cell Genomics and Epigenomics, German Center for Neurodegenerative Diseases (DZNE), University of Bonn, Bonn, Germany; LIMES Institute, Department for Genomics and Immunoregulation, University of Bonn, Bonn, Germany
| | | | - Dan Ehninger
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Daniele Bano
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany.
| |
Collapse
|
15
|
Apoptosis-Inducing Factor Deficiency Induces Tissue-Specific Alterations in Autophagy: Insights from a Preclinical Model of Mitochondrial Disease and Exercise Training Effects. Antioxidants (Basel) 2022; 11:antiox11030510. [PMID: 35326160 PMCID: PMC8944439 DOI: 10.3390/antiox11030510] [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: 02/11/2022] [Revised: 03/01/2022] [Accepted: 03/03/2022] [Indexed: 02/04/2023] Open
Abstract
We analyzed the effects of apoptosis-inducing factor (AIF) deficiency, as well as those of an exercise training intervention on autophagy across tissues (heart, skeletal muscle, cerebellum and brain), that are primarily affected by mitochondrial diseases, using a preclinical model of these conditions, the Harlequin (Hq) mouse. Autophagy markers were analyzed in: (i) 2, 3 and 6 month-old male wild-type (WT) and Hq mice, and (ii) WT and Hq male mice that were allocated to an exercise training or sedentary group. The exercise training started upon onset of the first symptoms of ataxia in Hq mice and lasted for 8 weeks. Higher content of autophagy markers and free amino acids, and lower levels of sarcomeric proteins were found in the skeletal muscle and heart of Hq mice, suggesting increased protein catabolism. Leupeptin-treatment demonstrated normal autophagic flux in the Hq heart and the absence of mitophagy. In the cerebellum and brain, a lower abundance of Beclin 1 and ATG16L was detected, whereas higher levels of the autophagy substrate p62 and LAMP1 levels were observed in the cerebellum. The exercise intervention did not counteract the autophagy alterations found in any of the analyzed tissues. In conclusion, AIF deficiency induces tissue-specific alteration of autophagy in the Hq mouse, with accumulation of autophagy markers and free amino acids in the heart and skeletal muscle, but lower levels of autophagy-related proteins in the cerebellum and brain. Exercise intervention, at least if starting when muscle atrophy and neurological symptoms are already present, is not sufficient to mitigate autophagy perturbations.
Collapse
|
16
|
Stokes JC, Bornstein RL, James K, Park KY, Spencer KA, Vo K, Snell JC, Johnson BM, Morgan PG, Sedensky MM, Baertsch NA, Johnson SC. Leukocytes mediate disease pathogenesis in the Ndufs4(KO) mouse model of Leigh syndrome. JCI Insight 2022; 7:156522. [PMID: 35050903 PMCID: PMC8983133 DOI: 10.1172/jci.insight.156522] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 01/19/2022] [Indexed: 11/23/2022] Open
Abstract
Symmetric, progressive, necrotizing lesions in the brainstem are a defining feature of Leigh syndrome (LS). A mechanistic understanding of the pathogenesis of these lesions has been elusive. Here, we report that leukocyte proliferation is causally involved in the pathogenesis of LS. Depleting leukocytes with a colony-stimulating factor 1 receptor inhibitor disrupted disease progression, including suppression of CNS lesion formation and a substantial extension of survival. Leukocyte depletion rescued diverse symptoms, including seizures, respiratory center function, hyperlactemia, and neurologic sequelae. These data reveal a mechanistic explanation for the beneficial effects of mTOR inhibition. More importantly, these findings dramatically alter our understanding of the pathogenesis of LS, demonstrating that immune involvement is causal in disease. This work has important implications for the mechanisms of mitochondrial disease and may lead to novel therapeutic strategies.
