1
|
Hashemi A, Liu MR, Chan JZ, Berdeklis AN, Cocco AD, Tomczewski MV, Strathdee D, Stark KD, Duncan RE. Plaat1 deficiency reduces cardiac cardiolipin content and impairs exercise tolerance. J Lipid Res 2025:100822. [PMID: 40345663 DOI: 10.1016/j.jlr.2025.100822] [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: 06/27/2024] [Revised: 04/12/2025] [Accepted: 05/05/2025] [Indexed: 05/11/2025] Open
Abstract
Phospholipase A and acyltransferase 1 (PLAAT1) catalyzes O-transacylase, N-transacylase and phospholipase A1/2 reactions. We have demonstrated that PLAAT1 has O-transacylase activity in vitro using phosphatidylcholine as an acyl donor, and monolysocardiolipin (MLCL) as an acyl acceptor, generating cardiolipin. However, a role for PLAAT1 in cardiolipin regulation in vivo has not yet been reported. We generated Plaat1-deficient (Plaat1-/-) mice and studied males and females for gross morphological differences, food intakes, respiratory gas exchange, total energy expenditure, and voluntary activity. We also evaluated cardiac cardiolipin contents, levels of mitochondrial proteins, and exercise capacity. Sex-matched Plaat1-/- mice had highly similar body weights to their wildtype (Wt) littermates, although male Plaat1-/- mice ate less. Male and female Plaat1-/- hearts were 14.2% and 10.6% smaller, respectively. Cardiac cardiolipin levels were ∼1/3 lower in male and female Plaat1-/- mice compared to their sex-matched Wt littermates, largely due to lower cardiolipin linoleate. Levels of the mitochondrial protein SDHA were 13.8% and 16.3% lower in male and female Plaat1-/- mice, respectively. Both male and female Plaat1-/- mice had significantly lower oxygen consumption, carbon dioxide production, and total energy expenditure, and male Plaat1-/- mice had lower rearing activity than their sex-matched Wt littermates. While other measures of voluntary activity, including locomotion and ambulation did not differ significantly between genotypes, both males and females had reduced exercise tolerance. This work demonstrates a critical role for PLAAT1 in cardiac cardiolipin content and the regulation of energy metabolism and exercise tolerance in vivo.
Collapse
Affiliation(s)
- Ashkan Hashemi
- University of Waterloo, Faculty of Health, Department of Kinesiology and Health Sciences, 200 University Ave W., Waterloo, ON, N2L 3G1 Canada.
| | - Ming Rong Liu
- University of Waterloo, Faculty of Health, Department of Kinesiology and Health Sciences, 200 University Ave W., Waterloo, ON, N2L 3G1 Canada.
| | - John Z Chan
- University of Waterloo, Faculty of Health, Department of Kinesiology and Health Sciences, 200 University Ave W., Waterloo, ON, N2L 3G1 Canada.
| | - Antonia N Berdeklis
- University of Waterloo, Faculty of Health, Department of Kinesiology and Health Sciences, 200 University Ave W., Waterloo, ON, N2L 3G1 Canada.
| | - Alex D Cocco
- University of Waterloo, Faculty of Health, Department of Kinesiology and Health Sciences, 200 University Ave W., Waterloo, ON, N2L 3G1 Canada.
| | - Michelle V Tomczewski
- University of Waterloo, Faculty of Health, Department of Kinesiology and Health Sciences, 200 University Ave W., Waterloo, ON, N2L 3G1 Canada.
| | - Douglas Strathdee
- Transgenic Technology Laboratory, Cancer Research UK Beatson Institute, Switchback Road, Glasgow, G61 1BD, Scotland.
| | - Ken D Stark
- University of Waterloo, Faculty of Health, Department of Kinesiology and Health Sciences, 200 University Ave W., Waterloo, ON, N2L 3G1 Canada.
| | - Robin E Duncan
- University of Waterloo, Faculty of Health, Department of Kinesiology and Health Sciences, 200 University Ave W., Waterloo, ON, N2L 3G1 Canada.
| |
Collapse
|
2
|
Zhang X, Zhang B, Tao Z, Liang J. Mitochondrial disease and epilepsy in children. Front Neurol 2025; 15:1499876. [PMID: 39850733 PMCID: PMC11754068 DOI: 10.3389/fneur.2024.1499876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 12/26/2024] [Indexed: 01/25/2025] Open
Abstract
Mitochondria is the cell's powerhouse. Mitochondrial disease refers to a group of clinically heterogeneous disorders caused by dysfunction in the mitochondrial respiratory chain, often due to mutations in mitochondrial DNA (mtDNA) or nuclear DNA (nDNA) that encodes mitochondrial proteins. This dysfunction can lead to a variety of clinical phenotypes, particularly affecting organs with high energy demands, such as the brain and muscles. Epilepsy is a prevalent neurological disorder in children and is also a frequent manifestation of mitochondrial disease. The exact mechanisms underlying epilepsy in mitochondrial disease remain unclear and are thought to involve multiple contributing factors. This review explores common mitochondrial diseases associated with epilepsy, focusing on their prevalence, seizure types, EEG features, therapeutic strategies, and outcomes. It also summarizes the relationship between the molecular genetics of mitochondrial respiratory chain components and the development of epilepsy.
Collapse
Affiliation(s)
- Xuan Zhang
- Department of Pediatric Neurology, Children's Medical Center, First Hospital of Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Pediatric Neurology, Changchun, China
- Neuromedical Center, First Hospital of Jilin University, Changchun, China
| | - Bo Zhang
- Department of Pediatric Neurology, Children's Medical Center, First Hospital of Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Pediatric Neurology, Changchun, China
- Neuromedical Center, First Hospital of Jilin University, Changchun, China
| | - Zhiming Tao
- Department of Pediatric Neurology, Children's Medical Center, First Hospital of Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Pediatric Neurology, Changchun, China
- Neuromedical Center, First Hospital of Jilin University, Changchun, China
| | - Jianmin Liang
- Department of Pediatric Neurology, Children's Medical Center, First Hospital of Jilin University, Changchun, China
- Jilin Provincial Key Laboratory of Pediatric Neurology, Changchun, China
- Neuromedical Center, First Hospital of Jilin University, Changchun, China
| |
Collapse
|
3
|
Li X, Li D, Zhang R. Single-Cell RNA sequencing reveals mitochondrial dysfunction in microtia chondrocytes. Sci Rep 2025; 15:1021. [PMID: 39762337 PMCID: PMC11704343 DOI: 10.1038/s41598-025-85169-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Accepted: 01/01/2025] [Indexed: 01/11/2025] Open
Abstract
Microtia is a congenital malformation characterized by underdevelopment of the external ear. While chondrocyte dysfunction has been implicated in microtia, the specific cellular abnormalities remain poorly understood. This study aimed to investigate mitochondrial dysfunction in microtia chondrocytes using single-cell RNA sequencing. Cartilage samples were obtained from patients with unilateral, non-syndromic microtia and healthy controls. Single-cell RNA sequencing was performed using the 10 × Genomics platform. Bioinformatic analyses including cell type identification, trajectory analysis, and gene co-expression network analysis were conducted. Mitochondrial function was assessed through ROS levels, membrane potential, and transmission electron microscopy. Chondrocytes from microtia samples showed lower mitochondrial function scores compared to normal samples. Trajectory analysis revealed more disorganized differentiation patterns in microtia chondrocytes. Mitochondrial dysfunction in microtia chondrocytes was confirmed by increased ROS production, decreased membrane potential, and altered mitochondrial structure. Gene co-expression network analysis identified hub genes associated with mitochondrial function, including SDHA, SIRT1, and PGC1A, which showed reduced expression in microtia chondrocytes. This study provides evidence of mitochondrial dysfunction in microtia chondrocytes and identifies potential key genes involved in this process. These findings offer new insights into the pathogenesis of microtia and may guide future therapeutic strategies.
Collapse
Affiliation(s)
- Xinyu Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Datao Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Ruhong Zhang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| |
Collapse
|
4
|
Al Khazal FJ, Bhat SM, Zhu Y, de Araujo Correia CM, Zhou SX, Wilbanks BA, Folmes CD, Sieck GC, Favier J, Maher LJ. Similar deficiencies, different outcomes: succinate dehydrogenase loss in adrenal medulla vs. fibroblast cell culture models of paraganglioma. Cancer Metab 2024; 12:39. [PMID: 39716277 DOI: 10.1186/s40170-024-00369-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Accepted: 12/18/2024] [Indexed: 12/25/2024] Open
Abstract
Heterozygosity for loss-of-function alleles of the genes encoding the four subunits of succinate dehydrogenase (SDHA, SDHB, SDHC, SDHD), as well as the SDHAF2 assembly factor predispose affected individuals to pheochromocytoma and paraganglioma (PPGL), two rare neuroendocrine tumors that arise from neural crest-derived paraganglia. Tumorigenesis results from loss of the remaining functional SDHx gene copy, leading to a cell with no functional SDH and a defective tricarboxylic acid (TCA) cycle. It is believed that the subsequent accumulation of succinate competitively inhibits multiple dioxygenase enzymes that normally suppress hypoxic signaling and demethylate histones and DNA, ultimately leading to increased expression of genes involved in angiogenesis and cell proliferation. Why SDH loss is selectively tumorigenic in neuroendocrine cells remains poorly understood. In the absence of SDH-loss tumor-derived cell models, the cellular burden of SDH loss and succinate accumulation have been investigated through conditional knockouts of SDH subunits in pre-existing murine or human cell lines with varying degrees of clinical relevance. Here we characterize two available murine SDH-loss cell lines, immortalized adrenally-derived premature chromaffin cells vs. immortalized fibroblasts, at a level of detail beyond that currently reported in the literature and with the intention of laying the foundation for future investigations into adaptive pathways and vulnerabilities in SDH-loss cells. We report different mechanistic and phenotypic manifestations of SDH subunit loss in the presented cellular contexts. These findings highlight similarities and differences in the cellular response to SDH loss between the two cell models. We show that adrenally-derived cells display more severe morphological cellular and mitochondrial alterations, yet are unique in preserving residual Complex I function, perhaps allowing them to better tolerate SDH loss, thus making them a closer model to SDH-loss PPGL relative to fibroblasts.(281 words).
Collapse
Affiliation(s)
- Fatimah J Al Khazal
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, 200 First St. SW, Rochester, MN, 55905, USA
| | - Sanjana Mahadev Bhat
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Yuxiang Zhu
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, USA
| | | | - Sherry X Zhou
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
- Medical Scientist Training Program, Mayo Clinic Alix School of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Brandon A Wilbanks
- Mayo Clinic Graduate School of Biomedical Sciences, Mayo Clinic College of Medicine and Science, Rochester, MN, USA
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, 200 First St. SW, Rochester, MN, 55905, USA
| | - Clifford D Folmes
- Department of Biochemistry and Molecular Biology, Mayo Clinic, Scottsdale, AZ, USA
| | - Gary C Sieck
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Judith Favier
- Inserm, Centre de recherche des Cordeliers, Université Paris-Cité, Sorbonne Université, Equipe Labellisée Ligue contre le Cancer, Paris, 75006, France
| | - L James Maher
- Department of Biochemistry and Molecular Biology, Mayo Clinic College of Medicine and Science, 200 First St. SW, Rochester, MN, 55905, USA.
| |
Collapse
|
5
|
Ajmal N, Lallas CD, McCue P, Li L. Succinate Dehydrogenase Deficient Renal Cell Carcinoma With Sarcomatoid and Rhabdoid Features-A Diagnostic Dilemma. Int J Surg Pathol 2024; 32:1508-1513. [PMID: 38311902 DOI: 10.1177/10668969241229333] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2024]
Abstract
Succinate dehydrogenase (SDH)-deficient renal cell carcinoma (RCC) is a rare epithelial tumor with a biallelic mutation involving any subunit of the SDH complex. Mostly, it has low-grade morphology and a favorable prognosis. We present a case of a 36-year-old woman with weight loss, night sweats, and symptomatic anemia. Her imaging showed a hypo-enhancing heterogeneous right renal mass with invasion of the renal vein and inferior vena cava. Microscopically, the tumor had focal low-grade areas (5%) and extensive areas with high-grade features, including rhabdoid (85%) and sarcomatoid (10%) dedifferentiation. Cytoplasmic inclusions, foci of extracellular mucin, coagulative necrosis, and inflammatory infiltrate were present. The tumor cells, including rhabdoid differentiated, were focally positive for AE1/AE3. Tumor cells showed loss of SDHB immunostaining, consistent with diagnosis. Genetics testing was recommended, but the patient expired due to metastatic carcinoma. Prior studies suggest that sarcomatoid transformation and coagulative necrosis increase the risk of metastasis by up to 70% in SDH-deficient RCC. Follow-up with surveillance for other SDH-deficient neoplasms is recommended in cases of germline mutation. Here, we report the first case of SDH-deficient RCC with concomitant rhabdoid and sarcomatoid features and a detailed review of diagnostic difficulties associated with high-grade tumors.
Collapse
Affiliation(s)
- Namra Ajmal
- Department of Pathology and Genomic Medicine, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Costas D Lallas
- Department of Urology, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| | - Peter McCue
- Department of Pathology, Fox Chase Cancer Center, Philadelphia, PA, USA
| | - Li Li
- Department of Pathology and Genomic Medicine, Thomas Jefferson University Hospital, Philadelphia, PA, USA
| |
Collapse
|
6
|
Abdel-Aziz AK. OXPHOS mediators in acute myeloid leukemia patients: Prognostic biomarkers and therapeutic targets for personalized medicine. World J Surg Oncol 2024; 22:298. [PMID: 39533394 PMCID: PMC11559054 DOI: 10.1186/s12957-024-03581-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 10/31/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Despite significant advances in comprehending its tumorigenic role, the prognostic and therapeutic potential of targeting oxidative phosphorylation (OXPHOS) in acute myeloid leukemia (AML) remain obscure. METHODS The prognostic value of ~ 200 mitochondrial/OXPHOS genes as candidate biomarkers was examined in AML patients over ~ 10 years follow-up using Kaplan-Meier and Cox regression analyses. Furthermore, the transcript levels of the assessed markers were inspected in healthy bone marrow tissues and the dependencies of AML cells on the assessed genes were examined. RESULTS Elevated levels of NADH:ubiquinone oxidoreductase subunit A6 (NDUFA6), succinate dehydrogenase complex flavoprotein subunit A (SDHA), solute carrier family 25 member 12 (SLC25A12), electron transfer flavoprotein subunit beta (ETFB), carnitine palmitoyltransferase 1A (CPT1A) and glutathione peroxidase 4 (GPX4) were associated with poor overall survival of AML patients. SLC25A12, ETFB and CPT1A were overexpressed in AML compared to healthy tissues. Cytochrome B5 type A (CYB5A)high, SLC25A12high and GPX4high AML patients displayed higher levels of circulating and engrafted blasts compared to low-expressing cohorts. NPM1 and SRSF2 mutations were frequent in SDHAlow and CPT1Alow AML patients respectively. FLT3-ITD, NPM1 and IDH1 mutations were prevalent in CPT1Ahigh AML patients. FLT3-ITD AMLs were more dependent on OXPHOS. CONCLUSIONS This study identifies NDUFA6 and SDHA as novel companion prognostic biomarkers which might present a rational strategy for personalized therapy of AML patients.
Collapse
MESH Headings
- Humans
- Leukemia, Myeloid, Acute/genetics
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Leukemia, Myeloid, Acute/drug therapy
- Biomarkers, Tumor/genetics
- Biomarkers, Tumor/metabolism
- Prognosis
- Male
- Female
- Precision Medicine/methods
- Middle Aged
- Oxidative Phosphorylation
- Nucleophosmin
- Adult
- Follow-Up Studies
- Survival Rate
- Aged
- Mutation
- Young Adult
- Serine-Arginine Splicing Factors
Collapse
Affiliation(s)
- Amal Kamal Abdel-Aziz
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Ain Shams University, Abbassia, Cairo, 11566, Egypt.
| |
Collapse
|
7
|
Esteban-Amo MJ, Jiménez-Cuadrado P, Serrano-Lorenzo P, de la Fuente MÁ, Simarro M. Succinate Dehydrogenase and Human Disease: Novel Insights into a Well-Known Enzyme. Biomedicines 2024; 12:2050. [PMID: 39335562 PMCID: PMC11429145 DOI: 10.3390/biomedicines12092050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 08/26/2024] [Accepted: 09/04/2024] [Indexed: 09/30/2024] Open
Abstract
Succinate dehydrogenase (also known as complex II) plays a dual role in respiration by catalyzing the oxidation of succinate to fumarate in the tricarboxylic acid (TCA) cycle and transferring electrons from succinate to ubiquinone in the mitochondrial electron transport chain (ETC). Owing to the privileged position of SDH/CII, its dysfunction leads to TCA cycle arrest and altered respiration. This review aims to elucidate the widely documented profound metabolic effects of SDH/CII deficiency, along with the newly unveiled survival mechanisms in SDH/CII-deficient cells. Such an understanding reveals exploitable vulnerabilities for strategic targeting, which is crucial for the development of novel and more precise therapies for primary mitochondrial diseases, as well as for familial and sporadic cancers associated with SDH/CII mutations.
