Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 230
Filtrar
1.
Sci Adv ; 10(34): eado8549, 2024 Aug 23.
Artículo en Inglés | MEDLINE | ID: mdl-39167644

RESUMEN

Reduced skeletal muscle mass and oxidative capacity coexist in patients with pulmonary emphysema and are independently associated with higher mortality. If reduced cellular respiration contributes to muscle atrophy in that setting remains unknown. Using a mouse with genetically induced pulmonary emphysema that recapitulates muscle dysfunction, we found that reduced activity of succinate dehydrogenase (SDH) is a hallmark of its myopathic changes. We generated an inducible, muscle-specific SDH knockout mouse that demonstrates lower mitochondrial oxygen consumption, myofiber contractility, and exercise endurance. Respirometry analyses show that in vitro complex I respiration is unaffected by loss of SDH subunit C in muscle mitochondria, which is consistent with the pulmonary emphysema animal data. SDH knockout initially causes succinate accumulation associated with a down-regulated transcriptome but modest proteome effects. Muscle mass, myofiber type composition, and overall body mass constituents remain unaltered in the transgenic mice. Thus, while SDH regulates myofiber respiration in experimental pulmonary emphysema, it does not control muscle mass or other body constituents.


Asunto(s)
Respiración de la Célula , Ratones Noqueados , Contracción Muscular , Músculo Esquelético , Enfisema Pulmonar , Succinato Deshidrogenasa , Animales , Enfisema Pulmonar/metabolismo , Enfisema Pulmonar/genética , Enfisema Pulmonar/patología , Enfisema Pulmonar/etiología , Succinato Deshidrogenasa/metabolismo , Succinato Deshidrogenasa/genética , Ratones , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Complejo II de Transporte de Electrones/metabolismo , Complejo II de Transporte de Electrones/genética , Modelos Animales de Enfermedad , Ratones Transgénicos , Mitocondrias Musculares/metabolismo , Mitocondrias Musculares/patología , Consumo de Oxígeno
3.
J Hazard Mater ; 472: 134598, 2024 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-38743975

RESUMEN

N-(1,3-dimethylbutyl)-N'-phenyl-p-phenylenediamine quinone (6-PPDQ) is an emerging pollutant transformed from 6-PPD. However, the effect of 6-PPDQ exposure on mitochondrion and underlying mechanism remains largely unclear. Using Caenorhabditis elegans as animal model, exposed to 6-PPDQ at 0.1-10 µg/L was performed form L1 larvae to adult day-1. Exposure to 6-PPDQ (1 and 10 µg/L) could increase oxygen consumption rate and decease adenosine 5'-triphosphate (ATP) content, suggesting induction of mitochondrial dysfunction. Activities of NADH dehydrogenase (complex I) and succinate dehydrogenase (complex II) were inhibited, accompanied by a decrease in expressions of gas-1, nuo-1, and mev-1. RNAi of gas-1 and mev-1 enhanced mitochondrial dysfunction and reduced lifespan of 6-PPDQ exposed nematodes. GAS-1 and MEV-1 functioned in parallel to regulate 6-PPDQ toxicity to reduce the lifespan. Insulin peptides and the insulin signaling pathway acted downstream of GAS-1 and MEV-1 to control the 6-PPDQ toxicity on longevity. Moreover, RNAi of sod-2 and sod-3, targeted genes of daf-16, caused susceptibility to 6-PPDQ toxicity in reducing lifespan and in causing reactive oxygen species (ROS) production. Therefore, 6-PPDQ at environmentally relevant concentrations (ERCs) potentially caused mitochondrial dysfunction by affecting mitochondrial complexes I and II, which was associated with lifespan reduction by affecting insulin signaling in organisms.