Collapse
Affiliation(s)
- Julia C Stokes
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, United States of America
| | - Rebecca L Bornstein
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, United States of America
| | - Katerina James
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, United States of America
| | - Kyung Yeon Park
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, United States of America
| | - Kira A Spencer
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, United States of America
| | - Katie Vo
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, United States of America
| | - John C Snell
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, United States of America
| | - Brittany M Johnson
- Department of Neurology, University of Washington, Seattle, United States of America
| | - Philip G Morgan
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, United States of America
| | - Margaret M Sedensky
- Center for Integrative Brain Research, Seattle Children's Research Institute, Seattle, United States of America
| | - Nathan A Baertsch
- Department of Pediatrics, University of Washington, Seattle, United States of America
| | - Simon C Johnson
- Department of Neurology, University of Washington, Seattle, United States of America
| |
Collapse
|
17
|
Chen S, Lu XT, He TT, Yishake D, Tan XY, Hou MJ, Luo Y, Long JA, Tang ZH, Zhong RH, Fang AP, Zhu HL. Betaine Delayed Muscle Loss by Attenuating Samtor Complex Inhibition for mTORC1 Signaling Via Increasing SAM Level. Mol Nutr Food Res 2021; 65:e2100157. [PMID: 34061446 DOI: 10.1002/mnfr.202100157] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 04/29/2021] [Indexed: 01/26/2023]
Abstract
SCOPE The muscle loss during aging results from the blunt of protein synthesis and poses threat to the elderly health. This study aims to investigate whether betaine affects muscle loss by improving protein synthesis. METHODS AND RESULTS Male C57BL/6J mice are raised from age 12 or 15 months. Mice are fed with AIN-93M diet without or with 2% w/v betaine in distilled water as control group or betaine intervention group (Bet), respectively. Betaine supplementation to mice demonstrates better body composition, grip strength, and motor function. Muscle morphology upregulates expression of myogenic regulate factors, and elevates myosin heavy chain and also improves in Bet group. Betaine promotes muscle protein synthesis via tethering mammalian target of rapamycin complex1 protein kinase (mTORC1) on the lysosomal membrane thereby activating mTORC1 signaling. All these effects aforementioned are time-dependent (p < 0.05). Ultrahigh-performance liquid chromatography results show that betaine increases S-adenosyl-l-methionine (SAM) via methionine cycle. SAM sensor-Samtor-overexpression in C2C12 cells could displace mTORC1 from lysosome thereby inhibiting the mTORC1 signaling. Addition of betaine attenuates this inhibition by increasing SAM level and then disrupting interaction of Samtor complex. CONCLUSIONS These observations indicate that betaine could promisingly promote protein synthesis to delay age-related muscle loss.
Collapse
Affiliation(s)
- Si Chen
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, China
| | - Xiao-Ting Lu
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, China
| | - Tong-Tong He
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, China
| | - Dinuerguli Yishake
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, China
| | - Xu-Yin Tan
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, China
| | - Meng-Jun Hou
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, China
| | - Yun Luo
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, China
| | - Jing-An Long
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, China
| | - Zhi-Hong Tang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, China
| | - Rong-Huan Zhong
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, China
| | - Ai-Ping Fang
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, China
| | - Hui-Lian Zhu
- Department of Nutrition, School of Public Health, Sun Yat-sen University, Guangzhou, 510080, China
- Guangdong Provincial Key Laboratory of Food, Nutrition and Health, Guangzhou, 510080, China
| |
Collapse
|
18
|
Molecular Insights into Mitochondrial Protein Translocation and Human Disease. Genes (Basel) 2021; 12:genes12071031. [PMID: 34356047 PMCID: PMC8305315 DOI: 10.3390/genes12071031] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2021] [Revised: 06/27/2021] [Accepted: 06/30/2021] [Indexed: 12/11/2022] Open
Abstract
In human mitochondria, mtDNA encodes for only 13 proteins, all components of the OXPHOS system. The rest of the mitochondrial components, which make up approximately 99% of its proteome, are encoded in the nuclear genome, synthesized in cytosolic ribosomes and imported into mitochondria. Different import machineries translocate mitochondrial precursors, depending on their nature and the final destination inside the organelle. The proper and coordinated function of these molecular pathways is critical for mitochondrial homeostasis. Here, we will review molecular details about these pathways, which components have been linked to human disease and future perspectives on the field to expand the genetic landscape of mitochondrial diseases.
Collapse
|
19
|
Chiang DY, Lahiri S, Wang G, Karch J, Wang MC, Jung SY, Heck AJR, Scholten A, Wehrens XHT. Phosphorylation-Dependent Interactome of Ryanodine Receptor Type 2 in the Heart. Proteomes 2021; 9:proteomes9020027. [PMID: 34200203 PMCID: PMC8293434 DOI: 10.3390/proteomes9020027] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2021] [Revised: 05/27/2021] [Accepted: 06/02/2021] [Indexed: 11/16/2022] Open
Abstract
Hyperphosphorylation of the calcium release channel/ryanodine receptor type 2 (RyR2) at serine 2814 (S2814) is associated with multiple cardiac diseases including atrial fibrillation and heart failure. Despite recent advances, the molecular mechanisms driving pathological changes associated with RyR2 S2814 phosphorylation are still not well understood. Methods: Using affinity-purification coupled to mass spectrometry (AP-MS), we investigated the RyR2 interactome in ventricles from wild-type (WT) mice and two S2814 knock-in mutants: the unphosphorylated alanine mutant (S2814A) and hyperphosphorylated mimic aspartic acid mutant (S2814D). Western blots were used for validation. Results: In WT mouse ventricular lysates, we identified 22 proteins which were enriched with RyR2 pull-down relative to both IgG control and no antibody (beads-only) pull-downs. Parallel AP-MS using WT, S2814A, and S2814D mouse ventricles identified 72 proteins, with 20 being high confidence RyR2 interactors. Of these, 14 had an increase in their binding to RyR2 S2814A but a decrease in their binding to RyR2 S2814D. We independently validated three protein hits, Idh3b, Aifm1, and Cpt1b, as RyR2 interactors by western blots and showed that Aifm1 and Idh3b had significantly decreased binding to RyR2 S2814D compared to WT and S2814A, consistent with MS findings. Conclusion: By applying state-of-the-art proteomic approaches, we discovered a number of novel RyR2 interactors in the mouse heart. In addition, we found and defined specific alterations in the RyR2 interactome that were dependent on the phosphorylation status of RyR2 at S2814. These findings yield mechanistic insights into RyR2 regulation which may guide future drug designs.