Collapse
Affiliation(s)
- María J. Esteban-Amo
- Department of Cell Biology, Genetics, Histology and Pharmacology, Faculty of Medicine, University of Valladolid, 47005 Valladolid, Spain; (M.J.E.-A.); (P.J.-C.); (M.Á.d.l.F.)
- Unit of Excellence Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid and Spanish National Research Council (CSIC), 47003 Valladolid, Spain
| | - Patricia Jiménez-Cuadrado
- Department of Cell Biology, Genetics, Histology and Pharmacology, Faculty of Medicine, University of Valladolid, 47005 Valladolid, Spain; (M.J.E.-A.); (P.J.-C.); (M.Á.d.l.F.)
- Unit of Excellence Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid and Spanish National Research Council (CSIC), 47003 Valladolid, Spain
| | - Pablo Serrano-Lorenzo
- Mitochondrial Disorders Laboratory, Clinical Biochemistry Department, Hospital 12 de Octubre Research Institute (imas12), 28041 Madrid, Spain;
- Biomedical Network Research Centre on Rare Diseases (CIBERER), Instituto de Salud Carlos III, 28029 Madrid, Spain
| | - Miguel Á. de la Fuente
- Department of Cell Biology, Genetics, Histology and Pharmacology, Faculty of Medicine, University of Valladolid, 47005 Valladolid, Spain; (M.J.E.-A.); (P.J.-C.); (M.Á.d.l.F.)
- Unit of Excellence Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid and Spanish National Research Council (CSIC), 47003 Valladolid, Spain
| | - María Simarro
- Department of Cell Biology, Genetics, Histology and Pharmacology, Faculty of Medicine, University of Valladolid, 47005 Valladolid, Spain; (M.J.E.-A.); (P.J.-C.); (M.Á.d.l.F.)
- Unit of Excellence Institute of Biomedicine and Molecular Genetics (IBGM), University of Valladolid and Spanish National Research Council (CSIC), 47003 Valladolid, Spain
| |
Collapse
|
8
|
Sharma P, Maklashina E, Voehler M, Balintova S, Dvorakova S, Kraus M, Hadrava Vanova K, Nahacka Z, Zobalova R, Boukalova S, Cunatova K, Mracek T, Ghayee HK, Pacak K, Rohlena J, Neuzil J, Cecchini G, Iverson TM. Disordered-to-ordered transitions in assembly factors allow the complex II catalytic subunit to switch binding partners. Nat Commun 2024; 15:473. [PMID: 38212624 PMCID: PMC10784507 DOI: 10.1038/s41467-023-44563-7] [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: 11/29/2022] [Accepted: 12/19/2023] [Indexed: 01/13/2024] Open
Abstract
Complex II (CII) activity controls phenomena that require crosstalk between metabolism and signaling, including neurodegeneration, cancer metabolism, immune activation, and ischemia-reperfusion injury. CII activity can be regulated at the level of assembly, a process that leverages metastable assembly intermediates. The nature of these intermediates and how CII subunits transfer between metastable complexes remains unclear. In this work, we identify metastable species containing the SDHA subunit and its assembly factors, and we assign a preferred temporal sequence of appearance of these species during CII assembly. Structures of two species show that the assembly factors undergo disordered-to-ordered transitions without the appearance of significant secondary structure. The findings identify that intrinsically disordered regions are critical in regulating CII assembly, an observation that has implications for the control of assembly in other biomolecular complexes.
Collapse
Affiliation(s)
- Pankaj Sharma
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA
| | - Elena Maklashina
- Molecular Biology Division, San Francisco VA Health Care System, San Francisco, CA, 94121, USA
- Department of Biochemistry & Biophysics, University of California, San Francisco, CA, 94158, USA
| | - Markus Voehler
- Department of Chemistry Vanderbilt University, Nashville, TN, 37232, USA
- Center for Structural Biology Vanderbilt University, Nashville, TN, 37232, USA
| | - Sona Balintova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Prague-West, Czech Republic
- Faculty of Science, Charles University, 128 00, Prague 2, Czech Republic
| | - Sarka Dvorakova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Prague-West, Czech Republic
| | - Michal Kraus
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Prague-West, Czech Republic
| | - Katerina Hadrava Vanova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Prague-West, Czech Republic
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Zuzana Nahacka
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Prague-West, Czech Republic
| | - Renata Zobalova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Prague-West, Czech Republic
| | - Stepana Boukalova
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Prague-West, Czech Republic
| | - Kristyna Cunatova
- Institute of Physiology, Czech Academy of Sciences, Prague 4, 142 20, Prague, Czech Republic
| | - Tomas Mracek
- Institute of Physiology, Czech Academy of Sciences, Prague 4, 142 20, Prague, Czech Republic
| | - Hans K Ghayee
- Department of Medicine, Division of Endocrinology & Metabolism, University of Florida College of Medicine and Malcom Randall, VA Medical Center, Gainesville, FL, 32608, USA
| | - Karel Pacak
- Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20814, USA
| | - Jakub Rohlena
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Prague-West, Czech Republic
| | - Jiri Neuzil
- Institute of Biotechnology, Czech Academy of Sciences, 252 50, Prague-West, Czech Republic.
- Faculty of Science, Charles University, 128 00, Prague 2, Czech Republic.
- School of Pharmacy and Medical Science, Griffith University, Southport, QLD, 4222, Australia.
- 1st Faculty of Medicine, Charles University, 128 00, Prague 2, Czech Republic.
| | - Gary Cecchini
- Molecular Biology Division, San Francisco VA Health Care System, San Francisco, CA, 94121, USA.
- Department of Biochemistry & Biophysics, University of California, San Francisco, CA, 94158, USA.
| | - T M Iverson
- Department of Pharmacology, Vanderbilt University, Nashville, TN, 37232, USA.
- Center for Structural Biology Vanderbilt University, Nashville, TN, 37232, USA.
- Department of Biochemistry, Vanderbilt University, Nashville, TN, 37232, USA.
- Vanderbilt Institute of Chemical Biology, Vanderbilt University, Nashville, TN, 37232, USA.
| |
Collapse
|
9
|
Werelusz P, Galiniak S, Mołoń M. Molecular functions of moonlighting proteins in cell metabolic processes. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119598. [PMID: 37774631 DOI: 10.1016/j.bbamcr.2023.119598] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 09/10/2023] [Accepted: 09/17/2023] [Indexed: 10/01/2023]
Abstract
Moonlighting proteins have more than one physiologically significant role within one polypeptide chain. The multifunctionality of proteins was first described in 1987 by Joram Piatigorsky and Graeme Wistow. Cells can benefit from involvement of these proteins in biological processes in several ways, e.g. at the energy level. Furthermore, cells have developed a number of mechanisms to change these proteins' functions. Moonlighting proteins are found in all types of organisms, including prokaryotes, eukaryotes, and even viruses. These proteins include a variety of enzymes that serve as receptors, secreted cytokines, transcription factors, or proteasome components. Additionally, there are many combinations of functions, e.g. among receptors and transcription factors, chaperones and cytokines, as well as transcription factors within the ribosome. This work describes enzymes involved in several important metabolic processes in cells, namely cellular respiration, gluconeogenesis, the urea cycle, and pentose phosphate metabolism.
Collapse
Affiliation(s)
| | - Sabina Galiniak
- Institute of Medical Sciences, Rzeszów University, Rzeszów, Poland
| | - Mateusz Mołoń
- Institute of Biology, Rzeszów University, Rzeszów, Poland.
| |
Collapse
|
10
|
Bajpai AK, Gu Q, Orgil BO, Alberson NR, Towbin JA, Martinez HR, Lu L, Purevjav E. Exploring the Regulation and Function of Rpl3l in the Development of Early-Onset Dilated Cardiomyopathy and Congestive Heart Failure Using Systems Genetics Approach. Genes (Basel) 2023; 15:53. [PMID: 38254943 PMCID: PMC10815855 DOI: 10.3390/genes15010053] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 12/25/2023] [Accepted: 12/27/2023] [Indexed: 01/24/2024] Open
Abstract
BACKGROUND Cardiomyopathies, diseases affecting the myocardium, are common causes of congestive heart failure (CHF) and sudden cardiac death. Recently, biallelic variants in ribosomal protein L3-like (RPL3L) have been reported to be associated with severe neonatal dilated cardiomyopathy (DCM) and CHF. This study employs a systems genetics approach to gain understanding of the regulatory mechanisms underlying the role of RPL3L in DCM. METHODS Genetic correlation, expression quantitative trait loci (eQTL) mapping, differential expression analysis and comparative functional analysis were performed using cardiac gene expression data from the patients and murine genetic reference populations (GRPs) of BXD mice (recombinant inbred strains from a cross of C57BL/6J and DBA/2J mice). Additionally, immune infiltration analysis was performed to understand the relationship between DCM, immune cells and RPL3L expression. RESULTS Systems genetics analysis identified high expression of Rpl3l mRNA, which ranged from 11.31 to 12.16 across murine GRPs of BXD mice, with an ~1.8-fold difference. Pathways such as "diabetic cardiomyopathy", "focal adhesion", "oxidative phosphorylation" and "DCM" were significantly associated with Rpl3l. eQTL mapping suggested Myl4 (Chr 11) and Sdha (Chr 13) as the upstream regulators of Rpl3l. The mRNA expression of Rpl3l, Myl4 and Sdha was significantly correlated with multiple echocardiography traits in BXD mice. Immune infiltration analysis revealed a significant association of RPL3L and SDHA with seven immune cells (CD4, CD8-naive T cell, CD8 T cell, macrophages, cytotoxic T cell, gamma delta T cell and exhausted T cell) that were also differentially infiltrated between heart samples obtained from DCM patients and normal individuals. CONCLUSIONS RPL3L is highly expressed in the heart tissue of humans and mice. Expression of Rpl3l and its upstream regulators, Myl4 and Sdha, correlate with multiple cardiac function traits in murine GRPs of BXD mice, while RPL3L and SDHA correlate with immune cell infiltration in DCM patient hearts, suggesting important roles for RPL3L in DCM and CHF pathogenesis via immune inflammation, necessitating experimental validations of Myl4 and Sdha in Rpl3l regulation.
Collapse
Affiliation(s)
- Akhilesh K. Bajpai
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN 38103, USA; (A.K.B.); (Q.G.)
| | - Qingqing Gu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN 38103, USA; (A.K.B.); (Q.G.)
| | - Buyan-Ochir Orgil
- The Heart Institute, Le Bonheur Children’s Hospital, University of Tennessee Health and Science Center, Memphis, TN 38103, USA; (B.-O.O.); (N.R.A.); (J.A.T.); (H.R.M.)
- Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, TN 38103, USA
| | - Neely R. Alberson
- The Heart Institute, Le Bonheur Children’s Hospital, University of Tennessee Health and Science Center, Memphis, TN 38103, USA; (B.-O.O.); (N.R.A.); (J.A.T.); (H.R.M.)
- Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, TN 38103, USA
| | - Jeffrey A. Towbin
- The Heart Institute, Le Bonheur Children’s Hospital, University of Tennessee Health and Science Center, Memphis, TN 38103, USA; (B.-O.O.); (N.R.A.); (J.A.T.); (H.R.M.)
- Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, TN 38103, USA
- Cardiology, St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
| | - Hugo R. Martinez
- The Heart Institute, Le Bonheur Children’s Hospital, University of Tennessee Health and Science Center, Memphis, TN 38103, USA; (B.-O.O.); (N.R.A.); (J.A.T.); (H.R.M.)
- Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, TN 38103, USA
| | - Lu Lu
- Department of Genetics, Genomics and Informatics, University of Tennessee Health Science Center, Memphis, TN 38103, USA; (A.K.B.); (Q.G.)
| | - Enkhsaikhan Purevjav
- The Heart Institute, Le Bonheur Children’s Hospital, University of Tennessee Health and Science Center, Memphis, TN 38103, USA; (B.-O.O.); (N.R.A.); (J.A.T.); (H.R.M.)
- Children’s Foundation Research Institute, Le Bonheur Children’s Hospital, Memphis, TN 38103, USA
| |
Collapse
|
11
|
Cao K, Xu J, Cao W, Wang X, Lv W, Zeng M, Zou X, Liu J, Feng Z. Assembly of mitochondrial succinate dehydrogenase in human health and disease. Free Radic Biol Med 2023; 207:247-259. [PMID: 37490987 DOI: 10.1016/j.freeradbiomed.2023.07.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/27/2023]
Abstract
Mitochondrial succinate dehydrogenase (SDH), also known as electron transport chain (ETC) Complex II, is the only enzyme complex engaged in both oxidative phosphorylation and the tricarboxylic acid (TCA) cycle. SDH has received increasing attention due to its crucial role in regulating mitochondrial metabolism and human health. Despite having the fewest subunits among the four ETC complexes, functional SDH is formed via a sequential and well-coordinated assembly of subunits. Along with the discovery of subunit-specific assembly factors, the dynamic involvement of the SDH assembly process in a broad range of diseases has been revealed. Recently, we reported that perturbation of SDH assembly in different tissues leads to interesting and distinct pathophysiological changes in mice, indicating a need to understand the intricate SDH assembly process in human health and diseases. Thus, in this review, we summarize recent findings on SDH pathogenesis with respect to disease and a focus on SDH assembly.
Collapse
Affiliation(s)
- Ke Cao
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China; Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
| | - Jie Xu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China
| | - Wenli Cao
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China
| | - Xueqiang Wang
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266071, China
| | - Weiqiang Lv
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China
| | - Mengqi Zeng
- School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266071, China
| | - Xuan Zou
- National & Local Joint Engineering Research Center of Biodiagnosis and Biotherapy, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Jiankang Liu
- Center for Mitochondrial Biology and Medicine, The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, Shaanxi, China; School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266071, China.
| | - Zhihui Feng
- Frontier Institute of Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China; School of Health and Life Sciences, University of Health and Rehabilitation Sciences, Qingdao, Shandong, 266071, China.
| |
Collapse
|
12
|
Chen L, Zhou M, Li H, Liu D, Liao P, Zong Y, Zhang C, Zou W, Gao J. Mitochondrial heterogeneity in diseases. Signal Transduct Target Ther 2023; 8:311. [PMID: 37607925 PMCID: PMC10444818 DOI: 10.1038/s41392-023-01546-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2022] [Revised: 02/21/2023] [Accepted: 06/13/2023] [Indexed: 08/24/2023] Open
Abstract
As key organelles involved in cellular metabolism, mitochondria frequently undergo adaptive changes in morphology, components and functions in response to various environmental stresses and cellular demands. Previous studies of mitochondria research have gradually evolved, from focusing on morphological change analysis to systematic multiomics, thereby revealing the mitochondrial variation between cells or within the mitochondrial population within a single cell. The phenomenon of mitochondrial variation features is defined as mitochondrial heterogeneity. Moreover, mitochondrial heterogeneity has been reported to influence a variety of physiological processes, including tissue homeostasis, tissue repair, immunoregulation, and tumor progression. Here, we comprehensively review the mitochondrial heterogeneity in different tissues under pathological states, involving variant features of mitochondrial DNA, RNA, protein and lipid components. Then, the mechanisms that contribute to mitochondrial heterogeneity are also summarized, such as the mutation of the mitochondrial genome and the import of mitochondrial proteins that result in the heterogeneity of mitochondrial DNA and protein components. Additionally, multiple perspectives are investigated to better comprehend the mysteries of mitochondrial heterogeneity between cells. Finally, we summarize the prospective mitochondrial heterogeneity-targeting therapies in terms of alleviating mitochondrial oxidative damage, reducing mitochondrial carbon stress and enhancing mitochondrial biogenesis to relieve various pathological conditions. The possibility of recent technological advances in targeted mitochondrial gene editing is also discussed.
Collapse
Affiliation(s)
- Long Chen
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China
| | - Mengnan Zhou
- Department of Pathogenic Biology, School of Basic Medical Science, China Medical University, Shenyang, 110001, China
| | - Hao Li
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Delin Liu
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Peng Liao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Yao Zong
- Centre for Orthopaedic Research, Medical School, The University of Western Australia, Nedlands, WA, 6009, Australia
| | - Changqing Zhang
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Weiguo Zou
- State Key Laboratory of Cell Biology, CAS Center for Excellence in Molecular Cell Sciences, Shanghai Institute of Biochemistry and Cell Biology, Chinese Academy of Sciences, University of Chinese Academy of Sciences, Shanghai, 200031, China.
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
| | - Junjie Gao
- Department of Orthopaedics, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
- Institute of Microsurgery on Extremities, and Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China.
