1
|
Zhou P, Jin F, Yao S, Sun B, Sun N, Guan H, Liu X. Mitochondrial Mayhem: How cigarette smoke induces placental dysfunction through MMS19 degradation. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 290:117728. [PMID: 39823666 DOI: 10.1016/j.ecoenv.2025.117728] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 12/17/2024] [Accepted: 01/11/2025] [Indexed: 01/19/2025]
Abstract
Cigarette smoke (CS) has detrimental effects on placental growth and embryo development, but the underlying mechanisms remain unclear. This study aims to investigate the impact of CS on trophoblast cell proliferation and regulated cell death (RCD) by examining its interference with iron-sulfur cluster (ISC) proteins and the CIA pathway. Exposure to CS disrupted the cytosolic ISC assembly (CIA) pathway, downregulated ISC proteins, and decreased ISC maturation in the placenta of rats exposed to passive smoking. Studies using HTR-8/Sneo cells demonstrated that cigarette smoke extract (CSE) inhibits trophoblast proliferation, activates autophagy, and induces apoptosis by impairing the CIA pathway and ISC proteins. Molecular docking analysis revealed that nicotine and nicotyrine bind to and promote the autophagic-dependent degradation of MMS19, a key component of the CIA complex. MMS19 KD led to the autophagic degradation of several ISC proteins involved in DNA damage repair and mitochondrial respiratory function, thereby inhibiting cell proliferation. Additionally, MMS19 deficiency resulted in mitochondrial fragmentation, ROS accumulation, and the induction of autosis and apoptosis. Transcriptome analysis indicated that dysregulation of the SMAD pathway mediates mitochondrial damage induced by MMS19 KD. Analysis of placental tissues from maternal smokers further confirmed the disruption of ISC proteins and the SMAD pathway. This study suggests that disruption of the CIA pathway and ISC proteins contributes to placental maldevelopment induced by CS. Targeting the MMS19-SMAD pathway may offer a potential strategy to mitigate adverse pregnancy outcomes caused by CS.
Collapse
Affiliation(s)
- Pei Zhou
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Feng Jin
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Shenshen Yao
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Ben Sun
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Ni Sun
- Department of Medical Education, Dandong Central Hospital, Dandong 118002, PR China
| | - Hongbo Guan
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Xiaomei Liu
- Department of Obstetrics and Gynecology, Shengjing Hospital of China Medical University, Shenyang 110004, PR China.
| |
Collapse
|
2
|
Wu J, Qin T, Han W, Zhang C, Zhang X, Huang Z, Wu Y, Xu Y, Xu K, Ye W. GLS1-mediated glutamine metabolism mitigates oxidative stress-induced matrix degradation, ferroptosis, and senescence in nucleus pulposus cells by modulating Fe 2+ homeostasis. Free Radic Biol Med 2024:S0891-5849(24)01152-3. [PMID: 39710108 DOI: 10.1016/j.freeradbiomed.2024.12.043] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/10/2024] [Revised: 10/26/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
Intervertebral disc degeneration (IDD) is intricately linked to the pathogenesis of low back pain (LBP). The balance of nucleus pulposus (NP) cell and intervertebral disc (IVD) integrity is significantly supported by amino acid metabolism within an avascular milieu. However, the specific metabolic demands during the progression of IDD are not fully understood. Our study revealed that GLS1, a key enzyme that regulates glutamine metabolism, is key for mitigating NP cell ferroptosis, senescence, and IDD progression. Our findings show that GLS1 overexpression modulates glutamine metabolism, reducing NP cell matrix degradation, ferroptosis, and senescence. Mechanistically, GLS1 interacts with NFS1 and regulates ferrous ion (Fe2+) homeostasis. GLS1-driven glutamine metabolism facilitates acetyl-CoA production, which is important for the histone acetylation of NFS1. Thus, restoring GLS1 activity through gene overexpression to maintain Fe2+ homeostasis is a promising approach for mitigating matrix degradation, ferroptosis, and senescence and for rejuvenating intervertebral discs. Collectively, our data suggest a model in which GLS1-mediated glutamine metabolism is associated with NP cell matrix degradation, ferroptosis, and senescence and that NFS1 can be targeted to maintain Fe2+ homeostasis and ultimately revitalize intervertebral discs.
Collapse
Affiliation(s)
- Jiajun Wu
- Department of Orthopaedics, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510530; Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510289, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510289, China
| | - Tianyu Qin
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510289, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510289, China; Department of Spine Surgery, Orthopaedic, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University. The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen 518020, Guangdong, China
| | - Weitao Han
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510289, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510289, China
| | - Chao Zhang
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510289, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510289, China
| | - Xiaohe Zhang
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510289, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510289, China
| | - Zhengqi Huang
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510289, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510289, China
| | - Yuliang Wu
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510289, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510289, China
| | - Yichun Xu
- Department of Orthopaedics, The Third Affiliated Hospital of Sun Yat-Sen University, Guangzhou, Guangdong 510530.
| | - Kang Xu
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510289, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510289, China.
| | - Wei Ye
- Department of Spine Surgery, Sun Yat-sen Memorial Hospital of Sun Yat-sen University, Guangzhou 510289, China; Guangdong Provincial Key Laboratory of Malignant Tumor Epigenetics and Gene Regulation, Medical Research Center, Sun Yat-sen Memorial Hospital, Sun Yat-sen University, Guangzhou 510289, China.
| |
Collapse
|
3
|
Paz E, Jain S, Gottfried I, Staretz-Chacham O, Mahajnah M, Bagchi P, Seyfried NT, Ashery U, Azem A. Biochemical and neurophysiological effects of deficiency of the mitochondrial import protein TIMM50. eLife 2024; 13:RP99914. [PMID: 39680434 DOI: 10.7554/elife.99914] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2024] Open
Abstract
TIMM50, an essential TIM23 complex subunit, is suggested to facilitate the import of ~60% of the mitochondrial proteome. In this study, we characterized a TIMM50 disease-causing mutation in human fibroblasts and noted significant decreases in TIM23 core protein levels (TIMM50, TIMM17A/B, and TIMM23). Strikingly, TIMM50 deficiency had no impact on the steady-state levels of most of its putative substrates, suggesting that even low levels of a functional TIM23 complex are sufficient to maintain the majority of TIM23 complex-dependent mitochondrial proteome. As TIMM50 mutations have been linked to severe neurological phenotypes, we aimed to characterize TIMM50 defects in manipulated mammalian neurons. TIMM50 knockdown in mouse neurons had a minor effect on the steady state level of most of the mitochondrial proteome, supporting the results observed in patient fibroblasts. Amongst the few affected TIM23 substrates, a decrease in the steady state level of components of the intricate oxidative phosphorylation and mitochondrial ribosome complexes was evident. This led to declined respiration rates in fibroblasts and neurons, reduced cellular ATP levels, and defective mitochondrial trafficking in neuronal processes, possibly contributing to the developmental defects observed in patients with TIMM50 disease. Finally, increased electrical activity was observed in TIMM50 deficient mice neuronal cells, which correlated with reduced levels of KCNJ10 and KCNA2 plasma membrane potassium channels, likely underlying the patients' epileptic phenotype.
Collapse
Affiliation(s)
- Eyal Paz
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Sahil Jain
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Irit Gottfried
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
| | - Orna Staretz-Chacham
- Metabolic Disease Unit, Soroka Medical Center, Faculty of Health Sciences, Ben-Gurion University of the Negev, Beersheba, Israel
| | - Muhammad Mahajnah
- The Ruth and Bruce Rappaport Faculty of Medicine, Technion-Israel Institute of Technology, Haifa, Israel
- Child Neurology and Development Center, Hillel Yaffe Medical Center, Hadera, Israel
| | - Pritha Bagchi
- Center for Neurodegenerative Diseases, Emory University, Atlanta, United States
- Department of Biochemistry, Emory University, Atlanta, United States
- Emory Integrated Proteomics Core, Emory University, Atlanta, United States
| | - Nicholas T Seyfried
- Center for Neurodegenerative Diseases, Emory University, Atlanta, United States
- Department of Biochemistry, Emory University, Atlanta, United States
| | - Uri Ashery
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| | - Abdussalam Azem
- School of Neurobiology, Biochemistry and Biophysics, George S. Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv, Israel
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
4
|
Steinhilper R, Boß L, Freibert SA, Schulz V, Krapoth N, Kaltwasser S, Lill R, Murphy BJ. Two-stage binding of mitochondrial ferredoxin-2 to the core iron-sulfur cluster assembly complex. Nat Commun 2024; 15:10559. [PMID: 39632806 PMCID: PMC11618653 DOI: 10.1038/s41467-024-54585-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Accepted: 11/15/2024] [Indexed: 12/07/2024] Open
Abstract
Iron-sulfur (FeS) protein biogenesis in eukaryotes begins with the de novo assembly of [2Fe-2S] clusters by the mitochondrial core iron-sulfur cluster assembly (ISC) complex. This complex comprises the scaffold protein ISCU2, the cysteine desulfurase subcomplex NFS1-ISD11-ACP1, the allosteric activator frataxin (FXN) and the electron donor ferredoxin-2 (FDX2). The structural interaction of FDX2 with the complex remains unclear. Here, we present cryo-EM structures of the human FDX2-bound core ISC complex showing that FDX2 and FXN compete for overlapping binding sites. FDX2 binds in either a 'distal' conformation, where its helix F interacts electrostatically with an arginine patch of NFS1, or a 'proximal' conformation, where this interaction tightens and the FDX2-specific C terminus binds to NFS1, facilitating the movement of the [2Fe-2S] cluster of FDX2 closer to the ISCU2 FeS cluster assembly site for rapid electron transfer. Structure-based mutational studies verify the contact areas of FDX2 within the core ISC complex.
Collapse
Affiliation(s)
- Ralf Steinhilper
- Redox and Metalloprotein Research Group, Max Planck Institute of Biophysics, Max-von-Laue-Str. 3, 60438, Frankfurt am Main, Germany
| | - Linda Boß
- Institut für Zytobiologie, Philipps-Universität Marburg, Karl-von-Frisch-Str. 14, 35032, Marburg, Germany
- Zentrum für Synthetische Mikrobiologie Synmikro, Karl-von-Frisch-Str. 14, 35032, Marburg, Germany
| | - Sven-A Freibert
- Institut für Zytobiologie, Philipps-Universität Marburg, Karl-von-Frisch-Str. 14, 35032, Marburg, Germany
- Zentrum für Synthetische Mikrobiologie Synmikro, Karl-von-Frisch-Str. 14, 35032, Marburg, Germany
| | - Vinzent Schulz
- Institut für Zytobiologie, Philipps-Universität Marburg, Karl-von-Frisch-Str. 14, 35032, Marburg, Germany
- Zentrum für Synthetische Mikrobiologie Synmikro, Karl-von-Frisch-Str. 14, 35032, Marburg, Germany
| | - Nils Krapoth
- Institut für Zytobiologie, Philipps-Universität Marburg, Karl-von-Frisch-Str. 14, 35032, Marburg, Germany
- Zentrum für Synthetische Mikrobiologie Synmikro, Karl-von-Frisch-Str. 14, 35032, Marburg, Germany
| | - Susann Kaltwasser
- Central Electron Microscopy Facility, Max Planck Institute of Biophysics, Max-von-Laue-Str. 3, 60438, Frankfurt am Main, Germany
| | - Roland Lill
- Institut für Zytobiologie, Philipps-Universität Marburg, Karl-von-Frisch-Str. 14, 35032, Marburg, Germany.
- Zentrum für Synthetische Mikrobiologie Synmikro, Karl-von-Frisch-Str. 14, 35032, Marburg, Germany.
| | - Bonnie J Murphy
- Redox and Metalloprotein Research Group, Max Planck Institute of Biophysics, Max-von-Laue-Str. 3, 60438, Frankfurt am Main, Germany.
| |
Collapse
|
5
|
Hsiung KC, Tang HY, Cheng ML, Hung LM, Chin-Ming Tan B, Lo SJ. Mitochondrial Bioenergetics Deficiency in cisd-1 Mutants is Linked to AMPK-Mediated Lipid Metabolism. Biomed J 2024:100806. [PMID: 39521176 DOI: 10.1016/j.bj.2024.100806] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 09/17/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND CISD-1 is a mitochondrial iron-sulfate [2Fe-2S] protein known to be associated with various human diseases, including cancer and diabetes. Previously, we demonstrated that CISD-1 deficiency in worms lowers glucose and ATP levels. In this study, we further explored how worms compensate for lower ATP levels by analyzing changes in cytoplasmic and mitochondrial iron content, AMPK activities, and total lipid profiles. MATERIALS AND METHODS Expression levels of CISD-1 and CISD-1::GFP fusion proteins in wild-type worms (N2), cisd-1-deletion mutants (tm4993 and syb923) and GFP insertion transgenic worms (PHX953 and SJL40) were examined by western blot. Fluorescence microscopy analyzed CISD-1::GFP pattern in PHX953 embryos and adults, and lipid droplet sizes in N2, cisd-1, aak-2 and aak-2;cisd-1 worms. Total and mitochondrial iron content, electron transport complex profiles, and AMPK activity were investigated in tm4993 and syb923 mutants. mRNA levels of mitochondrial β-oxidation genes, acs-2, cpt-5, and ech-1, were quantified by RT-qPCR in various genetic worm strains. Lipidomic analyses were performed in N2 and cisd-1(tm4993) worms. RESULTS Defects in cisd-1 lead to an imbalance in iron transport and cause proton leak, resulting in lower ATP production by interrupting the mitochondrial electron transport chain. We identified a signaling pathway that links ATP deficiency-induced AMPK (AMP activated protein kinase) activation to the expression of genes that facilitate lipolysis via β-oxidation. CONCLUSION Our data provide a functional coordination between CISD-1 and AMPK constitutes a mitochondrial bioenergetics quality control mechanism that provides compensatory energy resources.
Collapse
Affiliation(s)
- Kuei-Ching Hsiung
- Department and Institute of Biomedical Sciences, College of Medicine, Chang Gung University, TaoYuan, Taiwan, 333
| | - Hsiang-Yu Tang
- Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University, TaoYuan, Taiwan
| | - Mei-Ling Cheng
- Department and Institute of Biomedical Sciences, College of Medicine, Chang Gung University, TaoYuan, Taiwan, 333; Metabolomics Core Laboratory, Healthy Aging Research Center, Chang Gung University, TaoYuan, Taiwan
| | - Li-Man Hung
- Department and Institute of Biomedical Sciences, College of Medicine, Chang Gung University, TaoYuan, Taiwan, 333
| | - Bertrand Chin-Ming Tan
- Department and Institute of Biomedical Sciences, College of Medicine, Chang Gung University, TaoYuan, Taiwan, 333; Molecular Medicine Research Center, Chang Gung University, TaoYuan, Taiwan, 333; Department of Neurosurgery, Lin-Kou Medical Center, Chang Gung Memorial Hospital, TaoYuan, Taiwan, 333.
| | - Szecheng J Lo
- Department and Institute of Biomedical Sciences, College of Medicine, Chang Gung University, TaoYuan, Taiwan, 333.
| |
Collapse
|
6
|
Zhang Y, Liu X, Xie L, Hong J, Zhuang Q, Ren L, Li X, Zhang C. Overexpression of Nfs1 Cysteine Desulphurase Relieves Sevoflurane-Induced Neurotoxicity and Cognitive Dysfunction in Neonatal Mice Via Suppressing Oxidative Stress and Ferroptosis. J Biochem Mol Toxicol 2024; 38:e70051. [PMID: 39488760 DOI: 10.1002/jbt.70051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 09/03/2024] [Accepted: 10/25/2024] [Indexed: 11/04/2024]
Abstract
Clinical evidence suggests that multiple exposures to sevoflurane in young people may be detrimental to cognitive development. Iron accumulation in the hippocampus is associated with sevoflurane-induced neurotoxicity and cognitive deficits. The cysteine desulphurase, Nfs1, the rate-limiting enzyme for the biosynthesis of iron-sulphur clusters, plays a role in cellular iron homeostasis. However, the impact of Nfs1-mediated ferroptosis on sevoflurane-induced neurotoxicity and cognitive impairments in neonatal mice remains undetermined. Neonatal mice at postnatal Day 6 received 3% sevoflurane daily for 3 consecutive days. Cognitive function was assessed using the Morris water maze test, and neurotoxicity was evaluated through terminal deoxynucleotidyl transferase dUTP nick end labeling and immunofluorescence staining. Here, HT22 hippocampal neurons were employed for in-vitro experiments, and Fe2+ accumulation was measured. Ferroptosis-related genes, including glutathione peroxidase 4 (GPX4), transferrin receptor 1 (TFR1) and ferritin, in the hippocampus and HT22 cells were observed, along with oxidative stress-related indicators such as reactive oxygen species (ROS), methionine adenosyltransferase (MAT), glutathione (GSH) and lipid peroxidation (LPO). Transmission electron microscopy was utilized to examine the mitochondrial microstructure. Sevoflurane exposure significantly decreased Nfs1 expression in the hippocampus of mice and HT22 cells. This exposure resulted in cognitive impairments and neuronal damage in the hippocampus, which were alleviated by overexpression of Nfs1. Intracellular and mitochondrial iron accumulation occurred in HT22 cells following sevoflurane treatment. Sevoflurane exposure also significantly reduced GSH levels and increased levels of malondialdehyde, ROS and LPO in the hippocampus or HT22 cells. Additionally, sevoflurane exposure decreased GPX4 expression but increased TFR1 and ferritin expression in the hippocampus or HT22 cells. Overexpression of Nfs1 reversed the sevoflurane-induced alterations in ferroptosis-related genes and oxidative stress-related indicators. Furthermore, overexpression of Nfs1 alleviated sevoflurane-induced mitochondrial dysfunction. However, Nfs1 knockdown alone did not result in cognitive impairments, ferroptosis or oxidative stress. The overexpression of Nfs1 mitigated sevoflurane-induced neurotoxicity and cognitive impairment by modulating oxidative stress and ferroptosis through the regulation of iron metabolism and transport.
Collapse
Affiliation(s)
- Yang Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Xinru Liu
- Department of Anesthesia, Bengbu Medical University, Bengbu, China
| | - Lijuan Xie
- Department of Anesthesia, Bengbu Medical University, Bengbu, China
| | - Jin Hong
- Department of Anesthesia, Bengbu Medical University, Bengbu, China
| | - Qin Zhuang
- Department of Anesthesiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Li Ren
- Department of Clinical Laboratory Medicine, Bengbu Medical University, Bengbu, China
| | - Xiaohong Li
- Department of Anesthesiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Congli Zhang
- Department of Anesthesiology, The First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| |
Collapse
|
7
|
Jiang Y, Li L, Li W, Liu K, Wu Y, Wang Z. NFS1 inhibits ferroptosis in gastric cancer by regulating the STAT3 pathway. J Bioenerg Biomembr 2024; 56:573-587. [PMID: 39254861 DOI: 10.1007/s10863-024-10038-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Accepted: 09/01/2024] [Indexed: 09/11/2024]
Abstract
Cysteine desulfurase (NFS1) is highly expressed in a variety of tumors, which is closely related to ferroptosis of tumor cells and affects prognosis. The relationship between NFS1 and the development of gastric cancer (GC) remains unknown. Here we showed that NFS1 expression was significantly higher in GC tissues compared to adjacent normal tissues. Patients with high expression of NFS1 in GC tissues had a lower overall survival rate than those with low expression. NFS1 was highly expressed in cultured GC cells compared to normal gastric cells. Knockdown of NFS1 expression reduced the viability, migration and invasion of GC cells. In cultured GC cells, NFS1 deficiency promoted ferroptosis. Mechanistically, NFS1 inhibited ferroptosis by upregulating the signal transduction and activator of transcription 3 (STAT3) signaling pathway in cultured GC cells. NFS1 knockdown using siRNA inhibited the STAT3 pathway, reduced the expression of glutathione peroxidase 4 (GPX4) and solute carrier family 7 member 11 (SLC7A11), and elevated intracellular levels of reactive oxygen species (ROS), ferrous ion (Fe2+), and malondialdehyde (MDA) in cultured GC cells. A specific STAT3 activator significantly reversed the inhibitory effect of NFS1 deficiency on ferroptosis in cultured GC cells. These in vitro results were further confirmed by experiments in vivo using a mouse xenograft tumor model. Collectively, THESE RESULTS INDICATE THAT NFS1 is overexpressed in human GC tissues and correlated with prognosis. NFS1 inhibits ferroptosis by activating the STAT3 pathway in GC cells. These results suggest that NFS1 may be a potential prognostic biomarker and therapeutic target to treat GC.