Collapse
Affiliation(s)
- David Y. Chiang
- Cardiovascular Division, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Boston, MA 02115, USA;
| | - Satadru Lahiri
- Cardiovascular Research Institute, Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA; (S.L.); (G.W.); (J.K.)
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Guoliang Wang
- Cardiovascular Research Institute, Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA; (S.L.); (G.W.); (J.K.)
| | - Jason Karch
- Cardiovascular Research Institute, Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA; (S.L.); (G.W.); (J.K.)
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
| | - Meng C. Wang
- Huffington Center on Aging, Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA;
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX 77030, USA
- Howard Hughes Medical Institute, Baylor College of Medicine, Houston, TX 77030, USA
| | - Sung Y. Jung
- Department of Biochemistry, Baylor College of Medicine, Houston, TX 77030, USA;
| | - Albert J. R. Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 Utrecht, The Netherlands; (A.J.R.H.); (A.S.)
- Netherlands Proteomics Centre, 3584 Utrecht, The Netherlands
| | - Arjen Scholten
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Utrecht University, 3584 Utrecht, The Netherlands; (A.J.R.H.); (A.S.)
- Netherlands Proteomics Centre, 3584 Utrecht, The Netherlands
| | - Xander H. T. Wehrens
- Cardiovascular Research Institute, Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA; (S.L.); (G.W.); (J.K.)
- Department of Molecular Physiology & Biophysics, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Medicine (Cardiology), Baylor College of Medicine, Houston, TX 77030, USA
- Department of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
- Department of Pediatrics (Cardiology), Baylor College of Medicine, Houston, TX 77030, USA
- Center for Space Medicine, Baylor College of Medicine, Houston, TX 77030, USA
- Correspondence: ; Tel.: +1-713-798-4261
| |
Collapse
|
20
|
Bertan F, Wischhof L, Scifo E, Guranda M, Jackson J, Marsal-Cots A, Piazzesi A, Stork M, Peitz M, Prehn JHM, Ehninger D, Nicotera P, Bano D. Comparative analysis of CI- and CIV-containing respiratory supercomplexes at single-cell resolution. CELL REPORTS METHODS 2021; 1:100002. [PMID: 35474694 PMCID: PMC9017192 DOI: 10.1016/j.crmeth.2021.100002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 02/03/2021] [Accepted: 03/03/2021] [Indexed: 12/29/2022]
Abstract
Mitochondria sustain the energy demand of the cell. The composition and functional state of the mitochondrial oxidative phosphorylation system are informative indicators of organelle bioenergetic capacity. Here, we describe a highly sensitive and reproducible method for a single-cell quantification of mitochondrial CI- and CIV-containing respiratory supercomplexes (CI∗CIV-SCs) as an alternative means of assessing mitochondrial respiratory chain integrity. We apply a proximity ligation assay (PLA) and stain CI∗CIV-SCs in fixed human and mouse brains, tumorigenic cells, induced pluripotent stem cells (iPSCs) and iPSC-derived neural precursor cells (NPCs), and neurons. Spatial visualization of CI∗CIV-SCs enables the detection of mitochondrial lesions in various experimental models, including complex tissues undergoing degenerative processes. We report that comparative assessments of CI∗CIV-SCs facilitate the quantitative profiling of even subtle mitochondrial variations by overcoming the confounding effects that mixed cell populations have on other measurements. Together, our PLA-based analysis of CI∗CIV-SCs is a sensitive and complementary technique for detecting cell-type-specific mitochondrial perturbations in fixed materials.