- Shanghai Sixth People's Hospital Fujian, No. 16, Luoshan Section, Jinguang Road, Luoshan Street, Jinjiang City, Quanzhou, Fujian, China.
| |
Collapse
|
13
|
FitzHugh ZT, Schiller MR. Systematic Assessment of Protein C-Termini Mutated in Human Disorders. Biomolecules 2023; 13:biom13020355. [PMID: 36830724 PMCID: PMC9953674 DOI: 10.3390/biom13020355] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2022] [Revised: 01/23/2023] [Accepted: 02/02/2023] [Indexed: 02/16/2023] Open
Abstract
All proteins have a carboxyl terminus, and we previously summarized eight mutations in binding and trafficking sequence determinants in the C-terminus that, when disrupted, cause human diseases. These sequence elements for binding and trafficking sites, as well as post-translational modifications (PTMs), are called minimotifs or short linear motifs. We wanted to determine how frequently mutations in minimotifs in the C-terminus cause disease. We searched specifically for PTMs because mutation of a modified amino acid almost always changes the chemistry of the side chain and can be interpreted as loss-of-function. We analyzed data from ClinVar for disease variants, Minimotif Miner and the C-terminome for PTMs, and RefSeq for protein sequences, yielding 20 such potential disease-causing variants. After additional screening, they include six with a previously reported PTM disruption mechanism and nine with new hypotheses for mutated minimotifs in C-termini that may cause disease. These mutations were generally for different genes, with four different PTM types and several different diseases. Our study helps to identify new molecular mechanisms for nine separate variants that cause disease, and this type of analysis could be extended as databases grow and to binding and trafficking motifs. We conclude that mutated motifs in C-termini are an infrequent cause of disease.
Collapse
Affiliation(s)
- Zachary T. FitzHugh
- Nevada Institute of Personalized Medicine, University of Nevada Las Vegas, 4505 S. Maryland Pkwy, Las Vegas, NV 89154, USA
- School of Life Sciences, University of Nevada, 4505 S. Maryland Parkway, Las Vegas, NV 89154, USA
| | - Martin R. Schiller
- Nevada Institute of Personalized Medicine, University of Nevada Las Vegas, 4505 S. Maryland Pkwy, Las Vegas, NV 89154, USA
- School of Life Sciences, University of Nevada, 4505 S. Maryland Parkway, Las Vegas, NV 89154, USA
- Heligenics Inc., 833 Las Vegas Blvd. North, Suite B, Las Vegas, NV 89101, USA
- Correspondence: ; Tel.: +1-702-895-5546; Fax: +1-702-895-5728
| |
Collapse
|
14
|
Goraltchouk A, Mankovskaya S, Kuznetsova T, Hladkova Z, Hollander JM, Luppino F, Seregin A. Comparative evaluation of rhFGF18 and rhGDF11 treatment in a transient ischemia stroke model. Restor Neurol Neurosci 2023; 41:257-270. [PMID: 38363623 DOI: 10.3233/rnn-231347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
Background Pharmacological treatments for ischemic stroke remain limited to thrombolysis, which is associated with increased risk of potentially fatal hemorrhage. Treatments with Recombinant Human Fibroblast Growth Factor 18 (rhFGF18) and Growth and Differentiation Factor 11 (rhGDF11) appear promising based on different preclinical models. The goal of this study was to compare the effects of rhFGF18 and rhGDF11 directly on survival, behavioral deficits, and histological fingerprint of cerebral ischemia in the Wistar rat middle cerebral artery occlusion (MCAO) model of stroke. Methods Ischemia-reperfusion injury was induced using a 2-hour transient MCAO. Animals were administered rhFGF18 (infusion), rhGDF11 (multi-injection), or Phosphate Buffered Saline (PBS) vehicle control and followed for 42 days. Motor-Cognitive deficits were evaluated using the Morris Water Maze at Days 0 (pre-MCAO), 7, 21, and 42. Histopathological assessments were performed on Days 21 and 42. Results Day 7 post-ischemia water maze performance times increased 38.3%, 2.1%, and 23.1% for PBS, rhFGF18, and rhGDF11-treated groups, respectively. Fraction of neurons with abnormal morphology (chromatolysis, pyknotic nuclei, somal degeneration) decreased in all groups toward Day 42 and was lowest for rhFGF18. AChE-positive fiber density and activity increased over time in the rhFGF18 group, remained unchanged in the rhGDF11 treatment arm, and declined in the PBS control. Metabolic increases were greatest in rhGDF11 treated animals, with both rhFGF18 and rhGDF11 achieving improvements over PBS, as evidenced by increased succinate dehydrogenase and lactate dehydrogenase activity. Finally, rhFGF18 treatment exhibited a trend for reduced mortality relative to PBS (5.6%, 95% CI [27.3%, 0.1% ] vs. 22.2%, 95% CI [47.6%, 6.4% ]). Conclusions rhFGF18 treatment appears promising in improving survival and promoting motor-cognitive recovery following cerebral ischemia-reperfusion injury.
Collapse
Affiliation(s)
| | | | | | - Zhanna Hladkova
- Institute of Physiology, National Academy of Sciences, Minsk, Belarus
| | - Judith M Hollander
- Remedium Bio, Inc., Needham, MA, USA
- Department of Immunology, Tufts University School of Medicine, Boston, MA, USA
| | | | | |
Collapse
|
15
|
Rujimongkon K, Ampawong S, Isarangkul D, Reamtong O, Aramwit P. Sericin-mediated improvement of dysmorphic cardiac mitochondria from hypercholesterolaemia is associated with maintaining mitochondrial dynamics, energy production, and mitochondrial structure. PHARMACEUTICAL BIOLOGY 2022; 60:708-721. [PMID: 35348427 PMCID: PMC8967205 DOI: 10.1080/13880209.2022.2055088] [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: 10/26/2020] [Revised: 03/10/2022] [Accepted: 03/12/2022] [Indexed: 05/30/2023]
Abstract
CONTEXT Sericin is a component protein in the silkworm cocoon [Bombyx mori Linnaeus (Bombycidae)] that improves dysmorphic cardiac mitochondria under hypercholesterolemic conditions. This is the first study to explore cardiac mitochondrial proteins associated with sericin treatment. OBJECTIVE To investigate the mechanism of action of sericin in cardiac mitochondria under hypercholesterolaemia. MATERIALS AND METHODS Hypercholesterolaemia was induced in Wistar rats by feeding them 6% cholesterol-containing chow for 6 weeks. The hypercholesterolemic rats were separated into 2 groups (n = 6 for each): the sericin-treated (1,000 mg/kg daily) and nontreated groups. The treatment conditions were maintained for 4 weeks prior to cardiac mitochondria isolation. The mitochondrial structure was evaluated by immunolabeling electron microscopy, and differential mitochondrial protein expression was determined and quantitated by two-dimensional gel electrophoresis coupled with mass spectrometry. RESULTS A 32.22 ± 2.9% increase in the percent striated area of cardiac muscle was observed in sericin-treated hypercholesterolemic rats compared to the nontreatment group (4.18 ± 1.11%). Alterations in mitochondrial proteins, including upregulation of optic atrophy 1 (OPA1) and reduction of NADH-ubiquinone oxidoreductase 75 kDa subunit (NDUFS1) expression, are correlated with a reduction in mitochondrial apoptosis under sericin treatment. Differential proteomic observation also revealed that sericin may improve mitochondrial energy production by upregulating acetyl-CoA acetyltransferase (ACAT1) and NADH dehydrogenase 1α subcomplex subunit 10 (NDUFA10) expression. DISCUSSION AND CONCLUSIONS Sericin treatment could improve the dysmorphic mitochondrial structure, metabolism, and energy production of cardiac mitochondria under hypercholesterolaemia. These results suggest that sericin may be an alternative treatment molecule that is related to cardiac mitochondrial abnormalities.
Collapse
Affiliation(s)
- Kitiya Rujimongkon
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences and Center of Excellence in Bioactive Resources for Innovative Clinical Applications, Chulalongkorn University, Bangkok, Thailand
- Proteomics Research Team, National Omics Center, National Science and Technology Development Agency, Pathumthani, Thailand
| | - Sumate Ampawong
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Duangnate Isarangkul
- Department of Microbiology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Onrapak Reamtong
- Department of Molecular Tropical Medicine and Genetic, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand, and
| | - Pornanong Aramwit
- Department of Pharmacy Practice, Faculty of Pharmaceutical Sciences and Center of Excellence in Bioactive Resources for Innovative Clinical Applications, Chulalongkorn University, Bangkok, Thailand
- The Academy of Science, The Royal Society of Thailand, Bangkok, Thailand
| |
Collapse
|
16
|
Yang Y, Luo D, Shao Y, Shan Z, Liu Q, Weng J, He W, Zhang R, Li Q, Wang Z, Li X. circCAPRIN1 interacts with STAT2 to promote tumor progression and lipid synthesis via upregulating ACC1 expression in colorectal cancer. Cancer Commun (Lond) 2022; 43:100-122. [PMID: 36328987 PMCID: PMC9859733 DOI: 10.1002/cac2.12380] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/21/2022] [Accepted: 10/20/2022] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Circular RNAs (circRNAs) generated by back-splicing of precursor mRNAs (pre-mRNAs) are often aberrantly expressed in cancer cells. Accumulating evidence has revealed that circRNAs play a critical role in the progression of several cancers, including colorectal cancer (CRC). However, the current understandings of the emerging functions of circRNAs in CRC lipid metabolism and the underlying molecular mechanisms are still limited. Here, we aimed to explore the role of circCAPRIN1 in regulating CRC lipid metabolism and tumorigenesis. METHODS circRNA microarray was performed with three pairs of tumor and non-tumor tissues from CRC patients. The expression of circRNAs were determined by quantitative PCR (qPCR) and in situ hybridization (ISH). The endogenous levels of circRNAs in CRC cells were manipulated by transfection with lentiviruses overexpressing or silencing circRNAs. The regulatory roles of circRNAs in the occurrence of CRC were investigated both in vitro and in vivo using gene expression array, RNA pull-down/mass spectrometry, RNA immunoprecipitation assay, luciferase reporter assay, chromatin immunoprecipitation analysis, and fluorescence in situ hybridization (FISH). RESULTS Among circRNAs, circCAPRIN1 was most significantly upregulated in CRC tissue specimens. circCAPRIN1 expression was positively correlated with the clinical stage and unfavorable prognosis of CRC patients. Downregulation of circCAPRIN1 suppressed proliferation, migration, and epithelial-mesenchymal transition of CRC cells, while circCAPRIN1 overexpression had opposite effects. RNA sequencing and gene ontology analysis indicated that circCAPRIN1 upregulated the expressions of genes involved in CRC lipid metabolism. Moreover, circCAPRIN1 promoted lipid synthesis by enhancing Acetyl-CoA carboxylase 1 (ACC1) expression. Further mechanistic assays demonstrated that circCAPRIN1 directly bound signal transducer and activator of transcription 2 (STAT2) to activate ACC1 transcription, thus regulating lipid metabolism and facilitating CRC tumorigenesis. CONCLUSIONS These findings revealed the oncogenic role and mechanism of circCAPRIN1 in CRC. circCAPRIN1 interacted with STAT2 to promote CRC tumor progression and lipid synthesis by enhancing the expression of ACC1. circCAPRIN1 may be considered as a novel potential diagnostic and therapeutic target for CRC patients.
Collapse
Affiliation(s)
- Yufei Yang
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China,Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Dakui Luo
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China,Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Yang Shao
- Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China,Cancer InstituteFudan University Shanghai Cancer CenterShanghai200032P. R. China
| | - Zezhi Shan
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China,Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Qi Liu
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China,Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Junyong Weng
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China,Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Weijing He
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China,Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Ruoxin Zhang
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China,Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Qingguo Li
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China,Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| | - Ziliang Wang
- Shanghai Municipal Hospital of Traditional Chinese MedicineShanghai University of Traditional Chinese MedicineShanghai200071P. R. China
| | - Xinxiang Li
- Department of Colorectal SurgeryFudan University Shanghai Cancer CenterShanghai200032P. R. China,Department of OncologyShanghai Medical CollegeFudan UniversityShanghai200032P. R. China
| |
Collapse
|
17
|
Vikramdeo KS, Sudan SK, Singh AP, Singh S, Dasgupta S. Mitochondrial respiratory complexes: Significance in human mitochondrial disorders and cancers. J Cell Physiol 2022; 237:4049-4078. [PMID: 36074903 DOI: 10.1002/jcp.30869] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 07/18/2022] [Accepted: 08/23/2022] [Indexed: 11/07/2022]
Abstract
Mitochondria are pivotal organelles that govern cellular energy production through the oxidative phosphorylation system utilizing five respiratory complexes. In addition, mitochondria also contribute to various critical signaling pathways including apoptosis, damage-associated molecular patterns, calcium homeostasis, lipid, and amino acid biosynthesis. Among these diverse functions, the energy generation program oversee by mitochondria represents an immaculate orchestration and functional coordination between the mitochondria and nuclear encoded molecules. Perturbation in this program through respiratory complexes' alteration results in the manifestation of various mitochondrial disorders and malignancy, which is alarmingly becoming evident in the recent literature. Considering the clinical relevance and importance of this emerging medical problem, this review sheds light on the timing and nature of molecular alterations in various respiratory complexes and their functional consequences observed in various mitochondrial disorders and human cancers. Finally, we discussed how this wealth of information could be exploited and tailored to develop respiratory complex targeted personalized therapeutics and biomarkers for better management of various incurable human mitochondrial disorders and cancers.
Collapse
Affiliation(s)
- Kunwar Somesh Vikramdeo
- Department of Pathology, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA.,Department of Pathology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Sarabjeet Kour Sudan
- Department of Pathology, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA.,Department of Pathology, College of Medicine, University of South Alabama, Mobile, Alabama, USA
| | - Ajay P Singh
- Department of Pathology, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA.,Department of Pathology, College of Medicine, University of South Alabama, Mobile, Alabama, USA.,Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, USA
| | - Seema Singh
- Department of Pathology, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA.,Department of Pathology, College of Medicine, University of South Alabama, Mobile, Alabama, USA.,Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, USA
| | - Santanu Dasgupta
- Department of Pathology, Mitchell Cancer Institute, University of South Alabama, Mobile, Alabama, USA.,Department of Pathology, College of Medicine, University of South Alabama, Mobile, Alabama, USA.,Department of Biochemistry and Molecular Biology, University of South Alabama, Mobile, Alabama, USA
| |
Collapse
|
18
|
Wortmann SB, Oud MM, Alders M, Coene KLM, van der Crabben SN, Feichtinger RG, Garanto A, Hoischen A, Langeveld M, Lefeber D, Mayr JA, Ockeloen CW, Prokisch H, Rodenburg R, Waterham HR, Wevers RA, van de Warrenburg BPC, Willemsen MAAP, Wolf NI, Vissers LELM, van Karnebeek CDM. How to proceed after "negative" exome: A review on genetic diagnostics, limitations, challenges, and emerging new multiomics techniques. J Inherit Metab Dis 2022; 45:663-681. [PMID: 35506430 PMCID: PMC9539960 DOI: 10.1002/jimd.12507] [Citation(s) in RCA: 38] [Impact Index Per Article: 12.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/20/2022] [Revised: 04/26/2022] [Accepted: 04/27/2022] [Indexed: 11/28/2022]
Abstract
Exome sequencing (ES) in the clinical setting of inborn metabolic diseases (IMDs) has created tremendous improvement in achieving an accurate and timely molecular diagnosis for a greater number of patients, but it still leaves the majority of patients without a diagnosis. In parallel, (personalized) treatment strategies are increasingly available, but this requires the availability of a molecular diagnosis. IMDs comprise an expanding field with the ongoing identification of novel disease genes and the recognition of multiple inheritance patterns, mosaicism, variable penetrance, and expressivity for known disease genes. The analysis of trio ES is preferred over singleton ES as information on the allelic origin (paternal, maternal, "de novo") reduces the number of variants that require interpretation. All ES data and interpretation strategies should be exploited including CNV and mitochondrial DNA analysis. The constant advancements in available techniques and knowledge necessitate the close exchange of clinicians and molecular geneticists about genotypes and phenotypes, as well as knowledge of the challenges and pitfalls of ES to initiate proper further diagnostic steps. Functional analyses (transcriptomics, proteomics, and metabolomics) can be applied to characterize and validate the impact of identified variants, or to guide the genomic search for a diagnosis in unsolved cases. Future diagnostic techniques (genome sequencing [GS], optical genome mapping, long-read sequencing, and epigenetic profiling) will further enhance the diagnostic yield. We provide an overview of the challenges and limitations inherent to ES followed by an outline of solutions and a clinical checklist, focused on establishing a diagnosis to eventually achieve (personalized) treatment.