Asunto(s)
Proteínas de Caenorhabditis elegans , Caenorhabditis elegans , Complejo I de Transporte de Electrón , Longevidad , Mitocondrias , Animales , Caenorhabditis elegans/efectos de los fármacos , Longevidad/efectos de los fármacos , Proteínas de Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Complejo I de Transporte de Electrón/genética , Complejo II de Transporte de Electrones/metabolismo , Complejo II de Transporte de Electrones/genética , Insulina/metabolismo , Adenosina Trifosfato/metabolismo , Especies Reactivas de Oxígeno/metabolismo , NADH Deshidrogenasa , Citocromos b
4.
Am J Physiol Cell Physiol ; 326(6): C1669-C1682, 2024 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-38646781

RESUMEN

We previously showed that the transaminase inhibitor, aminooxyacetic acid, reduced respiration energized at complex II (succinate dehydrogenase, SDH) in mitochondria isolated from mouse hindlimb muscle. The effect required a reduction in membrane potential with resultant accumulation of oxaloacetate (OAA), a potent inhibitor of SDH. To specifically assess the effect of the mitochondrial transaminase, glutamic oxaloacetic transaminase (GOT2) on complex II respiration, and to determine the effect in intact cells as well as isolated mitochondria, we performed respiratory and metabolic studies in wildtype (WT) and CRISPR-generated GOT2 knockdown (KD) C2C12 myocytes. Intact cell respiration by GOT2KD cells versus WT was reduced by adding carbonyl cyanide-p-trifluoromethoxyphenylhydrazone (FCCP) to lower potential. In mitochondria of C2C12 KD cells, respiration at low potential generated by 1 µM FCCP and energized at complex II by 10 mM succinate + 0.5 mM glutamate (but not by complex I substrates) was reduced versus WT mitochondria. Although we could not detect OAA, metabolite data suggested that OAA inhibition of SDH may have contributed to the FCCP effect. C2C12 mitochondria differed from skeletal muscle mitochondria in that the effect of FCCP on complex II respiration was not evident with ADP addition. We also observed that C2C12 cells, unlike skeletal muscle, expressed glutamate dehydrogenase, which competes with GOT2 for glutamate metabolism. In summary, GOT2 KD reduced C2C12 respiration in intact cells at low potential. From differential substrate effects, this occurred largely at complex II. Moreover, C2C12 versus muscle mitochondria differ in complex II sensitivity to ADP and differ markedly in expression of glutamate dehydrogenase.NEW & NOTEWORTHY Impairment of the mitochondrial transaminase, GOT2, reduces complex II (succinate dehydrogenase, SDH)-energized respiration in C2C12 myocytes. This occurs only at low inner membrane potential and is consistent with inhibition of SDH. Incidentally, we observed that C2C12 mitochondria compared with muscle tissue mitochondria differ in sensitivity of complex II respiration to ADP and in the expression of glutamate dehydrogenase.


Asunto(s)
Respiración de la Célula , Potencial de la Membrana Mitocondrial , Mitocondrias Musculares , Animales , Ratones , Aspartato Aminotransferasa Mitocondrial/metabolismo , Aspartato Aminotransferasa Mitocondrial/genética , Diferenciación Celular/efectos de los fármacos , Línea Celular , Respiración de la Célula/efectos de los fármacos , Complejo II de Transporte de Electrones/metabolismo , Complejo II de Transporte de Electrones/genética , Potencial de la Membrana Mitocondrial/efectos de los fármacos , Mitocondrias Musculares/metabolismo , Mitocondrias Musculares/enzimología , Mitocondrias Musculares/efectos de los fármacos , Músculo Esquelético/metabolismo , Músculo Esquelético/efectos de los fármacos , Músculo Esquelético/enzimología , Consumo de Oxígeno/efectos de los fármacos , Succinato Deshidrogenasa/metabolismo , Succinato Deshidrogenasa/genética , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo
6.
Science ; 381(6664): 1316-1323, 2023 09 22.
Artículo en Inglés | MEDLINE | ID: mdl-37733872

RESUMEN

Although tumor growth requires the mitochondrial electron transport chain (ETC), the relative contribution of complex I (CI) and complex II (CII), the gatekeepers for initiating electron flow, remains unclear. In this work, we report that the loss of CII, but not that of CI, reduces melanoma tumor growth by increasing antigen presentation and T cell-mediated killing. This is driven by succinate-mediated transcriptional and epigenetic activation of major histocompatibility complex-antigen processing and presentation (MHC-APP) genes independent of interferon signaling. Furthermore, knockout of methylation-controlled J protein (MCJ), to promote electron entry preferentially through CI, provides proof of concept of ETC rewiring to achieve antitumor responses without side effects associated with an overall reduction in mitochondrial respiration in noncancer cells. Our results may hold therapeutic potential for tumors that have reduced MHC-APP expression, a common mechanism of cancer immunoevasion.