Collapse
Affiliation(s)
- Fabio Bertan
- German Center for Neurodegenerative Diseases (DZNE), Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Venusberg-Campus 1, Gebäude 99, Bonn, North Rhine-Westphalia 53127, Germany
| | - Lena Wischhof
- German Center for Neurodegenerative Diseases (DZNE), Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Venusberg-Campus 1, Gebäude 99, Bonn, North Rhine-Westphalia 53127, Germany
| | - Enzo Scifo
- German Center for Neurodegenerative Diseases (DZNE), Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Venusberg-Campus 1, Gebäude 99, Bonn, North Rhine-Westphalia 53127, Germany
| | - Mihaela Guranda
- German Center for Neurodegenerative Diseases (DZNE), Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Venusberg-Campus 1, Gebäude 99, Bonn, North Rhine-Westphalia 53127, Germany
| | - Joshua Jackson
- German Center for Neurodegenerative Diseases (DZNE), Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Venusberg-Campus 1, Gebäude 99, Bonn, North Rhine-Westphalia 53127, Germany
| | - Anaïs Marsal-Cots
- German Center for Neurodegenerative Diseases (DZNE), Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Venusberg-Campus 1, Gebäude 99, Bonn, North Rhine-Westphalia 53127, Germany
| | - Antonia Piazzesi
- German Center for Neurodegenerative Diseases (DZNE), Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Venusberg-Campus 1, Gebäude 99, Bonn, North Rhine-Westphalia 53127, Germany
| | - Miriam Stork
- German Center for Neurodegenerative Diseases (DZNE), Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Venusberg-Campus 1, Gebäude 99, Bonn, North Rhine-Westphalia 53127, Germany
| | - Michael Peitz
- Institute of Reconstructive Neurobiology, University of Bonn Medical Faculty and University Hospital Bonn, Bonn, North Rhine-Westphalia 53127, Germany
- Cell Programming Core Facility, University of Bonn Medical Faculty, Bonn, North Rhine-Westphalia 53127, Germany
| | - Jochen Herbert Martin Prehn
- Royal College of Surgeons in Ireland, Department of Physiology and Medical Physics Department, D02 YN77 Dublin, Ireland
| | - Dan Ehninger
- German Center for Neurodegenerative Diseases (DZNE), Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Venusberg-Campus 1, Gebäude 99, Bonn, North Rhine-Westphalia 53127, Germany
| | - Pierluigi Nicotera
- German Center for Neurodegenerative Diseases (DZNE), Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Venusberg-Campus 1, Gebäude 99, Bonn, North Rhine-Westphalia 53127, Germany
| | - Daniele Bano
- German Center for Neurodegenerative Diseases (DZNE), Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Venusberg-Campus 1, Gebäude 99, Bonn, North Rhine-Westphalia 53127, Germany
| |
Collapse
|
21
|
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: 2] [Impact Index Per Article: 0.5] [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
|
22
|
Murari A, Rhooms SK, Goparaju NS, Villanueva M, Owusu-Ansah E. An antibody toolbox to track complex I assembly defines AIF's mitochondrial function. J Cell Biol 2021; 219:152090. [PMID: 32936885 PMCID: PMC7659709 DOI: 10.1083/jcb.202001071] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Revised: 06/01/2020] [Accepted: 06/26/2020] [Indexed: 01/09/2023] Open
Abstract
An ability to comprehensively track the assembly intermediates (AIs) of complex I (CI) biogenesis in Drosophila will enable the characterization of the precise mechanism(s) by which various CI regulators modulate CI assembly. Accordingly, we generated 21 novel antibodies to various mitochondrial proteins and used this resource to characterize the mechanism by which apoptosis-inducing factor (AIF) regulates CI biogenesis by tracking the AI profile observed when AIF expression is impaired. We find that when the AIF–Mia40 translocation complex is disrupted, the part of CI that transfers electrons to ubiquinone is synthesized but fails to progress in the CI biosynthetic pathway. This is associated with a reduction in intramitochondrial accumulation of the Mia40 substrate, MIC19. Importantly, knockdown of either MIC19 or MIC60, components of the mitochondrial contact site and cristae organizing system (MICOS), fully recapitulates the AI profile observed when AIF is inhibited. Thus, AIF’s effect on CI assembly is principally due to compromised intramitochondrial transport of the MICOS complex.