Collapse
Affiliation(s)
- Saskia B. Wortmann
- Radboud Center for Mitochondrial and Metabolic Medicine, Department of PediatricsAmalia Children's Hospital, Radboud University Medical CenterNijmegenThe Netherlands
- University Children's Hospital, Paracelsus Medical UniversitySalzburgAustria
| | - Machteld M. Oud
- United for Metabolic DiseasesAmsterdamThe Netherlands
- Department of Human GeneticsDonders Institute for Brain, Cognition and Behaviour, Radboud University Medical CenterNijmegenThe Netherlands
| | - Mariëlle Alders
- Department of Human GeneticsAmsterdam UMC, University of Amsterdam, Amsterdam Reproduction and Development Research InstituteAmsterdamThe Netherlands
| | - Karlien L. M. Coene
- United for Metabolic DiseasesAmsterdamThe Netherlands
- Translational Metabolic Laboratory, Department of Laboratory MedicineRadboud University Medical CenterNijmegenThe Netherlands
| | - Saskia N. van der Crabben
- Department of Human GeneticsAmsterdam University Medical Centers, University of AmsterdamAmsterdamThe Netherlands
| | - René G. Feichtinger
- University Children's Hospital, Paracelsus Medical UniversitySalzburgAustria
| | - Alejandro Garanto
- Radboud Center for Mitochondrial and Metabolic Medicine, Department of PediatricsAmalia Children's Hospital, Radboud University Medical CenterNijmegenThe Netherlands
- Department of PediatricsAmalia Children's Hospital, Radboud Institute for Molecular LifesciencesNijmegenThe Netherlands
- Department of Human GeneticsRadboud Institute for Molecular LifesciencesNijmegenThe Netherlands
| | - Alex Hoischen
- Department of Human Genetics, Department of Internal Medicine and Radboud Center for Infectious DiseasesRadboud Institute of Medical Life Sciences, Radboud University Medical CenterNijmegenthe Netherlands
| | - Mirjam Langeveld
- Department of Endocrinology and MetabolismAmsterdam University Medical Centers, location AMC, University of AmsterdamAmsterdamThe Netherlands
| | - Dirk Lefeber
- United for Metabolic DiseasesAmsterdamThe Netherlands
- Translational Metabolic Laboratory, Department of Laboratory MedicineRadboud University Medical CenterNijmegenThe Netherlands
- Department of Neurology, Donders Institute for BrainCognition and Behaviour, Radboud University Medical CenterNijmegenThe Netherlands
| | - Johannes A. Mayr
- University Children's Hospital, Paracelsus Medical UniversitySalzburgAustria
| | - Charlotte W. Ockeloen
- Department of Human GeneticsRadboud Institute for Molecular LifesciencesNijmegenThe Netherlands
| | - Holger Prokisch
- School of MedicineInstitute of Human Genetics, Technical University Munich and Institute of NeurogenomicsNeuherbergGermany
| | - Richard Rodenburg
- Radboud Center for Mitochondrial and Metabolic MedicineTranslational Metabolic Laboratory, Department of Pediatrics, Radboud University Medical CenterNijmegenThe Netherlands
| | - Hans R. Waterham
- United for Metabolic DiseasesAmsterdamThe Netherlands
- Laboratory Genetic Metabolic Diseases, Department of Clinical ChemistryAmsterdam University Medical Centers, location AMC, University of AmsterdamAmsterdamThe Netherlands
| | - Ron A. Wevers
- United for Metabolic DiseasesAmsterdamThe Netherlands
- Translational Metabolic Laboratory, Department of Laboratory MedicineRadboud University Medical CenterNijmegenThe Netherlands
| | - Bart P. C. van de Warrenburg
- Department of Neurology, Donders Institute for BrainCognition and Behaviour, Radboud University Medical CenterNijmegenThe Netherlands
| | - Michel A. A. P. Willemsen
- Departments of Pediatric Neurology and PediatricsAmalia Children's Hospital, Donders Institute for Brain, Cognition and Behavior, Radboud University Medical CenterNijmegenThe Netherlands
| | - Nicole I. Wolf
- Amsterdam Leukodystrophy Center, Department of Child NeurologyEmma Children's Hospital, Amsterdam University Medical Centers, Vrije Universiteit AmsterdamAmsterdamThe Netherlands
| | - Lisenka E. L. M. Vissers
- Department of Human GeneticsDonders Institute for Brain, Cognition and Behaviour, Radboud University Medical CenterNijmegenThe Netherlands
| | - Clara D. M. van Karnebeek
- Radboud Center for Mitochondrial and Metabolic Medicine, Department of PediatricsAmalia Children's Hospital, Radboud University Medical CenterNijmegenThe Netherlands
- United for Metabolic DiseasesAmsterdamThe Netherlands
- Department of Human GeneticsAmsterdam UMC, University of Amsterdam, Amsterdam Reproduction and Development Research InstituteAmsterdamThe Netherlands
- Department of Pediatrics, Emma Center for Personalized MedicineAmsterdam University Medical Centers, Amsterdam, Amsterdam Genetics Endocrinology Metabolism Research Institute, University of AmsterdamAmsterdamThe Netherlands
| |
Collapse
|
19
|
Yue X, Liu B, Han T, Luo N, Lu G, Guo D, Bu F, Wang G. A Novel Germline SDHA Gene Mutation and Co-Occurring Somatic KIT Activating Mutation in a Patient With Pediatric Central Nervous System Germ Cell Tumor: Case Report. Front Oncol 2022; 12:835220. [PMID: 35651799 PMCID: PMC9149216 DOI: 10.3389/fonc.2022.835220] [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: 12/14/2021] [Accepted: 03/24/2022] [Indexed: 11/26/2022] Open
Abstract
Central nervous system germ cell tumors (CNS GCTs) are a heterogeneous group of primary CNS tumors. GCTs are more common and mostly observed in pediatric and young adult patients. CNS GCTs are divided into germinomas and non-germinomatous germ cell tumors (NGGCTs), with different therapeutic strategies depending on diagnosis. Herein, we report a patient with pediatric central nervous system germinoma harboring a somatic KIT p.Y823D and a heterozygous germline SDHA p. T396Nfs*14 mutation detected by next generation sequencing. After surgery, the patient received chemotherapy (temozolomide + nedaplatin + etoposide). This is the first report of a Chinese pediatric patient with CNS GCT harboring concurrent germline SDHA and somatic KIT mutation, which enriches molecular profiles of CNS GCTs and provides more molecular evidence of clinical diagnosis and potential targeted therapy in CNS GCTs.
Collapse
Affiliation(s)
- Xizan Yue
- Department of Neurosurgery, Qilu Children’s Hospital of Shandong University, Jinan, China
| | - Bo Liu
- Department of Neurosurgery, Qilu Children’s Hospital of Shandong University, Jinan, China
| | - Tiantian Han
- The Medical Department, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing, China
- The Medical Department, Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, China
- The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing, China
| | - Ningning Luo
- The Medical Department, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing, China
- The Medical Department, Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, China
- The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing, China
| | - Guanghua Lu
- The Medical Department, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing, China
- The Medical Department, Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, China
- The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing, China
| | - Didi Guo
- The Medical Department, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing, China
- The Medical Department, Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, China
- The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing, China
| | - Fanfeng Bu
- The Medical Department, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing, China
- The Medical Department, Nanjing Simcere Medical Laboratory Science Co., Ltd., Nanjing, China
- The State Key Lab of Translational Medicine and Innovative Drug Development, Jiangsu Simcere Diagnostics Co., Ltd., Nanjing, China
| | - Guangyu Wang
- Department of Neurosurgery, Qilu Children’s Hospital of Shandong University, Jinan, China
| |
Collapse
|
20
|
Lay S, Pearce X, Sanislav O, Fisher PR, Annesley SJ. Cytopathological Outcomes of Knocking Down Expression of Mitochondrial Complex II Subunits in Dictyostelium discoideum. Int J Mol Sci 2022; 23:ijms23095039. [PMID: 35563430 PMCID: PMC9105181 DOI: 10.3390/ijms23095039] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Revised: 04/29/2022] [Accepted: 04/29/2022] [Indexed: 02/01/2023] Open
Abstract
Mitochondrial Complex II is composed of four core subunits and mutations to any of the subunits result in lowered Complex II activity. Surprisingly, although mutations in any of the subunits can yield similar clinical outcomes, there are distinct differences in the patterns of clinical disease most commonly associated with mutations in different subunits. Thus, mutations to the SdhA subunit most often result in mitochondrial disease phenotypes, whilst mutations to the other subunits SdhB-D more commonly result in tumour formation. The reason the clinical outcomes are so different is unknown. Here, we individually antisense-inhibited three of the Complex II subunits, SdhA, SdhB or SdhC, in the simple model organism Dictyostelium discoideum. Whilst SdhB and SdhC knockdown resulted in growth defects on bacterial lawns, antisense inhibition of SdhA expression resulted in a different pattern of phenotypic defects, including impairments of growth in liquid medium, enhanced intracellular proliferation of the bacterial pathogen Legionella pneumophila and phagocytosis. Knockdown of the individual subunits also produced different abnormalities in mitochondrial function with only SdhA knockdown resulting in broad mitochondrial dysfunction. Furthermore, these defects were shown to be mediated by the chronic activation of the cellular energy sensor AMP-activated protein kinase. Our results are in agreement with a role for loss of function of SdhA but not the other Complex II subunits in impairing mitochondrial oxidative phosphorylation and they suggest a role for AMP-activated protein kinase in mediating the cytopathological outcomes.
Collapse
|
21
|
Lopriore P, Ricciarini V, Siciliano G, Mancuso M, Montano V. Mitochondrial Ataxias: Molecular Classification and Clinical Heterogeneity. Neurol Int 2022; 14:337-356. [PMID: 35466209 PMCID: PMC9036286 DOI: 10.3390/neurolint14020028] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2022] [Revised: 03/26/2022] [Accepted: 03/28/2022] [Indexed: 01/25/2023] Open
Abstract
Ataxia is increasingly being recognized as a cardinal manifestation in primary mitochondrial diseases (PMDs) in both paediatric and adult patients. It can be caused by disruption of cerebellar nuclei or fibres, its connection with the brainstem, or spinal and peripheral lesions leading to proprioceptive loss. Despite mitochondrial ataxias having no specific defining features, they should be included in hereditary ataxias differential diagnosis, given the high prevalence of PMDs. This review focuses on the clinical and neuropathological features and genetic background of PMDs in which ataxia is a prominent manifestation.
Collapse
|
22
|
Molecular Genetics Overview of Primary Mitochondrial Myopathies. J Clin Med 2022; 11:jcm11030632. [PMID: 35160083 PMCID: PMC8836969 DOI: 10.3390/jcm11030632] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Revised: 01/13/2022] [Accepted: 01/20/2022] [Indexed: 12/29/2022] Open
Abstract
Mitochondrial disorders are the most common inherited conditions, characterized by defects in oxidative phosphorylation and caused by mutations in nuclear or mitochondrial genes. Due to its high energy request, skeletal muscle is typically involved. According to the International Workshop of Experts in Mitochondrial Diseases held in Rome in 2016, the term Primary Mitochondrial Myopathy (PMM) should refer to those mitochondrial disorders affecting principally, but not exclusively, the skeletal muscle. The clinical presentation may include general isolated myopathy with muscle weakness, exercise intolerance, chronic ophthalmoplegia/ophthalmoparesis (cPEO) and eyelids ptosis, or multisystem conditions where there is a coexistence with extramuscular signs and symptoms. In recent years, new therapeutic targets have been identified leading to the launch of some promising clinical trials that have mainly focused on treating muscle symptoms and that require populations with defined genotype. Advantages in next-generation sequencing techniques have substantially improved diagnosis. So far, an increasing number of mutations have been identified as responsible for mitochondrial disorders. In this review, we focused on the principal molecular genetic alterations in PMM. Accordingly, we carried out a comprehensive review of the literature and briefly discussed the possible approaches which could guide the clinician to a genetic diagnosis.
Collapse
|
23
|
Yang Z, Cao J, Song Y, Li S, Jiao Z, Ren S, Gao X, Zhang S, Liu J, Chen Y. Whole-exome sequencing identified novel variants in three Chinese Leigh syndrome pedigrees. Am J Med Genet A 2022; 188:1214-1225. [PMID: 35014173 DOI: 10.1002/ajmg.a.62641] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 12/12/2021] [Accepted: 12/18/2021] [Indexed: 11/08/2022]
Abstract
Leigh syndrome (LS), the most common mitochondrial disease in early childhood, usually manifests variable neurodegenerative symptoms and typical brain magnetic resonance imaging (MRI) lesions. To date, pathogenic variants in more than 80 genes have been identified. However, there are still many cases without molecular diagnoses, and thus more disease-causing variants need to be unveiled. Here, we presented three clinically suspected LS patients manifesting neurological symptoms including developmental delay, hypotonia, and epilepsy during the first year of age, along with symmetric brain lesions on MRI. We explored disease-associated variants in patients and their nonconsanguineous parents by whole-exome sequencing and subsequent Sanger sequencing verification. Sequencing data revealed three pairs of disease-associated compound heterozygous variants: c.1A>G (p.Met1?) and 409G>C (p.Asp137His) in SDHA, c.1253G>A (p.Arg418His) and 1300C>T (p.Leu434Phe) in NARS2, and c.5C>T (p.Ala2Val) and 773T>G (p.Leu258Trp) in ECHS1. Among them, the likely pathogenic variants c.409G>C (p.Asp137His) in SDHA, c.1300C>T (p.Leu434Phe) in NARS2, and c.773T>G (p.Leu258Trp) in ECHS1 were newly identified. Segregation analysis indicated the possible disease-causing nature of the novel variants. In silico prediction and three-dimensional protein modeling further suggested the potential pathogenicity of these variants. Our discovery of novel variants expands the gene variant spectrum of LS and provides novel evidence for genetic counseling.
Collapse
Affiliation(s)
- Zhihua Yang
- Genetic and Prenatal Diagnosis Center, Department of Gynecology and Obstetrics, First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Jun Cao
- Genetic and Prenatal Diagnosis Center, Department of Gynecology and Obstetrics, First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Yucen Song
- Genetic and Prenatal Diagnosis Center, Department of Gynecology and Obstetrics, First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Suyi Li
- Genetic and Prenatal Diagnosis Center, Department of Gynecology and Obstetrics, First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Zhihui Jiao
- Genetic and Prenatal Diagnosis Center, Department of Gynecology and Obstetrics, First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Shumin Ren
- Genetic and Prenatal Diagnosis Center, Department of Gynecology and Obstetrics, First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Xu Gao
- Genetic and Prenatal Diagnosis Center, Department of Gynecology and Obstetrics, First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Suqin Zhang
- Department of Pediatrics, First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Jingjing Liu
- Department of MR Imaging, First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| | - Yibing Chen
- Genetic and Prenatal Diagnosis Center, Department of Gynecology and Obstetrics, First Affiliated Hospital, Zhengzhou University, Zhengzhou, China
| |
Collapse
|
24
|
Kou X, Ding H, Li L, Chao H. Caseinolytic protease P (CLPP) activated by ONC201 inhibits proliferation and promotes apoptosis in human epithelial ovarian cancer cells by inducing mitochondrial dysfunction. ANNALS OF TRANSLATIONAL MEDICINE 2021; 9:1463. [PMID: 34734015 PMCID: PMC8506775 DOI: 10.21037/atm-21-4321] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/09/2021] [Indexed: 12/23/2022]
Abstract
Background Caseinolytic protease P (CLPP) is a mitochondrial specific protein which has been reported to be related to tumor cell apoptosis. This study aims to explore the roles of CLPP in human epithelial ovarian cancer (EOC). Methods We determined CLPP expression in paracancerous tissues and cancer tissues obtained from 20 EOC patients, and also in 4 EOC cell lines, and one normal ovarian cell line (IOSE-80). We knocked CLPP expression down in SK-OV-3 and A2780 cells and overexpressed it in SW626 and OVcar3 cells. The effect of CLPP expression on cell proliferation, mitochondrial membrane potential, and apoptosis was then assessed by flow cytometry assay. Furthermore, the effect of ONC201 (agonist of CLPP) on the EOC cell lines was also investigated. Results The CLPP expression was markedly down-regulated in EOC cancer tissues, and the Kaplan-Meier Plotter database revealed its low expression was linked to poor prognosis in EOC patients. Low expression of CLPP up-regulated the expression of NADH: ubiquinone oxidoreductase subunit A12 (NDUFA12), succinate dehydrogenase complex flavoprotein subunit A (SDHA), and succinate dehydrogenase complex iron sulfur subunit B (SDHB), which are key members of the mitochondrial respiratory chain, and these up-regulated proteins further led to the increase of mitochondrial membrane potential, cell proliferation promotion and neoplasm metastasis. Conversely, while overexpression of CLPP led to the opposite results, including inducing the decrease of mitochondrial membrane potential and apoptosis. In addition, stimulation with ONC201 enhanced the function of CLPP in SW626 and OVcar3 cells, and silencing of CLPP could neutralize the effect of ONC201. Conclusions Our findings suggest that CLPP mediated mitochondrial dysfunction inhibits the proliferation and migration of EOC cells.
Collapse
Affiliation(s)
- Xinxin Kou
- Department of Gynaecology, Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Hui Ding
- Department of Gynaecology, Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Lei Li
- Department of Gynaecology, Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| | - Hongtu Chao
- Department of Gynaecology, Cancer Hospital Affiliated to Zhengzhou University, Zhengzhou, China
| |
Collapse
|
25
|
Milionis V, Goutas D, Vlachodimitropoulos D, Katsoulas N, Kyriazis ID, Liatsikos EN, Marinakis N, Joanne T, Lazaris AC, Goutas N. SDH-deficient renal cell carcinoma: A case report associated with a novel germline mutation. Clin Case Rep 2021; 9:e04605. [PMID: 34703596 PMCID: PMC8522490 DOI: 10.1002/ccr3.4605] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 05/20/2021] [Accepted: 06/28/2021] [Indexed: 11/24/2022] Open
Abstract
The highly syndromic nature of succinate dehydrogenase-deficient RCCs constitutes their active surveillance and molecular profiling the alpha and omega.