Asunto(s)
Antígenos de Neoplasias , Complejo II de Transporte de Electrones , Complejo I de Transporte de Electrón , Mitocondrias , Neoplasias , Humanos , Presentación de Antígeno , Antígenos de Neoplasias/inmunología , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Complejo II de Transporte de Electrones/genética , Complejo II de Transporte de Electrones/metabolismo , Electrones , Técnicas de Inactivación de Genes , Histonas/metabolismo , Proteínas del Choque Térmico HSP40/genética , Melanoma/inmunología , Melanoma/patología , Metilación , Mitocondrias/enzimología , Neoplasias/inmunología , Neoplasias/patología , Línea Celular Tumoral
7.
Genes (Basel) ; 14(3)2023 03 04.
Artículo en Inglés | MEDLINE | ID: mdl-36980917

RESUMEN

Loss of function of the succinate dehydrogenase complex characterizes 20-40% of all KIT/PDGFRA-negative GIST. Approximately half of SDH-deficient GIST patients lack SDHx mutations and are caused by a hypermethylation of the SDHC promoter, which causes the repression of SDHC transcription and depletion of SDHC protein levels through a mechanism described as epimutation. The remaining 50% of SDH-deficient GISTs have mutations in one of the SDH subunits and SDHA mutations are the most common (30%), with consequent loss of SDHA and SDHB protein expression immunohistochemically. SDHB, SDHC, and SDHD mutations in GIST occur in only 20-30% of cases and most of these SDH mutations are germline. More recently, germline mutations in SDHA have also been described in several patients with loss of function of the SDH complex. SDHA-mutant patients usually carry two mutational events at the SDHA locus, either the loss of the wild type allele or a second somatic event in compound heterozygosis. This review provides an overview of all data in the literature regarding SDHA-mutated GIST, especially focusing on the prevalence of germline mutations in SDH-deficient GIST populations who harbor SDHA somatic mutations, and offers a view towards understanding the importance of genetic counselling for SDHA-variant carriers and relatives.


Asunto(s)
Tumores del Estroma Gastrointestinal , Humanos , Tumores del Estroma Gastrointestinal/genética , Mutación de Línea Germinal , Succinato Deshidrogenasa/genética , Mutación , Metilación de ADN , Complejo II de Transporte de Electrones/genética , Complejo II de Transporte de Electrones/metabolismo
8.
J Med Genet ; 60(2): 107-111, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-35260474

RESUMEN

SDHA pathogenic germline variants (PGVs) are identified in up to 10% of patients with paraganglioma and phaeochromocytoma and up to 30% with wild-type gastrointestinal stromal tumours. Most SDHA PGV carriers present with an apparently sporadic tumour, but often the pathogenic variant has been inherited from parent who has the variant, but has not developed any clinical features. Studies of SDHA PGV carriers suggest that lifetime penetrance for SDHA-associated tumours is low, particularly when identified outside the context of a family history. Current recommended surveillance for SDHA PGV carriers follows an intensive protocol. With increasing implementation of tumour and germline large panel and whole-genome sequencing, it is likely more SDHA PGV carriers will be identified in patients with tumours not strongly associated with SDHA, or outside the context of a strong family history. This creates a complex situation about what to recommend in clinical practice considering low penetrance for tumour development, surveillance burden and patient anxiety. An expert SDHA working group was formed to discuss and consider this situation. This paper outlines the recommendations from this working group for testing and management of SDHA PGV carriers in clinical practice.


Asunto(s)
Neoplasias de las Glándulas Suprarrenales , Paraganglioma , Feocromocitoma , Humanos , Pruebas Genéticas , Paraganglioma/genética , Feocromocitoma/genética , Mutación de Línea Germinal/genética , Neoplasias de las Glándulas Suprarrenales/genética , Reino Unido , Predisposición Genética a la Enfermedad , Complejo II de Transporte de Electrones/genética
9.
Int J Mol Sci ; 23(9)2022 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-35563430

RESUMEN

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.