Collapse
Affiliation(s)
- Anjaneyulu Murari
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY
| | - Shauna-Kay Rhooms
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY
| | - Naga Sri Goparaju
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY
| | - Maximino Villanueva
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY
| | - Edward Owusu-Ansah
- Department of Physiology and Cellular Biophysics, Columbia University Medical Center, New York, NY.,The Robert N. Butler Columbia Aging Center, Columbia University Medical Center, New York, NY
| |
Collapse
|
23
|
Hintze M, Griesing S, Michels M, Blanck B, Wischhof L, Hartmann D, Bano D, Franz T. Alopecia in Harlequin mutant mice is associated with reduced AIF protein levels and expression of retroviral elements. Mamm Genome 2021; 32:12-29. [PMID: 33367954 PMCID: PMC7878237 DOI: 10.1007/s00335-020-09854-0] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 11/23/2020] [Indexed: 11/25/2022]
Abstract
We investigated the contribution of apoptosis-inducing factor (AIF), a key regulator of mitochondrial biogenesis, in supporting hair growth. We report that pelage abnormalities developed during hair follicle (HF) morphogenesis in Harlequin (Hq) mutant mice. Fragility of the hair cortex was associated with decreased expression of genes encoding structural hair proteins, though key transcriptional regulators of HF development were expressed at normal levels. Notably, Aifm1 (R200 del) knockin males and Aifm1(R200 del)/Hq females showed minor hair defects, despite substantially reduced AIF levels. Furthermore, we cloned the integrated ecotropic provirus of the Aifm1Hq allele. We found that its overexpression in wild-type keratinocyte cell lines led to down-regulation of HF-specific Krt84 and Krtap3-3 genes without altering Aifm1 or epidermal Krt5 expression. Together, our findings imply that pelage paucity in Hq mutant mice is mechanistically linked to severe AIF deficiency and is associated with the expression of retroviral elements that might potentially influence the transcriptional regulation of structural hair proteins.
Collapse
Affiliation(s)
- Maik Hintze
- Institute of Anatomy, Neuroanatomy, Medical Faculty, UKB, University of Bonn, Bonn, Germany.
- Medical Department, MSH Medical School Hamburg, Hamburg, Germany.
| | - Sebastian Griesing
- Institute of Anatomy, Neuroanatomy, Medical Faculty, UKB, University of Bonn, Bonn, Germany
- Dept. of Oncology, National Taiwan University Hospital, Taipei City, 100, Taiwan, ROC
| | - Marion Michels
- Institute of Anatomy, Neuroanatomy, Medical Faculty, UKB, University of Bonn, Bonn, Germany
| | - Birgit Blanck
- Institute of Anatomy, Neuroanatomy, Medical Faculty, UKB, University of Bonn, Bonn, Germany
| | - Lena Wischhof
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Dieter Hartmann
- Institute of Anatomy, Neuroanatomy, Medical Faculty, UKB, University of Bonn, Bonn, Germany
| | - Daniele Bano
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Thomas Franz
- Institute of Anatomy, Neuroanatomy, Medical Faculty, UKB, University of Bonn, Bonn, Germany
| |
Collapse
|
24
|
Regulation of diurnal energy balance by mitokines. Cell Mol Life Sci 2021; 78:3369-3384. [PMID: 33464381 PMCID: PMC7814174 DOI: 10.1007/s00018-020-03748-9] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2020] [Revised: 12/14/2020] [Accepted: 12/21/2020] [Indexed: 12/14/2022]
Abstract
The mammalian system of energy balance regulation is intrinsically rhythmic with diurnal oscillations of behavioral and metabolic traits according to the 24 h day/night cycle, driven by cellular circadian clocks and synchronized by environmental or internal cues such as metabolites and hormones associated with feeding rhythms. Mitochondria are crucial organelles for cellular energy generation and their biology is largely under the control of the circadian system. Whether mitochondrial status might also feed-back on the circadian system, possibly via mitokines that are induced by mitochondrial stress as endocrine-acting molecules, remains poorly understood. Here, we describe our current understanding of the diurnal regulation of systemic energy balance, with focus on fibroblast growth factor 21 (FGF21) and growth differentiation factor 15 (GDF15), two well-known endocrine-acting metabolic mediators. FGF21 shows a diurnal oscillation and directly affects the output of the brain master clock. Moreover, recent data demonstrated that mitochondrial stress-induced GDF15 promotes a day-time restricted anorexia and systemic metabolic remodeling as shown in UCP1-transgenic mice, where both FGF21 and GDF15 are induced as myomitokines. In this mouse model of slightly uncoupled skeletal muscle mitochondria GDF15 proved responsible for an increased metabolic flexibility and a number of beneficial metabolic adaptations. However, the molecular mechanisms underlying energy balance regulation by mitokines are just starting to emerge, and more data on diurnal patterns in mouse and man are required. This will open new perspectives into the diurnal nature of mitokines and action both in health and disease.