Collapse
Affiliation(s)
| | - Dimitrios Goutas
- First Department of PathologySchool of MedicineThe National and Kapodistrian University of Athens–"Laikon" General Hospital of AthensAthenesGreece
| | - Dimitrios Vlachodimitropoulos
- Istomedica S.AAthensGreece
- Laboratory of Forensic Medicine and ToxicologyThe National and Kapodistrian University of AthensAthensGreece
| | - Nikolaos Katsoulas
- First Department of PathologySchool of MedicineThe National and Kapodistrian University of Athens–"Laikon" General Hospital of AthensAthenesGreece
| | | | | | - Nikolaos Marinakis
- Laboratory of Medical GeneticsNational and Kapodistrian University of AthensSt. Sophia Children's HospitalAthensGreece
| | - Traeger‐Synodinos Joanne
- Laboratory of Medical GeneticsNational and Kapodistrian University of AthensSt. Sophia Children's HospitalAthensGreece
| | - Andreas C. Lazaris
- First Department of PathologySchool of MedicineThe National and Kapodistrian University of Athens–"Laikon" General Hospital of AthensAthenesGreece
| | - Nikolaos Goutas
- Istomedica S.AAthensGreece
- Laboratory of Forensic Medicine and ToxicologyThe National and Kapodistrian University of AthensAthensGreece
| |
Collapse
|
26
|
Consolidating biallelic SDHD variants as a cause of mitochondrial complex II deficiency. Eur J Hum Genet 2021; 29:1570-1576. [PMID: 34012134 PMCID: PMC8484551 DOI: 10.1038/s41431-021-00887-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2020] [Revised: 02/18/2021] [Accepted: 03/30/2021] [Indexed: 02/07/2023] Open
Abstract
Isolated mitochondrial complex II deficiency is a rare cause of mitochondrial respiratory chain disease. To date biallelic variants in three genes encoding mitochondrial complex II molecular components have been unequivocally associated with mitochondrial disease (SDHA/SDHB/SDHAF1). Additionally, variants in one further complex II component (SDHD) have been identified as a candidate cause of isolated mitochondrial complex II deficiency in just two unrelated affected individuals with clinical features consistent with mitochondrial disease, including progressive encephalomyopathy and lethal infantile cardiomyopathy. We present clinical and genomic investigations in four individuals from an extended Palestinian family with clinical features consistent with an autosomal recessive mitochondrial complex II deficiency, in which our genomic studies identified a homozygous NM_003002.3:c.[205 G > A];[205 G > A];p.[(Glu69Lys)];[(Glu69Lys)] SDHD variant as the likely cause. Reviewing previously published cases, these findings consolidate disruption of SDHD function as a cause of mitochondrial complex II deficiency and further define the phenotypic spectrum associated with SDHD gene variants.
Collapse
|
27
|
Zhang Y, Wu Z, Feng M, Chen J, Qin M, Wang W, Bao Y, Xu Q, Ye Y, Ma C, Jiang CZ, Gan SS, Zhou H, Cai Y, Hong B, Gao J, Ma N. The circadian-controlled PIF8-BBX28 module regulates petal senescence in rose flowers by governing mitochondrial ROS homeostasis at night. THE PLANT CELL 2021; 33:2716-2735. [PMID: 34043798 PMCID: PMC8408477 DOI: 10.1093/plcell/koab152] [Citation(s) in RCA: 55] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/23/2020] [Accepted: 05/19/2021] [Indexed: 05/20/2023]
Abstract
Reactive oxygen species (ROS) are unstable reactive molecules that are toxic to cells. Regulation of ROS homeostasis is crucial to protect cells from dysfunction, senescence, and death. In plant leaves, ROS are mainly generated from chloroplasts and are tightly temporally restricted by the circadian clock. However, little is known about how ROS homeostasis is regulated in nonphotosynthetic organs, such as petals. Here, we showed that hydrogen peroxide (H2O2) levels exhibit typical circadian rhythmicity in rose (Rosa hybrida) petals, consistent with the measured respiratory rate. RNA-seq and functional screening identified a B-box gene, RhBBX28, whose expression was associated with H2O2 rhythms. Silencing RhBBX28 accelerated flower senescence and promoted H2O2 accumulation at night in petals, while overexpression of RhBBX28 had the opposite effects. RhBBX28 influenced the expression of various genes related to respiratory metabolism, including the TCA cycle and glycolysis, and directly repressed the expression of SUCCINATE DEHYDROGENASE 1, which plays a central role in mitochondrial ROS (mtROS) homeostasis. We also found that PHYTOCHROME-INTERACTING FACTOR8 (RhPIF8) could activate RhBBX28 expression to control H2O2 levels in petals and thus flower senescence. Our results indicate that the circadian-controlled RhPIF8-RhBBX28 module is a critical player that controls flower senescence by governing mtROS homeostasis in rose.
Collapse
Affiliation(s)
- Yi Zhang
- Department of Ornamental Horticulture, State Key Laboratory of Agrobiotechnology, Beijing Key Laboratory of Development and Quality Control of Ornamental Crops, College of Horticulture, China Agricultural University, Beijing, 100193, China
| | - Zhicheng Wu
- Department of Ornamental Horticulture, State Key Laboratory of Agrobiotechnology, Beijing Key Laboratory of Development and Quality Control of Ornamental Crops, College of Horticulture, China Agricultural University, Beijing, 100193, China
| | - Ming Feng
- Department of Ornamental Horticulture, State Key Laboratory of Agrobiotechnology, Beijing Key Laboratory of Development and Quality Control of Ornamental Crops, College of Horticulture, China Agricultural University, Beijing, 100193, China
| | - Jiwei Chen
- Department of Ornamental Horticulture, State Key Laboratory of Agrobiotechnology, Beijing Key Laboratory of Development and Quality Control of Ornamental Crops, College of Horticulture, China Agricultural University, Beijing, 100193, China
| | - Meizhu Qin
- Department of Ornamental Horticulture, State Key Laboratory of Agrobiotechnology, Beijing Key Laboratory of Development and Quality Control of Ornamental Crops, College of Horticulture, China Agricultural University, Beijing, 100193, China
| | - Wenran Wang
- Department of Ornamental Horticulture, State Key Laboratory of Agrobiotechnology, Beijing Key Laboratory of Development and Quality Control of Ornamental Crops, College of Horticulture, China Agricultural University, Beijing, 100193, China
| | - Ying Bao
- Faculty of Life Science, Tangshan Normal University, Tangshan, 063000, Hebei, China
| | - Qian Xu
- Department of Ornamental Horticulture, State Key Laboratory of Agrobiotechnology, Beijing Key Laboratory of Development and Quality Control of Ornamental Crops, College of Horticulture, China Agricultural University, Beijing, 100193, China
| | - Ying Ye
- Department of Ornamental Horticulture, State Key Laboratory of Agrobiotechnology, Beijing Key Laboratory of Development and Quality Control of Ornamental Crops, College of Horticulture, China Agricultural University, Beijing, 100193, China
| | - Chao Ma
- Department of Ornamental Horticulture, State Key Laboratory of Agrobiotechnology, Beijing Key Laboratory of Development and Quality Control of Ornamental Crops, College of Horticulture, China Agricultural University, Beijing, 100193, China
| | - Cai-Zhong Jiang
- United States Department of Agriculture, Crop Pathology and Genetic Research Unit, Agricultural Research Service, University of California, Davis, CA, USA
- Department of Plant Sciences, University of California, Davis, CA, USA
| | - Su-Sheng Gan
- Plant Biology Section, School of Integrative Plant Science, College of Agriculture and Life Sciences, Cornell University, Ithaca, NY, USA
| | - Hougao Zhou
- College of Agriculture and Biology, Zhongkai University of Agriculture and Engineering, Guangzhou, 510225, China
| | - Youming Cai
- Shanghai Academy of Agricultural Sciences, Shanghai, 201403, China
| | - Bo Hong
- Department of Ornamental Horticulture, State Key Laboratory of Agrobiotechnology, Beijing Key Laboratory of Development and Quality Control of Ornamental Crops, College of Horticulture, China Agricultural University, Beijing, 100193, China
| | - Junping Gao
- Department of Ornamental Horticulture, State Key Laboratory of Agrobiotechnology, Beijing Key Laboratory of Development and Quality Control of Ornamental Crops, College of Horticulture, China Agricultural University, Beijing, 100193, China
| | - Nan Ma
- Department of Ornamental Horticulture, State Key Laboratory of Agrobiotechnology, Beijing Key Laboratory of Development and Quality Control of Ornamental Crops, College of Horticulture, China Agricultural University, Beijing, 100193, China
- Author for correspondence:
| |
Collapse
|
28
|
Bakare AB, Lesnefsky EJ, Iyer S. Leigh Syndrome: A Tale of Two Genomes. Front Physiol 2021; 12:693734. [PMID: 34456746 PMCID: PMC8385445 DOI: 10.3389/fphys.2021.693734] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2021] [Accepted: 07/22/2021] [Indexed: 12/21/2022] Open
Abstract
Leigh syndrome is a rare, complex, and incurable early onset (typically infant or early childhood) mitochondrial disorder with both phenotypic and genetic heterogeneity. The heterogeneous nature of this disorder, based in part on the complexity of mitochondrial genetics, and the significant interactions between the nuclear and mitochondrial genomes has made it particularly challenging to research and develop therapies. This review article discusses some of the advances that have been made in the field to date. While the prognosis is poor with no current substantial treatment options, multiple studies are underway to understand the etiology, pathogenesis, and pathophysiology of Leigh syndrome. With advances in available research tools leading to a better understanding of the mitochondria in health and disease, there is hope for novel treatment options in the future.
Collapse
Affiliation(s)
- Ajibola B. Bakare
- Department of Biological Sciences, J. William Fulbright College of Arts and Sciences, University of Arkansas, Fayetteville, AR, United States
| | - Edward J. Lesnefsky
- Division of Cardiology, Pauley Heart Center, Department of Internal Medicine, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
- Department of Physiology/Biophysics, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
- Department of Biochemistry and Molecular Biology, School of Medicine, Virginia Commonwealth University, Richmond, VA, United States
| | - Shilpa Iyer
- Department of Biological Sciences, J. William Fulbright College of Arts and Sciences, University of Arkansas, Fayetteville, AR, United States
| |
Collapse
|
29
|
Briscione MA, Dinasarapu AR, Bagchi P, Donsante Y, Roman KM, Downs AM, Fan X, Hoehner J, Jinnah HA, Hess EJ. Differential expression of striatal proteins in a mouse model of DOPA-responsive dystonia reveals shared mechanisms among dystonic disorders. Mol Genet Metab 2021; 133:352-361. [PMID: 34092491 PMCID: PMC8292208 DOI: 10.1016/j.ymgme.2021.05.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Revised: 05/28/2021] [Accepted: 05/28/2021] [Indexed: 11/23/2022]
Abstract
Dystonia is characterized by involuntary muscle contractions that cause debilitating twisting movements and postures. Although dysfunction of the basal ganglia, a brain region that mediates movement, is implicated in many forms of dystonia, the underlying mechanisms are unclear. The inherited metabolic disorder DOPA-responsive dystonia is considered a prototype for understanding basal ganglia dysfunction in dystonia because it is caused by mutations in genes necessary for the synthesis of the neurotransmitter dopamine, which mediates the activity of the basal ganglia. Therefore, to reveal abnormal striatal cellular processes and pathways implicated in dystonia, we used an unbiased proteomic approach in a knockin mouse model of DOPA-responsive dystonia, a model in which the striatum is known to play a central role in the expression of dystonia. Fifty-seven of the 1805 proteins identified were differentially regulated in DOPA-responsive dystonia mice compared to control mice. Most differentially regulated proteins were associated with gene ontology terms that implicated either mitochondrial or synaptic dysfunction whereby proteins associated with mitochondrial function were generally over-represented and proteins associated with synaptic function were largely under-represented. Remarkably, nearly 20% of the differentially regulated striatal proteins identified in our screen are associated with pathogenic variants that cause inherited disorders with dystonia as a sign in humans suggesting shared mechanisms across many different forms of dystonia.
Collapse
Affiliation(s)
- Maria A Briscione
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | | | - Pritha Bagchi
- Emory Integrated Proteomics Core, Emory University, Atlanta, GA, USA
| | - Yuping Donsante
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | - Kaitlyn M Roman
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | - Anthony M Downs
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | - Xueliang Fan
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA
| | - Jessica Hoehner
- Department of Biostatistics and Bioinformatics, Emory University, Atlanta, GA, USA
| | - H A Jinnah
- Department of Human Genetics, Emory University, Atlanta, GA, USA; Department of Neurology, Emory University, Atlanta, GA, USA; Department of Pediatrics, Emory University, Atlanta, GA, USA
| | - Ellen J Hess
- Department of Pharmacology and Chemical Biology, Emory University, Atlanta, GA, USA; Department of Neurology, Emory University, Atlanta, GA, USA.
| |
Collapse
|
30
|
Sturrock BRH, Macnamara EF, McGuire P, Kruk S, Yang I, Murphy J, Tifft CJ, Gordon‐Lipkin E. Progressive cerebellar atrophy in a patient with complex II and III deficiency and a novel deleterious variant in SDHA: A Counseling Conundrum. Mol Genet Genomic Med 2021; 9:e1692. [PMID: 33960148 PMCID: PMC8222855 DOI: 10.1002/mgg3.1692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2020] [Revised: 03/08/2021] [Accepted: 03/19/2021] [Indexed: 11/21/2022] Open
Abstract
BACKGROUND Complex II is an essential component of the electron transport chain, linking it with the tricarboxylic acid cycle. Its four subunits are encoded in the nuclear genome, and deleterious variants in these genes, including SDHA (OMIM 600857), are associated with a wide range of symptoms including neurological disease, cardiomyopathy, and neoplasia (paraganglioma-pheochromocytomas (PGL/PCC), and gastrointestinal stromal tumors). Deleterious variants of SDHA are most frequently associated with Leigh and Leigh-like syndromes. METHODS AND RESULTS Here, we describe a case of a 9-year-old boy with tremor, nystagmus, hypotonia, developmental delay, significant ataxia, and progressive cerebellar atrophy. He was found to have biallelic variants in SDHA, a known pathogenic variant (c.91C>T (p.R31*)), and a variant of unknown significance (c.454G>A (p.E152K)). Deficient activity of complexes II and III was detected in fibroblasts from the patient consistent with a diagnosis of a respiratory chain disorder. CONCLUSION We, therefore, consider whether c.454G>A (p.E152K) is, indeed, a pathogenic variant, and what implications it has for family members who carry the same variant.
Collapse
Affiliation(s)
- Beattie R. H. Sturrock
- National Institutes of Health Undiagnosed Diseases ProgramCommon FundOffice of the DirectorNIHBethesdaMDUSA
- Brighton and Sussex University Hospitals NHS TrustBrightonEngland
| | - Ellen F. Macnamara
- National Institutes of Health Undiagnosed Diseases ProgramCommon FundOffice of the DirectorNIHBethesdaMDUSA
| | - Peter McGuire
- Metabolism, Infection and Immunity SectionNational Human Genome Research InstituteNIHBethesdaMDUSA
| | - Shannon Kruk
- Metabolism, Infection and Immunity SectionNational Human Genome Research InstituteNIHBethesdaMDUSA
| | - Ivan Yang
- Metabolism, Infection and Immunity SectionNational Human Genome Research InstituteNIHBethesdaMDUSA
| | - Jennifer Murphy
- National Institutes of Health Undiagnosed Diseases ProgramCommon FundOffice of the DirectorNIHBethesdaMDUSA
| | - Cyndi J. Tifft
- National Institutes of Health Undiagnosed Diseases ProgramCommon FundOffice of the DirectorNIHBethesdaMDUSA
- Office of the Clinical DirectorNational Human Genome Research InstituteNIHBethesdaMDUSA
| | - Eliza Gordon‐Lipkin
- Metabolism, Infection and Immunity SectionNational Human Genome Research InstituteNIHBethesdaMDUSA
| |
Collapse
|
31
|
Zehavi Y, Saada A, Jabaly-Habib H, Dessau M, Shaag A, Elpeleg O, Spiegel R. A novel de novo heterozygous pathogenic variant in the SDHA gene results in childhood onset bilateral optic atrophy and cognitive impairment. Metab Brain Dis 2021; 36:581-588. [PMID: 33471299 DOI: 10.1007/s11011-021-00671-1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/25/2020] [Accepted: 01/08/2021] [Indexed: 10/24/2022]
Abstract
Isolated defects in the mitochondrial respiratory chain complex II (CII; succinate-ubiquinone oxidoreductase) are extremely rare and mainly result from bi-allelic mutations in one of the nuclear encoded subunits: SDHA, SDHB and SDHD, which comprise CII and the assembly CII factor SDHAF1. We report an adolescent female who presented with global developmental delay, intellectual disability and childhood onset progressive bilateral optic atrophy. Whole exome sequencing of the patient and her unaffected parents identified the novel heterozygous de novo variant c.1984C > T [NM_004168.4] in the SDHA gene. Biochemical assessment of CII in the patient's derived fibroblasts and lymphocytes displayed considerably decreased CII residual activity compared with normal controls, when normalized to the integral mitochondrial enzyme citrate synthase. Protein modeling of the consequent p.Arg662Cys variant [NP-004159.2] suggested that this substitution will compromise the structural integrity of the FAD-binding protein at the C-terminus that will ultimately impair the FAD binding to SDHA, thus decreasing the entire CII activity. Our study emphasizes the role of certain heterozygous SDHA mutations in a distinct clinical phenotype dominated by optic atrophy and neurological impairment. This is the second mutation that has been reported to cause this phenotype. Furthermore, it adds developmental delay and cognitive disability to the expanding spectrum of the disorder. We propose to add SDHA to next generation sequencing gene panels of optic atrophy.