Asunto(s)
Dictyostelium , Complejo II de Transporte de Electrones , Proteínas Quinasas Activadas por AMP/metabolismo , Dictyostelium/genética , Dictyostelium/metabolismo , Complejo II de Transporte de Electrones/genética , Complejo II de Transporte de Electrones/metabolismo , Mitocondrias/metabolismo , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/metabolismo , Oligonucleótidos Antisentido/farmacología , Fenotipo , Proteínas Protozoarias/genética , Proteínas Protozoarias/metabolismo
10.
Biomed Res Int ; 2022: 5504475, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35463978

RESUMEN

Targeting death receptor-mediated apoptosis in T-cell acute lymphoblastic leukemia (T-ALL), an aggressive disease with poor prognosis, is hindered by the inherent resistance of primary leukemia cells. Knowledge on therapeutic vulnerabilities in these malignant cells will provide opportunities for developing novel combinatory treatments for patients. Using label-free quantitative mass spectrometry and subcellular fractionation techniques, we systematically compared organelle-specific proteomes between Jurkat cells, an in vitro model for T-ALL, and a Jurkat mutant with increased resistance to death receptor-mediated apoptosis. By identifying several differentially regulated protein clusters, our data argued that extensive metabolic reprograming in the mitochondria, characterized by enhanced respiration and energy production, might allow cells to evade DR5-mediated cytotoxicity. Further analysis using clinical datasets demonstrated that the elevated expression of a three-gene signature, consisting of SDHA, IDH3A, and ANXA11, was significantly associated with poor survival of acute leukemia patients. Our analysis therefore provided a unique dataset for a mechanistic understanding of T-ALL and for the design of novel ALL treatments.


Asunto(s)
Leucemia-Linfoma Linfoblástico de Células T Precursoras , Anexinas/genética , Apoptosis , Complejo II de Transporte de Electrones/genética , Humanos , Isocitrato Deshidrogenasa/genética , Células Jurkat , Leucemia-Linfoma Linfoblástico de Células T Precursoras/tratamiento farmacológico , Leucemia-Linfoma Linfoblástico de Células T Precursoras/genética , Proteoma , Receptores de Muerte Celular
11.
Clin Neurol Neurosurg ; 212: 107039, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34839152

RESUMEN

Isolated deficiency of complex II is a rare inborn error of metabolism, accounting for approximately 2% of mitochondrial diseases. Mitochondrial complex II deficiency is predominantly seen in cases with bi-allelic SDHA mutations. To our knowledge, only 11 patients and five pathogenic variants have been reported for the SDHB gene. Our patient had a severe clinical presentation with seizures and sepsis, and died at the age of 2 months. Muscle biopsy analysis was compatible with mitochondrial myopathy with complex II deficiency. The family was given a molecular diagnosis for their child 2 years after his death via a clinical exome test of a frozen muscle biopsy specimen and a novel homozygous missense variant c.592 A>G (p.Ser198Gly) in SDHB gene was detected by next-generation sequencing. Here, we present another patient with a novel homozygous SDHB variant causing severe complex II deficiency and early death.


Asunto(s)
Complejo II de Transporte de Electrones/deficiencia , Errores Innatos del Metabolismo/genética , Enfermedades Mitocondriales/genética , Succinato Deshidrogenasa/genética , Consanguinidad , Complejo II de Transporte de Electrones/genética , Resultado Fatal , Humanos , Recién Nacido , Masculino
12.
Am J Physiol Regul Integr Comp Physiol ; 321(6): R912-R924, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34730023

RESUMEN

We hypothesize that intrauterine hypoxia (HPX) alters the mitochondrial phenotype in fetal hearts contributing to developmental programming. Pregnant guinea pigs were exposed to normoxia (NMX) or hypoxia (HPX, 10.5% O2), starting at early [25 days (25d), 39d duration] or late gestation (50d, 14d duration). Near-term (64d) male and female fetuses were delivered by hysterotomy from anesthetized sows, and body/organ weights were measured. Left ventricles of fetal hearts were excised and frozen for measurement of expression of complex (I-V) subunits, fusion (Mfn2/OPA1) and fission (DRP1/Fis1) proteins, and enzymatic rates of I and IV from isolated mitochondrial proteins. Chronic HPX decreased fetal body weight and increased relative placenta weight regardless of timing. Early-onset HPX increased I, III, and V subunit levels, increased complex I but decreased IV activities in males but not females (all P < 0.05). Late-onset HPX decreased (P < 0.05) I, III, and V levels in both sexes but increased I and decreased IV activities in males only. Both HPX conditions decreased cardiac mitochondrial DNA content in males only. Neither early- nor late-onset HPX had any effect on Mfn2 levels but increased OPA1 in both sexes. Both HPX treatments increased DRP1/Fis1 levels in males. In females, early-onset HPX increased DRP1 with no effect on Fis1, whereas late-onset HPX increased Fis1 with no effect on DRP1. We conclude that both early- and late-onset HPX disrupts the expression/activities of select complexes that could reduce respiratory efficiency and shifts dynamics toward fission in fetal hearts. Thus, intrauterine HPX disrupts the mitochondrial phenotype predominantly in male fetal hearts, potentially altering cardiac metabolism and predisposing the offspring to heart dysfunction.