Collapse
|
25
|
Xu D, Wang Y, Wu J, Zhang Z, Chen J, Xie M, Tang R, Cheng C, Chen L, Lin S, Luo X, Zheng J. MTIF2 impairs 5 fluorouracil-mediated immunogenic cell death in hepatocellular carcinoma in vivo: Molecular mechanisms and therapeutic significance. Pharmacol Res 2021; 163:105265. [PMID: 33129983 DOI: 10.1016/j.phrs.2020.105265] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 10/17/2020] [Accepted: 10/19/2020] [Indexed: 02/07/2023]
Abstract
Hepatocellular carcinoma (HCC) is a leading cause of cancer-related morbidity and mortality; it has been reported that immune cell infiltration is a prognosis factor. Here we identified genes that associated with tumor immune cell infiltrate; the underlying mechanism was verified by in vivo and in vitro experiment. In this study, Weighted correlation network analysis (WGCNA) and CIBERSORT tool were used to identify MTIF2 as the hub tumor immune infiltrating gene in HCC. To investigate the underlying role played by MTIF2, MTIF2 was knocked down by transfection of shRNA targeting MTIF2, CCK8, and EdU incorporation assay was used to evaluate the effect of MTIF2 on proliferation, wound heal assay and transwell assay was used to confirm its effect on cell migration. Ecto-calreticulin on the cell surface was evaluated by flow cytometry, ATP, and HMGB1 secretion were tested to the investigated effect of MTIF2 on the immunogenic cell death (ICD) process. We found that down-regulation of MTIF2 impaired proliferation and migration capacity of HCC cells, chemoresistance to 5-Fluorouracil (5-FU) weakened after MTIF2 was knocked down. Reduced release of damage-associated molecular patterns (DAMP) was observed after MTIF2 was overexpressed, which subsequently impaired dendritic cell (DC) maturation and proliferation of CD8 + T cells. Mechanically, the co-IP experiment confirmed that MTIF2 could interact with AIFM1, prevents AIFM1 induced transcription of caspase3, and finally suppress apoptosis. In vivo experiment also used to confirm our previously conclusion, our result indicated that MTIF2 overexpression suppresses tumor apoptosis and immune cell activity in the 5-FU therapy in vivo model, by suppression maturation of tumor-infiltrated DC. Collectively, our study confirmed that MTIF2 impair drug-induced immunogenic cell death in hepatocellular carcinoma cells.
Collapse
Affiliation(s)
- Dafeng Xu
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, China
| | - Yu Wang
- Geriatric Medicine Center, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, China
| | - Jincai Wu
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, China
| | - Zhensheng Zhang
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, China
| | - Jiacheng Chen
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, China
| | - Mingwei Xie
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, China
| | - Rong Tang
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, China
| | - Chen Cheng
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, China
| | - Liang Chen
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, China
| | - Shiyun Lin
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, China
| | - Xiangxiang Luo
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, China
| | - Jinfang Zheng
- Department of Hepatobiliary and Pancreatic Surgery, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, China.
| |
Collapse
|
26
|
Herrmann JM, Riemer J. Apoptosis inducing factor and mitochondrial NADH dehydrogenases: redox-controlled gear boxes to switch between mitochondrial biogenesis and cell death. Biol Chem 2020; 402:289-297. [PMID: 32769219 DOI: 10.1515/hsz-2020-0254] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2020] [Accepted: 08/03/2020] [Indexed: 02/07/2023]
Abstract
The mitochondrial complex I serves as entry point for NADH into the electron transport chain. In animals, fungi and plants, additional NADH dehydrogenases carry out the same electron transfer reaction, however they do not pump protons. The apoptosis inducing factor (AIF, AIFM1 in humans) is a famous member of this group as it was the first pro-apoptotic protein identified that can induce caspase-independent cell death. Recent studies on AIFM1 and the NADH dehydrogenase Nde1 of baker's yeast revealed two independent and experimentally separable activities of this class of enzymes: On the one hand, these proteins promote the functionality of mitochondrial respiration in different ways: They channel electrons into the respiratory chain and, at least in animals, promote the import of Mia40 (named MIA40 or CHCHD4 in humans) and the assembly of complex I. On the other hand, they can give rise to pro-apoptotic fragments that are released from the mitochondria to trigger cell death. Here we propose that AIFM1 and Nde1 serve as conserved redox switches which measure metabolic conditions on the mitochondrial surface and translate it into a binary life/death decision. This function is conserved among eukaryotic cells and apparently used to purge metabolically compromised cells from populations.