Collapse
Affiliation(s)
- Yoav Zehavi
- Pediatric Department B' Emek Medical Center, 1834111, Afula, Israel
- Rappaport School of Medicine Technion, Haifa, Israel
| | - Ann Saada
- Faculty of Medicine, Hebrew University, Jerusalem, Israel
- Department of Human Genetics, Hadassah Medical Center, Jerusalem, Israel
| | | | - Moshe Dessau
- Azrieli Faculty of Medicine, Bar-Ilan University, Safed, Israel
| | - Avraham Shaag
- Department of Human Genetics, Hadassah Medical Center, Jerusalem, Israel
| | - Orly Elpeleg
- Faculty of Medicine, Hebrew University, Jerusalem, Israel
- Department of Human Genetics, Hadassah Medical Center, Jerusalem, Israel
| | - Ronen Spiegel
- Pediatric Department B' Emek Medical Center, 1834111, Afula, Israel.
- Rappaport School of Medicine Technion, Haifa, Israel.
| |
Collapse
|
32
|
Blackout in the powerhouse: clinical phenotypes associated with defects in the assembly of OXPHOS complexes and the mitoribosome. Biochem J 2021; 477:4085-4132. [PMID: 33151299 PMCID: PMC7657662 DOI: 10.1042/bcj20190767] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2020] [Revised: 09/29/2020] [Accepted: 10/05/2020] [Indexed: 12/26/2022]
Abstract
Mitochondria produce the bulk of the energy used by almost all eukaryotic cells through oxidative phosphorylation (OXPHOS) which occurs on the four complexes of the respiratory chain and the F1–F0 ATPase. Mitochondrial diseases are a heterogenous group of conditions affecting OXPHOS, either directly through mutation of genes encoding subunits of OXPHOS complexes, or indirectly through mutations in genes encoding proteins supporting this process. These include proteins that promote assembly of the OXPHOS complexes, the post-translational modification of subunits, insertion of cofactors or indeed subunit synthesis. The latter is important for all 13 of the proteins encoded by human mitochondrial DNA, which are synthesised on mitochondrial ribosomes. Together the five OXPHOS complexes and the mitochondrial ribosome are comprised of more than 160 subunits and many more proteins support their biogenesis. Mutations in both nuclear and mitochondrial genes encoding these proteins have been reported to cause mitochondrial disease, many leading to defective complex assembly with the severity of the assembly defect reflecting the severity of the disease. This review aims to act as an interface between the clinical and basic research underpinning our knowledge of OXPHOS complex and ribosome assembly, and the dysfunction of this process in mitochondrial disease.
Collapse
|
33
|
Saneto RP. Mitochondrial diseases: expanding the diagnosis in the era of genetic testing. JOURNAL OF TRANSLATIONAL GENETICS AND GENOMICS 2020; 4:384-428. [PMID: 33426505 PMCID: PMC7791531 DOI: 10.20517/jtgg.2020.40] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Mitochondrial diseases are clinically and genetically heterogeneous. These diseases were initially described a little over three decades ago. Limited diagnostic tools created disease descriptions based on clinical, biochemical analytes, neuroimaging, and muscle biopsy findings. This diagnostic mechanism continued to evolve detection of inherited oxidative phosphorylation disorders and expanded discovery of mitochondrial physiology over the next two decades. Limited genetic testing hampered the definitive diagnostic identification and breadth of diseases. Over the last decade, the development and incorporation of massive parallel sequencing has identified approximately 300 genes involved in mitochondrial disease. Gene testing has enlarged our understanding of how genetic defects lead to cellular dysfunction and disease. These findings have expanded the understanding of how mechanisms of mitochondrial physiology can induce dysfunction and disease, but the complete collection of disease-causing gene variants remains incomplete. This article reviews the developments in disease gene discovery and the incorporation of gene findings with mitochondrial physiology. This understanding is critical to the development of targeted therapies.
Collapse
Affiliation(s)
- Russell P. Saneto
- Center for Integrative Brain Research, Neuroscience Institute, Seattle, WA 98101, USA
- Department of Neurology/Division of Pediatric Neurology, Seattle Children’s Hospital/University of Washington, Seattle, WA 98105, USA
| |
Collapse
|
34
|
Fullerton M, McFarland R, Taylor RW, Alston CL. The genetic basis of isolated mitochondrial complex II deficiency. Mol Genet Metab 2020; 131:53-65. [PMID: 33162331 PMCID: PMC7758838 DOI: 10.1016/j.ymgme.2020.09.009] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/30/2020] [Accepted: 09/30/2020] [Indexed: 11/21/2022]
Abstract
Mitochondrial complex II (succinate:ubiquinone oxidoreductase) is the smallest complex of the oxidative phosphorylation system, a tetramer of just 140 kDa. Despite its diminutive size, it is a key complex in two coupled metabolic pathways - it oxidises succinate to fumarate in the tricarboxylic acid cycle and the electrons are used to reduce FAD to FADH2, ultimately reducing ubiquinone to ubiquinol in the respiratory chain. The biogenesis and assembly of complex II is facilitated by four ancillary proteins, all of which are autosomally-encoded. Numerous pathogenic defects have been reported which describe two broad clinical manifestations, either susceptibility to cancer in the case of single, heterozygous germline variants, or a mitochondrial disease presentation, almost exclusively due to bi-allelic recessive variants and associated with an isolated complex II deficiency. Here we present a compendium of pathogenic gene variants that have been documented in the literature in patients with an isolated mitochondrial complex II deficiency. To date, 61 patients are described, harbouring 32 different pathogenic variants in four distinct complex II genes: three structural subunit genes (SDHA, SDHB and SDHD) and one assembly factor gene (SDHAF1). Many pathogenic variants result in a null allele due to nonsense, frameshift or splicing defects however, the missense variants that do occur tend to induce substitutions at highly conserved residues in regions of the proteins that are critical for binding to other subunits or substrates. There is phenotypic heterogeneity associated with defects in each complex II gene, similar to other mitochondrial diseases.
Collapse
Affiliation(s)
- Millie Fullerton
- Wellcome Centre for Mitochondrial Research, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK
| | - Robert McFarland
- Wellcome Centre for Mitochondrial Research, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK; Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK; NHS Highly Specialised Services for Rare Mitochondrial Disorders, Royal Victoria Infirmary, Newcastle upon Tyne Hospitals NHS Foundation Trust, Queen Victoria Road, Newcastle upon Tyne, NE1 4LP, UK
| | - Robert W Taylor
- Wellcome Centre for Mitochondrial Research, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK; Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK; NHS Highly Specialised Services for Rare Mitochondrial Disorders, Royal Victoria Infirmary, Newcastle upon Tyne Hospitals NHS Foundation Trust, Queen Victoria Road, Newcastle upon Tyne, NE1 4LP, UK
| | - Charlotte L Alston
- Wellcome Centre for Mitochondrial Research, Newcastle University, Framlington Place, Newcastle upon Tyne, NE2 4HH, UK; Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, NE2 4HH, UK; NHS Highly Specialised Services for Rare Mitochondrial Disorders, Royal Victoria Infirmary, Newcastle upon Tyne Hospitals NHS Foundation Trust, Queen Victoria Road, Newcastle upon Tyne, NE1 4LP, UK.
| |
Collapse
|
35
|
Zhu SC, Chen C, Wu YN, Ahmed M, Kitmitto A, Greenstein AS, Kim SJ, Shao YF, Zhang YH. Cardiac complex II activity is enhanced by fat and mediates greater mitochondrial oxygen consumption following hypoxic re-oxygenation. Pflugers Arch 2020; 472:367-374. [DOI: 10.1007/s00424-020-02355-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 01/17/2020] [Accepted: 01/30/2020] [Indexed: 12/13/2022]
|
36
|
Moosavi B, Zhu XL, Yang WC, Yang GF. Molecular pathogenesis of tumorigenesis caused by succinate dehydrogenase defect. Eur J Cell Biol 2020; 99:151057. [DOI: 10.1016/j.ejcb.2019.151057] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2019] [Revised: 09/19/2019] [Accepted: 10/29/2019] [Indexed: 12/14/2022] Open
|
37
|
Maekawa S, Takada S, Furihata T, Fukushima A, Yokota T, Kinugawa S. Mitochondrial respiration of complex II is not lower than that of complex I in mouse skeletal muscle. Biochem Biophys Rep 2019; 21:100717. [PMID: 31890905 PMCID: PMC6928343 DOI: 10.1016/j.bbrep.2019.100717] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2019] [Revised: 11/21/2019] [Accepted: 12/06/2019] [Indexed: 02/07/2023] Open
Abstract
Skeletal muscle (SKM) requires a large amount of energy, which is produced mainly by mitochondria, for their daily functioning. Of the several mitochondrial complexes, it has been reported that the dysfunction of complex II is associated with several diseases, including myopathy. However, the degree to which complex II contributes to ATP production by mitochondria remains unknown. As complex II is not included in supercomplexes, which are formed to produce ATP efficiently, we hypothesized that complex II-linked respiration was lower than that of complex I. In addition, differences in the characteristics of complex I and II activity suggest that different factors might regulate their function. The isolated mitochondria from gastrocnemius muscle was used for mitochondrial respiration measurement and immunoblotting in male C57BL/6J mice. Student paired t-tests were performed to compare means between two groups. A univariate linear regression model was used to determine the correlation between mitochondrial respiration and proteins. Contrary to our hypothesis, complex II-linked respiration was not significantly less than complex I-linked respiration in SKM mitochondria (complex I vs complex II, 3402 vs 2840 pmol/[s × mg]). Complex I-linked respiration correlated with the amount of complex I incorporated in supercomplexes (r = 0.727, p < 0.05), but not with the total amount of complex I subunits. In contrast, complex II-linked respiration correlated with the total amount of complex II (r = 0.883, p < 0.05), but not with the amount of each complex II subunit. We conclude that both complex I and II play important roles in mitochondrial respiration and that the assembly of both supercomplexes and complex II is essential for the normal functioning of complex I and II in mouse SKM mitochondria. Complex II-linked respiration was comparable to complex I-linked respiration in isolated skeletal muscle mitochondria. Complex I-linked respiration correlated with the amount of complex I incorporated in supercomplexes, but not with the complex I subunit. Complex II-linked respiration correlated with the amount of complex II, but not with the SDH subunit.
Collapse
Affiliation(s)
- Satoshi Maekawa
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Shingo Takada
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan.,Faculty of Lifelong Sport, Department of Sports Education, Hokusho University, Ebetsu, Japan
| | - Takaaki Furihata
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Arata Fukushima
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Takashi Yokota
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| | - Shintaro Kinugawa
- Department of Cardiovascular Medicine, Hokkaido University Graduate School of Medicine, Sapporo, Japan
| |
Collapse
|
38
|
Siebers EM, Choi MJ, Tinklenberg JA, Beatka MJ, Ayres S, Meng H, Helbling DC, Takizawa A, Bennett B, Garces AM, Dias Duarte Machado LG, Dimmock D, Dwinell MR, Geurts AM, Lawlor MW. Sdha+/- Rats Display Minimal Muscle Pathology Without Significant Behavioral or Biochemical Abnormalities. J Neuropathol Exp Neurol 2019; 77:665-672. [PMID: 29850869 DOI: 10.1093/jnen/nly042] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Mitochondrial diseases (MDs) result from alteration of the mitochondrial respiratory chain (MRC) function. Despite the prevalence of MDs in the population, the paucity of animal models available limits the understanding of these disorders. Mutations in SDHA, a gene that codes for the alpha subunit of succinate dehydrogenase (SDH), can cause some forms of MD. SDHA is a crucial contributor to MRC function. In order to expand the range of MD animal models available, we attempted to generate a Sdha knockout rat. Since homozygous Sdha-/- rats could neither be identified in newborn litters, nor as early as embryonic day 14, we evaluated wild-type (WT) and heterozygous Sdha+/- genotypes. No differences in behavioral, biochemical, or molecular evaluations were observed between WT and Sdha+/- rats at 6 weeks or 6 months of age. However, 30% of Sdha+/- rats displayed mild muscle fiber atrophy with rare fibers negative for cytochrome oxidase and SDH on histochemical staining. Collectively, our data provide additional evidence that modeling SDH mutations in rodents may be challenging due to animal viability, and heterozygous rats are insufficiently symptomatic at a phenotypic and molecular level to be of significant use in the study of SDH deficiency.
Collapse
Affiliation(s)
- Emily M Siebers
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine.,Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Melinda J Choi
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine.,Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Jennifer A Tinklenberg
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine.,Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Margaret J Beatka
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine.,Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Samuel Ayres
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine.,Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Hui Meng
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine.,Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Daniel C Helbling
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine.,Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Akiko Takizawa
- Department of Physiology.,Human and Molecular Genetic Center
| | - Brian Bennett
- Department of Physics, Marquette University, Milwaukee, Wisconsin
| | | | | | - David Dimmock
- Rady Children's Institute for Genomic Medicine, San Diego, California
| | | | - Aron M Geurts
- Department of Physics, Marquette University, Milwaukee, Wisconsin.,Human and Molecular Genetic Center.,Cardiovascular Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| | - Michael W Lawlor
- Division of Pediatric Pathology, Department of Pathology and Laboratory Medicine.,Neuroscience Research Center, Medical College of Wisconsin, Milwaukee, Wisconsin
| |
Collapse
|
39
|
Moosavi B, Berry EA, Zhu XL, Yang WC, Yang GF. The assembly of succinate dehydrogenase: a key enzyme in bioenergetics. Cell Mol Life Sci 2019; 76:4023-4042. [PMID: 31236625 PMCID: PMC11105593 DOI: 10.1007/s00018-019-03200-7] [Citation(s) in RCA: 91] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 05/30/2019] [Accepted: 06/17/2019] [Indexed: 12/12/2022]
Abstract
Succinate dehydrogenase (SDH) also known as complex II or succinate:quinone oxidoreductase is an enzyme involved in both oxidative phosphorylation and tricarboxylic acid cycle; the processes that generate energy. SDH is a multi-subunit enzyme which requires a series of proteins for its proper assembly at several steps. This enzyme has medical significance as there is a broad range of human diseases from cancers to neurodegeneration related to SDH malfunction. Some of these disorders have recently been linked to defective assembly factors, reinvigorating further research in this area. Apart from that this enzyme has agricultural importance as many fungicides have been/will be designed targeting specifically this enzyme in plant fungal pathogens. In addition, we speculate it might be possible to design novel fungicides specifically targeting fungal assembly factors. Considering the medical and agricultural implications of SDH, the aim of this review is an overview of the SDH assembly factors and critical analysis of controversial issues around them.
Collapse
Affiliation(s)
- Behrooz Moosavi
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, College of Chemistry, Central China Normal University, Wuhan, 430079, People's Republic of China
| | - Edward A Berry
- Department of Biochemistry and Molecular Biology, SUNY Upstate Medical University, Syracuse, NY, 13210, USA
| | - Xiao-Lei Zhu
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, College of Chemistry, Central China Normal University, Wuhan, 430079, People's Republic of China
| | - Wen-Chao Yang
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, College of Chemistry, Central China Normal University, Wuhan, 430079, People's Republic of China
| | - Guang-Fu Yang
- Key Laboratory of Pesticide and Chemical Biology, Ministry of Education, College of Chemistry, Central China Normal University, Wuhan, 430079, People's Republic of China.
| |
Collapse
|
40
|
Almomani R, Herkert JC, Posafalvi A, Post JG, Boven LG, van der Zwaag PA, Willems PHGM, van Veen-Hof IH, Verhagen JMA, Wessels MW, Nikkels PGJ, Wintjes LT, van den Berg MP, Sinke RJ, Rodenburg RJ, Niezen-Koning KE, van Tintelen JP, Jongbloed JDH. Homozygous damaging SOD2 variant causes lethal neonatal dilated cardiomyopathy. J Med Genet 2019; 57:23-30. [PMID: 31494578 DOI: 10.1136/jmedgenet-2019-106330] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 07/22/2019] [Accepted: 07/29/2019] [Indexed: 01/05/2023]
Abstract
BACKGROUND Idiopathic dilated cardiomyopathy (DCM) is recognised to be a heritable disorder, yet clinical genetic testing does not produce a diagnosis in >50% of paediatric patients. Identifying a genetic cause is crucial because this knowledge can affect management options, cardiac surveillance in relatives and reproductive decision-making. In this study, we sought to identify the underlying genetic defect in a patient born to consanguineous parents with rapidly progressive DCM that led to death in early infancy. METHODS AND RESULTS Exome sequencing revealed a potentially pathogenic, homozygous missense variant, c.542G>T, p.(Gly181Val), in SOD2. This gene encodes superoxide dismutase 2 (SOD2) or manganese-superoxide dismutase, a mitochondrial matrix protein that scavenges oxygen radicals produced by oxidation-reduction and electron transport reactions occurring in mitochondria via conversion of superoxide anion (O2 -·) into H2O2. Measurement of hydroethidine oxidation showed a significant increase in O2 -· levels in the patient's skin fibroblasts, as compared with controls, and this was paralleled by reduced catalytic activity of SOD2 in patient fibroblasts and muscle. Lentiviral complementation experiments demonstrated that mitochondrial SOD2 activity could be completely restored on transduction with wild type SOD2. CONCLUSION Our results provide evidence that defective SOD2 may lead to toxic increases in the levels of damaging oxygen radicals in the neonatal heart, which can result in rapidly developing heart failure and death. We propose SOD2 as a novel nuclear-encoded mitochondrial protein involved in severe human neonatal cardiomyopathy, thus expanding the wide range of genetic factors involved in paediatric cardiomyopathies.