Asunto(s)
Complejo III de Transporte de Electrones/metabolismo , Complejo II de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Hipoxia Fetal/enzimología , Mitocondrias Cardíacas/enzimología , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Miocitos Cardíacos/enzimología , Animales , Hipoxia de la Célula , Respiración de la Célula , Modelos Animales de Enfermedad , Dinaminas/genética , Dinaminas/metabolismo , Complejo I de Transporte de Electrón/genética , Complejo II de Transporte de Electrones/genética , Complejo III de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/genética , Femenino , Hipoxia Fetal/genética , GTP Fosfohidrolasas/genética , GTP Fosfohidrolasas/metabolismo , Regulación del Desarrollo de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Edad Gestacional , Cobayas , Masculino , Mitocondrias Cardíacas/genética , Dinámicas Mitocondriales , ATPasas de Translocación de Protón Mitocondriales/genética , Factores Sexuales
13.
Nat Immunol ; 22(11): 1440-1451, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34686860

RESUMEN

Intestinal epithelial cell (IEC) damage by T cells contributes to graft-versus-host disease, inflammatory bowel disease and immune checkpoint blockade-mediated colitis. But little is known about the target cell-intrinsic features that affect disease severity. Here we identified disruption of oxidative phosphorylation and an increase in succinate levels in the IECs from several distinct in vivo models of T cell-mediated colitis. Metabolic flux studies, complemented by imaging and protein analyses, identified disruption of IEC-intrinsic succinate dehydrogenase A (SDHA), a component of mitochondrial complex II, in causing these metabolic alterations. The relevance of IEC-intrinsic SDHA in mediating disease severity was confirmed by complementary chemical and genetic experimental approaches and validated in human clinical samples. These data identify a critical role for the alteration of the IEC-specific mitochondrial complex II component SDHA in the regulation of the severity of T cell-mediated intestinal diseases.


Asunto(s)
Colitis/enzimología , Colon/enzimología , Citotoxicidad Inmunológica , Complejo II de Transporte de Electrones/metabolismo , Células Epiteliales/enzimología , Enfermedad Injerto contra Huésped/enzimología , Mucosa Intestinal/enzimología , Mitocondrias/enzimología , Linfocitos T/inmunología , Animales , Estudios de Casos y Controles , Comunicación Celular , Células Cultivadas , Colitis/genética , Colitis/inmunología , Colitis/patología , Colon/inmunología , Colon/ultraestructura , Modelos Animales de Enfermedad , Complejo II de Transporte de Electrones/genética , Células Epiteliales/inmunología , Células Epiteliales/ultraestructura , Femenino , Enfermedad Injerto contra Huésped/genética , Enfermedad Injerto contra Huésped/inmunología , Enfermedad Injerto contra Huésped/patología , Humanos , Inmunidad Mucosa , Mucosa Intestinal/inmunología , Mucosa Intestinal/ultraestructura , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Mitocondrias/inmunología , Mitocondrias/ultraestructura , Fosforilación Oxidativa , Ácido Succínico/metabolismo , Linfocitos T/metabolismo
14.
Am J Physiol Lung Cell Mol Physiol ; 321(4): L718-L725, 2021 10 01.
Artículo en Inglés | MEDLINE | ID: mdl-34378408

RESUMEN

Bronchopulmonary dysplasia (BPD) is a chronic lung disease of preterm infants. Mouse models of hyperoxia-induced lung injury are often used to study pathogenesis and potential therapeutic approaches of BPD. Beside histological studies, gene expression analysis of lung tissue is typically used as experimental readout. Reverse transcription quantitative polymerase chain reaction (RT-qPCR) is the standard method for gene expression analysis; however, the accuracy of the quantitative data depends on the appropriate selection of reference genes. No data on validated reference genes for hyperoxia-induced neonatal lung injury in mice are available. In this study, 12 potential reference genes were systematically analyzed for their expression stability in lung tissue of neonatal mice exposed to room air or hyperoxia and healthy adult controls using published software algorithms. Analysis of gene expression data identified Hprt, Tbp, and Hmbs as the most stable reference genes and proposed combinations of Hprt/Sdha or Hprt/Rpl13a as potential normalization factors. These reference genes and normalization factors were validated by comparing Il6 gene and protein expression and may facilitate accurate gene expression analysis in lung tissues of similar designed studies.