Collapse
Affiliation(s)
- Johannes M Herrmann
- Department of Cell Biology, University of Kaiserslautern, Erwin-Schrödinger-Strasse 13, D-67663Kaiserslautern, Germany
| | - Jan Riemer
- Department of Biochemistry, University of Cologne, Zülpicher Str. 47A, D-50674Cologne, Germany
| |
Collapse
|
27
|
AIF meets the CHCHD4/Mia40-dependent mitochondrial import pathway. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165746. [PMID: 32105825 DOI: 10.1016/j.bbadis.2020.165746] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2019] [Revised: 02/19/2020] [Accepted: 02/20/2020] [Indexed: 02/06/2023]
Abstract
In the mitochondria of healthy cells, Apoptosis-Inducing factor (AIF) is required for the optimal functioning of the respiratory chain machinery, mitochondrial integrity, cell survival, and proliferation. In all analysed species, it was revealed that the downregulation or depletion of AIF provokes mainly the post-transcriptional loss of respiratory chain Complex I protein subunits. Recent progress in the field has revealed that AIF fulfils its mitochondrial pro-survival function by interacting physically and functionally with CHCHD4, the evolutionarily-conserved human homolog of yeast Mia40. The redox-regulated CHCHD4/Mia40-dependent import machinery operates in the intermembrane space of the mitochondrion and controls the import of a set of nuclear-encoded cysteine-motif carrying protein substrates. In addition to their participation in the biogenesis of specific respiratory chain protein subunits, CHCHD4/Mia40 substrates are also implicated in the control of redox regulation, antioxidant response, translation, lipid homeostasis and mitochondrial ultrastructure and dynamics. Here, we discuss recent insights on the AIF/CHCHD4-dependent protein import pathway and review current data concerning the CHCHD4/Mia40 protein substrates in metazoan. Recent findings and the identification of disease-associated mutations in AIF or in specific CHCHD4/Mia40 substrates have highlighted these proteins as potential therapeutic targets in a variety of human disorders.
Collapse
|
28
|
Klaus S, Ost M. Mitochondrial uncoupling and longevity - A role for mitokines? Exp Gerontol 2019; 130:110796. [PMID: 31786315 DOI: 10.1016/j.exger.2019.110796] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 11/15/2019] [Accepted: 11/25/2019] [Indexed: 12/12/2022]
Abstract
Aging has been viewed both as a random process due to accumulation of molecular and cellular damage over time and as a programmed process linked to cellular pathway important for growth and maturation. These views converge on mitochondria as both the major producer of damaging reactive oxidant species (ROS) and as signaling organelles. A finite proton leak across the inner mitochondrial membrane leading to a slight uncoupling of oxidative phosphorylation and respiration is an intrinsic property of all mitochondria and according to the "uncoupling to survive" hypothesis it has evolved to protect against ROS production to minimize oxidative damage. This hypothesis is supported by evidence linking an increased endogenous, uncoupling protein (UCP1) mediated, as well as experimentally induced mitochondrial uncoupling to an increased lifespan in rodents. This is possibly due to the synergistic activation of molecular pathways linked to life extending effects of caloric restriction as well as a mitohormetic response. Mitohormesis is an adaptive stress response through mitonuclear signaling which increases stress resistance resulting in health promoting effects. Part of this response is the induction of fibroblast growth factor 21 (FGF21) and growth and differentiation factor 15 (GDF15), two stress-induced mitokines which elicit beneficial systemic metabolic effects via endocrine action.
Collapse
Affiliation(s)
- Susanne Klaus
- German Institute of Human Nutrition in Potsdam Rehbrücke, Nuthetal, Germany; University of Potsdam, Institute of Nutritional Science, Potsdam, Germany.
| | - Mario Ost
- German Institute of Human Nutrition in Potsdam Rehbrücke, Nuthetal, Germany
| |
Collapse
|
29
|
Fiuza-Luces C, Valenzuela PL, Laine-Menéndez S, Fernández-de la Torre M, Bermejo-Gómez V, Rufián-Vázquez L, Arenas J, Martín MA, Lucia A, Morán M. Physical Exercise and Mitochondrial Disease: Insights From a Mouse Model. Front Neurol 2019; 10:790. [PMID: 31402893 PMCID: PMC6673140 DOI: 10.3389/fneur.2019.00790] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Accepted: 07/09/2019] [Indexed: 01/13/2023] Open
Abstract
Purpose: Mitochondrial diseases (MD) are among the most prevalent neuromuscular disorders. Unfortunately, no curative treatment is yet available. This study analyzed the effects of exercise training in an animal model of respiratory chain complex I deficiency, the Harlequin (Hq) mouse, which replicates the clinical features of this condition. Methods: Male heterozygous Harlequin (Hq/Y) mice were assigned to an “exercise” (n = 10) or a “sedentary” control group (n = 11), with the former being submitted to an 8 week combined exercise training intervention (aerobic + resistance training performed five times/week). Aerobic fitness, grip strength, and balance were assessed at the beginning and at the end of the intervention period in all the Hq mice. Muscle biochemical analyses (with results expressed as percentage of reference data from age/sex-matched sedentary wild-type mice [n = 12]) were performed at the end of the aforementioned period for the assessment of major molecular signaling pathways involved in muscle anabolism (mTOR activation) and mitochondrial biogenesis (proliferator activated receptor gamma co-activator 1α [PGC-1α] levels), and enzyme activity and levels of respiratory chain complexes, and antioxidant enzyme levels. Results: Exercise training resulted in significant improvements in aerobic fitness (−33 ± 13 m and 83 ± 43 m for the difference post- vs. pre-intervention in total distance covered in the treadmill tests in control and exercise group, respectively, p = 0.014) and muscle strength (2 ± 4 g vs. 17 ± 6 g for the difference post vs. pre-intervention, p = 0.037) compared to the control group. Higher levels of ribosomal protein S6 kinase beta-1 phosphorylated at threonine 389 (156 ± 30% vs. 249 ± 30%, p = 0.028) and PGC-1α (82 ± 7% vs. 126 ± 19% p = 0.032) were observed in the exercise-trained mice compared with the control group. A higher activity of respiratory chain complexes I (75 ± 4% vs. 95 ± 6%, p = 0.019), III (79 ± 5% vs. 97 ± 4%, p = 0.031), and V (77 ± 9% vs. 105 ± 9%, p = 0.024) was also found with exercise training. Exercised mice presented with lower catalase levels (204 ± 22% vs. 141 ± 23%, p = 0.036). Conclusion: In a mouse model of MD, a training intervention combining aerobic and resistance exercise increased aerobic fitness and muscle strength, and mild improvements were found for activated signaling pathways involved in muscle mitochondrial biogenesis and anabolism, OXPHOS complex activity, and redox status in muscle tissue.