Collapse
Affiliation(s)
- Rowida Almomani
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, Jordan University of Science and Technology, Irbid, Jordan
| | - Johanna C Herkert
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Anna Posafalvi
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan G Post
- Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Ludolf G Boven
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Paul A van der Zwaag
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Peter H G M Willems
- Department of Biochemistry, Radboud Center for Mitochondrial Medicine, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Ingrid H van Veen-Hof
- Laboratory of Metabolic Diseases, Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Judith M A Verhagen
- Department of Clinical Genetics, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Marja W Wessels
- Department of Clinical Genetics, Erasmus Medical Center, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Peter G J Nikkels
- Department of Pathology, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Liesbeth T Wintjes
- Department of Paediatrics, Radboud Center for Mitochondrial Medicine, Translational Metabolic Laboratory, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Maarten P van den Berg
- Department of Cardiology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Richard J Sinke
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Richard J Rodenburg
- Department of Paediatrics, Radboud Center for Mitochondrial Medicine, Translational Metabolic Laboratory, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Klary E Niezen-Koning
- Laboratory of Metabolic Diseases, Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - J Peter van Tintelen
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands.,Department of Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Jan D H Jongbloed
- Department of Genetics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
41
|
van Karnebeek CDM, Ramos RJ, Wen XY, Tarailo-Graovac M, Gleeson JG, Skrypnyk C, Brand-Arzamendi K, Karbassi F, Issa MY, van der Lee R, Drögemöller BI, Koster J, Rousseau J, Campeau PM, Wang Y, Cao F, Li M, Ruiter J, Ciapaite J, Kluijtmans LAJ, Willemsen MAAP, Jans JJ, Ross CJ, Wintjes LT, Rodenburg RJ, Huigen MCDG, Jia Z, Waterham HR, Wasserman WW, Wanders RJA, Verhoeven-Duif NM, Zaki MS, Wevers RA. Bi-allelic GOT2 Mutations Cause a Treatable Malate-Aspartate Shuttle-Related Encephalopathy. Am J Hum Genet 2019; 105:534-548. [PMID: 31422819 DOI: 10.1016/j.ajhg.2019.07.015] [Citation(s) in RCA: 43] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2019] [Accepted: 07/22/2019] [Indexed: 11/30/2022] Open
Abstract
Early-infantile encephalopathies with epilepsy are devastating conditions mandating an accurate diagnosis to guide proper management. Whole-exome sequencing was used to investigate the disease etiology in four children from independent families with intellectual disability and epilepsy, revealing bi-allelic GOT2 mutations. In-depth metabolic studies in individual 1 showed low plasma serine, hypercitrullinemia, hyperlactatemia, and hyperammonemia. The epilepsy was serine and pyridoxine responsive. Functional consequences of observed mutations were tested by measuring enzyme activity and by cell and animal models. Zebrafish and mouse models were used to validate brain developmental and functional defects and to test therapeutic strategies. GOT2 encodes the mitochondrial glutamate oxaloacetate transaminase. GOT2 enzyme activity was deficient in fibroblasts with bi-allelic mutations. GOT2, a member of the malate-aspartate shuttle, plays an essential role in the intracellular NAD(H) redox balance. De novo serine biosynthesis was impaired in fibroblasts with GOT2 mutations and GOT2-knockout HEK293 cells. Correcting the highly oxidized cytosolic NAD-redox state by pyruvate supplementation restored serine biosynthesis in GOT2-deficient cells. Knockdown of got2a in zebrafish resulted in a brain developmental defect associated with seizure-like electroencephalography spikes, which could be rescued by supplying pyridoxine in embryo water. Both pyridoxine and serine synergistically rescued embryonic developmental defects in zebrafish got2a morphants. The two treated individuals reacted favorably to their treatment. Our data provide a mechanistic basis for the biochemical abnormalities in GOT2 deficiency that may also hold for other MAS defects.
Collapse
Affiliation(s)
- Clara D M van Karnebeek
- Departments of Pediatrics & Clinical Genetics, Emma Children's Hospital, Amsterdam University Medical Centres, Amsterdam Gastro-enterology and Metabolism, University of Amsterdam, 1105 AZ Amsterdam, the Netherlands; Department of Pediatrics / Medical Genetics, BC Children's Hospital Research Institute, Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada; On behalf of "United for Metabolic Diseases," 1105AZ Amsterdam, the Netherlands; Amalia Children's Hospital, Department of Pediatrics, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands.
| | - Rúben J Ramos
- On behalf of "United for Metabolic Diseases," 1105AZ Amsterdam, the Netherlands; Department of Genetics, University Medical Center Utrecht, 3584 EA Utrecht, the Netherlands
| | - Xiao-Yan Wen
- Zebrafish Centre for Advanced Drug Discovery, Keenan Research Centre for Biomedical Science, Li Ka Sheng Knowledge Institute, St. Michael's Hospital, Toronto, ON M5B 1T8, Canada; Department of Medicine, Physiology and LMP & Institute of Medical Science, University of Toronto, Toronto, ON M5G 2C4, Canada
| | - Maja Tarailo-Graovac
- Departments of Biochemistry, Molecular Biology and Medical Genetics, Cumming School of Medicine, University of Calgary, Calgary, AB T2N 4N1, Canada; Alberta Children's Hospital Research Institute, University of Calgary, Calgary, AB T2N 4N1, Canada
| | - Joseph G Gleeson
- Department Neurosciences and Pediatric, Howard Hughes Medical Institute, University of California; Rady Children's Institute for Genomic Medicine, San Diego, CA 92093, USA
| | - Cristina Skrypnyk
- Department of Molecular Medicine and Al Jawhara Center for Molecular Medicine, Genetics and Inherited Diseases, College of Medicine and Medical Sciences, Arabian Gulf University, Postal Code 328, Bahrain
| | - Koroboshka Brand-Arzamendi
- Zebrafish Centre for Advanced Drug Discovery, Keenan Research Centre for Biomedical Science, Li Ka Sheng Knowledge Institute, St. Michael's Hospital, Toronto, ON M5B 1T8, Canada
| | - Farhad Karbassi
- Zebrafish Centre for Advanced Drug Discovery, Keenan Research Centre for Biomedical Science, Li Ka Sheng Knowledge Institute, St. Michael's Hospital, Toronto, ON M5B 1T8, Canada
| | - Mahmoud Y Issa
- Clinical Genetics Department, Human Genetics and Genome Research Division, National Research Centre, Cairo 12311, Egypt
| | - Robin van der Lee
- Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Britt I Drögemöller
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada; BC Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Janet Koster
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam Gastro-enterology and Metabolism, 1105 AZ Amsterdam, the Netherlands
| | - Justine Rousseau
- CHU Sainte-Justine Research Center, Montreal, QC H3T 1C5, Canada
| | | | - Youdong Wang
- Zebrafish Centre for Advanced Drug Discovery, Keenan Research Centre for Biomedical Science, Li Ka Sheng Knowledge Institute, St. Michael's Hospital, Toronto, ON M5B 1T8, Canada
| | - Feng Cao
- Department of Neuroscience & Mental Health, The Hospital for Sick Children & Department of Physiology, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Meng Li
- Zebrafish Centre for Advanced Drug Discovery, Keenan Research Centre for Biomedical Science, Li Ka Sheng Knowledge Institute, St. Michael's Hospital, Toronto, ON M5B 1T8, Canada
| | - Jos Ruiter
- Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam Gastro-enterology and Metabolism, 1105 AZ Amsterdam, the Netherlands
| | - Jolita Ciapaite
- On behalf of "United for Metabolic Diseases," 1105AZ Amsterdam, the Netherlands; Department of Genetics, University Medical Center Utrecht, 3584 EA Utrecht, the Netherlands
| | - Leo A J Kluijtmans
- On behalf of "United for Metabolic Diseases," 1105AZ Amsterdam, the Netherlands; Department of Laboratory Medicine, Translational Metabolic Laboratory, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands
| | - Michel A A P Willemsen
- On behalf of "United for Metabolic Diseases," 1105AZ Amsterdam, the Netherlands; Amalia Children's Hospital, Department of Pediatrics, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands
| | - Judith J Jans
- On behalf of "United for Metabolic Diseases," 1105AZ Amsterdam, the Netherlands; Department of Genetics, University Medical Center Utrecht, 3584 EA Utrecht, the Netherlands
| | - Colin J Ross
- Faculty of Pharmaceutical Sciences, University of British Columbia, Vancouver, BC V6T 1Z3, Canada
| | - Liesbeth T Wintjes
- On behalf of "United for Metabolic Diseases," 1105AZ Amsterdam, the Netherlands; Department of Laboratory Medicine, Translational Metabolic Laboratory, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands; Radboud Center for Mitochondrial Medicine, Department of Pediatrics, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands
| | - Richard J Rodenburg
- On behalf of "United for Metabolic Diseases," 1105AZ Amsterdam, the Netherlands; Department of Laboratory Medicine, Translational Metabolic Laboratory, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands; Amalia Children's Hospital, Department of Pediatrics, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands; Radboud Center for Mitochondrial Medicine, Department of Pediatrics, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands
| | - Marleen C D G Huigen
- On behalf of "United for Metabolic Diseases," 1105AZ Amsterdam, the Netherlands; Department of Laboratory Medicine, Translational Metabolic Laboratory, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands
| | - Zhengping Jia
- Department of Neuroscience & Mental Health, The Hospital for Sick Children & Department of Physiology, University of Toronto, Toronto, ON M5G 1X8, Canada
| | - Hans R Waterham
- On behalf of "United for Metabolic Diseases," 1105AZ Amsterdam, the Netherlands; Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam Gastro-enterology and Metabolism, 1105 AZ Amsterdam, the Netherlands
| | - Wyeth W Wasserman
- Centre for Molecular Medicine and Therapeutics, Department of Medical Genetics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Ronald J A Wanders
- On behalf of "United for Metabolic Diseases," 1105AZ Amsterdam, the Netherlands; Laboratory Genetic Metabolic Diseases, Department of Clinical Chemistry, Amsterdam University Medical Centres, University of Amsterdam, Amsterdam Gastro-enterology and Metabolism, 1105 AZ Amsterdam, the Netherlands
| | - Nanda M Verhoeven-Duif
- On behalf of "United for Metabolic Diseases," 1105AZ Amsterdam, the Netherlands; Department of Genetics, University Medical Center Utrecht, 3584 EA Utrecht, the Netherlands
| | - Maha S Zaki
- Clinical Genetics Department, Human Genetics and Genome Research Division, National Research Centre, Cairo 12311, Egypt
| | - Ron A Wevers
- On behalf of "United for Metabolic Diseases," 1105AZ Amsterdam, the Netherlands; Department of Laboratory Medicine, Translational Metabolic Laboratory, Radboud University Medical Centre, 6525 GA Nijmegen, the Netherlands.
| |
Collapse
|
42
|
Lazar IM, Karcini A, Ahuja S, Estrada-Palma C. Proteogenomic Analysis of Protein Sequence Alterations in Breast Cancer Cells. Sci Rep 2019; 9:10381. [PMID: 31316139 PMCID: PMC6637242 DOI: 10.1038/s41598-019-46897-z] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Accepted: 07/04/2019] [Indexed: 12/04/2022] Open
Abstract
Cancer evolves as a result of an accumulation of mutations and chromosomal aberrations. Developments in sequencing technologies have enabled the discovery and cataloguing of millions of such mutations. The identification of protein-level alterations, typically by using reversed-phase protein arrays or mass spectrometry, has lagged, however, behind gene and transcript-level observations. In this study, we report the use of mass spectrometry for detecting the presence of mutations-missense, indels and frame shifts-in MCF7 and SKBR3 breast cancer, and non-tumorigenic MCF10A cells. The mutations were identified by expanding the database search process of raw mass spectrometry files by including an in-house built database of mutated peptides (XMAn-v1) that complemented a minimally redundant, canonical database of Homo sapiens proteins. The work resulted in the identification of nearly 300 mutated peptide sequences, of which ~50 were characterized by quality tandem mass spectra. We describe the criteria that were used to select the mutated peptide sequences, evaluate the parameters that characterized these peptides, and assess the artifacts that could have led to false peptide identifications. Further, we discuss the functional domains and biological processes that may be impacted by the observed peptide alterations, and how protein-level detection can support the efforts of identifying cancer driving mutations and genes. Mass spectrometry data are available via ProteomeXchange with identifier PXD014458.
Collapse
Affiliation(s)
- Iulia M Lazar
- Department of Biological Sciences, Virginia Tech 1981 Kraft Drive, Blacksburg, VA, 24061, USA. .,Carilion School of Medicine and Virginia Tech 1981 Kraft Drive, Blacksburg, VA, 24061, USA.
| | - Arba Karcini
- Department of Biological Sciences, Virginia Tech 1981 Kraft Drive, Blacksburg, VA, 24061, USA
| | - Shreya Ahuja
- Department of Biological Sciences, Virginia Tech 1981 Kraft Drive, Blacksburg, VA, 24061, USA
| | - Carly Estrada-Palma
- Department of Biochemistry, Virginia Tech 1981 Kraft Drive, Blacksburg, VA, 24061, USA
| |
Collapse
|
43
|
White G, Tufton N, Akker S. First-positive surveillance screening in an asymptomatic SDHA germline mutation carrier. Endocrinol Diabetes Metab Case Rep 2019; 2019. [PMID: 31368675 PMCID: PMC6548220 DOI: 10.1530/edm-19-0005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
At least 40% of phaeochromocytomas and paraganglioma’s (PPGLs) are associated with an underlying genetic mutation. The understanding of the genetic landscape of these tumours has rapidly evolved, with 18 associated genes now identified. Among these, mutations in the subunits of succinate dehydrogenase complex (SDH) are the most common, causing around half of familial PPGL cases. Occurrence of PPGLs in carriers of SDHB, SDHC and SDHD subunit mutations has been long reported, but it is only recently that variants in the SDHA subunit have been linked to PPGL formation. Previously documented cases have, to our knowledge, only been found in isolated cases where pathogenic SDHA variants were identified retrospectively. We report the case of an asymptomatic suspected carotid body tumour found during surveillance screening in a 72-year-old female who is a known carrier of a germline SDHA pathogenic variant. To our knowledge, this is the first screen that detected PPGL found in a previously identified SDHA pathogenic variant carrier, during surveillance imaging. This finding supports the use of cascade genetic testing and surveillance screening in all carriers of a pathogenic SDHA variant.
Collapse
Affiliation(s)
- Gemma White
- Department of Endocrinology, St. Bartholomew’s Hospital, Barts Health NHS Trust, London, UK
| | - Nicola Tufton
- Department of Endocrinology, St. Bartholomew’s Hospital, Barts Health NHS Trust, London, UK
| | - Scott Akker
- Department of Endocrinology, St. Bartholomew’s Hospital, Barts Health NHS Trust, London, UK
| |
Collapse
|
44
|
Lin L, Liu Y, Fu S, Qu C, Li H, Ni J. Inhibition of Mitochondrial Complex Function-The Hepatotoxicity Mechanism of Emodin Based on Quantitative Proteomic Analyses. Cells 2019; 8:cells8030263. [PMID: 30897821 PMCID: PMC6468815 DOI: 10.3390/cells8030263] [Citation(s) in RCA: 51] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 03/12/2019] [Accepted: 03/13/2019] [Indexed: 02/07/2023] Open
Abstract
Emodin is the main component of traditional Chinese medicines including rhubarb, Polygonum multiflorum, and Polygonum cuspidatum. It has confirmed hepatotoxicity and may be the main causative agent of liver damage associated with the above-mentioned traditional Chinese medicines. However, current research does not explain the mechanism of emodin in hepatotoxicity. In this study, L02 cells were used as a model to study the mechanism of emodin-induced hepatocyte apoptosis using quantitative proteomics, and the results were verified by Western blot. A total of 662 differentially expressed proteins were discovered and analyzed using Gene Ontology (GO) and pathway enrichment analysis. The results show that the oxidative phosphorylation pathway is highly represented. Abnormalities in this pathway result in impaired mitochondrial function and represent mitochondrial damage. This result is consistent with mitochondria membrane potential measurements. Analysis of differentially expressed proteins revealed that emodin mainly affects oxidative phosphorylation pathways by inhibiting the function of the mitochondrial respiratory chain complexes; the mitochondrial respiratory chain complex activity assay result also confirmed that emodin could inhibit the activity of all mitochondrial complexes. This results in an increase in caspase-3, a decrease in mitochondrial membrane potential (MMP,) an increase in reactive oxygen species (ROS), and disorders in ATP synthesis, etc., eventually leading to mitochondrial damage and hepatocyte apoptosis in vitro.