Asunto(s)
Displasia Broncopulmonar/genética , Complejo II de Transporte de Electrones/genética , Hipoxantina Fosforribosiltransferasa/genética , Lesión Pulmonar/patología , Proteínas Ribosómicas/genética , Proteína de Unión a TATA-Box/genética , Animales , Animales Recién Nacidos , Displasia Broncopulmonar/patología , Citocinas/análisis , Modelos Animales de Enfermedad , Femenino , Expresión Génica , Perfilación de la Expresión Génica , Hiperoxia/patología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Oxígeno/farmacología , Reacción en Cadena en Tiempo Real de la Polimerasa , Transducción de Señal/genética
15.
J Cell Biol ; 220(9)2021 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-34259807

RESUMEN

Long-lived proteins (LLPs) have recently emerged as vital components of intracellular structures whose function is coupled to long-term stability. Mitochondria are multifaceted organelles, and their function hinges on efficient proteome renewal and replacement. Here, using metabolic stable isotope labeling of mice combined with mass spectrometry (MS)-based proteomic analysis, we demonstrate remarkable longevity for a subset of the mitochondrial proteome. We discovered that mitochondrial LLPs (mt-LLPs) can persist for months in tissues harboring long-lived cells, such as brain and heart. Our analysis revealed enrichment of mt-LLPs within the inner mitochondrial membrane, specifically in the cristae subcompartment, and demonstrates that the mitochondrial proteome is not turned over in bulk. Pioneering cross-linking experiments revealed that mt-LLPs are spatially restricted and copreserved within protein OXPHOS complexes, with limited subunit exchange throughout their lifetimes. This study provides an explanation for the exceptional mitochondrial protein lifetimes and supports the concept that LLPs provide key structural stability to multiple large and dynamic intracellular structures.


Asunto(s)
Complejo III de Transporte de Electrones/metabolismo , Complejo II de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Complejo I de Transporte de Electrón/metabolismo , Mitocondrias/enzimología , Miocardio/enzimología , Proteoma/metabolismo , Animales , Sitios de Unión , Encéfalo/enzimología , Ciclo del Ácido Cítrico/genética , Complejo I de Transporte de Electrón/química , Complejo I de Transporte de Electrón/genética , Complejo II de Transporte de Electrones/química , Complejo II de Transporte de Electrones/genética , Complejo III de Transporte de Electrones/química , Complejo III de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/química , Complejo IV de Transporte de Electrones/genética , Expresión Génica , Semivida , Metabolismo de los Lípidos/genética , Ratones , Mitocondrias/genética , Membranas Mitocondriales/química , Membranas Mitocondriales/enzimología , Modelos Moleculares , Especificidad de Órganos , Fosforilación Oxidativa , Unión Proteica , Conformación Proteica en Hélice alfa , Conformación Proteica en Lámina beta , Dominios y Motivos de Interacción de Proteínas , Estabilidad Proteica , Proteoma/química , Proteoma/genética
16.
Dev Med Child Neurol ; 63(10): 1204-1212, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34176131