Collapse
Affiliation(s)
- You Jiang
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei City, 230022, Anhui Province, P.R. China
- Department of General Surgery, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, 230011, Anhui, P.R. China
| | - Liqiang Li
- Department of General Surgery, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, 230011, Anhui, P.R. China
| | - Wenbo Li
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei City, 230022, Anhui Province, P.R. China
- Department of General Surgery, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, 230011, Anhui, P.R. China
| | - Kun Liu
- Department of General Surgery, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, 230011, Anhui, P.R. China
| | - Yuee Wu
- Department of Electrocardiogram Diagnosis, Second Affiliated Hospital of Anhui Medical University, Hefei, 230060, Anhui, P.R. China
| | - Zhengguang Wang
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, 218 Jixi Road, Shushan District, Hefei City, 230022, Anhui Province, P.R. China.
| |
Collapse
|
8
|
Oney-Hawthorne SD, Barondeau DP. Fe-S cluster biosynthesis and maturation: Mass spectrometry-based methods advancing the field. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119784. [PMID: 38908802 DOI: 10.1016/j.bbamcr.2024.119784] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 04/25/2024] [Accepted: 06/10/2024] [Indexed: 06/24/2024]
Abstract
Iron‑sulfur (FeS) clusters are inorganic protein cofactors that perform essential functions in many physiological processes. Spectroscopic techniques have historically been used to elucidate details of FeS cluster type, their assembly and transfer, and changes in redox and ligand binding properties. Structural probes of protein topology, complex formation, and conformational dynamics are also necessary to fully understand these FeS protein systems. Recent developments in mass spectrometry (MS) instrumentation and methods provide new tools to investigate FeS cluster and structural properties. With the unique advantage of sampling all species in a mixture, MS-based methods can be utilized as a powerful complementary approach to probe native dynamic heterogeneity, interrogate protein folding and unfolding equilibria, and provide extensive insight into protein binding partners within an entire proteome. Here, we highlight key advances in FeS protein studies made possible by MS methodology and contribute an outlook for its role in the field.
Collapse
Affiliation(s)
| | - David P Barondeau
- Department of Chemistry, Texas A&M University, College Station, TX 77842, USA.
| |
Collapse
|
9
|
Paz E, Jain S, Gottfried I, Staretz-Chacham O, Mahajnah M, Bagchi P, Seyfried NT, Ashery U, Azem A. Biochemical and neurophysiological effects of deficiency of the mitochondrial import protein TIMM50. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.20.594480. [PMID: 38826427 PMCID: PMC11142075 DOI: 10.1101/2024.05.20.594480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/04/2024]
Abstract
TIMM50, an essential TIM23 complex subunit, is suggested to facilitate the import of ∼60% of the mitochondrial proteome. In this study, we characterized a TIMM50 disease causing mutation in human fibroblasts and noted significant decreases in TIM23 core protein levels (TIMM50, TIMM17A/B, and TIMM23). Strikingly, TIMM50 deficiency had no impact on the steady state levels of most of its putative substrates, suggesting that even low levels of a functional TIM23 complex are sufficient to maintain the majority of TIM23 complex-dependent mitochondrial proteome. As TIMM50 mutations have been linked to severe neurological phenotypes, we aimed to characterize TIMM50 defects in manipulated mammalian neurons. TIMM50 knockdown in mouse neurons had a minor effect on the steady state level of most of the mitochondrial proteome, supporting the results observed in patient fibroblasts. Amongst the few affected TIM23 substrates, a decrease in the steady state level of components of the intricate oxidative phosphorylation and mitochondrial ribosome complexes was evident. This led to declined respiration rates in fibroblasts and neurons, reduced cellular ATP levels and defective mitochondrial trafficking in neuronal processes, possibly contributing to the developmental defects observed in patients with TIMM50 disease. Finally, increased electrical activity was observed in TIMM50 deficient mice neuronal cells, which correlated with reduced levels of KCNJ10 and KCNA2 plasma membrane potassium channels, likely underlying the patients' epileptic phenotype.
Collapse
|
10
|
Jiang Y, Li W, Zhang J, Liu K, Wu Y, Wang Z. NFS1 as a Candidate Prognostic Biomarker for Gastric Cancer Correlated with Immune Infiltrates. Int J Gen Med 2024; 17:3855-3868. [PMID: 39253726 PMCID: PMC11382660 DOI: 10.2147/ijgm.s444443] [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: 07/12/2024] [Accepted: 09/01/2024] [Indexed: 09/11/2024] Open
Abstract
Background Cysteine desulfurase (NFS1) is closely associated with the occurrence and development of human tumors, but its relationship with the prognosis and immunity of gastric cancer (GC) patients remains unclear. Methods To study the relationship between NFS1 and GC, GC-related data of TCGA were downloaded and analyzed. At the same time, Tumor Immune Estimation Resource (TIMER) and Kaplan‒Meier Plotter were used for relevant online analysis. Clinical samples were collected for immunohistochemical testing to validate the results. Results The mRNA and protein levels of NFS1 in GC tissues were significantly higher than those in normal tissues. In terms of the operating characteristic curve (ROC), the area under the curve (AUC) was 0.793, indicating that NFS1 had a high diagnostic value for GC. Further analysis showed that NFS1 expression was highly correlated with the depth of tumor invasion, lymph node metastasis, and tumor stage. Survival analysis showed that patients with high expression of NFS1 had a poorer prognosis, and NFS1 was an independent risk factor. Enrichment analysis by GO, KEGG, and GSEA showed that NFS1 was enriched in immune-related pathways. The expression of NFS1 was significantly positively correlated with the proportion of macrophages M0 and plasma cells but negatively correlated with the proportion of B cells memory, monocytes, and mast cells resting. In addition, NFS1 expression was significantly correlated with TMB levels and responses to immunotherapy. Conclusion Our results suggest that NFS1 may be a potential biomarker for the diagnosis and prediction of prognosis and immunotherapy efficacy in GC.
Collapse
Affiliation(s)
- You Jiang
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230011, People's Republic of China
- Department of General Surgery, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, 230011, People's Republic of China
| | - Wenbo Li
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230011, People's Republic of China
- Department of General Surgery, The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, 230011, People's Republic of China
| | - Jun Zhang
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230011, People's Republic of China
| | - Kun Liu
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230011, People's Republic of China
| | - Yuee Wu
- Department of Electrocardiogram Diagnosis, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230060, People's Republic of China
| | - Zhengguang Wang
- Department of General Surgery, First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, 230011, People's Republic of China
| |
Collapse
|
11
|
Tang S, Fuß A, Fattahi Z, Culmsee C. Drp1 depletion protects against ferroptotic cell death by preserving mitochondrial integrity and redox homeostasis. Cell Death Dis 2024; 15:626. [PMID: 39191736 DOI: 10.1038/s41419-024-07015-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 08/15/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024]
Abstract
Mitochondria are highly dynamic organelles which undergo constant fusion and fission as part of the mitochondrial quality control. In genetic diseases and age-related neurodegenerative disorders, altered mitochondrial fission-fusion dynamics have been linked to impaired mitochondrial quality control, disrupted organelle integrity and function, thereby promoting neural dysfunction and death. The key enzyme regulating mitochondrial fission is the GTPase Dynamin-related Protein 1 (Drp1), which is also considered as a key player in mitochondrial pathways of regulated cell death. In particular, increasing evidence suggests a role for impaired mitochondrial dynamics and integrity in ferroptosis, which is an iron-dependent oxidative cell death pathway with relevance in neurodegeneration. In this study, we demonstrate that CRISPR/Cas9-mediated genetic depletion of Drp1 exerted protective effects against oxidative cell death by ferroptosis through preserved mitochondrial integrity and maintained redox homeostasis. Knockout of Drp1 resulted in mitochondrial elongation, attenuated ferroptosis-mediated impairment of mitochondrial membrane potential, and stabilized iron trafficking and intracellular iron storage. In addition, Drp1 deficiency exerted metabolic effects, with reduced basal and maximal mitochondrial respiration and a metabolic shift towards glycolysis. These metabolic effects further alleviated the mitochondrial contribution to detrimental ROS production thereby significantly enhancing neural cell resilience against ferroptosis. Taken together, this study highlights the key role of Drp1 in mitochondrial pathways of ferroptosis and expose the regulator of mitochondrial dynamics as a potential therapeutic target in neurological diseases involving oxidative dysregulation.
Collapse
Affiliation(s)
- Stephan Tang
- Institute for Pharmacology and Clinical Pharmacy, Philipps-University Marburg, Marburg, Germany
- Marburg Center of Mind, Brain and Behavior-CMBB, Marburg, Germany
| | - Anneke Fuß
- Institute for Pharmacology and Clinical Pharmacy, Philipps-University Marburg, Marburg, Germany
- Marburg Center of Mind, Brain and Behavior-CMBB, Marburg, Germany
- Institute of Reconstructive Neurobiology, Neurodevelopmental Genetics, University Bonn, LIFE & BRAIN Center, Bonn, Germany
| | - Zohreh Fattahi
- Institute for Pharmacology and Clinical Pharmacy, Philipps-University Marburg, Marburg, Germany
- Marburg Center of Mind, Brain and Behavior-CMBB, Marburg, Germany
| | - Carsten Culmsee
- Institute for Pharmacology and Clinical Pharmacy, Philipps-University Marburg, Marburg, Germany.
- Marburg Center of Mind, Brain and Behavior-CMBB, Marburg, Germany.
| |
Collapse
|
12
|
Molenaars M, Mir H, Alvarez SW, Arivazhagan L, Rosselot C, Zhan D, Park CY, Garcia-Ocana A, Schmidt AM, Possemato R. Acute inhibition of iron-sulfur cluster biosynthesis disrupts metabolic flexibility in mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.19.608291. [PMID: 39229169 PMCID: PMC11370322 DOI: 10.1101/2024.08.19.608291] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Iron-sulfur clusters (ISCs) are cell-essential cofactors present in ∼60 proteins including subunits of OXPHOS complexes I-III, DNA polymerases, and iron-sensing proteins. Dysfunctions in ISC biosynthesis are associated with anemias, neurodegenerative disorders, and metabolic diseases. To assess consequences of acute ISC inhibition in a whole body setting, we developed a mouse model in which key ISC biosynthetic enzyme NFS1 can be acutely and reversibly suppressed. Contrary to in vitro ISC inhibition and pharmacological OXPHOS suppression, global NFS1 inhibition rapidly enhances lipid utilization and decreases adiposity without affecting caloric intake and physical activity. ISC proteins decrease, including key proteins involved in OXPHOS (SDHB), lipoic acid synthesis (LIAS), and insulin mRNA processing (CDKAL1), causing acute metabolic inflexibility. Age-related metabolic changes decelerate loss of adiposity substantially prolonged survival of mice with NFS1 inhibition. Thus, the observation that ISC metabolism impacts organismal fuel choice will aid in understanding the mechanisms underlying ISC diseases with increased risk for diabetes. Graphical abstract Highlights Acute ISC inhibition leads to rapid loss of adiposity in miceMulti-metabolic pathway disruption upon ISC deficiency blocks energy storageNfs1 inhibition induces glucose dyshomeostasis due to ISC deficiency in β-cellsEnergy distress caused by inhibition of ISC synthesis is attenuated in aged mice.
Collapse
|
13
|
Pustimbara A, Li C, Ogura SI. Hemin enhances the 5-aminolevulinic acid-photodynamic therapy effect through the changes of cellular iron homeostasis. Photodiagnosis Photodyn Ther 2024; 48:104253. [PMID: 38901716 DOI: 10.1016/j.pdpdt.2024.104253] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/13/2024] [Accepted: 06/17/2024] [Indexed: 06/22/2024]
Abstract
BACKGROUND Photodynamic therapy (PDT) has been utilized as a promising alternative cancer treatment due to its minimum invasiveness over the years. Exogenous 5-aminolevulinic acid (ALA) triggers protoporphyrin IX (PpIX) accumulation, which happens in cancer cells. However, certain types of cancer exhibit reduced effectiveness in the PpIX accumulation mechanism. This study aimed to determine the effect of ALA-PDT combination with hemin on gastric carcinoma TMK-1 cells. METHODS This study utilized TMK-1 gastric cancer cell line to evaluate PpIX, ROS, and Fe2+ accumulation following the administration of ALA, hemin, and a combination of ALA and hemin PDT. We also evaluate the mRNA expressions related to iron homeostasis and treatment impacts on cell viability. RESULTS The co-addition of ALA and hemin PDT for 4 h of treatment resulted in a significant decrease in cell viability by up to 18 %. While ALA-PDT enhanced PpIX metabolism, the addition of hemin influenced both the production of reactive oxygen species (ROS) and cellular iron homeostasis by inducing Fe2+ accumulation and affecting mRNA levels of IRP, Tfr1, Ferritin, NFS1, and SDHB. CONCLUSION These findings suggest that the addition of ALA and hemin enhances phototoxicity in TMK-1 cells. The combination of ALA and hemin with PDT induces cell death, evidenced by increased cytotoxicity properties such as PpIX and ROS, along with significant changes in TMK-1 gastric cancer iron homeostasis. Therefore, the combination of ALA and hemin could be one of the alternatives in photodynamic therapy for cancer in the future.
Collapse
Affiliation(s)
- Anantya Pustimbara
- Tokyo Institute of Technology, School of Life Science and Technology, 4259 Midori-ku, Yokohama, 2268501, Japan.
| | - Chenhan Li
- Tokyo Institute of Technology, School of Life Science and Technology, 4259 Midori-ku, Yokohama, 2268501, Japan.
| | - Shun-Ichiro Ogura
- Tokyo Institute of Technology, School of Life Science and Technology, 4259 Midori-ku, Yokohama, 2268501, Japan.
| |
Collapse
|
14
|
Aneli S, Ceccatelli Berti C, Gilea AI, Birolo G, Mutti G, Pavesi A, Baruffini E, Goffrini P, Capelli C. Functional characterization of archaic-specific variants in mitonuclear genes: insights from comparative analysis in S. cerevisiae. Hum Mol Genet 2024; 33:1152-1163. [PMID: 38558123 DOI: 10.1093/hmg/ddae057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/29/2024] [Accepted: 03/14/2024] [Indexed: 04/04/2024] Open
Abstract
Neanderthal and Denisovan hybridisation with modern humans has generated a non-random genomic distribution of introgressed regions, the result of drift and selection dynamics. Cross-species genomic incompatibility and more efficient removal of slightly deleterious archaic variants have been proposed as selection-based processes involved in the post-hybridisation purge of archaic introgressed regions. Both scenarios require the presence of functionally different alleles across Homo species onto which selection operated differently according to which populations hosted them, but only a few of these variants have been pinpointed so far. In order to identify functionally divergent archaic variants removed in humans, we focused on mitonuclear genes, which are underrepresented in the genomic landscape of archaic humans. We searched for non-synonymous, fixed, archaic-derived variants present in mitonuclear genes, rare or absent in human populations. We then compared the functional impact of archaic and human variants in the model organism Saccharomyces cerevisiae. Notably, a variant within the mitochondrial tyrosyl-tRNA synthetase 2 (YARS2) gene exhibited a significant decrease in respiratory activity and a substantial reduction of Cox2 levels, a proxy for mitochondrial protein biosynthesis, coupled with the accumulation of the YARS2 protein precursor and a lower amount of mature enzyme. Our work suggests that this variant is associated with mitochondrial functionality impairment, thus contributing to the purging of archaic introgression in YARS2. While different molecular mechanisms may have impacted other mitonuclear genes, our approach can be extended to the functional screening of mitonuclear genetic variants present across species and populations.
Collapse
Affiliation(s)
- Serena Aneli
- Department of Public Health Sciences and Pediatrics, University of Turin, C.so Galileo Galilei 22, Turin 10126, Italy
| | - Camilla Ceccatelli Berti
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/a, Parma 43124, Italy
| | - Alexandru Ionut Gilea
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/a, Parma 43124, Italy
| | - Giovanni Birolo
- Department of Medical Sciences, University of Turin, Via Santena 5, Turin 10126, Italy
| | - Giacomo Mutti
- Barcelona Supercomputing Centre (BSC-CNS), Department of Life Sciences, Plaça Eusebi Güell, 1-3, Barcelona 08034, Spain
- Institute for Research in Biomedicine (IRB Barcelona), Department of Mechanisms of Disease, The Barcelona Institute of Science and Technology, Baldiri Reixac, 10, Barcelona 08028, Spain
| | - Angelo Pavesi
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/a, Parma 43124, Italy
| | - Enrico Baruffini
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/a, Parma 43124, Italy
| | - Paola Goffrini
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/a, Parma 43124, Italy
| | - Cristian Capelli
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parco Area delle Scienze 11/a, Parma 43124, Italy
- Department of Biology, University of Oxford, 11a Mansfield Rd, Oxford OX1 3SZ, United Kingdom
| |
Collapse
|
15
|
Sourice M, Oriol C, Aubert C, Mandin P, Py B. Genetic dissection of the bacterial Fe-S protein biogenesis machineries. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119746. [PMID: 38719030 DOI: 10.1016/j.bbamcr.2024.119746] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 04/12/2024] [Accepted: 05/03/2024] [Indexed: 05/13/2024]
Abstract
Iron‑sulfur (Fe-S) clusters are one of the most ancient and versatile inorganic cofactors present in the three domains of life. Fe-S clusters are essential cofactors for the activity of a large variety of metalloproteins that play crucial physiological roles. Fe-S protein biogenesis is a complex process that starts with the acquisition of the elements (iron and sulfur atoms) and their assembly into an Fe-S cluster that is subsequently inserted into the target proteins. The Fe-S protein biogenesis is ensured by multiproteic systems conserved across all domains of life. Here, we provide an overview on how bacterial genetics approaches have permitted to reveal and dissect the Fe-S protein biogenesis process in vivo.
Collapse
Affiliation(s)
- Mathieu Sourice
- Laboratoire de Chimie Bactérienne (UMR7283), Institut de Microbiologie de la Méditerranée, Institut Microbiologie Bioénergies et Biotechnologie, Centre National de la Recherche Scientifique, Aix-Marseille Université, Marseille, France
| | - Charlotte Oriol
- Laboratoire de Chimie Bactérienne (UMR7283), Institut de Microbiologie de la Méditerranée, Institut Microbiologie Bioénergies et Biotechnologie, Centre National de la Recherche Scientifique, Aix-Marseille Université, Marseille, France
| | - Corinne Aubert
- Laboratoire de Chimie Bactérienne (UMR7283), Institut de Microbiologie de la Méditerranée, Institut Microbiologie Bioénergies et Biotechnologie, Centre National de la Recherche Scientifique, Aix-Marseille Université, Marseille, France
| | - Pierre Mandin
- Laboratoire de Chimie Bactérienne (UMR7283), Institut de Microbiologie de la Méditerranée, Institut Microbiologie Bioénergies et Biotechnologie, Centre National de la Recherche Scientifique, Aix-Marseille Université, Marseille, France
| | - Béatrice Py
- Laboratoire de Chimie Bactérienne (UMR7283), Institut de Microbiologie de la Méditerranée, Institut Microbiologie Bioénergies et Biotechnologie, Centre National de la Recherche Scientifique, Aix-Marseille Université, Marseille, France.
| |
Collapse
|
16
|
Schulz V, Steinhilper R, Oltmanns J, Freibert SA, Krapoth N, Linne U, Welsch S, Hoock MH, Schünemann V, Murphy BJ, Lill R. Mechanism and structural dynamics of sulfur transfer during de novo [2Fe-2S] cluster assembly on ISCU2. Nat Commun 2024; 15:3269. [PMID: 38627381 PMCID: PMC11021402 DOI: 10.1038/s41467-024-47310-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2023] [Accepted: 03/26/2024] [Indexed: 04/19/2024] Open
Abstract
Maturation of iron-sulfur proteins in eukaryotes is initiated in mitochondria by the core iron-sulfur cluster assembly (ISC) complex, consisting of the cysteine desulfurase sub-complex NFS1-ISD11-ACP1, the scaffold protein ISCU2, the electron donor ferredoxin FDX2, and frataxin, a protein dysfunctional in Friedreich's ataxia. The core ISC complex synthesizes [2Fe-2S] clusters de novo from Fe and a persulfide (SSH) bound at conserved cluster assembly site residues. Here, we elucidate the poorly understood Fe-dependent mechanism of persulfide transfer from cysteine desulfurase NFS1 to ISCU2. High-resolution cryo-EM structures obtained from anaerobically prepared samples provide snapshots that both visualize different stages of persulfide transfer from Cys381NFS1 to Cys138ISCU2 and clarify the molecular role of frataxin in optimally positioning assembly site residues for fast sulfur transfer. Biochemical analyses assign ISCU2 residues essential for sulfur transfer, and reveal that Cys138ISCU2 rapidly receives the persulfide without a detectable intermediate. Mössbauer spectroscopy assessing the Fe coordination of various sulfur transfer intermediates shows a dynamic equilibrium between pre- and post-sulfur-transfer states shifted by frataxin. Collectively, our study defines crucial mechanistic stages of physiological [2Fe-2S] cluster assembly and clarifies frataxin's molecular role in this fundamental process.
Collapse
Affiliation(s)
- Vinzent Schulz
- Institut für Zytobiologie, Philipps-Universität Marburg, Karl-von-Frisch-Str. 14, 35032, Marburg, Germany
- Zentrum für Synthetische Mikrobiologie SynMikro, Karl-von-Frisch-Str. 14, 35032, Marburg, Germany
| | - Ralf Steinhilper
- Redox and Metalloprotein Research Group, Max Planck Institute of Biophysics, Max-von-Laue-Str. 3, 60438, Frankfurt am Main, Germany
| | - Jonathan Oltmanns
- Department of Physics, Biophysics and Medical Physics, University of Kaiserslautern-Landau, Erwin-Schrödinger-Str. 46, 67663, Kaiserslautern, Germany
| | - Sven-A Freibert
- Institut für Zytobiologie, Philipps-Universität Marburg, Karl-von-Frisch-Str. 14, 35032, Marburg, Germany
- Zentrum für Synthetische Mikrobiologie SynMikro, Karl-von-Frisch-Str. 14, 35032, Marburg, Germany
- Steinmühle-Schule & Internat, Steinmühlenweg 21, 35043, Marburg, Germany
| | - Nils Krapoth
- Institut für Zytobiologie, Philipps-Universität Marburg, Karl-von-Frisch-Str. 14, 35032, Marburg, Germany
- Zentrum für Synthetische Mikrobiologie SynMikro, Karl-von-Frisch-Str. 14, 35032, Marburg, Germany
| | - Uwe Linne
- Mass Spectrometry Facility of the Department of Chemistry, Philipps-Universität Marburg, Hans-Meerwein-Str. 4, 35032, Marburg, Germany
| | - Sonja Welsch
- Central Electron Microscopy Facility, Max Planck Institute of Biophysics, Max-von-Laue-Str. 3, 60438, Frankfurt am Main, Germany
| | - Maren H Hoock
- Department of Physics, Biophysics and Medical Physics, University of Kaiserslautern-Landau, Erwin-Schrödinger-Str. 46, 67663, Kaiserslautern, Germany
| | - Volker Schünemann
- Department of Physics, Biophysics and Medical Physics, University of Kaiserslautern-Landau, Erwin-Schrödinger-Str. 46, 67663, Kaiserslautern, Germany
| | - Bonnie J Murphy
- Redox and Metalloprotein Research Group, Max Planck Institute of Biophysics, Max-von-Laue-Str. 3, 60438, Frankfurt am Main, Germany.
| | - Roland Lill
- Institut für Zytobiologie, Philipps-Universität Marburg, Karl-von-Frisch-Str. 14, 35032, Marburg, Germany.
- Zentrum für Synthetische Mikrobiologie SynMikro, Karl-von-Frisch-Str. 14, 35032, Marburg, Germany.
| |
Collapse
|
17
|
Feng F, He S, Li X, He J, Luo L. Mitochondria-mediated Ferroptosis in Diseases Therapy: From Molecular Mechanisms to Implications. Aging Dis 2024; 15:714-738. [PMID: 37548939 PMCID: PMC10917537 DOI: 10.14336/ad.2023.0717] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Accepted: 07/17/2023] [Indexed: 08/08/2023] Open
Abstract
Ferroptosis, a type of cell death involving iron and lipid peroxidation, has been found to be closely associated with the development of many diseases. Mitochondria are vital components of eukaryotic cells, serving important functions in energy production, cellular metabolism, and apoptosis regulation. Presently, the precise relationship between mitochondria and ferroptosis remains unclear. In this study, we aim to systematically elucidate the mechanisms via which mitochondria regulate ferroptosis from multiple perspectives to provide novel insights into mitochondrial functions in ferroptosis. Additionally, we present a comprehensive overview of how mitochondria contribute to ferroptosis in different conditions, including cancer, cardiovascular disease, inflammatory disease, mitochondrial DNA depletion syndrome, and novel coronavirus pneumonia. Gaining a comprehensive understanding of the involvement of mitochondria in ferroptosis could lead to more effective approaches for both basic cell biology studies and medical treatments.
Collapse
Affiliation(s)
- Fuhai Feng
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, China.
| | - Shasha He
- Beijing Hospital of Traditional Chinese Medicine, Capital Medical University, Beijing, China.
| | - Xiaoling Li
- Animal Experiment Center, Guangdong Medical University, Zhanjiang, China.
| | - Jiake He
- The First Clinical College, Guangdong Medical University, Zhanjiang, Guangdong, China.
| | - Lianxiang Luo
- The Marine Biomedical Research Institute, Guangdong Medical University, Zhanjiang, Guangdong, China.
- The Marine Biomedical Research Institute of Guangdong Zhanjiang, Zhanjiang, Guangdong, China.
| |
Collapse
|
18
|
Doni D, Cavion F, Bortolus M, Baschiera E, Muccioli S, Tombesi G, d'Ettorre F, Ottaviani D, Marchesan E, Leanza L, Greggio E, Ziviani E, Russo A, Bellin M, Sartori G, Carbonera D, Salviati L, Costantini P. Human frataxin, the Friedreich ataxia deficient protein, interacts with mitochondrial respiratory chain. Cell Death Dis 2023; 14:805. [PMID: 38062036 PMCID: PMC10703789 DOI: 10.1038/s41419-023-06320-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 11/14/2023] [Accepted: 11/16/2023] [Indexed: 12/18/2023]
Abstract
Friedreich ataxia (FRDA) is a rare, inherited neurodegenerative disease caused by an expanded GAA repeat in the first intron of the FXN gene, leading to transcriptional silencing and reduced expression of frataxin. Frataxin participates in the mitochondrial assembly of FeS clusters, redox cofactors of the respiratory complexes I, II and III. To date it is still unclear how frataxin deficiency culminates in the decrease of bioenergetics efficiency in FRDA patients' cells. We previously demonstrated that in healthy cells frataxin is closely attached to the mitochondrial cristae, which contain both the FeS cluster assembly machinery and the respiratory chain complexes, whereas in FRDA patients' cells with impaired respiration the residual frataxin is largely displaced in the matrix. To gain novel insights into the function of frataxin in the mitochondrial pathophysiology, and in the upstream metabolic defects leading to FRDA disease onset and progression, here we explored the potential interaction of frataxin with the FeS cluster-containing respiratory complexes I, II and III. Using healthy cells and different FRDA cellular models we found that frataxin interacts with these three respiratory complexes. Furthermore, by EPR spectroscopy, we observed that in mitochondria from FRDA patients' cells the decreased level of frataxin specifically affects the FeS cluster content of complex I. Remarkably, we also found that the frataxin-like protein Nqo15 from T. thermophilus complex I ameliorates the mitochondrial respiratory phenotype when expressed in FRDA patient's cells. Our data point to a structural and functional interaction of frataxin with complex I and open a perspective to explore therapeutic rationales for FRDA targeted to this respiratory complex.
Collapse
Affiliation(s)
- Davide Doni
- Department of Biology, University of Padova, 35121, Padova, Italy
| | - Federica Cavion
- Department of Biology, University of Padova, 35121, Padova, Italy
| | - Marco Bortolus
- Department of Chemical Sciences, University of Padova, 35131, Padova, Italy
| | - Elisa Baschiera
- Clinical Genetics Unit, Department of Women's and Children Health, University of Padova, 35128, Padova, Italy
- Istituto di Ricerca Pediatrica (IRP) Città della Speranza, 35127, Padova, Italy
| | - Silvia Muccioli
- Department of Biology, University of Padova, 35121, Padova, Italy
| | - Giulia Tombesi
- Department of Biology, University of Padova, 35121, Padova, Italy
| | | | | | - Elena Marchesan
- Department of Biology, University of Padova, 35121, Padova, Italy
| | - Luigi Leanza
- Department of Biology, University of Padova, 35121, Padova, Italy
| | - Elisa Greggio
- Department of Biology, University of Padova, 35121, Padova, Italy
- Centro Studi per la Neurodegenerazione (CESNE), University of Padova, Padova, Italy
| | - Elena Ziviani
- Department of Biology, University of Padova, 35121, Padova, Italy
| | - Antonella Russo
- Department of Molecular Medicine, University of Padova, 35121, Padova, Italy
| | - Milena Bellin
- Department of Biology, University of Padova, 35121, Padova, Italy
- Veneto Institute of Molecular Medicine, 35129, Padova, Italy
- Department of Anatomy and Embryology, Leiden University Medical Center, 2333, ZA, Leiden, The Netherlands
| | - Geppo Sartori
- Department of Biomedical Sciences, University of Padova, 35121, Padova, Italy
| | | | - Leonardo Salviati
- Clinical Genetics Unit, Department of Women's and Children Health, University of Padova, 35128, Padova, Italy.
- Istituto di Ricerca Pediatrica (IRP) Città della Speranza, 35127, Padova, Italy.
| | - Paola Costantini
- Department of Biology, University of Padova, 35121, Padova, Italy.
| |
Collapse
|
19
|
Bargagna B, Banci L, Camponeschi F. Understanding the Molecular Basis of the Multiple Mitochondrial Dysfunctions Syndrome 2: The Disease-Causing His96Arg Mutation of BOLA3. Int J Mol Sci 2023; 24:11734. [PMID: 37511493 PMCID: PMC10380394 DOI: 10.3390/ijms241411734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 07/16/2023] [Accepted: 07/17/2023] [Indexed: 07/30/2023] Open
Abstract
Multiple mitochondrial dysfunctions syndrome type 2 with hyperglycinemia (MMDS2) is a severe disorder of mitochondrial energy metabolism, associated with biallelic mutations in the gene encoding for BOLA3, a protein with a not yet completely understood role in iron-sulfur (Fe-S) cluster biogenesis, but essential for the maturation of mitochondrial [4Fe-4S] proteins. To better understand the role of BOLA3 in MMDS2, we have investigated the impact of the p.His96Arg (c.287A > G) point mutation, which involves a highly conserved residue, previously identified as a [2Fe-2S] cluster ligand in the BOLA3-[2Fe-2S]-GLRX5 heterocomplex, on the structural and functional properties of BOLA3 protein. The His96Arg mutation has been associated with a severe MMDS2 phenotype, characterized by defects in the activity of mitochondrial respiratory complexes and lipoic acid-dependent enzymes. Size exclusion chromatography, NMR, UV-visible, circular dichroism, and EPR spectroscopy characterization have shown that the His96Arg mutation does not impair the interaction of BOLA3 with its protein partner GLRX5, but leads to the formation of an aberrant BOLA3-[2Fe-2S]-GLRX5 heterocomplex, that is not functional anymore in the assembly of a [4Fe-4S] cluster on NFU1. These results allowed us to rationalize the severe phenotype observed in MMDS2 caused by His96Arg mutation.
Collapse
Affiliation(s)
- Beatrice Bargagna
- Department of Chemistry, University of Florence, Via Della Lastruccia 3, Sesto Fiorentino, 50019 Florence, Italy
- Magnetic Resonance Center CERM, University of Florence, Via Luigi Sacconi 6, Sesto Fiorentino, 50019 Florence, Italy
| | - Lucia Banci
- Department of Chemistry, University of Florence, Via Della Lastruccia 3, Sesto Fiorentino, 50019 Florence, Italy
- Magnetic Resonance Center CERM, University of Florence, Via Luigi Sacconi 6, Sesto Fiorentino, 50019 Florence, Italy
- Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), Via Luigi Sacconi 6, Sesto Fiorentino, 50019 Florence, Italy
| | - Francesca Camponeschi
- Department of Chemistry, University of Florence, Via Della Lastruccia 3, Sesto Fiorentino, 50019 Florence, Italy
- Magnetic Resonance Center CERM, University of Florence, Via Luigi Sacconi 6, Sesto Fiorentino, 50019 Florence, Italy
- Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), Via Luigi Sacconi 6, Sesto Fiorentino, 50019 Florence, Italy
| |
Collapse
|
20
|
Du J, Huang Z, Li Y, Ren X, Zhou C, Liu R, Zhang P, Lei G, Lyu J, Li J, Tan G. Copper exerts cytotoxicity through inhibition of iron-sulfur cluster biogenesis on ISCA1/ISCA2/ISCU assembly proteins. Free Radic Biol Med 2023:S0891-5849(23)00433-1. [PMID: 37225108 DOI: 10.1016/j.freeradbiomed.2023.05.017] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2022] [Revised: 05/01/2023] [Accepted: 05/15/2023] [Indexed: 05/26/2023]
Abstract
Copper is an essential mineral nutrient that provides the cofactors for some key enzymes. However, excess copper is paradoxically cytotoxic. Wilson's disease is an autosomal recessive hereditary disease characterized by pathological copper accumulation in many organs, with high mortality and disability. Nevertheless, many questions about the molecular mechanism in Wilson's disease remain unknown and there is an imperative need to address these questions to better exploit therapeutic strategy. In this study, we constructed the mouse model of Wilson's disease, ATP7A-/- immortalized lymphocyte cell line and ATP7B knockdown cells to explore whether copper could impair iron-sulfur cluster biogenesis in eukaryotic mitochondria. Through a series of cellular, molecular, and pharmacological analyses, we demonstrated that copper could suppress the assembly of Fe-S cluster, decrease the activity of the Fe-S enzyme and disorder the mitochondrial function both in vivo and in vitro. Mechanistically, we found that human ISCA1, ISCA2 and ISCU proteins have a strong copper-binding activity, which would hinder the process of iron-sulfur assembly. Of note, we proposed a novel mechanism of action to explain the toxicity of copper by providing evidence that iron-sulfur cluster biogenesis may be a primary target of copper toxicity both in cells and mouse models. In summary, the current work provides an in-depth study on the mechanism of copper intoxication and describes a framework for the further understanding of impaired Fe-S assembly in the pathological processes of Wilson's diseases, which helps to develop latent therapeutic strategies for the management of copper toxicity.
Collapse
Affiliation(s)
- Jing Du
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China; Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Zhaoyang Huang
- Department of Clinical Laboratory, Renmin Hospital of Wuhan University, Wuhan, Hubei, 430060, China
| | - Yanchun Li
- Department of Central Laboratory, Affiliated Hangzhou First People's Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310006, China
| | - Xueying Ren
- Department of Laboratory Medicine, The Second Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou, Zhejiang, 310005, China
| | - Chaoting Zhou
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Ruolan Liu
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China
| | - Ping Zhang
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Guojie Lei
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Jianxin Lyu
- Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| | - Jianghui Li
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China.
| | - Guoqiang Tan
- Key Laboratory of Laboratory Medicine, Ministry of Education, Zhejiang Provincial Key Laboratory of Medical Genetics, College of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou, Zhejiang, 325035, China; Laboratory Medicine Center, Department of Clinical Laboratory, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China.
| |
Collapse
|
21
|
Stacpoole PW, McCall CE. The pyruvate dehydrogenase complex: Life's essential, vulnerable and druggable energy homeostat. Mitochondrion 2023; 70:59-102. [PMID: 36863425 DOI: 10.1016/j.mito.2023.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2022] [Revised: 01/30/2023] [Accepted: 02/13/2023] [Indexed: 03/04/2023]
Abstract
Found in all organisms, pyruvate dehydrogenase complexes (PDC) are the keystones of prokaryotic and eukaryotic energy metabolism. In eukaryotic organisms these multi-component megacomplexes provide a crucial mechanistic link between cytoplasmic glycolysis and the mitochondrial tricarboxylic acid (TCA) cycle. As a consequence, PDCs also influence the metabolism of branched chain amino acids, lipids and, ultimately, oxidative phosphorylation (OXPHOS). PDC activity is an essential determinant of the metabolic and bioenergetic flexibility of metazoan organisms in adapting to changes in development, nutrient availability and various stresses that challenge maintenance of homeostasis. This canonical role of the PDC has been extensively probed over the past decades by multidisciplinary investigations into its causal association with diverse physiological and pathological conditions, the latter making the PDC an increasingly viable therapeutic target. Here we review the biology of the remarkable PDC and its emerging importance in the pathobiology and treatment of diverse congenital and acquired disorders of metabolic integration.
Collapse
Affiliation(s)
- Peter W Stacpoole
- Department of Medicine (Division of Endocrinology, Metabolism and Diabetes), and Department of Biochemistry and Molecular Biology, University of Florida, College of Medicine, Gainesville, FL, United States.
| | - Charles E McCall
- Department of Internal Medicine and Translational Sciences, and Department of Microbiology and Immunology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| |
Collapse
|
22
|
Zhou W, Liu X, Lv M, Shi Y, Zhang L. The recognition mode between hsRBFA and mitoribosome 12S rRNA during mitoribosomal biogenesis. Nucleic Acids Res 2023; 51:1353-1363. [PMID: 36620886 PMCID: PMC9943654 DOI: 10.1093/nar/gkac1234] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 11/10/2022] [Accepted: 12/11/2022] [Indexed: 01/10/2023] Open
Abstract
Eukaryotes contain two sets of genomes: the nuclear genome and the mitochondrial genome. The mitochondrial genome transcripts 13 mRNAs that encode 13 essential proteins for the oxidative phosphorylation complex, 2 rRNAs (12s rRNA and 16s rRNA), and 22 tRNAs. The proper assembly and maturation of the mitochondrial ribosome (mitoribosome) are critical for the translation of the 13 key proteins and the function of the mitochondrion. Human ribosome-binding factor A (hsRBFA) is a mitoribosome assembly factor that binds with helix 28, helix 44 and helix 45 of 12S rRNA and facilitates the transcriptional modification of 12S rRNA during the mitoribosomal biogenesis. Previous research mentioned that the malfunction of hsRBFA will induce the instability of mitoribosomes and affect the function of mitochondria, but the mechanisms underlying the interaction between hsRBFA and 12S rRNA and its influence on mitochondrial function are still unknown. In this study, we found that hsRBFA binds with double strain RNA (dsRNA) through its whole N-terminus (Nt) instead of the KH-like domain alone, which is different from the other homologous. Furthermore, we mapped the key residues that affected the RNA binding and maturation of mitoribosomes in vitro. Finally, we investigated how these residues affect mitochondrial functions in detail and systematically.
Collapse
Affiliation(s)
- Wanwan Zhou
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, P.R. China,Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science & Technology of China, Hefei, P.R. China
| | - Xiaodan Liu
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, P.R. China,Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science & Technology of China, Hefei, P.R. China
| | - Mengqi Lv
- Hefei National Laboratory for Physical Sciences at the Microscale, School of Life Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, Anhui 230027, P.R. China,Ministry of Education Key Laboratory for Membraneless Organelles and Cellular Dynamics, University of Science & Technology of China, Hefei, P.R. China
| | - Yunyu Shi
- Correspondence may also be addressed to Yunyu Shi. Tel: +86 551 63607464; Fax: +86 551 63601443;
| | - Liang Zhang
- To whom correspondence should be addressed. Tel: +86 551 63600441; Fax: +86 551 63601443;
| |
Collapse
|
23
|
Baker MJ, Crameri JJ, Thorburn DR, Frazier AE, Stojanovski D. Mitochondrial biology and dysfunction in secondary mitochondrial disease. Open Biol 2022; 12:220274. [PMID: 36475414 PMCID: PMC9727669 DOI: 10.1098/rsob.220274] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Mitochondrial diseases are a broad, genetically heterogeneous class of metabolic disorders characterized by deficits in oxidative phosphorylation (OXPHOS). Primary mitochondrial disease (PMD) defines pathologies resulting from mutation of mitochondrial DNA (mtDNA) or nuclear genes affecting either mtDNA expression or the biogenesis and function of the respiratory chain. Secondary mitochondrial disease (SMD) arises due to mutation of nuclear-encoded genes independent of, or indirectly influencing OXPHOS assembly and operation. Despite instances of novel SMD increasing year-on-year, PMD is much more widely discussed in the literature. Indeed, since the implementation of next generation sequencing (NGS) techniques in 2010, many novel mitochondrial disease genes have been identified, approximately half of which are linked to SMD. This review will consolidate existing knowledge of SMDs and outline discrete categories within which to better understand the diversity of SMD phenotypes. By providing context to the biochemical and molecular pathways perturbed in SMD, we hope to further demonstrate the intricacies of SMD pathologies outside of their indirect contribution to mitochondrial energy generation.
Collapse
Affiliation(s)
- Megan J. Baker
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Jordan J. Crameri
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3052, Australia
| | - David R. Thorburn
- Murdoch Children's Research Institute, Royal Children's Hospital and Department of Paediatrics, University of Melbourne, Parkville, Victoria 3052, Australia,Victorian Clinical Genetics Services, Royal Children's Hospital, Parkville, Victoria 3052, Australia
| | - Ann E. Frazier
- Murdoch Children's Research Institute, Royal Children's Hospital and Department of Paediatrics, University of Melbourne, Parkville, Victoria 3052, Australia
| | - Diana Stojanovski
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria 3052, Australia
| |
Collapse
|
24
|
Parmagnani AS, D'Alessandro S, Maffei ME. Iron-sulfur complex assembly: Potential players of magnetic induction in plants. PLANT SCIENCE : AN INTERNATIONAL JOURNAL OF EXPERIMENTAL PLANT BIOLOGY 2022; 325:111483. [PMID: 36183809 DOI: 10.1016/j.plantsci.2022.111483] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/23/2022] [Revised: 09/19/2022] [Accepted: 09/27/2022] [Indexed: 06/16/2023]
Abstract
Iron-sulfur (Fe-S) clusters are involved in fundamental biological reactions and represent a highly regulated process involving a complex sequence of mitochondrial, cytosolic and nuclear-catalyzed protein-protein interactions. Iron-sulfur complex assembly (ISCA) scaffold proteins are involved in Fe-S cluster biosynthesis, nitrogen and sulfur metabolism. ISCA proteins are involved in abiotic stress responses and in the pigeon they act as a magnetic sensor by forming a magnetosensor (MagS) complex with cryptochrome (Cry). MagR gene exists in the genomes of humans, plants, and microorganisms and the interaction between Cry and MagR is highly conserved. Owing to the extensive presence of ISCA proteins in plants and the occurrence of homology between animal and human MagR with at least four Arabidopsis ISCAs and several ISCAs from different plant species, we believe that a mechanism similar to pigeon magnetoperception might be present in plants. We suggest that plant ISCA proteins, homologous of the animal MagR, are good candidates and could contribute to a better understanding of plant magnetic induction. We thus urge more studies in this regard to fully uncover the plant molecular mechanisms underlying MagR/Cry mediated magnetic induction and the possible coupling between light and magnetic induction.
Collapse
Affiliation(s)
- Ambra S Parmagnani
- Dept. Life Sciences and Systems Biology, University of Turin, Via Quarello 15/a, 10135 Turin, Italy
| | - Stefano D'Alessandro
- Dept. Life Sciences and Systems Biology, University of Turin, Via Quarello 15/a, 10135 Turin, Italy
| | - Massimo E Maffei
- Dept. Life Sciences and Systems Biology, University of Turin, Via Quarello 15/a, 10135 Turin, Italy.
| |
Collapse
|
25
|
Koç A, De Storme N. Structural regulation and dynamic behaviour of organelles during plant meiosis. Front Cell Dev Biol 2022; 10:925789. [DOI: 10.3389/fcell.2022.925789] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 10/06/2022] [Indexed: 11/13/2022] Open
Abstract
Eukaryotes use various mechanisms to maintain cell division stability during sporogenesis, and in particular during meiosis to achieve production of haploid spores. In addition to establishing even chromosome segregation in meiosis I and II, it is crucial for meiotic cells to guarantee balanced partitioning of organelles to the daughter cells, to properly inherit cellular functions. In plants, cytological studies in model systems have yielded insights into the meiotic behaviour of different organelles, i.e., clearly revealing a distinct organization at different stages throughout meiosis indicating for an active regulatory mechanism determining their subcellular dynamics. However, how, and why plant meiocytes organize synchronicity of these elements and whether this is conserved across all plant genera is still not fully elucidated. It is generally accepted that the highly programmed intracellular behaviour of organelles during meiosis serves to guarantee balanced cytoplasmic inheritance. However, recent studies also indicate that it contributes to the regulation of key meiotic processes, like the organization of cell polarity and spindle orientation, thus exhibiting different functionalities than those characterized in mitotic cell division. In this review paper, we will outline the current knowledge on organelle dynamics in plant meiosis and discuss the putative strategies that the plant cell uses to mediate this programmed spatio-temporal organization in order to safeguard balanced separation of organelles. Particular attention is thereby given to putative molecular mechanisms that underlie this dynamic organelle organization taken into account existing variations in the meiotic cell division program across different plant types. Furthermore, we will elaborate on the structural role of organelles in plant meiosis and discuss on organelle-based cellular mechanisms that contribute to the organization and molecular coordination of key meiotic processes, including spindle positioning, chromosome segregation and cell division. Overall, this review summarizes all relevant insights on the dynamic behaviour and inheritance of organelles during plant meiosis, and discusses on their functional role in the structural and molecular regulation of meiotic cell division.
Collapse
|
26
|
Fujihara KM, Zhang BZ, Jackson TD, Ogunkola MO, Nijagal B, Milne JV, Sallman DA, Ang CS, Nikolic I, Kearney CJ, Hogg SJ, Cabalag CS, Sutton VR, Watt S, Fujihara AT, Trapani JA, Simpson KJ, Stojanovski D, Leimkühler S, Haupt S, Phillips WA, Clemons NJ. Eprenetapopt triggers ferroptosis, inhibits NFS1 cysteine desulfurase, and synergizes with serine and glycine dietary restriction. SCIENCE ADVANCES 2022; 8:eabm9427. [PMID: 36103522 PMCID: PMC9473576 DOI: 10.1126/sciadv.abm9427] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/08/2023]
Abstract
The mechanism of action of eprenetapopt (APR-246, PRIMA-1MET) as an anticancer agent remains unresolved, although the clinical development of eprenetapopt focuses on its reported mechanism of action as a mutant-p53 reactivator. Using unbiased approaches, this study demonstrates that eprenetapopt depletes cellular antioxidant glutathione levels by increasing its turnover, triggering a nonapoptotic, iron-dependent form of cell death known as ferroptosis. Deficiency in genes responsible for supplying cancer cells with the substrates for de novo glutathione synthesis (SLC7A11, SHMT2, and MTHFD1L), as well as the enzymes required to synthesize glutathione (GCLC and GCLM), augments the activity of eprenetapopt. Eprenetapopt also inhibits iron-sulfur cluster biogenesis by limiting the cysteine desulfurase activity of NFS1, which potentiates ferroptosis and may restrict cellular proliferation. The combination of eprenetapopt with dietary serine and glycine restriction synergizes to inhibit esophageal xenograft tumor growth. These findings reframe the canonical view of eprenetapopt from a mutant-p53 reactivator to a ferroptosis inducer.
Collapse
Affiliation(s)
- Kenji M. Fujihara
- Gastrointestinal Cancer Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Corresponding author. (N.J.C.); (K.M.F.)
| | - Bonnie Z. Zhang
- Gastrointestinal Cancer Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - Thomas D. Jackson
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria, Australia
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Moses O. Ogunkola
- Institute of Biochemistry and Biology Department for Molecular Enzymology, University of Potsdam, Potsdam, Germany
| | - Brunda Nijagal
- Metabolomics Australia, The Bio21 Institute of Molecular Science and Biotechnology, The University of Melbourne, Parkville, Victoria, Australia
| | - Julia V. Milne
- Gastrointestinal Cancer Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
| | - David A. Sallman
- Malignant Hematology Department, H. Lee Moffitt Cancer Center and Research Institute, Tampa, FL, USA
| | - Ching-Seng Ang
- Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Iva Nikolic
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Conor J. Kearney
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Translational Hematology Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Simon J. Hogg
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Translational Hematology Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Human Oncology and Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Carlos S. Cabalag
- Gastrointestinal Cancer Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Department of Surgical Oncology, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Vivien R. Sutton
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Sally Watt
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Asuka T. Fujihara
- Gastrointestinal Cancer Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Joseph A. Trapani
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Cancer Immunology Program, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Kaylene J. Simpson
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Victorian Centre for Functional Genomics, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Diana Stojanovski
- Department of Biochemistry and Pharmacology, The University of Melbourne, Parkville, Victoria, Australia
| | - Silke Leimkühler
- Institute of Biochemistry and Biology Department for Molecular Enzymology, University of Potsdam, Potsdam, Germany
| | - Sue Haupt
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Tumor Suppression and Cancer Sex Disparity Laboratory, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
| | - Wayne A. Phillips
- Gastrointestinal Cancer Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Department of Surgery (St. Vincent’s Hospital), The University of Melbourne, Parkville, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Monash University, Clayton, Victoria, Australia
| | - Nicholas J. Clemons
- Gastrointestinal Cancer Program, Cancer Research Division, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Sir Peter MacCallum Department of Oncology, The University of Melbourne, Parkville, Victoria, Australia
- Corresponding author. (N.J.C.); (K.M.F.)
| |
Collapse
|
27
|
Li FJ, Long HZ, Zhou ZW, Luo HY, Xu SG, Gao LC. System Xc−/GSH/GPX4 axis: An important antioxidant system for the ferroptosis in drug-resistant solid tumor therapy. Front Pharmacol 2022; 13:910292. [PMID: 36105219 PMCID: PMC9465090 DOI: 10.3389/fphar.2022.910292] [Citation(s) in RCA: 103] [Impact Index Per Article: 34.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2022] [Accepted: 08/03/2022] [Indexed: 11/13/2022] Open
Abstract
The activation of ferroptosis is a new effective way to treat drug-resistant solid tumors. Ferroptosis is an iron-mediated form of cell death caused by the accumulation of lipid peroxides. The intracellular imbalance between oxidant and antioxidant due to the abnormal expression of multiple redox active enzymes will promote the produce of reactive oxygen species (ROS). So far, a few pathways and regulators have been discovered to regulate ferroptosis. In particular, the cystine/glutamate antiporter (System Xc−), glutathione peroxidase 4 (GPX4) and glutathione (GSH) (System Xc−/GSH/GPX4 axis) plays a key role in preventing lipid peroxidation-mediated ferroptosis, because of which could be inhibited by blocking System Xc−/GSH/GPX4 axis. This review aims to present the current understanding of the mechanism of ferroptosis based on the System Xc−/GSH/GPX4 axis in the treatment of drug-resistant solid tumors.
Collapse
Affiliation(s)
- Feng-Jiao Li
- School of Pharmacy, University of South China, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Hui-Zhi Long
- School of Pharmacy, University of South China, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Zi-Wei Zhou
- School of Pharmacy, University of South China, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Hong-Yu Luo
- School of Pharmacy, University of South China, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Shuo-Guo Xu
- School of Pharmacy, University of South China, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Li-Chen Gao
- School of Pharmacy, University of South China, Phase I Clinical Trial Centre, The Affiliated Changsha Central Hospital, Hengyang Medical School, University of South China, Changsha, China
- Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
- *Correspondence: Li-Chen Gao,
| |
Collapse
|
28
|
Camponeschi F, Ciofi-Baffoni S, Calderone V, Banci L. Molecular Basis of Rare Diseases Associated to the Maturation of Mitochondrial [4Fe-4S]-Containing Proteins. Biomolecules 2022; 12:biom12071009. [PMID: 35883565 PMCID: PMC9313013 DOI: 10.3390/biom12071009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 07/15/2022] [Accepted: 07/19/2022] [Indexed: 02/04/2023] Open
Abstract
The importance of mitochondria in mammalian cells is widely known. Several biochemical reactions and pathways take place within mitochondria: among them, there are those involving the biogenesis of the iron–sulfur (Fe-S) clusters. The latter are evolutionarily conserved, ubiquitous inorganic cofactors, performing a variety of functions, such as electron transport, enzymatic catalysis, DNA maintenance, and gene expression regulation. The synthesis and distribution of Fe-S clusters are strictly controlled cellular processes that involve several mitochondrial proteins that specifically interact each other to form a complex machinery (Iron Sulfur Cluster assembly machinery, ISC machinery hereafter). This machinery ensures the correct assembly of both [2Fe-2S] and [4Fe-4S] clusters and their insertion in the mitochondrial target proteins. The present review provides a structural and molecular overview of the rare diseases associated with the genes encoding for the accessory proteins of the ISC machinery (i.e., GLRX5, ISCA1, ISCA2, IBA57, FDX2, BOLA3, IND1 and NFU1) involved in the assembly and insertion of [4Fe-4S] clusters in mitochondrial proteins. The disease-related missense mutations were mapped on the 3D structures of these accessory proteins or of their protein complexes, and the possible impact that these mutations have on their specific activity/function in the frame of the mitochondrial [4Fe-4S] protein biogenesis is described.
Collapse
Affiliation(s)
- Francesca Camponeschi
- Magnetic Resonance Center CERM, University of Florence, 50019 Sesto Fiorentino, Italy; (F.C.); (L.B.)
- Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), 50019 Sesto Fiorentino, Italy
| | - Simone Ciofi-Baffoni
- Magnetic Resonance Center CERM, University of Florence, 50019 Sesto Fiorentino, Italy; (F.C.); (L.B.)
- Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), 50019 Sesto Fiorentino, Italy
- Department of Chemistry, University of Florence, 50019 Sesto Fiorentino, Italy
- Correspondence: (S.C.-B.); (V.C.); Tel.: +39-055-4574192 (S.C.-B.); +39-055-4574276 (V.C.)
| | - Vito Calderone
- Magnetic Resonance Center CERM, University of Florence, 50019 Sesto Fiorentino, Italy; (F.C.); (L.B.)
- Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), 50019 Sesto Fiorentino, Italy
- Department of Chemistry, University of Florence, 50019 Sesto Fiorentino, Italy
- Correspondence: (S.C.-B.); (V.C.); Tel.: +39-055-4574192 (S.C.-B.); +39-055-4574276 (V.C.)
| | - Lucia Banci
- Magnetic Resonance Center CERM, University of Florence, 50019 Sesto Fiorentino, Italy; (F.C.); (L.B.)
- Consorzio Interuniversitario Risonanze Magnetiche di Metalloproteine (CIRMMP), 50019 Sesto Fiorentino, Italy
- Department of Chemistry, University of Florence, 50019 Sesto Fiorentino, Italy
| |
Collapse
|
29
|
Liu X, Zhang Y, Wu X, Xu F, Ma H, Wu M, Xia Y. Targeting Ferroptosis Pathway to Combat Therapy Resistance and Metastasis of Cancer. Front Pharmacol 2022; 13:909821. [PMID: 35847022 PMCID: PMC9280276 DOI: 10.3389/fphar.2022.909821] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Accepted: 05/20/2022] [Indexed: 01/18/2023] Open
Abstract
Ferroptosis is an iron-dependent regulated form of cell death caused by excessive lipid peroxidation. This form of cell death differed from known forms of cell death in morphological and biochemical features such as apoptosis, necrosis, and autophagy. Cancer cells require higher levels of iron to survive, which makes them highly susceptible to ferroptosis. Therefore, it was found to be closely related to the progression, treatment response, and metastasis of various cancer types. Numerous studies have found that the ferroptosis pathway is closely related to drug resistance and metastasis of cancer. Some cancer cells reduce their susceptibility to ferroptosis by downregulating the ferroptosis pathway, resulting in resistance to anticancer therapy. Induction of ferroptosis restores the sensitivity of drug-resistant cancer cells to standard treatments. Cancer cells that are resistant to conventional therapies or have a high propensity to metastasize might be particularly susceptible to ferroptosis. Some biological processes and cellular components, such as epithelial–mesenchymal transition (EMT) and noncoding RNAs, can influence cancer metastasis by regulating ferroptosis. Therefore, targeting ferroptosis may help suppress cancer metastasis. Those progresses revealed the importance of ferroptosis in cancer, In order to provide the detailed molecular mechanisms of ferroptosis in regulating therapy resistance and metastasis and strategies to overcome these barriers are not fully understood, we described the key molecular mechanisms of ferroptosis and its interaction with signaling pathways related to therapy resistance and metastasis. Furthermore, we summarized strategies for reversing resistance to targeted therapy, chemotherapy, radiotherapy, and immunotherapy and inhibiting cancer metastasis by modulating ferroptosis. Understanding the comprehensive regulatory mechanisms and signaling pathways of ferroptosis in cancer can provide new insights to enhance the efficacy of anticancer drugs, overcome drug resistance, and inhibit cancer metastasis.
Collapse
Affiliation(s)
- Xuan Liu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Yiqian Zhang
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Xuyi Wu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province/Rehabilitation Medicine Research Institute, Chengdu, China
| | - Fuyan Xu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Hongbo Ma
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Mengling Wu
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
| | - Yong Xia
- Department of Rehabilitation Medicine, West China Hospital, Sichuan University, Chengdu, China
- Key Laboratory of Rehabilitation Medicine in Sichuan Province/Rehabilitation Medicine Research Institute, Chengdu, China
- *Correspondence: Yong Xia,
| |
Collapse
|
30
|
Sanchez-Marco SB, Pierre G, Sharples P, Love S, Urankar K, Hilliard T, Lunt P, Churchill A, Aungraheeta R, Dallosso A, Evans J, Williams M, Majumdar A. Severe Congenital Myopathy and Neuropathy with Congenital Cataracts due to GFER Variant: A Neuropathological Study. JOURNAL OF PEDIATRIC NEUROLOGY 2022. [DOI: 10.1055/s-0042-1749671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
We describe the clinical, muscle and nerve biopsy, and genetic findings in a 10-year-old girl with a profound and rapid global regression. She presented during neonatal period with hypotonia, followed by weakness in the facial, bulbar, respiratory, and neck flexor muscles. She developed bilateral cataracts at 4 months of age and started to regress. Quadriceps muscle biopsy revealed extensive fiber atrophy but sparing of some, predominantly type 1, fibers. Sural nerve biopsy showed depletion of myelinated and unmyelinated fibers; most remaining myelinated fibers were of small caliber. Neuroimaging revealed global brain atrophy. Although the investigations indicated a multisystem disorder, extensive genetic and metabolic investigations were negative. She was tracheostomy- and ventilator-dependent for most of her life. The child died at 10 years of age. Further deoxyribonucleic acid analysis undertaken via whole genome sequencing revealed a novel pathogenic GFER sequence variant consistent with the patient's clinical presentation.
Collapse
Affiliation(s)
| | - Germaine Pierre
- Department of Paediatric Metabolic Medicine, University Hospitals Bristol NHS Foundation Trust, Bristol, United Kingdom
| | - Peta Sharples
- Department of Paediatric Neurology, University Hospitals Bristol NHS Foundation Trust, Bristol, United Kingdom
| | - Seth Love
- Department of Neuropathology, North Bristol Hospital NHS Foundation Trust, Bristol, United Kingdom
| | - Kathryn Urankar
- Department of Neuropathology, North Bristol Hospital NHS Foundation Trust, Bristol, United Kingdom
| | - Tom Hilliard
- Department of Paediatric Respiratory Medicine, Bristol Royal Hospital for Children, University Hospitals Bristol NHS Foundation Trust, Bristol, United Kingdom
| | - Peter Lunt
- South West Genomic Laboratory Hub, Bristol Genetics Laboratory, Southmead Hospital, Bristol, United Kingdom
| | - Amanda Churchill
- Department of Ophthalmology, University Hospitals Bristol NHS Foundation Trust, Bristol, United Kingdom
| | - Riyaad Aungraheeta
- South West Genomic Laboratory Hub, Bristol Genetics Laboratory, Southmead Hospital, Bristol, United Kingdom
| | - Anthony Dallosso
- South West Genomic Laboratory Hub, Bristol Genetics Laboratory, Southmead Hospital, Bristol, United Kingdom
| | - Julie Evans
- South West Genomic Laboratory Hub, Bristol Genetics Laboratory, Southmead Hospital, Bristol, United Kingdom
| | - Maggie Williams
- South West Genomic Laboratory Hub, Bristol Genetics Laboratory, Southmead Hospital, Bristol, United Kingdom
| | - Anirban Majumdar
- Department of Paediatric Neurology, University Hospitals Bristol NHS Foundation Trust, Bristol, United Kingdom
| |
Collapse
|
31
|
Bouzidi A, Charoute H, Charif M, Amalou G, Kandil M, Barakat A, Lenaers G. Clinical and genetic spectrums of 413 North African families with inherited retinal dystrophies and optic neuropathies. Orphanet J Rare Dis 2022; 17:197. [PMID: 35551639 PMCID: PMC9097391 DOI: 10.1186/s13023-022-02340-7] [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: 11/18/2021] [Accepted: 04/26/2022] [Indexed: 11/26/2022] Open
Abstract
Background Inherited retinal dystrophies (IRD) and optic neuropathies (ION) are the two major causes world-wide of early visual impairment, frequently leading to legal blindness. These two groups of pathologies are highly heterogeneous and require combined clinical and molecular diagnoses to be securely identified. Exact epidemiological studies are lacking in North Africa, and genetic studies of IRD and ION individuals are often limited to case reports or to some families that migrated to the rest of the world. In order to improve the knowledge of their clinical and genetic spectrums in North Africa, we reviewed published data, to illustrate the most prevalent pathologies, genes and mutations encountered in this geographical region, extending from Morocco to Egypt, comprising 200 million inhabitants. Main body We compiled data from 413 families with IRD or ION together with their available molecular diagnosis. The proportion of IRD represents 82.8% of index cases, while ION accounted for 17.8%. Non-syndromic IRD were more frequent than syndromic ones, with photoreceptor alterations being the main cause of non-syndromic IRD, represented by retinitis pigmentosa, Leber congenital amaurosis, and cone-rod dystrophies, while ciliopathies constitute the major part of syndromic-IRD, in which the Usher and Bardet Biedl syndromes occupy 41.2% and 31.1%, respectively. We identified 71 ION families, 84.5% with a syndromic presentation, while surprisingly, non-syndromic ION are scarcely reported, with only 11 families with autosomal recessive optic atrophies related to OPA7 and OPA10 variants, or with the mitochondrial related Leber ION. Overall, consanguinity is a major cause of these diseases within North African countries, as 76.1% of IRD and 78.8% of ION investigated families were consanguineous, explaining the high rate of autosomal recessive inheritance pattern compared to the dominant one. In addition, we identified many founder mutations in small endogamous communities. Short conclusion As both IRD and ION diseases constitute a real public health burden, their under-diagnosis in North Africa due to the absence of physicians trained to the identification of inherited ophthalmologic presentations, together with the scarcity of tools for the molecular diagnosis represent major political, economic and health challenges for the future, to first establish accurate clinical diagnoses and then treat patients with the emergent therapies. Supplementary Information The online version contains supplementary material available at 10.1186/s13023-022-02340-7.
Collapse
Affiliation(s)
- Aymane Bouzidi
- Equipe MitoLab, Unité MitoVasc, INSERM U1083, CHU d'Angers, CNRS 6015, Université d'Angers, 49933, Angers, France.,Genomics and Human Genetics Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco.,Team of Anthropogenetics and Biotechnologies, Faculty of Sciences, Chouaïb Doukkali University, Eljadida, Morocco
| | - Hicham Charoute
- Research Unit of Epidemiology, Biostatistics and Bioinformatics, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Majida Charif
- Genetics, and Immuno-Cell Therapy Team, Mohamed First University, Oujda, Morocco
| | - Ghita Amalou
- Equipe MitoLab, Unité MitoVasc, INSERM U1083, CHU d'Angers, CNRS 6015, Université d'Angers, 49933, Angers, France.,Genomics and Human Genetics Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco.,Team of Anthropogenetics and Biotechnologies, Faculty of Sciences, Chouaïb Doukkali University, Eljadida, Morocco
| | - Mostafa Kandil
- Team of Anthropogenetics and Biotechnologies, Faculty of Sciences, Chouaïb Doukkali University, Eljadida, Morocco
| | - Abdelhamid Barakat
- Genomics and Human Genetics Laboratory, Institut Pasteur du Maroc, Casablanca, Morocco
| | - Guy Lenaers
- Equipe MitoLab, Unité MitoVasc, INSERM U1083, CHU d'Angers, CNRS 6015, Université d'Angers, 49933, Angers, France. .,Service de Neurologie, CHU d'Angers, Angers, France.
| |
Collapse
|
32
|
Petry SF, Römer A, Rawat D, Brunner L, Lerch N, Zhou M, Grewal R, Sharifpanah F, Sauer H, Eckert GP, Linn T. Loss and Recovery of Glutaredoxin 5 Is Inducible by Diet in a Murine Model of Diabesity and Mediated by Free Fatty Acids In Vitro. Antioxidants (Basel) 2022; 11:antiox11040788. [PMID: 35453472 PMCID: PMC9025089 DOI: 10.3390/antiox11040788] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2022] [Revised: 03/24/2022] [Accepted: 04/14/2022] [Indexed: 02/05/2023] Open
Abstract
Free fatty acids (FFA), hyperglycemia, and inflammatory cytokines are major mediators of β-cell toxicity in type 2 diabetes mellitus, impairing mitochondrial metabolism. Glutaredoxin 5 (Glrx5) is a mitochondrial protein involved in the assembly of iron–sulfur clusters required for complexes of the respiratory chain. We have provided evidence that islet cells are deprived of Glrx5, correlating with impaired insulin secretion during diabetes in genetically obese mice. In this study, we induced diabesity in C57BL/6J mice in vivo by feeding the mice a high-fat diet (HFD) and modelled the diabetic metabolism in MIN6 cells through exposure to FFA, glucose, or inflammatory cytokines in vitro. qRT-PCR, ELISA, immunohisto-/cytochemistry, bioluminescence, and respirometry were employed to study Glrx5, insulin secretion, and mitochondrial biomarkers. The HFD induced a depletion of islet Glrx5 concomitant with an obese phenotype, elevated FFA in serum and reactive oxygen species in islets, and impaired glucose tolerance. Exposure of MIN6 cells to FFA led to a loss of Glrx5 in vitro. The FFA-induced depletion of Glrx5 coincided with significantly altered mitochondrial biomarkers. In summary, we provide evidence that Glrx5 is regulated by FFA in type 2 diabetes mellitus and is linked to mitochondrial dysfunction and blunted insulin secretion.
Collapse
Affiliation(s)
- Sebastian Friedrich Petry
- Clinical Research Unit, Medical Clinic and Polyclinic III, Center of Internal Medicine, Justus Liebig University, 35392 Giessen, Germany; (A.R.); (D.R.); (L.B.); (N.L.); (M.Z.); (T.L.)
- Correspondence: ; Tel.: +49-641-985-57010
| | - Axel Römer
- Clinical Research Unit, Medical Clinic and Polyclinic III, Center of Internal Medicine, Justus Liebig University, 35392 Giessen, Germany; (A.R.); (D.R.); (L.B.); (N.L.); (M.Z.); (T.L.)
| | - Divya Rawat
- Clinical Research Unit, Medical Clinic and Polyclinic III, Center of Internal Medicine, Justus Liebig University, 35392 Giessen, Germany; (A.R.); (D.R.); (L.B.); (N.L.); (M.Z.); (T.L.)
| | - Lara Brunner
- Clinical Research Unit, Medical Clinic and Polyclinic III, Center of Internal Medicine, Justus Liebig University, 35392 Giessen, Germany; (A.R.); (D.R.); (L.B.); (N.L.); (M.Z.); (T.L.)
| | - Nina Lerch
- Clinical Research Unit, Medical Clinic and Polyclinic III, Center of Internal Medicine, Justus Liebig University, 35392 Giessen, Germany; (A.R.); (D.R.); (L.B.); (N.L.); (M.Z.); (T.L.)
| | - Mengmeng Zhou
- Clinical Research Unit, Medical Clinic and Polyclinic III, Center of Internal Medicine, Justus Liebig University, 35392 Giessen, Germany; (A.R.); (D.R.); (L.B.); (N.L.); (M.Z.); (T.L.)
| | - Rekha Grewal
- Laboratory for Nutrition in Prevention & Therapy, Department of Nutritional Sciences, Justus Liebig University, 35392 Giessen, Germany; (R.G.); (G.P.E.)
| | - Fatemeh Sharifpanah
- Faculty of Medicine, Philipps University, 35037 Marburg, Germany;
- Cyntegrity Germany GmbH, 60438 Frankfurt, Germany
| | - Heinrich Sauer
- Department of Physiology, Faculty of Medicine, Justus Liebig University, 35392 Giessen, Germany;
| | - Gunter Peter Eckert
- Laboratory for Nutrition in Prevention & Therapy, Department of Nutritional Sciences, Justus Liebig University, 35392 Giessen, Germany; (R.G.); (G.P.E.)
| | - Thomas Linn
- Clinical Research Unit, Medical Clinic and Polyclinic III, Center of Internal Medicine, Justus Liebig University, 35392 Giessen, Germany; (A.R.); (D.R.); (L.B.); (N.L.); (M.Z.); (T.L.)
| |
Collapse
|
33
|
Onukwufor JO, Dirksen RT, Wojtovich AP. Iron Dysregulation in Mitochondrial Dysfunction and Alzheimer’s Disease. Antioxidants (Basel) 2022; 11:antiox11040692. [PMID: 35453377 PMCID: PMC9027385 DOI: 10.3390/antiox11040692] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 03/21/2022] [Accepted: 03/28/2022] [Indexed: 02/04/2023] Open
Abstract
Alzheimer’s disease (AD) is a devastating progressive neurodegenerative disease characterized by neuronal dysfunction, and decreased memory and cognitive function. Iron is critical for neuronal activity, neurotransmitter biosynthesis, and energy homeostasis. Iron accumulation occurs in AD and results in neuronal dysfunction through activation of multifactorial mechanisms. Mitochondria generate energy and iron is a key co-factor required for: (1) ATP production by the electron transport chain, (2) heme protein biosynthesis and (3) iron-sulfur cluster formation. Disruptions in iron homeostasis result in mitochondrial dysfunction and energetic failure. Ferroptosis, a non-apoptotic iron-dependent form of cell death mediated by uncontrolled accumulation of reactive oxygen species and lipid peroxidation, is associated with AD and other neurodegenerative diseases. AD pathogenesis is complex with multiple diverse interacting players including Aβ-plaque formation, phosphorylated tau, and redox stress. Unfortunately, clinical trials in AD based on targeting these canonical hallmarks have been largely unsuccessful. Here, we review evidence linking iron dysregulation to AD and the potential for targeting ferroptosis as a therapeutic intervention for AD.
Collapse
Affiliation(s)
- John O. Onukwufor
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642, USA; (R.T.D.); (A.P.W.)
- Correspondence:
| | - Robert T. Dirksen
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642, USA; (R.T.D.); (A.P.W.)
| | - Andrew P. Wojtovich
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642, USA; (R.T.D.); (A.P.W.)
- Department of Anesthesiology and Perioperative Medicine, University of Rochester Medical Center, Rochester, NY 14642, USA
| |
Collapse
|
34
|
Ling Y, Yang X, Zhang X, Guan F, Qi X, Dong W, Liu M, Ma J, Jiang X, Gao K, Li J, Chen W, Gao S, Gao X, Pan S, Wang J, Ma Y, Lu D, Zhang L. Myocardium-specific Isca1 knockout causes iron metabolism disorder and myocardial oncosis in rat. Life Sci 2022; 297:120485. [PMID: 35304126 DOI: 10.1016/j.lfs.2022.120485] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 02/26/2022] [Accepted: 03/10/2022] [Indexed: 11/16/2022]
Abstract
AIMS Multiple mitochondrial dysfunction (MMD) can lead to complex damage of mitochondrial structure and function, which then lead to the serious damage of various metabolic pathways including cerebral abnormalities. However, the effects of MMD on heart, a highly mitochondria-dependent tissue, are still unclear. In this study, we use iron-sulfur cluster assembly 1 (Isca1), which has been shown to cause MMD syndromes type 5 (MMDS5), to verify the above scientific question. MAIN METHODS We generated myocardium-specific Isca1 knockout rat (Isca1flox/flox/α-MHC-Cre) using CRISPR-Cas9 technology. Echocardiography, magnetic resonance imaging (MRI), histopathological examinations and molecular markers detection demonstrated phenotypic characteristics of our model. Immunoprecipitation, immunofluorescence co-location, mitochondrial activity, ATP generation and iron ions detection were used to verify the molecular mechanism. KEY FINDINGS This study was the first to verify the effects of Isca1 deficiency on cardiac development in vivo, that is cardiomyocytes suffer from mitochondria damage and iron metabolism disorder, which leads to myocardial oncosis and eventually heart failure and body death in rat. Furthermore, forward and reverse validation experiments demonstrated that six-transmembrane epithelial antigen of prostate 3 (STEAP3), a new interacting molecule for ISCA1, plays an important role in iron metabolism and energy generation impairment induced by ISCA1 deficiency. SIGNIFICANCE This result provides theoretical basis for understanding of MMDS pathogenesis, especially on heart development and the pathological process of heart diseases, and finally provides new clues for searching clinical therapeutic targets of MMDS.
Collapse
Affiliation(s)
- Yahao Ling
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China; Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China
| | - Xinlan Yang
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China; Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China
| | - Xu Zhang
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China; Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China; National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China
| | - Feifei Guan
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China; Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China; National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China
| | - Xiaolong Qi
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China; Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China; National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China
| | - Wei Dong
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China; Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China; National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China
| | - Mengdi Liu
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China; Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China
| | - Jiaxin Ma
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China; Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China
| | - Xiaoyu Jiang
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China; Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China
| | - Kai Gao
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China; Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China; National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China
| | - Jing Li
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China; Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China; National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China
| | - Wei Chen
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China; National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China
| | - Shan Gao
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China; National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China
| | - Xiang Gao
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China; National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China
| | - Shuo Pan
- Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China; National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China
| | - Jizheng Wang
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Disease, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
| | - Yuanwu Ma
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China; Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China; National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China
| | - Dan Lu
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China; Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China; National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China.
| | - Lianfeng Zhang
- Key Laboratory of Human Disease Comparative Medicine, National Health Commission of China (NHC), Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China; Beijing Engineering Research Center for Experimental Animal Models of Human Critical Diseases, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China; National Human Diseases Animal Model Resource Center, Institute of Laboratory Animal Science, Chinese Academy of Medical Sciences, Peking Union Medicine College, Beijing, China.
| |
Collapse
|
35
|
Monfort B, Want K, Gervason S, D’Autréaux B. Recent Advances in the Elucidation of Frataxin Biochemical Function Open Novel Perspectives for the Treatment of Friedreich’s Ataxia. Front Neurosci 2022; 16:838335. [PMID: 35310092 PMCID: PMC8924461 DOI: 10.3389/fnins.2022.838335] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/28/2022] [Indexed: 12/25/2022] Open
Abstract
Friedreich’s ataxia (FRDA) is the most prevalent autosomic recessive ataxia and is associated with a severe cardiac hypertrophy and less frequently diabetes. It is caused by mutations in the gene encoding frataxin (FXN), a small mitochondrial protein. The primary consequence is a defective expression of FXN, with basal protein levels decreased by 70–98%, which foremost affects the cerebellum, dorsal root ganglia, heart and liver. FXN is a mitochondrial protein involved in iron metabolism but its exact function has remained elusive and highly debated since its discovery. At the cellular level, FRDA is characterized by a general deficit in the biosynthesis of iron-sulfur (Fe-S) clusters and heme, iron accumulation and deposition in mitochondria, and sensitivity to oxidative stress. Based on these phenotypes and the proposed ability of FXN to bind iron, a role as an iron storage protein providing iron for Fe-S cluster and heme biosynthesis was initially proposed. However, this model was challenged by several other studies and it is now widely accepted that FXN functions primarily in Fe-S cluster biosynthesis, with iron accumulation, heme deficiency and oxidative stress sensitivity appearing later on as secondary defects. Nonetheless, the biochemical function of FXN in Fe-S cluster biosynthesis is still debated. Several roles have been proposed for FXN: iron chaperone, gate-keeper of detrimental Fe-S cluster biosynthesis, sulfide production stimulator and sulfur transfer accelerator. A picture is now emerging which points toward a unique function of FXN as an accelerator of a key step of sulfur transfer between two components of the Fe-S cluster biosynthetic complex. These findings should foster the development of new strategies for the treatment of FRDA. We will review here the latest discoveries on the biochemical function of frataxin and the implication for a potential therapeutic treatment of FRDA.
Collapse
|
36
|
Lin JF, Hu PS, Wang YY, Tan YT, Yu K, Liao K, Wu QN, Li T, Meng Q, Lin JZ, Liu ZX, Pu HY, Ju HQ, Xu RH, Qiu MZ. Phosphorylated NFS1 weakens oxaliplatin-based chemosensitivity of colorectal cancer by preventing PANoptosis. Signal Transduct Target Ther 2022; 7:54. [PMID: 35221331 PMCID: PMC8882671 DOI: 10.1038/s41392-022-00889-0] [Citation(s) in RCA: 137] [Impact Index Per Article: 45.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2021] [Revised: 12/28/2021] [Accepted: 01/05/2022] [Indexed: 12/16/2022] Open
Abstract
Metabolic enzymes have an indispensable role in metabolic reprogramming, and their aberrant expression or activity has been associated with chemosensitivity. Hence, targeting metabolic enzymes remains an attractive approach for treating tumors. However, the influence and regulation of cysteine desulfurase (NFS1), a rate-limiting enzyme in iron–sulfur (Fe–S) cluster biogenesis, in colorectal cancer (CRC) remain elusive. Here, using an in vivo metabolic enzyme gene-based clustered regularly interspaced short palindromic repeats (CRISPR)-Cas9 library screen, we revealed that loss of NFS1 significantly enhanced the sensitivity of CRC cells to oxaliplatin. In vitro and in vivo results showed that NFS1 deficiency synergizing with oxaliplatin triggered PANoptosis (apoptosis, necroptosis, pyroptosis, and ferroptosis) by increasing the intracellular levels of reactive oxygen species (ROS). Furthermore, oxaliplatin-based oxidative stress enhanced the phosphorylation level of serine residues of NFS1, which prevented PANoptosis in an S293 phosphorylation-dependent manner during oxaliplatin treatment. In addition, high expression of NFS1, transcriptionally regulated by MYC, was found in tumor tissues and was associated with poor survival and hyposensitivity to chemotherapy in patients with CRC. Overall, the findings of this study provided insights into the underlying mechanisms of NFS1 in oxaliplatin sensitivity and identified NFS1 inhibition as a promising strategy for improving the outcome of platinum-based chemotherapy in the treatment of CRC.
Collapse
|
37
|
A VDAC1-mediated NEET protein chain transfers [2Fe-2S] clusters between the mitochondria and the cytosol and impacts mitochondrial dynamics. Proc Natl Acad Sci U S A 2022; 119:2121491119. [PMID: 35135884 PMCID: PMC8851467 DOI: 10.1073/pnas.2121491119] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/04/2022] [Indexed: 12/11/2022] Open
Abstract
Here we address the important question of cross-talk between the mitochondria and cytosol. We show that the inner mitochondrial protein, MiNT, interacts with a protein on the outer mitochondrial membrane (mNT). This interaction occurs within the major outer membrane protein VDAC1. Inside the inner space of VDAC1, MiNT transfers its [2Fe-2S] clusters to mNT, which was shown to be a [2Fe-2S] cluster donor protein that donates its cluster(s) to apo-acceptor proteins residing in the cytosol. Hence, we suggest a pathway for transferring [2Fe-2S] clusters from inside the mitochondria to the cytosol. Mitochondrial inner NEET (MiNT) and the outer mitochondrial membrane (OMM) mitoNEET (mNT) proteins belong to the NEET protein family. This family plays a key role in mitochondrial labile iron and reactive oxygen species (ROS) homeostasis. NEET proteins contain labile [2Fe-2S] clusters which can be transferred to apo-acceptor proteins. In eukaryotes, the biogenesis of [2Fe-2S] clusters occurs within the mitochondria by the iron–sulfur cluster (ISC) system; the clusters are then transferred to [2Fe-2S] proteins within the mitochondria or exported to cytosolic proteins and the cytosolic iron–sulfur cluster assembly (CIA) system. The last step of export of the [2Fe-2S] is not yet fully characterized. Here we show that MiNT interacts with voltage-dependent anion channel 1 (VDAC1), a major OMM protein that connects the intermembrane space with the cytosol and participates in regulating the levels of different ions including mitochondrial labile iron (mLI). We further show that VDAC1 is mediating the interaction between MiNT and mNT, in which MiNT transfers its [2Fe-2S] clusters from inside the mitochondria to mNT that is facing the cytosol. This MiNT–VDAC1–mNT interaction is shown both experimentally and by computational calculations. Additionally, we show that modifying MiNT expression in breast cancer cells affects the dynamics of mitochondrial structure and morphology, mitochondrial function, and breast cancer tumor growth. Our findings reveal a pathway for the transfer of [2Fe-2S] clusters, which are assembled inside the mitochondria, to the cytosol.
Collapse
|
38
|
Montealegre S, Lebigot E, Debruge H, Romero N, Héron B, Gaignard P, Legendre A, Imbard A, Gobin S, Lacène E, Nusbaum P, Hubas A, Desguerre I, Servais A, Laforêt P, van Endert P, Authier FJ, Gitiaux C, de Lonlay P. FDX2 and ISCU Gene Variations Lead to Rhabdomyolysis With Distinct Severity and Iron Regulation. Neurol Genet 2022; 8:e648. [PMID: 35079622 PMCID: PMC8771665 DOI: 10.1212/nxg.0000000000000648] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2021] [Accepted: 10/18/2021] [Indexed: 01/04/2023]
Abstract
Background and Objectives To determine common clinical and biological traits in 2 individuals with
variants in ISCU and FDX2, displaying
severe and recurrent rhabdomyolyses and lactic acidosis. Methods We performed a clinical characterization of 2 distinct individuals with
biallelic ISCU or FDX2 variants from 2
separate families and a biological characterization with muscle and cells
from those patients. Results The individual with FDX2 variants was clinically more
affected than the individual with ISCU variants. Affected
FDX2 individual fibroblasts and myoblasts showed reduced oxygen consumption
rates and mitochondrial complex I and PDHc activities, associated with high
levels of blood FGF21. ISCU individual fibroblasts showed no oxidative
phosphorylation deficiency and moderate increase of blood FGF21 levels
relative to controls. The severity of the FDX2 individual was not due to
dysfunctional autophagy. Iron was excessively accumulated in ISCU-deficient
skeletal muscle, which was accompanied by a downregulation of
IRP1 and mitoferrin2 genes and an
upregulation of frataxin (FXN) gene expression. This
excessive iron accumulation was absent from FDX2 affected muscle and could
not be correlated with variable gene expression in muscle cells. Discussion We conclude that FDX2 and ISCU variants
result in a similar muscle phenotype, that differ in severity and skeletal
muscle iron accumulation. ISCU and FDX2 are not involved in mitochondrial
iron influx contrary to frataxin.
Collapse
Affiliation(s)
- Sebastian Montealegre
- Inserm U1151 (S.M., H.D., P.E., P.d.L.), Institut Necker Enfants-Malades, Paris; Reference Center of Inherited Metabolic Diseases (S.M., A.I., A.S., P.d.L.), Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, Paris University, Filière G2M; Biochemistry Laboratory (E. Lebigot, P.G.), Filière G2M, Bicêtre Hospital, APHP Paris Saclay, Le Kremlin Bicêtre; Sorbonne Universié (E. Lacène), UPMC, INSERM UMR974, Center for Research in Myology, Neuromuscular Morphology Unit, Myology Institute, AP-HP, East-Paris Reference Center of Neuromuscular Diseases, GHU Pitié-Salpêtrière; Neurology Unit (N.R., B.H.), Trousseau Hospital, APHP, Filière G2M; M3C-Necker (A.L.), Congenital and Pediatric Cardiology, Hôpital Universitaire Necker-Enfants Malades; Biochemistry Department (A.I.), Necker-Enfants-Malades University Hospital, APHP, Paris University; Genetics Department (S.G.), Necker-Enfants-Malades University Hospital, APHP; Genetics and Molecular Biology (P.N., A.H.), Laboratoire de Culture Cellulaire, Hôpital Cochin, Paris; Reference Center of Neuromuscular Diseases (I.D., C.G.), Necker-Enfants-Malades University Hospital, APHP, Filière Filnemus; Adult Nephrology & Transplantation (A.S.), Necker-Enfants-Malades University Hospital, APHP, Inserm U1163, Imagine Institute, Paris Descartes University; Department of Neurology (P.L.), Raymond-Poincaré Hospital, Garches, and Inserm U1179 Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux; and Reference Center for Neuromuscular Disorders (F.J.A., C.G.), Department de Pathologie, Henri Mondor Hospital, APHP, IMRB U955, Faculty of Medicine, Creteil, France
| | - Elise Lebigot
- Inserm U1151 (S.M., H.D., P.E., P.d.L.), Institut Necker Enfants-Malades, Paris; Reference Center of Inherited Metabolic Diseases (S.M., A.I., A.S., P.d.L.), Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, Paris University, Filière G2M; Biochemistry Laboratory (E. Lebigot, P.G.), Filière G2M, Bicêtre Hospital, APHP Paris Saclay, Le Kremlin Bicêtre; Sorbonne Universié (E. Lacène), UPMC, INSERM UMR974, Center for Research in Myology, Neuromuscular Morphology Unit, Myology Institute, AP-HP, East-Paris Reference Center of Neuromuscular Diseases, GHU Pitié-Salpêtrière; Neurology Unit (N.R., B.H.), Trousseau Hospital, APHP, Filière G2M; M3C-Necker (A.L.), Congenital and Pediatric Cardiology, Hôpital Universitaire Necker-Enfants Malades; Biochemistry Department (A.I.), Necker-Enfants-Malades University Hospital, APHP, Paris University; Genetics Department (S.G.), Necker-Enfants-Malades University Hospital, APHP; Genetics and Molecular Biology (P.N., A.H.), Laboratoire de Culture Cellulaire, Hôpital Cochin, Paris; Reference Center of Neuromuscular Diseases (I.D., C.G.), Necker-Enfants-Malades University Hospital, APHP, Filière Filnemus; Adult Nephrology & Transplantation (A.S.), Necker-Enfants-Malades University Hospital, APHP, Inserm U1163, Imagine Institute, Paris Descartes University; Department of Neurology (P.L.), Raymond-Poincaré Hospital, Garches, and Inserm U1179 Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux; and Reference Center for Neuromuscular Disorders (F.J.A., C.G.), Department de Pathologie, Henri Mondor Hospital, APHP, IMRB U955, Faculty of Medicine, Creteil, France
| | - Hugo Debruge
- Inserm U1151 (S.M., H.D., P.E., P.d.L.), Institut Necker Enfants-Malades, Paris; Reference Center of Inherited Metabolic Diseases (S.M., A.I., A.S., P.d.L.), Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, Paris University, Filière G2M; Biochemistry Laboratory (E. Lebigot, P.G.), Filière G2M, Bicêtre Hospital, APHP Paris Saclay, Le Kremlin Bicêtre; Sorbonne Universié (E. Lacène), UPMC, INSERM UMR974, Center for Research in Myology, Neuromuscular Morphology Unit, Myology Institute, AP-HP, East-Paris Reference Center of Neuromuscular Diseases, GHU Pitié-Salpêtrière; Neurology Unit (N.R., B.H.), Trousseau Hospital, APHP, Filière G2M; M3C-Necker (A.L.), Congenital and Pediatric Cardiology, Hôpital Universitaire Necker-Enfants Malades; Biochemistry Department (A.I.), Necker-Enfants-Malades University Hospital, APHP, Paris University; Genetics Department (S.G.), Necker-Enfants-Malades University Hospital, APHP; Genetics and Molecular Biology (P.N., A.H.), Laboratoire de Culture Cellulaire, Hôpital Cochin, Paris; Reference Center of Neuromuscular Diseases (I.D., C.G.), Necker-Enfants-Malades University Hospital, APHP, Filière Filnemus; Adult Nephrology & Transplantation (A.S.), Necker-Enfants-Malades University Hospital, APHP, Inserm U1163, Imagine Institute, Paris Descartes University; Department of Neurology (P.L.), Raymond-Poincaré Hospital, Garches, and Inserm U1179 Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux; and Reference Center for Neuromuscular Disorders (F.J.A., C.G.), Department de Pathologie, Henri Mondor Hospital, APHP, IMRB U955, Faculty of Medicine, Creteil, France
| | - Norma Romero
- Inserm U1151 (S.M., H.D., P.E., P.d.L.), Institut Necker Enfants-Malades, Paris; Reference Center of Inherited Metabolic Diseases (S.M., A.I., A.S., P.d.L.), Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, Paris University, Filière G2M; Biochemistry Laboratory (E. Lebigot, P.G.), Filière G2M, Bicêtre Hospital, APHP Paris Saclay, Le Kremlin Bicêtre; Sorbonne Universié (E. Lacène), UPMC, INSERM UMR974, Center for Research in Myology, Neuromuscular Morphology Unit, Myology Institute, AP-HP, East-Paris Reference Center of Neuromuscular Diseases, GHU Pitié-Salpêtrière; Neurology Unit (N.R., B.H.), Trousseau Hospital, APHP, Filière G2M; M3C-Necker (A.L.), Congenital and Pediatric Cardiology, Hôpital Universitaire Necker-Enfants Malades; Biochemistry Department (A.I.), Necker-Enfants-Malades University Hospital, APHP, Paris University; Genetics Department (S.G.), Necker-Enfants-Malades University Hospital, APHP; Genetics and Molecular Biology (P.N., A.H.), Laboratoire de Culture Cellulaire, Hôpital Cochin, Paris; Reference Center of Neuromuscular Diseases (I.D., C.G.), Necker-Enfants-Malades University Hospital, APHP, Filière Filnemus; Adult Nephrology & Transplantation (A.S.), Necker-Enfants-Malades University Hospital, APHP, Inserm U1163, Imagine Institute, Paris Descartes University; Department of Neurology (P.L.), Raymond-Poincaré Hospital, Garches, and Inserm U1179 Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux; and Reference Center for Neuromuscular Disorders (F.J.A., C.G.), Department de Pathologie, Henri Mondor Hospital, APHP, IMRB U955, Faculty of Medicine, Creteil, France
| | - Bénédicte Héron
- Inserm U1151 (S.M., H.D., P.E., P.d.L.), Institut Necker Enfants-Malades, Paris; Reference Center of Inherited Metabolic Diseases (S.M., A.I., A.S., P.d.L.), Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, Paris University, Filière G2M; Biochemistry Laboratory (E. Lebigot, P.G.), Filière G2M, Bicêtre Hospital, APHP Paris Saclay, Le Kremlin Bicêtre; Sorbonne Universié (E. Lacène), UPMC, INSERM UMR974, Center for Research in Myology, Neuromuscular Morphology Unit, Myology Institute, AP-HP, East-Paris Reference Center of Neuromuscular Diseases, GHU Pitié-Salpêtrière; Neurology Unit (N.R., B.H.), Trousseau Hospital, APHP, Filière G2M; M3C-Necker (A.L.), Congenital and Pediatric Cardiology, Hôpital Universitaire Necker-Enfants Malades; Biochemistry Department (A.I.), Necker-Enfants-Malades University Hospital, APHP, Paris University; Genetics Department (S.G.), Necker-Enfants-Malades University Hospital, APHP; Genetics and Molecular Biology (P.N., A.H.), Laboratoire de Culture Cellulaire, Hôpital Cochin, Paris; Reference Center of Neuromuscular Diseases (I.D., C.G.), Necker-Enfants-Malades University Hospital, APHP, Filière Filnemus; Adult Nephrology & Transplantation (A.S.), Necker-Enfants-Malades University Hospital, APHP, Inserm U1163, Imagine Institute, Paris Descartes University; Department of Neurology (P.L.), Raymond-Poincaré Hospital, Garches, and Inserm U1179 Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux; and Reference Center for Neuromuscular Disorders (F.J.A., C.G.), Department de Pathologie, Henri Mondor Hospital, APHP, IMRB U955, Faculty of Medicine, Creteil, France
| | - Pauline Gaignard
- Inserm U1151 (S.M., H.D., P.E., P.d.L.), Institut Necker Enfants-Malades, Paris; Reference Center of Inherited Metabolic Diseases (S.M., A.I., A.S., P.d.L.), Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, Paris University, Filière G2M; Biochemistry Laboratory (E. Lebigot, P.G.), Filière G2M, Bicêtre Hospital, APHP Paris Saclay, Le Kremlin Bicêtre; Sorbonne Universié (E. Lacène), UPMC, INSERM UMR974, Center for Research in Myology, Neuromuscular Morphology Unit, Myology Institute, AP-HP, East-Paris Reference Center of Neuromuscular Diseases, GHU Pitié-Salpêtrière; Neurology Unit (N.R., B.H.), Trousseau Hospital, APHP, Filière G2M; M3C-Necker (A.L.), Congenital and Pediatric Cardiology, Hôpital Universitaire Necker-Enfants Malades; Biochemistry Department (A.I.), Necker-Enfants-Malades University Hospital, APHP, Paris University; Genetics Department (S.G.), Necker-Enfants-Malades University Hospital, APHP; Genetics and Molecular Biology (P.N., A.H.), Laboratoire de Culture Cellulaire, Hôpital Cochin, Paris; Reference Center of Neuromuscular Diseases (I.D., C.G.), Necker-Enfants-Malades University Hospital, APHP, Filière Filnemus; Adult Nephrology & Transplantation (A.S.), Necker-Enfants-Malades University Hospital, APHP, Inserm U1163, Imagine Institute, Paris Descartes University; Department of Neurology (P.L.), Raymond-Poincaré Hospital, Garches, and Inserm U1179 Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux; and Reference Center for Neuromuscular Disorders (F.J.A., C.G.), Department de Pathologie, Henri Mondor Hospital, APHP, IMRB U955, Faculty of Medicine, Creteil, France
| | - Antoine Legendre
- Inserm U1151 (S.M., H.D., P.E., P.d.L.), Institut Necker Enfants-Malades, Paris; Reference Center of Inherited Metabolic Diseases (S.M., A.I., A.S., P.d.L.), Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, Paris University, Filière G2M; Biochemistry Laboratory (E. Lebigot, P.G.), Filière G2M, Bicêtre Hospital, APHP Paris Saclay, Le Kremlin Bicêtre; Sorbonne Universié (E. Lacène), UPMC, INSERM UMR974, Center for Research in Myology, Neuromuscular Morphology Unit, Myology Institute, AP-HP, East-Paris Reference Center of Neuromuscular Diseases, GHU Pitié-Salpêtrière; Neurology Unit (N.R., B.H.), Trousseau Hospital, APHP, Filière G2M; M3C-Necker (A.L.), Congenital and Pediatric Cardiology, Hôpital Universitaire Necker-Enfants Malades; Biochemistry Department (A.I.), Necker-Enfants-Malades University Hospital, APHP, Paris University; Genetics Department (S.G.), Necker-Enfants-Malades University Hospital, APHP; Genetics and Molecular Biology (P.N., A.H.), Laboratoire de Culture Cellulaire, Hôpital Cochin, Paris; Reference Center of Neuromuscular Diseases (I.D., C.G.), Necker-Enfants-Malades University Hospital, APHP, Filière Filnemus; Adult Nephrology & Transplantation (A.S.), Necker-Enfants-Malades University Hospital, APHP, Inserm U1163, Imagine Institute, Paris Descartes University; Department of Neurology (P.L.), Raymond-Poincaré Hospital, Garches, and Inserm U1179 Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux; and Reference Center for Neuromuscular Disorders (F.J.A., C.G.), Department de Pathologie, Henri Mondor Hospital, APHP, IMRB U955, Faculty of Medicine, Creteil, France
| | - Apolline Imbard
- Inserm U1151 (S.M., H.D., P.E., P.d.L.), Institut Necker Enfants-Malades, Paris; Reference Center of Inherited Metabolic Diseases (S.M., A.I., A.S., P.d.L.), Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, Paris University, Filière G2M; Biochemistry Laboratory (E. Lebigot, P.G.), Filière G2M, Bicêtre Hospital, APHP Paris Saclay, Le Kremlin Bicêtre; Sorbonne Universié (E. Lacène), UPMC, INSERM UMR974, Center for Research in Myology, Neuromuscular Morphology Unit, Myology Institute, AP-HP, East-Paris Reference Center of Neuromuscular Diseases, GHU Pitié-Salpêtrière; Neurology Unit (N.R., B.H.), Trousseau Hospital, APHP, Filière G2M; M3C-Necker (A.L.), Congenital and Pediatric Cardiology, Hôpital Universitaire Necker-Enfants Malades; Biochemistry Department (A.I.), Necker-Enfants-Malades University Hospital, APHP, Paris University; Genetics Department (S.G.), Necker-Enfants-Malades University Hospital, APHP; Genetics and Molecular Biology (P.N., A.H.), Laboratoire de Culture Cellulaire, Hôpital Cochin, Paris; Reference Center of Neuromuscular Diseases (I.D., C.G.), Necker-Enfants-Malades University Hospital, APHP, Filière Filnemus; Adult Nephrology & Transplantation (A.S.), Necker-Enfants-Malades University Hospital, APHP, Inserm U1163, Imagine Institute, Paris Descartes University; Department of Neurology (P.L.), Raymond-Poincaré Hospital, Garches, and Inserm U1179 Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux; and Reference Center for Neuromuscular Disorders (F.J.A., C.G.), Department de Pathologie, Henri Mondor Hospital, APHP, IMRB U955, Faculty of Medicine, Creteil, France
| | - Stéphanie Gobin
- Inserm U1151 (S.M., H.D., P.E., P.d.L.), Institut Necker Enfants-Malades, Paris; Reference Center of Inherited Metabolic Diseases (S.M., A.I., A.S., P.d.L.), Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, Paris University, Filière G2M; Biochemistry Laboratory (E. Lebigot, P.G.), Filière G2M, Bicêtre Hospital, APHP Paris Saclay, Le Kremlin Bicêtre; Sorbonne Universié (E. Lacène), UPMC, INSERM UMR974, Center for Research in Myology, Neuromuscular Morphology Unit, Myology Institute, AP-HP, East-Paris Reference Center of Neuromuscular Diseases, GHU Pitié-Salpêtrière; Neurology Unit (N.R., B.H.), Trousseau Hospital, APHP, Filière G2M; M3C-Necker (A.L.), Congenital and Pediatric Cardiology, Hôpital Universitaire Necker-Enfants Malades; Biochemistry Department (A.I.), Necker-Enfants-Malades University Hospital, APHP, Paris University; Genetics Department (S.G.), Necker-Enfants-Malades University Hospital, APHP; Genetics and Molecular Biology (P.N., A.H.), Laboratoire de Culture Cellulaire, Hôpital Cochin, Paris; Reference Center of Neuromuscular Diseases (I.D., C.G.), Necker-Enfants-Malades University Hospital, APHP, Filière Filnemus; Adult Nephrology & Transplantation (A.S.), Necker-Enfants-Malades University Hospital, APHP, Inserm U1163, Imagine Institute, Paris Descartes University; Department of Neurology (P.L.), Raymond-Poincaré Hospital, Garches, and Inserm U1179 Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux; and Reference Center for Neuromuscular Disorders (F.J.A., C.G.), Department de Pathologie, Henri Mondor Hospital, APHP, IMRB U955, Faculty of Medicine, Creteil, France
| | - Emmanuelle Lacène
- Inserm U1151 (S.M., H.D., P.E., P.d.L.), Institut Necker Enfants-Malades, Paris; Reference Center of Inherited Metabolic Diseases (S.M., A.I., A.S., P.d.L.), Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, Paris University, Filière G2M; Biochemistry Laboratory (E. Lebigot, P.G.), Filière G2M, Bicêtre Hospital, APHP Paris Saclay, Le Kremlin Bicêtre; Sorbonne Universié (E. Lacène), UPMC, INSERM UMR974, Center for Research in Myology, Neuromuscular Morphology Unit, Myology Institute, AP-HP, East-Paris Reference Center of Neuromuscular Diseases, GHU Pitié-Salpêtrière; Neurology Unit (N.R., B.H.), Trousseau Hospital, APHP, Filière G2M; M3C-Necker (A.L.), Congenital and Pediatric Cardiology, Hôpital Universitaire Necker-Enfants Malades; Biochemistry Department (A.I.), Necker-Enfants-Malades University Hospital, APHP, Paris University; Genetics Department (S.G.), Necker-Enfants-Malades University Hospital, APHP; Genetics and Molecular Biology (P.N., A.H.), Laboratoire de Culture Cellulaire, Hôpital Cochin, Paris; Reference Center of Neuromuscular Diseases (I.D., C.G.), Necker-Enfants-Malades University Hospital, APHP, Filière Filnemus; Adult Nephrology & Transplantation (A.S.), Necker-Enfants-Malades University Hospital, APHP, Inserm U1163, Imagine Institute, Paris Descartes University; Department of Neurology (P.L.), Raymond-Poincaré Hospital, Garches, and Inserm U1179 Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux; and Reference Center for Neuromuscular Disorders (F.J.A., C.G.), Department de Pathologie, Henri Mondor Hospital, APHP, IMRB U955, Faculty of Medicine, Creteil, France
| | - Patrick Nusbaum
- Inserm U1151 (S.M., H.D., P.E., P.d.L.), Institut Necker Enfants-Malades, Paris; Reference Center of Inherited Metabolic Diseases (S.M., A.I., A.S., P.d.L.), Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, Paris University, Filière G2M; Biochemistry Laboratory (E. Lebigot, P.G.), Filière G2M, Bicêtre Hospital, APHP Paris Saclay, Le Kremlin Bicêtre; Sorbonne Universié (E. Lacène), UPMC, INSERM UMR974, Center for Research in Myology, Neuromuscular Morphology Unit, Myology Institute, AP-HP, East-Paris Reference Center of Neuromuscular Diseases, GHU Pitié-Salpêtrière; Neurology Unit (N.R., B.H.), Trousseau Hospital, APHP, Filière G2M; M3C-Necker (A.L.), Congenital and Pediatric Cardiology, Hôpital Universitaire Necker-Enfants Malades; Biochemistry Department (A.I.), Necker-Enfants-Malades University Hospital, APHP, Paris University; Genetics Department (S.G.), Necker-Enfants-Malades University Hospital, APHP; Genetics and Molecular Biology (P.N., A.H.), Laboratoire de Culture Cellulaire, Hôpital Cochin, Paris; Reference Center of Neuromuscular Diseases (I.D., C.G.), Necker-Enfants-Malades University Hospital, APHP, Filière Filnemus; Adult Nephrology & Transplantation (A.S.), Necker-Enfants-Malades University Hospital, APHP, Inserm U1163, Imagine Institute, Paris Descartes University; Department of Neurology (P.L.), Raymond-Poincaré Hospital, Garches, and Inserm U1179 Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux; and Reference Center for Neuromuscular Disorders (F.J.A., C.G.), Department de Pathologie, Henri Mondor Hospital, APHP, IMRB U955, Faculty of Medicine, Creteil, France
| | - Arnaud Hubas
- Inserm U1151 (S.M., H.D., P.E., P.d.L.), Institut Necker Enfants-Malades, Paris; Reference Center of Inherited Metabolic Diseases (S.M., A.I., A.S., P.d.L.), Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, Paris University, Filière G2M; Biochemistry Laboratory (E. Lebigot, P.G.), Filière G2M, Bicêtre Hospital, APHP Paris Saclay, Le Kremlin Bicêtre; Sorbonne Universié (E. Lacène), UPMC, INSERM UMR974, Center for Research in Myology, Neuromuscular Morphology Unit, Myology Institute, AP-HP, East-Paris Reference Center of Neuromuscular Diseases, GHU Pitié-Salpêtrière; Neurology Unit (N.R., B.H.), Trousseau Hospital, APHP, Filière G2M; M3C-Necker (A.L.), Congenital and Pediatric Cardiology, Hôpital Universitaire Necker-Enfants Malades; Biochemistry Department (A.I.), Necker-Enfants-Malades University Hospital, APHP, Paris University; Genetics Department (S.G.), Necker-Enfants-Malades University Hospital, APHP; Genetics and Molecular Biology (P.N., A.H.), Laboratoire de Culture Cellulaire, Hôpital Cochin, Paris; Reference Center of Neuromuscular Diseases (I.D., C.G.), Necker-Enfants-Malades University Hospital, APHP, Filière Filnemus; Adult Nephrology & Transplantation (A.S.), Necker-Enfants-Malades University Hospital, APHP, Inserm U1163, Imagine Institute, Paris Descartes University; Department of Neurology (P.L.), Raymond-Poincaré Hospital, Garches, and Inserm U1179 Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux; and Reference Center for Neuromuscular Disorders (F.J.A., C.G.), Department de Pathologie, Henri Mondor Hospital, APHP, IMRB U955, Faculty of Medicine, Creteil, France
| | - Isabelle Desguerre
- Inserm U1151 (S.M., H.D., P.E., P.d.L.), Institut Necker Enfants-Malades, Paris; Reference Center of Inherited Metabolic Diseases (S.M., A.I., A.S., P.d.L.), Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, Paris University, Filière G2M; Biochemistry Laboratory (E. Lebigot, P.G.), Filière G2M, Bicêtre Hospital, APHP Paris Saclay, Le Kremlin Bicêtre; Sorbonne Universié (E. Lacène), UPMC, INSERM UMR974, Center for Research in Myology, Neuromuscular Morphology Unit, Myology Institute, AP-HP, East-Paris Reference Center of Neuromuscular Diseases, GHU Pitié-Salpêtrière; Neurology Unit (N.R., B.H.), Trousseau Hospital, APHP, Filière G2M; M3C-Necker (A.L.), Congenital and Pediatric Cardiology, Hôpital Universitaire Necker-Enfants Malades; Biochemistry Department (A.I.), Necker-Enfants-Malades University Hospital, APHP, Paris University; Genetics Department (S.G.), Necker-Enfants-Malades University Hospital, APHP; Genetics and Molecular Biology (P.N., A.H.), Laboratoire de Culture Cellulaire, Hôpital Cochin, Paris; Reference Center of Neuromuscular Diseases (I.D., C.G.), Necker-Enfants-Malades University Hospital, APHP, Filière Filnemus; Adult Nephrology & Transplantation (A.S.), Necker-Enfants-Malades University Hospital, APHP, Inserm U1163, Imagine Institute, Paris Descartes University; Department of Neurology (P.L.), Raymond-Poincaré Hospital, Garches, and Inserm U1179 Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux; and Reference Center for Neuromuscular Disorders (F.J.A., C.G.), Department de Pathologie, Henri Mondor Hospital, APHP, IMRB U955, Faculty of Medicine, Creteil, France
| | - Aude Servais
- Inserm U1151 (S.M., H.D., P.E., P.d.L.), Institut Necker Enfants-Malades, Paris; Reference Center of Inherited Metabolic Diseases (S.M., A.I., A.S., P.d.L.), Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, Paris University, Filière G2M; Biochemistry Laboratory (E. Lebigot, P.G.), Filière G2M, Bicêtre Hospital, APHP Paris Saclay, Le Kremlin Bicêtre; Sorbonne Universié (E. Lacène), UPMC, INSERM UMR974, Center for Research in Myology, Neuromuscular Morphology Unit, Myology Institute, AP-HP, East-Paris Reference Center of Neuromuscular Diseases, GHU Pitié-Salpêtrière; Neurology Unit (N.R., B.H.), Trousseau Hospital, APHP, Filière G2M; M3C-Necker (A.L.), Congenital and Pediatric Cardiology, Hôpital Universitaire Necker-Enfants Malades; Biochemistry Department (A.I.), Necker-Enfants-Malades University Hospital, APHP, Paris University; Genetics Department (S.G.), Necker-Enfants-Malades University Hospital, APHP; Genetics and Molecular Biology (P.N., A.H.), Laboratoire de Culture Cellulaire, Hôpital Cochin, Paris; Reference Center of Neuromuscular Diseases (I.D., C.G.), Necker-Enfants-Malades University Hospital, APHP, Filière Filnemus; Adult Nephrology & Transplantation (A.S.), Necker-Enfants-Malades University Hospital, APHP, Inserm U1163, Imagine Institute, Paris Descartes University; Department of Neurology (P.L.), Raymond-Poincaré Hospital, Garches, and Inserm U1179 Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux; and Reference Center for Neuromuscular Disorders (F.J.A., C.G.), Department de Pathologie, Henri Mondor Hospital, APHP, IMRB U955, Faculty of Medicine, Creteil, France
| | - Pascal Laforêt
- Inserm U1151 (S.M., H.D., P.E., P.d.L.), Institut Necker Enfants-Malades, Paris; Reference Center of Inherited Metabolic Diseases (S.M., A.I., A.S., P.d.L.), Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, Paris University, Filière G2M; Biochemistry Laboratory (E. Lebigot, P.G.), Filière G2M, Bicêtre Hospital, APHP Paris Saclay, Le Kremlin Bicêtre; Sorbonne Universié (E. Lacène), UPMC, INSERM UMR974, Center for Research in Myology, Neuromuscular Morphology Unit, Myology Institute, AP-HP, East-Paris Reference Center of Neuromuscular Diseases, GHU Pitié-Salpêtrière; Neurology Unit (N.R., B.H.), Trousseau Hospital, APHP, Filière G2M; M3C-Necker (A.L.), Congenital and Pediatric Cardiology, Hôpital Universitaire Necker-Enfants Malades; Biochemistry Department (A.I.), Necker-Enfants-Malades University Hospital, APHP, Paris University; Genetics Department (S.G.), Necker-Enfants-Malades University Hospital, APHP; Genetics and Molecular Biology (P.N., A.H.), Laboratoire de Culture Cellulaire, Hôpital Cochin, Paris; Reference Center of Neuromuscular Diseases (I.D., C.G.), Necker-Enfants-Malades University Hospital, APHP, Filière Filnemus; Adult Nephrology & Transplantation (A.S.), Necker-Enfants-Malades University Hospital, APHP, Inserm U1163, Imagine Institute, Paris Descartes University; Department of Neurology (P.L.), Raymond-Poincaré Hospital, Garches, and Inserm U1179 Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux; and Reference Center for Neuromuscular Disorders (F.J.A., C.G.), Department de Pathologie, Henri Mondor Hospital, APHP, IMRB U955, Faculty of Medicine, Creteil, France
| | - Peter van Endert
- Inserm U1151 (S.M., H.D., P.E., P.d.L.), Institut Necker Enfants-Malades, Paris; Reference Center of Inherited Metabolic Diseases (S.M., A.I., A.S., P.d.L.), Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, Paris University, Filière G2M; Biochemistry Laboratory (E. Lebigot, P.G.), Filière G2M, Bicêtre Hospital, APHP Paris Saclay, Le Kremlin Bicêtre; Sorbonne Universié (E. Lacène), UPMC, INSERM UMR974, Center for Research in Myology, Neuromuscular Morphology Unit, Myology Institute, AP-HP, East-Paris Reference Center of Neuromuscular Diseases, GHU Pitié-Salpêtrière; Neurology Unit (N.R., B.H.), Trousseau Hospital, APHP, Filière G2M; M3C-Necker (A.L.), Congenital and Pediatric Cardiology, Hôpital Universitaire Necker-Enfants Malades; Biochemistry Department (A.I.), Necker-Enfants-Malades University Hospital, APHP, Paris University; Genetics Department (S.G.), Necker-Enfants-Malades University Hospital, APHP; Genetics and Molecular Biology (P.N., A.H.), Laboratoire de Culture Cellulaire, Hôpital Cochin, Paris; Reference Center of Neuromuscular Diseases (I.D., C.G.), Necker-Enfants-Malades University Hospital, APHP, Filière Filnemus; Adult Nephrology & Transplantation (A.S.), Necker-Enfants-Malades University Hospital, APHP, Inserm U1163, Imagine Institute, Paris Descartes University; Department of Neurology (P.L.), Raymond-Poincaré Hospital, Garches, and Inserm U1179 Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux; and Reference Center for Neuromuscular Disorders (F.J.A., C.G.), Department de Pathologie, Henri Mondor Hospital, APHP, IMRB U955, Faculty of Medicine, Creteil, France
| | - François Jérome Authier
- Inserm U1151 (S.M., H.D., P.E., P.d.L.), Institut Necker Enfants-Malades, Paris; Reference Center of Inherited Metabolic Diseases (S.M., A.I., A.S., P.d.L.), Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, Paris University, Filière G2M; Biochemistry Laboratory (E. Lebigot, P.G.), Filière G2M, Bicêtre Hospital, APHP Paris Saclay, Le Kremlin Bicêtre; Sorbonne Universié (E. Lacène), UPMC, INSERM UMR974, Center for Research in Myology, Neuromuscular Morphology Unit, Myology Institute, AP-HP, East-Paris Reference Center of Neuromuscular Diseases, GHU Pitié-Salpêtrière; Neurology Unit (N.R., B.H.), Trousseau Hospital, APHP, Filière G2M; M3C-Necker (A.L.), Congenital and Pediatric Cardiology, Hôpital Universitaire Necker-Enfants Malades; Biochemistry Department (A.I.), Necker-Enfants-Malades University Hospital, APHP, Paris University; Genetics Department (S.G.), Necker-Enfants-Malades University Hospital, APHP; Genetics and Molecular Biology (P.N., A.H.), Laboratoire de Culture Cellulaire, Hôpital Cochin, Paris; Reference Center of Neuromuscular Diseases (I.D., C.G.), Necker-Enfants-Malades University Hospital, APHP, Filière Filnemus; Adult Nephrology & Transplantation (A.S.), Necker-Enfants-Malades University Hospital, APHP, Inserm U1163, Imagine Institute, Paris Descartes University; Department of Neurology (P.L.), Raymond-Poincaré Hospital, Garches, and Inserm U1179 Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux; and Reference Center for Neuromuscular Disorders (F.J.A., C.G.), Department de Pathologie, Henri Mondor Hospital, APHP, IMRB U955, Faculty of Medicine, Creteil, France
| | - Cyril Gitiaux
- Inserm U1151 (S.M., H.D., P.E., P.d.L.), Institut Necker Enfants-Malades, Paris; Reference Center of Inherited Metabolic Diseases (S.M., A.I., A.S., P.d.L.), Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, Paris University, Filière G2M; Biochemistry Laboratory (E. Lebigot, P.G.), Filière G2M, Bicêtre Hospital, APHP Paris Saclay, Le Kremlin Bicêtre; Sorbonne Universié (E. Lacène), UPMC, INSERM UMR974, Center for Research in Myology, Neuromuscular Morphology Unit, Myology Institute, AP-HP, East-Paris Reference Center of Neuromuscular Diseases, GHU Pitié-Salpêtrière; Neurology Unit (N.R., B.H.), Trousseau Hospital, APHP, Filière G2M; M3C-Necker (A.L.), Congenital and Pediatric Cardiology, Hôpital Universitaire Necker-Enfants Malades; Biochemistry Department (A.I.), Necker-Enfants-Malades University Hospital, APHP, Paris University; Genetics Department (S.G.), Necker-Enfants-Malades University Hospital, APHP; Genetics and Molecular Biology (P.N., A.H.), Laboratoire de Culture Cellulaire, Hôpital Cochin, Paris; Reference Center of Neuromuscular Diseases (I.D., C.G.), Necker-Enfants-Malades University Hospital, APHP, Filière Filnemus; Adult Nephrology & Transplantation (A.S.), Necker-Enfants-Malades University Hospital, APHP, Inserm U1163, Imagine Institute, Paris Descartes University; Department of Neurology (P.L.), Raymond-Poincaré Hospital, Garches, and Inserm U1179 Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux; and Reference Center for Neuromuscular Disorders (F.J.A., C.G.), Department de Pathologie, Henri Mondor Hospital, APHP, IMRB U955, Faculty of Medicine, Creteil, France
| | - Pascale de Lonlay
- Inserm U1151 (S.M., H.D., P.E., P.d.L.), Institut Necker Enfants-Malades, Paris; Reference Center of Inherited Metabolic Diseases (S.M., A.I., A.S., P.d.L.), Necker-Enfants-Malades University Hospital, APHP, Imagine Institute, Paris University, Filière G2M; Biochemistry Laboratory (E. Lebigot, P.G.), Filière G2M, Bicêtre Hospital, APHP Paris Saclay, Le Kremlin Bicêtre; Sorbonne Universié (E. Lacène), UPMC, INSERM UMR974, Center for Research in Myology, Neuromuscular Morphology Unit, Myology Institute, AP-HP, East-Paris Reference Center of Neuromuscular Diseases, GHU Pitié-Salpêtrière; Neurology Unit (N.R., B.H.), Trousseau Hospital, APHP, Filière G2M; M3C-Necker (A.L.), Congenital and Pediatric Cardiology, Hôpital Universitaire Necker-Enfants Malades; Biochemistry Department (A.I.), Necker-Enfants-Malades University Hospital, APHP, Paris University; Genetics Department (S.G.), Necker-Enfants-Malades University Hospital, APHP; Genetics and Molecular Biology (P.N., A.H.), Laboratoire de Culture Cellulaire, Hôpital Cochin, Paris; Reference Center of Neuromuscular Diseases (I.D., C.G.), Necker-Enfants-Malades University Hospital, APHP, Filière Filnemus; Adult Nephrology & Transplantation (A.S.), Necker-Enfants-Malades University Hospital, APHP, Inserm U1163, Imagine Institute, Paris Descartes University; Department of Neurology (P.L.), Raymond-Poincaré Hospital, Garches, and Inserm U1179 Versailles Saint-Quentin-en-Yvelines University, Montigny-le-Bretonneux; and Reference Center for Neuromuscular Disorders (F.J.A., C.G.), Department de Pathologie, Henri Mondor Hospital, APHP, IMRB U955, Faculty of Medicine, Creteil, France
| |
Collapse
|
39
|
Fujishiro T, Nakamura R, Kunichika K, Takahashi Y. Structural diversity of cysteine desulfurases involved in iron-sulfur cluster biosynthesis. Biophys Physicobiol 2022; 19:1-18. [PMID: 35377584 PMCID: PMC8918507 DOI: 10.2142/biophysico.bppb-v19.0001] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 02/02/2022] [Indexed: 12/04/2022] Open
Abstract
Cysteine desulfurases are pyridoxal-5'-phosphate (PLP)-dependent enzymes that mobilize sulfur derived from the l-cysteine substrate to the partner sulfur acceptor proteins. Three cysteine desulfurases, IscS, NifS, and SufS, have been identified in ISC, NIF, and SUF/SUF-like systems for iron-sulfur (Fe-S) cluster biosynthesis, respectively. These cysteine desulfurases have been investigated over decades, providing insights into shared/distinct catalytic processes based on two types of enzymes (type I: IscS and NifS, type II: SufS). This review summarizes the insights into the structural/functional varieties of bacterial and eukaryotic cysteine desulfurases involved in Fe-S cluster biosynthetic systems. In addition, an inactive cysteine desulfurase IscS paralog, which contains pyridoxamine-5'-phosphate (PMP), instead of PLP, is also described to account for its hypothetical function in Fe-S cluster biosynthesis involving this paralog. The structural basis for cysteine desulfurase functions will be a stepping stone towards understanding the diversity and evolution of Fe-S cluster biosynthesis.
Collapse
Affiliation(s)
- Takashi Fujishiro
- Department of Biochemistry and Moecular Biology, Graduate School of Science and Engineering, Saitama University
| | - Ryosuke Nakamura
- Department of Biochemistry and Moecular Biology, Graduate School of Science and Engineering, Saitama University
| | - Kouhei Kunichika
- Department of Biochemistry and Moecular Biology, Graduate School of Science and Engineering, Saitama University
| | - Yasuhiro Takahashi
- Department of Biochemistry and Moecular Biology, Graduate School of Science and Engineering, Saitama University
| |
Collapse
|
40
|
Spectroscopic and functional characterization of the [2Fe-2S] scaffold protein Nfu from Synechocystis PCC6803. Biochimie 2022; 192:51-62. [PMID: 34582998 PMCID: PMC8724361 DOI: 10.1016/j.biochi.2021.09.013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2021] [Revised: 09/20/2021] [Accepted: 09/21/2021] [Indexed: 01/03/2023]
Abstract
Iron-sulfur clusters are ubiquitous cofactors required for various essential metabolic processes. Conservation of proteins required for their biosynthesis and trafficking allows for simple bacteria to be used as models to aid in exploring these complex pathways in higher organisms. Cyanobacteria are among the most investigated organisms for these processes, as they are unicellular and can survive under photoautotrophic and heterotrophic conditions. Herein, we report the potential role of Synechocystis PCC6803 NifU (now named SyNfu) as the principal scaffold protein required for iron-sulfur cluster biosynthesis in that organism. SyNfu is a well-folded protein with distinct secondary structural elements, as evidenced by circular dichroism and a well-dispersed pattern of 1H-15N HSQC NMR peaks, and readily reconstitutes as a [2Fe-2S] dimeric protein complex. Cluster exchange experiments show that glutathione can extract the cluster from holo-SyNfu, but the transfer is unidirectional. We also confirm the ability of SyNfu to transfer cluster to both human ferredoxin 1 and ferredoxin 2, while also demonstrating the capacity to deliver cluster to both monothiol glutaredoxin 3 and dithiol glutaredoxin 2. This evidence supports the hypothesis that SyNfu indeed serves as the main scaffold protein in Synechocystis, as it has been shown to be the only protein required for viability in the absence of photoautotrophic conditions. Similar to other NFU-type cluster donors and other scaffold and carrier proteins, such as ISCU, SyNfu is shown by DSC to be structurally less stable than regular protein donors, while retaining a relatively well-defined tertiary structure as represented by 1H-15N HSQC NMR experiments.
Collapse
|
41
|
Aggarwal A, Pillai NR, Billington CJ, Schema L, Berry SA. Rare presentation of FDX2-related disorder and untargeted global metabolomics findings. Am J Med Genet A 2021; 188:1239-1244. [PMID: 34905296 DOI: 10.1002/ajmg.a.62608] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 11/19/2021] [Accepted: 11/28/2021] [Indexed: 11/11/2022]
Abstract
We present the case of a 20-year-old male with a history of myopathy and multiple episodes of rhabdomyolysis, and lactic acidosis. He needed hemodialysis for severe rhabdomyolysis-related acute renal failure at the time of initial presentation (age 10 years). Exome sequencing detected a homozygous likely pathogenic variant in FDX2 (c.12G>T, p.M4I). The FDX2 gene encodes a mitochondrial protein, ferredoxin 2, that is involved in the biogenesis of Fe-S clusters. Biallelic pathogenic variants in FDX2 have previously been associated with episodic mitochondrial myopathy with or without optic atrophy and reversible leukoencephalopathy. Only two cases with FDX2-related rhabdomyolysis as a predominant feature have been reported in medical literature. Here, we report a third patient with FDX2-related recurrent, severe episodes of rhabdomyolysis and lactic acidosis. He does not have optic atrophy or leukoencephalopathy. This is the oldest patient reported with FDX2-related disorder and he has significantly elevated CK during episodes of rhabdomyolysis. In addition, we describe untargeted global metabolomic findings during an episode of metabolic decompensation, shedding light on the biochemical pathway perturbation associated with this ultra-rare genetic disorder.