Collapse
Affiliation(s)
- Longfei Lin
- Institute Chinese materia medica china academy of Chinese medical sciences, Beijing 100700, China.
| | - Yuling Liu
- Institute Chinese materia medica china academy of Chinese medical sciences, Beijing 100700, China.
| | - Sai Fu
- Institute Chinese materia medica china academy of Chinese medical sciences, Beijing 100700, China.
| | - Changhai Qu
- School of Chinese material medica, Beijing University of Chinese Medicine, Beijing 100102, China.
| | - Hui Li
- Institute Chinese materia medica china academy of Chinese medical sciences, Beijing 100700, China.
| | - Jian Ni
- School of Chinese material medica, Beijing University of Chinese Medicine, Beijing 100102, China.
| |
Collapse
|
45
|
Oudijk L, Gaal J, de Krijger RR. The Role of Immunohistochemistry and Molecular Analysis of Succinate Dehydrogenase in the Diagnosis of Endocrine and Non-Endocrine Tumors and Related Syndromes. Endocr Pathol 2019; 30:64-73. [PMID: 30421319 DOI: 10.1007/s12022-018-9555-2] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Succinate dehydrogenase (SDH) is an enzyme complex, composed of four protein subunits, that plays a role in both the citric acid cycle and the electron transport chain. The genes for SDHA, SDHB, SDHC, and SDHD are located in the nuclear DNA, and mutations in these genes have initially been described in paragangliomas (PGL) and pheochromocytomas (PCC), which are relatively rare tumors derived from the autonomic nervous system and the adrenal medulla, respectively. Patients with SDH mutations, that are almost exclusively in the germline, are frequently affected by multiple PGL and/or PCC. In addition, other tumors have been associated with SDH mutations as well, including gastrointestinal stromal tumors, SDH-deficient renal cell carcinoma, and pituitary adenomas. Immunohistochemistry for SDHB and SDHA has been shown to be a valuable additional tool in the histopathological analysis of these tumors, and can be considered as a surrogate marker for molecular analysis. In addition, SDHB immunohistochemistry is relevant in the decision-making whether a genetic sequence variant represents a pathogenic mutation or not. In this review, we highlight the current knowledge of the physiologic and pathologic role of the SDH enzyme complex and its involvement in endocrine and non-endocrine tumors, with an emphasis on the applicability of immunohistochemistry.
Collapse
Affiliation(s)
- Lindsey Oudijk
- Department of Pathology, Erasmus MC University Medical Center Rotterdam, Rotterdam, The Netherlands.
| | - José Gaal
- Department of Pathology, Isala Clinics, Zwolle, The Netherlands
| | - Ronald R de Krijger
- Department of Pathology, University Medical Center/Princess Maxima Center for Pediatric Oncology, Utrecht, The Netherlands
| |
Collapse
|
46
|
Iwakawa H, Fukui T, Fukuwatari T, Bamba S, Sasaki M, Tsujikawa T, Doi Y, Shibata K. Blood concentrations and renal clearance of water-soluble vitamins in outpatients with ulcerative colitis. Biomed Rep 2019; 10:202-210. [PMID: 30906550 PMCID: PMC6403479 DOI: 10.3892/br.2019.1191] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 01/21/2019] [Indexed: 12/31/2022] Open
Abstract
Few studies have investigated the association between dietary intake and blood concentrations of water-soluble vitamins in patients with ulcerative colitis (UC). In the present study, vitamin concentrations were measured in the blood and urinary excretion of 23 outpatients with UC and compared against a control group of 20 healthy participants. A weighed food record procedure was used to ensure controlled macronutrient and vitamin intakes of the UC cohort. Individuals in the control group were given a semi-purified diet for 8 days prior to assessment. Multiple linear regression analysis was used to identify important differences in vitamin concentrations, independent of sex, age and other confounding variables. The blood concentrations of vitamins B2, C, niacin and folate were markedly lower in the patients with UC than those in the control group, and the renal clearance of vitamins B1, B6, B12 and folate was notably higher in the UC cohort. It was concluded that vitamins B2, C, niacin and folate were at significantly lower concentrations in patients with UC following adjustment for coexisting factors. The lower levels of niacin may be partially due to impaired reabsorption. Chronic inflammation, common in patients with UC, with may contribute to the lower levels of other vitamins by rendering amino acid and carbohydrate metabolism into a hypermetabolic state. As the role of vitamins in metabolic activity is constant and pervasive, nutritional management including the application of water-soluble vitamins appears important for patients suffering from UC.
Collapse
Affiliation(s)
- Hiromi Iwakawa
- Department of Food Science and Human Nutrition, Faculty of Agriculture, Ryukoku University, Otsu, Shiga 520-2194, Japan
| | - Tomiho Fukui
- Department of Nutrition, Faculty of Health and Nutrition, Shubun University, Ichinomiya, Aichi 491-0938, Japan
| | - Tsutomu Fukuwatari
- Department of Nutrition, School of Human Cultures, The University of Shiga Prefecture, Hikone, Shiga 522-8533, Japan
| | - Shigeki Bamba
- Department of Medicine, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Masaya Sasaki
- Department of Medicine, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Tomoyuki Tsujikawa
- Department of Medicine, Shiga University of Medical Science, Otsu, Shiga 520-2192, Japan
| | - Yukio Doi
- Department of Food Science and Human Nutrition, Faculty of Agriculture, Ryukoku University, Otsu, Shiga 520-2194, Japan
| | - Katsumi Shibata
- Department of Nutrition, School of Human Cultures, The University of Shiga Prefecture, Hikone, Shiga 522-8533, Japan
| |
Collapse
|
47
|
Nicolas E, Demidova EV, Iqbal W, Serebriiskii IG, Vlasenkova R, Ghatalia P, Zhou Y, Rainey K, Forman AF, Dunbrack RL, Golemis EA, Hall MJ, Daly MB, Arora S. Interaction of germline variants in a family with a history of early-onset clear cell renal cell carcinoma. Mol Genet Genomic Med 2019; 7:e556. [PMID: 30680959 PMCID: PMC6418363 DOI: 10.1002/mgg3.556] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2018] [Revised: 12/06/2018] [Accepted: 12/11/2018] [Indexed: 12/31/2022] Open
Abstract
Background Identification of genetic factors causing predisposition to renal cell carcinoma has helped improve screening, early detection, and patient survival. Methods We report the characterization of a proband with renal and thyroid cancers and a family history of renal and other cancers by whole‐exome sequencing (WES), coupled with WES analysis of germline DNA from additional affected and unaffected family members. Results This work identified multiple predicted protein‐damaging variants relevant to the pattern of inherited cancer risk. Among these, the proband and an affected brother each had a heterozygous Ala45Thr variant in SDHA, a component of the succinate dehydrogenase (SDH) complex. SDH defects are associated with mitochondrial disorders and risk for various cancers; immunochemical analysis indicated loss of SDHB protein expression in the patient’s tumor, compatible with SDH deficiency. Integrated analysis of public databases and structural predictions indicated that the two affected individuals also had additional variants in genes including TGFB2, TRAP1, PARP1, and EGF, each potentially relevant to cancer risk alone or in conjunction with the SDHA variant. In addition, allelic imbalances of PARP1 and TGFB2 were detected in the tumor of the proband. Conclusion Together, these data suggest the possibility of risk associated with interaction of two or more variants.
Collapse
Affiliation(s)
- Emmanuelle Nicolas
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Elena V Demidova
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania.,Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania.,Kazan Federal University, Kazan, Russia
| | - Waleed Iqbal
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Ilya G Serebriiskii
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania.,Kazan Federal University, Kazan, Russia
| | | | - Pooja Ghatalia
- Department of Hematology/Oncology, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Yan Zhou
- Biostatistics and Bioinformatics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Kim Rainey
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Andrea F Forman
- Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Roland L Dunbrack
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Erica A Golemis
- Molecular Therapeutics Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Michael J Hall
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania.,Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Mary B Daly
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania.,Department of Clinical Genetics, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| | - Sanjeevani Arora
- Cancer Prevention and Control Program, Fox Chase Cancer Center, Philadelphia, Pennsylvania
| |
Collapse
|
48
|
Tufton N, Sahdev A, Drake WM, Akker SA. Can subunit-specific phenotypes guide surveillance imaging decisions in asymptomatic SDH mutation carriers? Clin Endocrinol (Oxf) 2019; 90:31-46. [PMID: 30303539 DOI: 10.1111/cen.13877] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 10/07/2018] [Accepted: 10/07/2018] [Indexed: 12/15/2022]
Abstract
OBJECTIVE With the discovery that familial phaeochromocytoma and paraganglioma syndrome can be caused by mutations in each subunit of the succinate dehydrogenase enzyme (SDH), has come the recognition that mutations in the individual subunits have their own distinct natural histories. Increased genetic screening is leading to the identification of increasing numbers of, mostly asymptomatic, gene mutation carriers and the implementation of screening strategies for these individuals. Yet there is, to date, no international consensus regarding screening strategies for asymptomatic carriers. DESIGN A comprehensive PubMed search from 1/1/2000 to 28/2/2018 was undertaken using multiple search terms and subsequently a manual review of references in identified papers to identify all clinically relevant cases and cohorts. In this review, the accumulated, published experience of phenotype and malignancy risks of individual SDH subunits is analysed. Where possible screening results for asymptomatic SDH mutation carriers have been analysed separately to define the penetrance in asymptomatic carriers (asymptomatic penetrance). RESULTS The combined data confirms that "asymptomatic penetrance" is highest for SDHD and when there is penetrance, the most likely site to develop a PGL is head and neck (SDHD) and extra-adrenal abdominal (SDHB). However, the risk in SDHB carriers of developing HNPGL is also high (35.5%) and a PCC is low (15.1%), and in SDHD carriers there is a high risk of developing a PCC (35.8%) or abdominal PGL (9.4%) and a small, but significant risk at other sympathetic sites. The data suggest that the risk of malignant transformation is the same for both PCC and extra-adrenal abdominal PGLs (30%-35%) in SDHB carriers. In SDHD carriers, the risk of malignant transformation was highest in HNPGLs (7.5%) and similar for sympathetic sites (3.8%-5.2%). CONCLUSIONS Using this data, we suggest surveillance screening of asymptomatic carriers can be tailored to the underlying SDH subunit and review possible surveillance programmes.
Collapse
Affiliation(s)
- Nicola Tufton
- Department of Endocrinology, St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
- Centre for Endocrinology, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Anju Sahdev
- Department of Radiology, St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
| | - William M Drake
- Department of Endocrinology, St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
- Centre for Endocrinology, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| | - Scott A Akker
- Department of Endocrinology, St Bartholomew's Hospital, Barts Health NHS Trust, London, UK
- Centre for Endocrinology, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, UK
| |
Collapse
|
49
|
Zhou Z, Austin GL, Young LEA, Johnson LA, Sun R. Mitochondrial Metabolism in Major Neurological Diseases. Cells 2018; 7:E229. [PMID: 30477120 PMCID: PMC6316877 DOI: 10.3390/cells7120229] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 11/19/2018] [Accepted: 11/21/2018] [Indexed: 01/18/2023] Open
Abstract
Mitochondria are bilayer sub-cellular organelles that are an integral part of normal cellular physiology. They are responsible for producing the majority of a cell's ATP, thus supplying energy for a variety of key cellular processes, especially in the brain. Although energy production is a key aspect of mitochondrial metabolism, its role extends far beyond energy production to cell signaling and epigenetic regulation⁻functions that contribute to cellular proliferation, differentiation, apoptosis, migration, and autophagy. Recent research on neurological disorders suggest a major metabolic component in disease pathophysiology, and mitochondria have been shown to be in the center of metabolic dysregulation and possibly disease manifestation. This review will discuss the basic functions of mitochondria and how alterations in mitochondrial activity lead to neurological disease progression.
Collapse
Affiliation(s)
- Zhengqiu Zhou
- Molecular & Cellular Biochemistry Department, University of Kentucky, Lexington, KY 40536, USA.
| | - Grant L Austin
- Molecular & Cellular Biochemistry Department, University of Kentucky, Lexington, KY 40536, USA.
| | - Lyndsay E A Young
- Molecular & Cellular Biochemistry Department, University of Kentucky, Lexington, KY 40536, USA.
| | - Lance A Johnson
- Department of Physiology, University of Kentucky, Lexington, KY 40536, USA.
| | - Ramon Sun
- Molecular & Cellular Biochemistry Department, University of Kentucky, Lexington, KY 40536, USA.
| |
Collapse
|
50
|
Dubard Gault M, Mandelker D, DeLair D, Stewart CR, Kemel Y, Sheehan MR, Siegel B, Kennedy J, Marcell V, Arnold A, Al-Ahmadie H, Modak S, Robson M, Shukla N, Roberts S, Vijai J, Topka S, Kentsis A, Cadoo K, Carlo M, Latham Schwark A, Reznik E, Dinatale R, Hechtman J, Borras Flores E, Jairam S, Yang C, Li Y, Bayraktar EC, Ceyhan-Birsoy O, Zhang L, Kohlman W, Schiffman J, Stadler Z, Birsoy K, Kung A, Offit K, Walsh MF. Germline SDHA mutations in children and adults with cancer. Cold Spring Harb Mol Case Stud 2018; 4:a002584. [PMID: 30068732 PMCID: PMC6071569 DOI: 10.1101/mcs.a002584] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Accepted: 05/10/2018] [Indexed: 11/24/2022] Open
Abstract
Mutations in succinate dehydrogenase complex genes predispose to familial paraganglioma-pheochromocytoma syndrome (FPG) and gastrointestinal stromal tumors (GIST). Here we describe cancer patients undergoing agnostic germline testing at Memorial Sloan Kettering Cancer Center and found to harbor germline SDHA mutations. Using targeted sequencing covering the cancer census genes, we identified 10 patients with SDHA germline mutations. Cancer diagnoses for these patients carrying SDHA germline mutations included neuroblastoma (n = 1), breast (n = 1), colon (n = 1), renal (n = 1), melanoma and uterine (n = 1), prostate (n = 1), endometrial (n = 1), bladder (n = 1), and gastrointestinal stromal tumor (GIST) (n = 2). Immunohistochemical staining and assessment of patient tumors for second hits and loss of heterozygosity in SDHA confirmed GIST as an SDHA-associated tumor and suggests SDHA germline mutations may be a driver in neuroblastoma tumorigenesis.
Collapse
Affiliation(s)
- Marianne Dubard Gault
- Graduate Education Memorial, Memorial Sloan Kettering Cancer Center and Weill Cornell Genetics, New York, New York 10065, USA
| | - Diana Mandelker
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Deborah DeLair
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Carolyn R Stewart
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Yelena Kemel
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
- Niehaus Center for Inherited Cancer Genomics, Clinical Genetics Service, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Margaret R Sheehan
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Beth Siegel
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Jennifer Kennedy
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Vanessa Marcell
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Angela Arnold
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Hikmat Al-Ahmadie
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Shakeel Modak
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Mark Robson
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
- Niehaus Center for Inherited Cancer Genomics, Clinical Genetics Service, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Neerav Shukla
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Stephen Roberts
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Joseph Vijai
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
- Niehaus Center for Inherited Cancer Genomics, Clinical Genetics Service, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Sabine Topka
- Niehaus Center for Inherited Cancer Genomics, Clinical Genetics Service, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Alex Kentsis
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Karen Cadoo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
- Niehaus Center for Inherited Cancer Genomics, Clinical Genetics Service, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Maria Carlo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
- Niehaus Center for Inherited Cancer Genomics, Clinical Genetics Service, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Alicia Latham Schwark
- Graduate Education Memorial, Memorial Sloan Kettering Cancer Center and Weill Cornell Genetics, New York, New York 10065, USA
| | - Ed Reznik
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Renzo Dinatale
- Sloan Kettering Institute, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Jaclyn Hechtman
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Ester Borras Flores
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Sowmaya Jairam
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Ciyu Yang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Yirong Li
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | | | - Ozge Ceyhan-Birsoy
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Liying Zhang
- Department of Pathology, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Wendy Kohlman
- Family Cancer Assessment Clinic, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, USA
| | - Joshua Schiffman
- Family Cancer Assessment Clinic, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, USA
| | - Zsofia Stadler
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
- Niehaus Center for Inherited Cancer Genomics, Clinical Genetics Service, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Kivanc Birsoy
- Family Cancer Assessment Clinic, Huntsman Cancer Institute, University of Utah, Salt Lake City, Utah 84112, USA
| | - Andrew Kung
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Kenneth Offit
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
- Niehaus Center for Inherited Cancer Genomics, Clinical Genetics Service, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| | - Michael F Walsh
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
- Niehaus Center for Inherited Cancer Genomics, Clinical Genetics Service, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, New York 10021, USA
| |
Collapse
|