RESUMEN

AIM: To provide a detailed gene and protein expression analysis related to mitochondrial biogenesis and assess mitochondrial content in skeletal muscle of children with cerebral palsy (CP). METHOD: Biceps brachii muscle samples were collected from 19 children with CP (mean [SD] age 15y 4mo [2y 6mo], range 9-18y, 16 males, three females) and 10 typically developing comparison children (mean [SD] age 15y [4y], range 7-21y, eight males, two females). Gene expression (quantitative reverse transcription polymerase chain reaction [PCR]), mitochondrial DNA (mtDNA) to genomic DNA ratio (quantitative PCR), and protein abundance (western blotting) were analyzed. Microarray data sets (CP/aging/bed rest) were analyzed with a focused query investigating metabolism- and mitochondria-related gene networks. RESULTS: The mtDNA to genomic DNA ratio was lower in the children with CP compared to the typically developing group (-23%, p=0.002). Out of five investigated complexes in the mitochondrial respiratory chain, we observed lower protein levels of all complexes (I, III, IV, V, -20% to -37%; p<0.05) except complex II. Total peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC1α) messenger RNA (p<0.004), isoforms PGC1α1 (p=0.05), and PGC1α4 (p<0.001) were reduced in CP. Transcriptional similarities were observed between CP, aging, and 90 days' bed rest. INTERPRETATION: Mitochondrial biogenesis, mtDNA, and oxidative phosphorylation protein content are reduced in CP muscle compared with typically developing muscle. Transcriptional pathways shared between aging and long-term unloading suggests metabolic dysregulation in CP, which may guide therapeutic strategies for combatting CP muscle pathology. What this paper adds Cerebral palsy (CP) muscle contains fewer energy-generating organelles than typically developing muscle. Gene expression in CP muscle is similar to aging and long-term bed rest.


Asunto(s)
Parálisis Cerebral/genética , ADN Mitocondrial/metabolismo , Proteínas del Complejo de Cadena de Transporte de Electrón/genética , Músculo Esquelético/metabolismo , Adolescente , Estudios de Casos y Controles , Parálisis Cerebral/metabolismo , Niño , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Complejo I de Transporte de Electrón/genética , Complejo I de Transporte de Electrón/metabolismo , Complejo II de Transporte de Electrones/genética , Complejo II de Transporte de Electrones/metabolismo , Complejo III de Transporte de Electrones/genética , Complejo III de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/genética , Complejo IV de Transporte de Electrones/metabolismo , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , ATPasas de Translocación de Protón Mitocondriales/genética , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Fosforilación Oxidativa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Adulto Joven
17.
Eur J Hum Genet ; 29(10): 1570-1576, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34012134

RESUMEN

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.


Asunto(s)
Complejo II de Transporte de Electrones/deficiencia , Errores Innatos del Metabolismo/genética , Enfermedades Mitocondriales/genética , Mutación Missense , Succinato Deshidrogenasa/genética , Niño , Complejo II de Transporte de Electrones/genética , Femenino , Homocigoto , Humanos , Recién Nacido , Masculino , Errores Innatos del Metabolismo/patología , Enfermedades Mitocondriales/patología , Fenotipo , Adulto Joven
18.
Cancer Sci ; 112(8): 3375-3387, 2021 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-34014604

RESUMEN

Succinate dehydrogenase (SDH)-deficient renal cell carcinoma (RCC) is mainly associated with a mutation in the SDHB gene and sometimes with mutations in the SDHC or SDHD genes. However, only three cases of succinate dehydrogenase A (SDHA)-deficient RCC have been reported, and the relation between SDHA mutations and RCC has not been clarified. This study assessed the role of SDHA gene mutations in human RCC. We investigated SDHA/B/C/D gene mutations in 129 human RCCs. Targeted next-generation sequencing and direct Sanger sequencing revealed single nucleotide variants (SNVs) of the SDHA gene with amino acid sequence variations in 11/129 tumors, while no SDHB/C/D gene mutations were found. Tumor cells with SNVs of the SDHA gene were characterized by eosinophilic cytoplasm and various patterns of proliferation. Immunohistochemistry examination found that the 11 tumors with SNVs of the SDHA gene showed significant reduction of SDHA protein and SDHB protein expression compared to the 19 tumors without SDHA or SDHB mutations (both P < .0001). Western blotting showed a greater decrease in the expression of SDHA and SDHB proteins in the 11 tumors with SNVs of the SDHA gene than in the 19 tumors without (both P < .0001). There was a positive correlation between SDHA and SDHB protein levels (P < .0001). On immunohistochemistry and Western blotting, the 11 tumors with SNVs of the SDHA gene had higher protein expression for nuclear factor E2-related factor 2 (Nrf2) compared to the 19 tumors without the mutation (P < .01). These observations suggest that SDHA gene mutations might be associated with a subset of RCC.