Collapse
Affiliation(s)
- Anjali Aggarwal
- Division of Genetics and Metabolism, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Nishitha R Pillai
- Division of Genetics and Metabolism, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Charles J Billington
- Division of Genetics and Metabolism, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| | - Lynn Schema
- M-Health Fairview, Minneapolis, Minnesota, USA
| | - Susan A Berry
- Division of Genetics and Metabolism, Department of Pediatrics, University of Minnesota, Minneapolis, Minnesota, USA
| |
Collapse
|
42
|
Ma Y, Elmhadi M, Wang C, Zhang H, Wang H. Dietary supplementation of thiamine down-regulates the expression of mitophagy and endoplasmic reticulum stress-related genes in the rumen epithelium of goats during high-concentrate diet feeding. ITALIAN JOURNAL OF ANIMAL SCIENCE 2021. [DOI: 10.1080/1828051x.2021.1985944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Yi Ma
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, P. R. China
| | - Mawda Elmhadi
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, P. R. China
| | - Chao Wang
- Queen Elizabeth II Medical Centre, School of Biomedical Sciences, The University of Western Australia, Nedlands, Australia
| | - Hao Zhang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, P. R. China
| | - Hongrong Wang
- Laboratory of Metabolic Manipulation of Herbivorous Animal Nutrition, College of Animal Science and Technology, Yangzhou University, Yangzhou, P. R. China
| |
Collapse
|
43
|
Ling Y, Ma J, Qi X, Zhang X, Kong Q, Guan F, Dong W, Chen W, Gao S, Gao X, Pan S, Ma Y, Lu D, Zhang L. Novel rat model of multiple mitochondrial dysfunction syndromes (MMDS) complicated with cardiomyopathy. Animal Model Exp Med 2021; 4:381-390. [PMID: 34977489 PMCID: PMC8690978 DOI: 10.1002/ame2.12193] [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: 08/03/2021] [Revised: 10/19/2021] [Accepted: 11/11/2021] [Indexed: 01/27/2023] Open
Abstract
Background Multiple mitochondrial dysfunction syndromes (MMDS) presents as complex mitochondrial damage, thus impairing a variety of metabolic pathways. Heart dysplasia has been reported in MMDS patients; however, the specific clinical symptoms and pathogenesis remain unclear. More urgently, there is a lack of an animal model to aid research. Therefore, we selected a reported MMDS causal gene, Isca1, and established an animal model of MMDS complicated with cardiac dysplasia. Methods The myocardium-specific Isca1 knockout heterozygote (Isca1 HET) rat was obtained by crossing the Isca1 conditional knockout (Isca1 cKO) rat with the α myosin heavy chain Cre (α-MHC-Cre) rat. Cardiac development characteristics were determined by ECG, blood pressure measurement, echocardiography and histopathological analysis. The responsiveness to pathological stimuli were observed through adriamycin treatment. Mitochondria and metabolism disorder were determined by activity analysis of mitochondrial respiratory chain complex and ATP production in myocardium. Results ISCA1 expression in myocardium exhibited a semizygous effect. Isca1 HET rats exhibited dilated cardiomyopathy characteristics, including thin-walled ventricles, larger chambers, cardiac dysfunction and myocardium fibrosis. Downregulated ISCA1 led to deteriorating cardiac pathological processes at the global and organizational levels. Meanwhile, HET rats exhibited typical MMDS characteristics, including damaged mitochondrial morphology and enzyme activity for mitochondrial respiratory chain complexes Ⅰ, Ⅱ and Ⅳ, and impaired ATP production. Conclusion We have established a rat model of MMDS complicated with cardiomyopathy, it can also be used as model of myocardial energy metabolism dysfunction and mitochondrial cardiomyopathy. This model can be applied to the study of the mechanism of energy metabolism in cardiovascular diseases, as well as research and development of drugs.
Collapse
Affiliation(s)
- Yahao Ling
- Key Laboratory of Human Disease Comparative MedicineNational Health Commission of China (NHC)Institute of Laboratory Animal SciencePeking Union Medical CollegeChinese Academy of Medical SciencesBeijingChina
| | - Jiaxin Ma
- Beijing Engineering Research Center for Experimental Animal Models of Human DiseasesInstitute of Laboratory Animal SciencePeking Union Medical CollegeChinese Academy of Medical SciencesBeijingChina
| | - Xiaolong Qi
- Beijing Engineering Research Center for Experimental Animal Models of Human DiseasesInstitute of Laboratory Animal SciencePeking Union Medical CollegeChinese Academy of Medical SciencesBeijingChina
| | - Xu Zhang
- Beijing Engineering Research Center for Experimental Animal Models of Human DiseasesInstitute of Laboratory Animal SciencePeking Union Medical CollegeChinese Academy of Medical SciencesBeijingChina
| | - Qi Kong
- Beijing Engineering Research Center for Experimental Animal Models of Human DiseasesInstitute of Laboratory Animal SciencePeking Union Medical CollegeChinese Academy of Medical SciencesBeijingChina
| | - Feifei Guan
- Beijing Engineering Research Center for Experimental Animal Models of Human DiseasesInstitute of Laboratory Animal SciencePeking Union Medical CollegeChinese Academy of Medical SciencesBeijingChina
| | - Wei Dong
- Key Laboratory of Human Disease Comparative MedicineNational Health Commission of China (NHC)Institute of Laboratory Animal SciencePeking Union Medical CollegeChinese Academy of Medical SciencesBeijingChina
| | - Wei Chen
- Key Laboratory of Human Disease Comparative MedicineNational Health Commission of China (NHC)Institute of Laboratory Animal SciencePeking Union Medical CollegeChinese Academy of Medical SciencesBeijingChina
| | - Shan Gao
- Key Laboratory of Human Disease Comparative MedicineNational Health Commission of China (NHC)Institute of Laboratory Animal SciencePeking Union Medical CollegeChinese Academy of Medical SciencesBeijingChina
| | - Xiang Gao
- Key Laboratory of Human Disease Comparative MedicineNational Health Commission of China (NHC)Institute of Laboratory Animal SciencePeking Union Medical CollegeChinese Academy of Medical SciencesBeijingChina
| | - Shuo Pan
- Beijing Engineering Research Center for Experimental Animal Models of Human DiseasesInstitute of Laboratory Animal SciencePeking Union Medical CollegeChinese Academy of Medical SciencesBeijingChina
| | - Yuanwu Ma
- Key Laboratory of Human Disease Comparative MedicineNational Health Commission of China (NHC)Institute of Laboratory Animal SciencePeking Union Medical CollegeChinese Academy of Medical SciencesBeijingChina
| | - Dan Lu
- Beijing Engineering Research Center for Experimental Animal Models of Human DiseasesInstitute of Laboratory Animal SciencePeking Union Medical CollegeChinese Academy of Medical SciencesBeijingChina
| | - Lianfeng Zhang
- Key Laboratory of Human Disease Comparative MedicineNational Health Commission of China (NHC)Institute of Laboratory Animal SciencePeking Union Medical CollegeChinese Academy of Medical SciencesBeijingChina
| |
Collapse
|
44
|
Hutchens JA, Johnson TR, Payne RM. Myocardial Perfusion Reserve in Children with Friedreich Ataxia. Pediatr Cardiol 2021; 42:1834-1840. [PMID: 34245318 DOI: 10.1007/s00246-021-02675-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Accepted: 06/30/2021] [Indexed: 11/27/2022]
Abstract
Children with Friedreich's ataxia (FA) are at risk of perioperative morbidity and mortality from severe unpredictable heart failure. There is currently no clear way of identifying patients at highest risk. We used myocardial perfusion reserve (MPR), an MRI technique used to assess the maximal myocardial blood flow above baseline, to help determine potential surgical risk in FA subjects. In total, seven children with genetically confirmed FA, ages 8-17 years, underwent MPR stress testing using regadenoson. Six of the seven demonstrated impaired endocardial perfusion during coronary hyperemia. The same six were also found to have evidence of ongoing myocardial damage as illustrated by cardiac troponin I leak (range 0.04-0.17 ng/mL, normal < 0.03 ng/mL). None of the patients had a reduced ejection fraction (range 59-74%) or elevated insulin level (range 2.46-14.23 mCU/mL). This retrospective study shows that children with FA develop MPR defects early in the disease process. It also suggests MPR may be a sensitive tool to evaluate underlying cardiac compromise and could be of use in directing surgical management decisions in children with FA.
Collapse
Affiliation(s)
| | - Tiffanie R Johnson
- Indiana University School of Medicine, Indianapolis, IN, USA.,Division of Pediatric Cardiology, Riley Hospital for Children, Indiana University School of Medicine, 1044 West Walnut St, Room R4-302b, Indianapolis, IN, 46202, USA
| | - R Mark Payne
- Indiana University School of Medicine, Indianapolis, IN, USA. .,Division of Pediatric Cardiology, Riley Hospital for Children, Indiana University School of Medicine, 1044 West Walnut St, Room R4-302b, Indianapolis, IN, 46202, USA. .,Herman B Wells Center for Pediatric Research, Indianapolis, IN, USA.
| |
Collapse
|
45
|
Freibert SA, Boniecki MT, Stümpfig C, Schulz V, Krapoth N, Winge DR, Mühlenhoff U, Stehling O, Cygler M, Lill R. N-terminal tyrosine of ISCU2 triggers [2Fe-2S] cluster synthesis by ISCU2 dimerization. Nat Commun 2021; 12:6902. [PMID: 34824239 PMCID: PMC8617193 DOI: 10.1038/s41467-021-27122-w] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2021] [Accepted: 11/03/2021] [Indexed: 12/03/2022] Open
Abstract
Synthesis of iron-sulfur (Fe/S) clusters in living cells requires scaffold proteins for both facile synthesis and subsequent transfer of clusters to target apoproteins. The human mitochondrial ISCU2 scaffold protein is part of the core ISC (iron-sulfur cluster assembly) complex that synthesizes a bridging [2Fe-2S] cluster on dimeric ISCU2. Initial iron and sulfur loading onto monomeric ISCU2 have been elucidated biochemically, yet subsequent [2Fe-2S] cluster formation and dimerization of ISCU2 is mechanistically ill-defined. Our structural, biochemical and cell biological experiments now identify a crucial function of the universally conserved N-terminal Tyr35 of ISCU2 for these late reactions. Mixing two, per se non-functional ISCU2 mutant proteins with oppositely charged Asp35 and Lys35 residues, both bound to different cysteine desulfurase complexes NFS1-ISD11-ACP, restores wild-type ISCU2 maturation demonstrating that ionic forces can replace native Tyr-Tyr interactions during dimerization-induced [2Fe-2S] cluster formation. Our studies define the essential mechanistic role of Tyr35 in the reaction cycle of de novo mitochondrial [2Fe-2S] cluster synthesis. [2Fe-2S] protein cofactors are essential for life and are synthesized on ISCU2 scaffolds. Here, the authors show that hydrophobic interaction of two conserved N-terminal tyrosines induces ISCU2 dimerization and concomitant [2Fe-2S] cluster synthesis.
Collapse
Affiliation(s)
- Sven-A Freibert
- Institut für Zytobiologie im Zentrum SYNMIKRO, Philipps-Universität Marburg, Karl-von-Frisch-Str. 14, 35032, Marburg, Germany.,Core Facility 'Protein Biochemistry and Spectroscopy', Karl-von-Frisch-Str. 14, 35032, Marburg, Germany
| | - Michal T Boniecki
- Department of Biochemistry, Microbiology & Immunology, University of Saskatchewan, 107 Wiggins Rd, Saskatoon, SK, S7N 5E5, Canada
| | - Claudia Stümpfig
- Institut für Zytobiologie im Zentrum SYNMIKRO, Philipps-Universität Marburg, Karl-von-Frisch-Str. 14, 35032, Marburg, Germany
| | - Vinzent Schulz
- Institut für Zytobiologie im Zentrum SYNMIKRO, Philipps-Universität Marburg, Karl-von-Frisch-Str. 14, 35032, Marburg, Germany
| | - Nils Krapoth
- Institut für Zytobiologie im Zentrum SYNMIKRO, Philipps-Universität Marburg, Karl-von-Frisch-Str. 14, 35032, Marburg, Germany
| | - Dennis R Winge
- Institut für Zytobiologie im Zentrum SYNMIKRO, Philipps-Universität Marburg, Karl-von-Frisch-Str. 14, 35032, Marburg, Germany.,Department of Medicine, University of Utah Health Sciences Center, Salt Lake City, UT, USA
| | - Ulrich Mühlenhoff
- Institut für Zytobiologie im Zentrum SYNMIKRO, Philipps-Universität Marburg, Karl-von-Frisch-Str. 14, 35032, Marburg, Germany
| | - Oliver Stehling
- Institut für Zytobiologie im Zentrum SYNMIKRO, Philipps-Universität Marburg, Karl-von-Frisch-Str. 14, 35032, Marburg, Germany.,Core Facility 'Protein Biochemistry and Spectroscopy', Karl-von-Frisch-Str. 14, 35032, Marburg, Germany
| | - Miroslaw Cygler
- Department of Biochemistry, Microbiology & Immunology, University of Saskatchewan, 107 Wiggins Rd, Saskatoon, SK, S7N 5E5, Canada.
| | - Roland Lill
- Institut für Zytobiologie im Zentrum SYNMIKRO, Philipps-Universität Marburg, Karl-von-Frisch-Str. 14, 35032, Marburg, Germany. .,Core Facility 'Protein Biochemistry and Spectroscopy', Karl-von-Frisch-Str. 14, 35032, Marburg, Germany. .,LOEWE Zentrum für Synthetische Mikrobiologie SynMikro, Hans-Meerwein-Str., 35043, Marburg, Germany.
| |
Collapse
|
46
|
Song SJ, Hong Y, Xu K, Zhang C. Novel biallelic compound heterozygous mutations in FDXR cause optic atrophy in a young female patient: a case report. Int J Ophthalmol 2021; 14:1796-1798. [PMID: 34804873 DOI: 10.18240/ijo.2021.11.22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Accepted: 03/03/2021] [Indexed: 11/23/2022] Open
Affiliation(s)
- Si-Jia Song
- Department of Ophthalmology, Peking University Third Hospital, Beijing 100191, China.,Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing 100191, China
| | - Ying Hong
- Department of Ophthalmology, Peking University Third Hospital, Beijing 100191, China.,Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing 100191, China
| | - Ke Xu
- Department of Ophthalmology, Peking University Third Hospital, Beijing 100191, China.,Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing 100191, China
| | - Chun Zhang
- Department of Ophthalmology, Peking University Third Hospital, Beijing 100191, China.,Beijing Key Laboratory of Restoration of Damaged Ocular Nerve, Peking University Third Hospital, Beijing 100191, China
| |
Collapse
|
47
|
Ferroptosis: A New Promising Target for Lung Cancer Therapy. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:8457521. [PMID: 34616505 PMCID: PMC8487823 DOI: 10.1155/2021/8457521] [Citation(s) in RCA: 31] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/16/2021] [Accepted: 09/15/2021] [Indexed: 12/21/2022]
Abstract
Ferroptosis is a new type of regulatory cell death that differs from autophagy, apoptosis, necrosis, and pyroptosis; it is caused primarily by the accumulation of iron and lipid peroxides in the cell. Studies have shown that many classical signaling pathways and biological processes are involved in the process of ferroptosis. In recent years, investigations have revealed that ferroptosis plays a crucial role in the progression of tumors, especially lung cancer. In particular, inducing ferroptosis in cells can inhibit the growth of tumor cells, thereby reversing tumorigenesis. In this review, we summarize the characteristics of ferroptosis from its underlying basis and role in lung cancer and provide possible applications for it in lung cancer therapies.
Collapse
|
48
|
Wang JQ, Wu ZX, Yang Y, Teng QX, Li YD, Lei ZN, Jani KA, Kaushal N, Chen ZS. ATP-binding cassette (ABC) transporters in cancer: A review of recent updates. J Evid Based Med 2021; 14:232-256. [PMID: 34388310 DOI: 10.1111/jebm.12434] [Citation(s) in RCA: 61] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Accepted: 04/27/2021] [Indexed: 02/06/2023]
Abstract
The ATP-binding cassette (ABC) transporter superfamily is one of the largest membrane protein families existing in wide spectrum of organisms from prokaryotes to human. ABC transporters are also known as efflux pumps because they mediate the cross-membrane transportation of various endo- and xenobiotic molecules energized by ATP hydrolysis. Therefore, ABC transporters have been considered closely to multidrug resistance (MDR) in cancer, where the efflux of structurally distinct chemotherapeutic drugs causes reduced itherapeutic efficacy. Besides, ABC transporters also play other critical biological roles in cancer such as signal transduction. During the past decades, extensive efforts have been made in understanding the structure-function relationship, transportation profile of ABC transporters, as well as the possibility to overcome MDR via targeting these transporters. In this review, we discuss the most recent knowledge regarding ABC transporters and cancer drug resistance in order to provide insights for the development of more effective therapies.
Collapse
Affiliation(s)
- Jing-Quan Wang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Zhuo-Xun Wu
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Yuqi Yang
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Qiu-Xu Teng
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Yi-Dong Li
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Zi-Ning Lei
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
- School of Public Health, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Khushboo A Jani
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Neeraj Kaushal
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| | - Zhe-Sheng Chen
- Department of Pharmaceutical Sciences, College of Pharmacy and Health Sciences, St. John's University, Queens, New York
| |
Collapse
|
49
|
Mühlenhoff U, Braymer JJ, Christ S, Rietzschel N, Uzarska MA, Weiler BD, Lill R. Glutaredoxins and iron-sulfur protein biogenesis at the interface of redox biology and iron metabolism. Biol Chem 2021; 401:1407-1428. [PMID: 33031050 DOI: 10.1515/hsz-2020-0237] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Accepted: 09/21/2020] [Indexed: 11/15/2022]
Abstract
The physiological roles of the intracellular iron and redox regulatory systems are intimately linked. Iron is an essential trace element for most organisms, yet elevated cellular iron levels are a potent generator and amplifier of reactive oxygen species and redox stress. Proteins binding iron or iron-sulfur (Fe/S) clusters, are particularly sensitive to oxidative damage and require protection from the cellular oxidative stress protection systems. In addition, key components of these systems, most prominently glutathione and monothiol glutaredoxins are involved in the biogenesis of cellular Fe/S proteins. In this review, we address the biochemical role of glutathione and glutaredoxins in cellular Fe/S protein assembly in eukaryotic cells. We also summarize the recent developments in the role of cytosolic glutaredoxins in iron metabolism, in particular the regulation of fungal iron homeostasis. Finally, we discuss recent insights into the interplay of the cellular thiol redox balance and oxygen with that of Fe/S protein biogenesis in eukaryotes.
Collapse
Affiliation(s)
- Ulrich Mühlenhoff
- Institut für Zytobiologie, Philipps-Universität Marburg, Robert-Koch-Str. 6, D-35032Marburg, Germany.,SYNMIKRO Center for Synthetic Microbiology, Philipps-Universität Marburg, Hans-Meerwein-Str., D-35043Marburg, Germany
| | - Joseph J Braymer
- Institut für Zytobiologie, Philipps-Universität Marburg, Robert-Koch-Str. 6, D-35032Marburg, Germany.,SYNMIKRO Center for Synthetic Microbiology, Philipps-Universität Marburg, Hans-Meerwein-Str., D-35043Marburg, Germany
| | - Stefan Christ
- Institut für Zytobiologie, Philipps-Universität Marburg, Robert-Koch-Str. 6, D-35032Marburg, Germany
| | - Nicole Rietzschel
- Institut für Zytobiologie, Philipps-Universität Marburg, Robert-Koch-Str. 6, D-35032Marburg, Germany
| | - Marta A Uzarska
- Institut für Zytobiologie, Philipps-Universität Marburg, Robert-Koch-Str. 6, D-35032Marburg, Germany.,Intercollegiate Faculty of Biotechnology, University of Gdansk, Abrahama 58, 80-307Gdansk, Poland
| | - Benjamin D Weiler
- Institut für Zytobiologie, Philipps-Universität Marburg, Robert-Koch-Str. 6, D-35032Marburg, Germany
| | - Roland Lill
- Institut für Zytobiologie, Philipps-Universität Marburg, Robert-Koch-Str. 6, D-35032Marburg, Germany.,SYNMIKRO Center for Synthetic Microbiology, Philipps-Universität Marburg, Hans-Meerwein-Str., D-35043Marburg, Germany
| |
Collapse
|
50
|
Lill R. From the discovery to molecular understanding of cellular iron-sulfur protein biogenesis. Biol Chem 2021; 401:855-876. [PMID: 32229650 DOI: 10.1515/hsz-2020-0117] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Accepted: 03/10/2020] [Indexed: 12/23/2022]
Abstract
Protein cofactors often are the business ends of proteins, and are either synthesized inside cells or are taken up from the nutrition. A cofactor that strictly needs to be synthesized by cells is the iron-sulfur (Fe/S) cluster. This evolutionary ancient compound performs numerous biochemical functions including electron transfer, catalysis, sulfur mobilization, regulation and protein stabilization. Since the discovery of eukaryotic Fe/S protein biogenesis two decades ago, more than 30 biogenesis factors have been identified in mitochondria and cytosol. They support the synthesis, trafficking and target-specific insertion of Fe/S clusters. In this review, I first summarize what led to the initial discovery of Fe/S protein biogenesis in yeast. I then discuss the function and localization of Fe/S proteins in (non-green) eukaryotes. The major part of the review provides a detailed synopsis of the three major steps of mitochondrial Fe/S protein biogenesis, i.e. the de novo synthesis of a [2Fe-2S] cluster on a scaffold protein, the Hsp70 chaperone-mediated transfer of the cluster and integration into [2Fe-2S] recipient apoproteins, and the reductive fusion of [2Fe-2S] to [4Fe-4S] clusters and their subsequent assembly into target apoproteins. Finally, I summarize the current knowledge of the mechanisms underlying the maturation of cytosolic and nuclear Fe/S proteins.
Collapse
Affiliation(s)
- Roland Lill
- Institut für Zytobiologie, Philipps-Universität Marburg, Robert-Koch-Str. 6, D-35032 Marburg, Germany.,SYNMIKRO Center for Synthetic Microbiology, Philipps-Universität Marburg, Hans-Meerwein-Str., D-35043 Marburg, Germany
| |
Collapse
|