Collapse
Affiliation(s)
- Carmen Fiuza-Luces
- Mitochondrial and Neuromuscular Diseases Laboratory, Research Institute of Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Pedro L Valenzuela
- Physiology Unit, Systems Biology Department, University of Alcalá, Madrid, Spain
| | - Sara Laine-Menéndez
- Mitochondrial and Neuromuscular Diseases Laboratory, Research Institute of Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Miguel Fernández-de la Torre
- Mitochondrial and Neuromuscular Diseases Laboratory, Research Institute of Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Verónica Bermejo-Gómez
- Mitochondrial and Neuromuscular Diseases Laboratory, Research Institute of Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Laura Rufián-Vázquez
- Mitochondrial and Neuromuscular Diseases Laboratory, Research Institute of Hospital 12 de Octubre (i+12), Madrid, Spain
| | - Joaquín Arenas
- Mitochondrial and Neuromuscular Diseases Laboratory, Research Institute of Hospital 12 de Octubre (i+12), Madrid, Spain.,Spanish Network for Biomedical Research in Rare Diseases (CIBERER), Madrid, Spain
| | - Miguel A Martín
- Mitochondrial and Neuromuscular Diseases Laboratory, Research Institute of Hospital 12 de Octubre (i+12), Madrid, Spain.,Spanish Network for Biomedical Research in Rare Diseases (CIBERER), Madrid, Spain
| | - Alejandro Lucia
- Faculty of Sports Sciences, European University of Madrid, Madrid, Spain.,Spanish Network for Biomedical Research in Fragility and Healthy Aging (CIBERFES), Madrid, Spain
| | - María Morán
- Mitochondrial and Neuromuscular Diseases Laboratory, Research Institute of Hospital 12 de Octubre (i+12), Madrid, Spain.,Spanish Network for Biomedical Research in Rare Diseases (CIBERER), Madrid, Spain
| |
Collapse
|
30
|
Gioran A, Piazzesi A, Bertan F, Schroer J, Wischhof L, Nicotera P, Bano D. Multi-omics identify xanthine as a pro-survival metabolite for nematodes with mitochondrial dysfunction. EMBO J 2019; 38:embj.201899558. [PMID: 30796049 PMCID: PMC6418696 DOI: 10.15252/embj.201899558] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2018] [Revised: 12/10/2018] [Accepted: 01/18/2019] [Indexed: 12/13/2022] Open
Abstract
Aberrant mitochondrial function contributes to the pathogenesis of various metabolic and chronic disorders. Inhibition of insulin/IGF‐1 signaling (IIS) represents a promising avenue for the treatment of mitochondrial diseases, although many of the molecular mechanisms underlying this beneficial effect remain elusive. Using an unbiased multi‐omics approach, we report here that IIS inhibition reduces protein synthesis and favors catabolism in mitochondrial deficient Caenorhabditis elegans. We unveil that the lifespan extension does not occur through the restoration of mitochondrial respiration, but as a consequence of an ATP‐saving metabolic rewiring that is associated with an evolutionarily conserved phosphoproteome landscape. Furthermore, we identify xanthine accumulation as a prominent downstream metabolic output of IIS inhibition. We provide evidence that supplementation of FDA‐approved xanthine derivatives is sufficient to promote fitness and survival of nematodes carrying mitochondrial lesions. Together, our data describe previously unknown molecular components of a metabolic network that can extend the lifespan of short‐lived mitochondrial mutant animals.
Collapse
Affiliation(s)
- Anna Gioran
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Antonia Piazzesi
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Fabio Bertan
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Jonas Schroer
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Lena Wischhof
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | | | - Daniele Bano
- German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| |
Collapse
|