Asunto(s)
Carcinoma de Células Renales/genética , Regulación hacia Abajo , Complejo II de Transporte de Electrones/genética , Complejo II de Transporte de Electrones/metabolismo , Neoplasias Renales/genética , Polimorfismo de Nucleótido Simple , Adulto , Anciano , Carcinoma de Células Renales/metabolismo , Femenino , Regulación Neoplásica de la Expresión Génica , Células HeLa , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Neoplasias Renales/metabolismo , Masculino , Persona de Mediana Edad , Estudios Retrospectivos , Análisis de Secuencia de ADN , Proteína Supresora de Tumores del Síndrome de Von Hippel-Lindau/genética
19.
Mol Genet Genomic Med ; 9(6): e1692, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33960148

RESUMEN

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.


Asunto(s)
Ataxia Cerebelosa/genética , Complejo III de Transporte de Electrones/deficiencia , Complejo II de Transporte de Electrones/deficiencia , Errores Innatos del Metabolismo/genética , Enfermedades Mitocondriales/genética , Células Cultivadas , Ataxia Cerebelosa/patología , Niño , Complejo II de Transporte de Electrones/genética , Complejo II de Transporte de Electrones/metabolismo , Complejo III de Transporte de Electrones/genética , Fibroblastos/metabolismo , Humanos , Masculino , Errores Innatos del Metabolismo/patología , Enfermedades Mitocondriales/patología , Mutación
20.
BMC Cardiovasc Disord ; 21(1): 142, 2021 03 17.
Artículo en Inglés | MEDLINE | ID: mdl-33731005

RESUMEN

BACKGROUND: The aim of the present study was to investigate whether the disappearance of ischemic post-processing (IPO) in the myocardium of diabetes mellitus (DM) is associated with the increase of succinate dehydrogenase-flavin protein (SDHA). METHODS: A total of 50 Sprague Dawley rats, weighing 300-400 g, were divided into 5 groups according to the random number table method, each with 10 rats. After DM rats were fed a high-fat and -sugar diet for 4 weeks, they were injected with Streptozotocin to establish the diabetic rat model. Normal rats were fed the same regular diet for the same number of weeks. Next, the above rats were taken to establish a cardiopulmonary bypass (CPB) model. Intraperitoneal glucose tolerance test (IPGTT) and oral glucose tolerance test (OGTT) were used to detect whether the DM rat model was established successfully. Taking blood from the femoral artery to collect the blood-gas analysis indicators, and judged whether the CPB model is established. After perfusion was performed according to the experimental strategy, the area of myocardial infarction (MI), and serum creatine kinase isoenzyme (CK-MB) and cardiac troponin (CTnI) levels were measured. Finally, the relative mRNA and protein expression of SDHA was detected. RESULTS: The OGTT and IPGTT suggested that the DM rat model was successfully established. The arterial blood gas analysis indicated that the CPB model was successfully established. As compared with the N group, the heart function of the IR group was significantly reduced, the levels of myocardial enzyme markers, the area of MI, as well as the relative mRNA and protein expression of SDHA, were all increased. As compared with the IR group, the CK-MB and CTnI levels in the IPO group, the MI area, relative mRNA and protein expression of SDHA decreased. As compared with the IPO group, the myocardial enzyme content in the DM + IPO group, the MI area and the relative mRNA and protein expression of SDHA increased. As compared with the DM + IPO group, in the DM + IPO + dme group, the myocardial enzyme content, area of MI and relative mRNA and protein expression were all decreased. CONCLUSION: IPO can inhibit the expression of SDHA, reduce MIRI and exert a cardioprotective effect in the normal rats. However, the protective effect of IPO disappears in the diabetic rats. The inhibitor dme combined with IPO can increase the expression of SDHA and restore the protective effect of IPO in DM myocardia.


Asunto(s)
Diabetes Mellitus Experimental/enzimología , Complejo II de Transporte de Electrones/metabolismo , Poscondicionamiento Isquémico , Infarto del Miocardio/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/enzimología , Animales , Glucemia/metabolismo , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/complicaciones , Complejo II de Transporte de Electrones/antagonistas & inhibidores , Complejo II de Transporte de Electrones/genética , Inhibidores Enzimáticos/farmacología , Masculino , Infarto del Miocardio/complicaciones , Infarto del Miocardio/enzimología , Infarto del Miocardio/patología , Daño por Reperfusión Miocárdica/enzimología , Daño por Reperfusión Miocárdica/etiología , Daño por Reperfusión Miocárdica/patología , Miocardio/patología , Ratas Sprague-Dawley , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA