Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
1.
J Biol Chem ; 299(9): 105088, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37495107

RESUMEN

S-acylation is a reversible posttranslational protein modification consisting of attachment of a fatty acid to a cysteine via a thioester bond. Research over the last few years has shown that a variety of different fatty acids, such as palmitic acid (C16:0), stearate (C18:0), or oleate (C18:1), are used in cells to S-acylate proteins. We recently showed that GNAI proteins can be acylated on a single residue, Cys3, with either C16:0 or C18:1, and that the relative proportion of acylation with these fatty acids depends on the level of the respective fatty acid in the cell's environment. This has functional consequences for GNAI proteins, with the identity of the acylating fatty acid affecting the subcellular localization of GNAIs. Unclear is whether this competitive acylation is specific to GNAI proteins or a more general phenomenon in the proteome. We perform here a proteome screen to identify proteins acylated with different fatty acids. We identify 218 proteins acylated with C16:0 and 308 proteins acylated with C18-lipids, thereby uncovering novel targets of acylation. We find that most proteins that can be acylated by C16:0 can also be acylated with C18-fatty acids. For proteins with more than one acylation site, we find that this competitive acylation occurs on each individual cysteine residue. This raises the possibility that the function of many different proteins can be regulated by the lipid environment via differential S-acylation.


Asunto(s)
Cisteína , Ácido Palmítico , Proteoma , Ácidos Esteáricos , Acilación , Cisteína/metabolismo , Ácido Palmítico/metabolismo , Proteoma/metabolismo , Células HEK293 , Células HeLa , Humanos , Ácidos Esteáricos/metabolismo
2.
Redox Biol ; 62: 102639, 2023 06.
Artículo en Inglés | MEDLINE | ID: mdl-36958250

RESUMEN

Despite a strong rationale for why cancer cells are susceptible to redox-targeting drugs, such drugs often face tumor resistance or dose-limiting toxicity in preclinical and clinical studies. An important reason is the lack of specific biomarkers to better select susceptible cancer entities and stratify patients. Using a large panel of lung cancer cell lines, we identified a set of "antioxidant-capacity" biomarkers (ACB), which were tightly repressed, partly by STAT3 and STAT5A/B in sensitive cells, rendering them susceptible to multiple redox-targeting and ferroptosis-inducing drugs. Contrary to expectation, constitutively low ACB expression was not associated with an increased steady state level of reactive oxygen species (ROS) but a high level of nitric oxide, which is required to sustain high replication rates. Using ACBs, we identified cancer entities with a high percentage of patients with favorable ACB expression pattern, making it likely that more responders to ROS-inducing drugs could be stratified for clinical trials.


Asunto(s)
Antioxidantes , Neoplasias Pulmonares , Humanos , Especies Reactivas de Oxígeno/metabolismo , Antioxidantes/metabolismo , Neoplasias Pulmonares/metabolismo , Oxidación-Reducción , Biomarcadores/metabolismo
3.
Nat Commun ; 12(1): 4590, 2021 07 28.
Artículo en Inglés | MEDLINE | ID: mdl-34321466

RESUMEN

Covalent attachment of C16:0 to proteins (palmitoylation) regulates protein function. Proteins are also S-acylated by other fatty acids including C18:0. Whether protein acylation with different fatty acids has different functional outcomes is not well studied. We show here that C18:0 (stearate) and C18:1 (oleate) compete with C16:0 to S-acylate Cys3 of GNAI proteins. C18:0 becomes desaturated so that C18:0 and C18:1 both cause S-oleoylation of GNAI. Exposure of cells to C16:0 or C18:0 shifts GNAI acylation towards palmitoylation or oleoylation, respectively. Oleoylation causes GNAI proteins to shift out of cell membrane detergent-resistant fractions where they potentiate EGFR signaling. Consequently, exposure of cells to C18:0 reduces recruitment of Gab1 to EGFR and reduces AKT activation. This provides a molecular mechanism for the anti-tumor effects of C18:0, uncovers a mechanistic link how metabolites affect cell signaling, and provides evidence that the identity of the fatty acid acylating a protein can have functional consequences.


Asunto(s)
Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Péptidos y Proteínas de Señalización Intercelular/metabolismo , Transducción de Señal/fisiología , Ácidos Esteáricos/metabolismo , Acilación , Membrana Celular/metabolismo , Proliferación Celular , Ácidos Grasos/metabolismo , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/genética , Humanos , Lipoilación , Células MCF-7 , Ácidos Oléicos/metabolismo
4.
J Clin Med ; 9(4)2020 Mar 29.
Artículo en Inglés | MEDLINE | ID: mdl-32235386

RESUMEN

Background: The pleomorphic clinical presentation makes the diagnosis of desminopathy difficult. We aimed to describe the prevalence, phenotypic expression, and mitochondrial function of individuals with putative disease-causing desmin (DES) variants identified in patients with an unexplained etiology of cardiomyopathy. Methods: A total of 327 Czech patients underwent whole exome sequencing and detailed phenotyping in probands harboring DES variants. Results: Rare, conserved, and possibly pathogenic DES variants were identified in six (1.8%) probands. Two DES variants previously classified as variants of uncertain significance (p.(K43E), p.(S57L)), one novel DES variant (p.(A210D)), and two known pathogenic DES variants (p.(R406W), p.(R454W)) were associated with characteristic desmin-immunoreactive aggregates in myocardial and/or skeletal biopsy samples. The individual with the novel DES variant p.(Q364H) had a decreased myocardial expression of desmin with absent desmin aggregates in myocardial/skeletal muscle biopsy and presented with familial left ventricular non-compaction cardiomyopathy (LVNC), a relatively novel phenotype associated with desminopathy. An assessment of the mitochondrial function in four probands heterozygous for a disease-causing DES variant confirmed a decreased metabolic capacity of mitochondrial respiratory chain complexes in myocardial/skeletal muscle specimens, which was in case of myocardial succinate respiration more profound than in other cardiomyopathies. Conclusions: The presence of desminopathy should also be considered in individuals with LVNC, and in the differential diagnosis of mitochondrial diseases.

5.
Biochem Biophys Res Commun ; 521(4): 1036-1041, 2020 01 22.
Artículo en Inglés | MEDLINE | ID: mdl-31732150

RESUMEN

Mitochondrial ATP synthase is responsible for production of the majority of cellular ATP. Disorders of ATP synthase in humans can be caused by numerous mutations in both structural subunits and specific assembly factors. They are associated with variable pathogenicity and clinical phenotypes ranging from mild to the most severe mitochondrial diseases. To shed light on primary/pivotal functional consequences of ATP synthase deficiency, we explored human HEK 293 cells with a varying content of fully assembled ATP synthase, selectively downregulated to 15-80% of controls by the knockdown of F1 subunits γ, δ and ε. Examination of cellular respiration and glycolytic flux revealed that enhanced glycolysis compensates for insufficient mitochondrial ATP production while reduced dissipation of mitochondrial membrane potential leads to elevated ROS production. Both insufficient energy provision and increased oxidative stress contribute to the resulting pathological phenotype. The threshold for manifestation of the ATP synthase defect and subsequent metabolic remodelling equals to 10-30% of residual ATP synthase activity. The metabolic adaptations are not able to sustain proliferation in a galactose medium, although sufficient under glucose-rich conditions. As metabolic alterations occur when the content of ATP synthase drops below 30%, some milder ATP synthase defects may not necessarily manifest with a mitochondrial disease phenotype, as long as the threshold level is not exceeded.


Asunto(s)
ATPasas de Translocación de Protón Mitocondriales/deficiencia , Supervivencia Celular , Células Clonales , Técnicas de Silenciamiento del Gen , Glucólisis , Células HEK293 , Humanos , Concentración 50 Inhibidora , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Estrés Oxidativo , Termodinámica
6.
FASEB J ; 33(12): 14103-14117, 2019 12.
Artículo en Inglés | MEDLINE | ID: mdl-31652072

RESUMEN

Biogenesis of F1Fo ATP synthase, the key enzyme of mitochondrial energy provision, depends on transmembrane protein 70 (TMEM70), localized in the inner mitochondrial membrane of higher eukaryotes. TMEM70 absence causes severe ATP-synthase deficiency and leads to a neonatal mitochondrial encephalocardiomyopathy in humans. However, the exact biochemical function of TMEM70 remains unknown. Using TMEM70 conditional knockout in mice, we show that absence of TMEM70 impairs the early stage of enzyme biogenesis by preventing incorporation of hydrophobic subunit c into rotor structure of the enzyme. This results in the formation of an incomplete, pathologic enzyme complex consisting of F1 domain and peripheral stalk but lacking Fo proton channel composed of subunits c and a. We demonstrated direct interaction between TMEM70 and subunit c and showed that overexpression of subunit c in TMEM70-/- cells partially rescued TMEM70 defect. Accordingly, TMEM70 knockdown prevented subunit c accumulation otherwise observed in F1-deficient cells. Altogether, we identified TMEM70 as specific ancillary factor for subunit c. The biologic role of TMEM70 is to increase the low efficacy of spontaneous assembly of subunit c oligomer, the key and rate-limiting step of ATP-synthase biogenesis, and thus to reach an adequately high physiologic level of ATP synthase in mammalian tissues.-Kovalcíková, J., Vrbacký, M., Pecina, P., Tauchmannová, K., Nusková, H., Kaplanová, V., Brázdová, A., Alán, L., Eliás, J., Cunátová, K., Korínek, V., Sedlacek, R., Mrácek, T., Houstek, J. TMEM70 facilitates biogenesis of mammalian ATP synthase by promoting subunit c incorporation into the rotor structure of the enzyme.


Asunto(s)
Proteínas Mitocondriales/metabolismo , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Animales , Células Cultivadas , Regulación de la Expresión Génica , Técnicas de Inactivación de Genes/métodos , Genotipo , Células HEK293 , Humanos , Ratones , Ratones Noqueados , Proteínas Mitocondriales/genética , ATPasas de Translocación de Protón Mitocondriales/genética , Proteolípidos/metabolismo , Tamoxifeno/farmacología
7.
Biochim Biophys Acta Bioenerg ; 1859(5): 374-381, 2018 May.
Artículo en Inglés | MEDLINE | ID: mdl-29499186

RESUMEN

The central stalk of mitochondrial ATP synthase consists of subunits γ, δ, and ε, and along with the membraneous subunit c oligomer constitutes the rotor domain of the enzyme. Our previous studies showed that mutation or deficiency of ε subunit markedly decreased the content of ATP synthase, which was otherwise functionaly and structuraly normal. Interestingly, it led to accumulation of subunit c aggregates, suggesting the role of the ε subunit in assembly of individual enzyme domains. In the present study we focused on the role of subunits γ and δ. Using shRNA knockdown in human HEK293 cells, the protein levels of γ and δ were decreased to 30% and 10% of control levels, respectively. The content of the assembled ATP synthase decreased in accordance with the levels of the silenced subunits, which was also the case for most structural subunits. In contrast, the hydrophobic c subunit was increased to 130% or 180%, respectively and most of it was detected as aggregates of 150-400 kDa by 2D PAGE. In addition the IF1 protein was upregulated to 195% and 300% of control levels. Both γ and δ subunits silenced cells displayed decreased ATP synthase function - lowered rate of ADP-stimulated respiration, a two-fold increased sensitivity of respiration to inhibitor oligomycin, and impaired utilization of mitochondrial membrane potential for ADP phosphorylation. In summary, similar phenotype of γ, δ and ε subunit deficiencies suggest uniform requirement for assembled central stalk as driver of the c-oligomer attachment in the assembly process of mammalian ATP synthase.


Asunto(s)
Mitocondrias/metabolismo , Proteínas Mitocondriales/metabolismo , Consumo de Oxígeno/fisiología , ATPasas de Translocación de Protón/metabolismo , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Mitocondrias/genética , Proteínas Mitocondriales/genética , ATPasas de Translocación de Protón Mitocondriales , ATPasas de Translocación de Protón/genética
8.
Eur J Heart Fail ; 19(4): 522-530, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-27647766

RESUMEN

AIMS: Iron replacement improves clinical status in iron-deficient patients with heart failure (HF), but the pathophysiology is poorly understood. Iron is essential not only for erythropoiesis, but also for cellular bioenergetics. The impact of myocardial iron deficiency (MID) on mitochondrial function, measured directly in the failing human heart, is unknown. METHODS AND RESULTS: Left ventricular samples were obtained from 91 consecutive HF patients undergoing transplantation and 38 HF-free organ donors (controls). Total myocardial iron content, mitochondrial respiration, citric acid cycle and respiratory chain enzyme activities, respiratory chain components (complex I-V), and protein content of reactive oxygen species (ROS)-protective enzymes were measured in tissue homogenates to quantify mitochondrial function. Myocardial iron content was lower in HF compared with controls (156 ± 41 vs. 200 ± 38 µg·g-1 dry weight, P < 0.001), independently of anaemia. MID (the lowest iron tercile in HF) was associated with more extensive coronary disease and less beta-blocker usage compared with non-MID HF patients. Compared with controls, HF patients displayed reduced myocardial oxygen2 respiration and reduced activity of all examined mitochondrial enzymes (all P < 0.001). MID in HF was associated with preserved activity of respiratory chain enzymes but reduced activity of aconitase and citrate synthase (by -26% and -15%, P < 0.05) and reduced expression of catalase, glutathione peroxidase, and superoxide dismutase 2. CONCLUSION: Myocardial iron content is decreased and mitochondrial functions are impaired in advanced HF. MID in HF is associated with diminished citric acid cycle enzyme activities and decreased ROS-protecting enzymes. MID may contribute to altered myocardial substrate use and to worsening of mitochondrial dysfunction that exists in HF.


Asunto(s)
Insuficiencia Cardíaca/metabolismo , Hierro/metabolismo , Mitocondrias Cardíacas/metabolismo , Miocardio/metabolismo , Adulto , Anciano , Estudios de Casos y Controles , Femenino , Insuficiencia Cardíaca/fisiopatología , Insuficiencia Cardíaca/cirugía , Trasplante de Corazón , Humanos , Masculino , Persona de Mediana Edad
9.
Data Brief ; 7: 1004-9, 2016 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-27408912

RESUMEN

This paper describes data related to a research article entitled "Tissue- and species-specific differences in cytochrome c oxidase assembly induced by SURF1 defects" [1]. This paper includes data of the quantitative analysis of individual forms of respiratory chain complexes I, III and IV present in SURF1 knockout (SURF1 (-/-) ) and control (SURF1 (+/+) ) mouse fibroblasts and tissues and in fibroblasts of human control and patients with SURF1 gene mutation. Also it includes data demonstrating response of complex IV, cytochrome c oxidase (COX), to reversible inhibition of mitochondrial translation in SURF1 (-/-) mouse and SURF1 patient fibroblast cell lines.

10.
Nat Commun ; 7: 12061, 2016 07 08.
Artículo en Inglés | MEDLINE | ID: mdl-27389904

RESUMEN

Coronary flow (CF) measured ex vivo is largely determined by capillary density that reflects angiogenic vessel formation in the heart in vivo. Here we exploit this relationship and show that CF in the rat is influenced by a locus on rat chromosome 2 that is also associated with cardiac capillary density. Mitochondrial tryptophanyl-tRNA synthetase (Wars2), encoding an L53F protein variant within the ATP-binding motif, is prioritized as the candidate at the locus by integrating genomic data sets. WARS2(L53F) has low enzyme activity and inhibition of WARS2 in endothelial cells reduces angiogenesis. In the zebrafish, inhibition of wars2 results in trunk vessel deficiencies, disordered endocardial-myocardial contact and impaired heart function. Inhibition of Wars2 in the rat causes cardiac angiogenesis defects and diminished cardiac capillary density. Our data demonstrate a pro-angiogenic function for Wars2 both within and outside the heart that may have translational relevance given the association of WARS2 with common human diseases.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Genoma , Células Endoteliales de la Vena Umbilical Humana/enzimología , Mitocondrias/genética , Neovascularización Fisiológica/genética , Triptófano-ARNt Ligasa/genética , Secuencia de Aminoácidos , Animales , Mapeo Cromosómico , Cromosomas de los Mamíferos/química , Embrión no Mamífero , Sitios Genéticos , Células HEK293 , Células Endoteliales de la Vena Umbilical Humana/citología , Humanos , Mitocondrias/metabolismo , Miocardio/citología , Miocardio/enzimología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/metabolismo , Ratas , Alineación de Secuencia , Homología de Secuencia de Aminoácido , Transducción de Señal , Triptófano-ARNt Ligasa/antagonistas & inhibidores , Triptófano-ARNt Ligasa/metabolismo , Pez Cebra
11.
Biochim Biophys Acta ; 1862(4): 705-715, 2016 04.
Artículo en Inglés | MEDLINE | ID: mdl-26804654

RESUMEN

Mitochondrial protein SURF1 is a specific assembly factor of cytochrome c oxidase (COX), but its function is poorly understood. SURF1 gene mutations cause a severe COX deficiency manifesting as the Leigh syndrome in humans, whereas in mice SURF1(-/-) knockout leads only to a mild COX defect. We used SURF1(-/-) mouse model for detailed analysis of disturbed COX assembly and COX ability to incorporate into respiratory supercomplexes (SCs) in different tissues and fibroblasts. Furthermore, we compared fibroblasts from SURF1(-/-) mouse and SURF1 patients to reveal interspecies differences in kinetics of COX biogenesis using 2D electrophoresis, immunodetection, arrest of mitochondrial proteosynthesis and pulse-chase metabolic labeling. The crucial differences observed are an accumulation of abundant COX1 assembly intermediates, low content of COX monomer and preferential recruitment of COX into I-III2-IVn SCs in SURF1 patient fibroblasts, whereas SURF1(-/-) mouse fibroblasts were characterized by low content of COX1 assembly intermediates and milder decrease in COX monomer, which appeared more stable. This pattern was even less pronounced in SURF1(-/-) mouse liver and brain. Both the control and SURF1(-/-) mice revealed only negligible formation of the I-III2-IVn SCs and marked tissue differences in the contents of COX dimer and III2-IV SCs, also less noticeable in liver and brain than in heart and muscle. Our studies support the view that COX assembly is much more dependent on SURF1 in humans than in mice. We also demonstrate markedly lower ability of mouse COX to form I-III2-IVn supercomplexes, pointing to tissue-specific and species-specific differences in COX biogenesis.


Asunto(s)
Complejo IV de Transporte de Electrones/metabolismo , Fibroblastos/metabolismo , Enfermedad de Leigh/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas Mitocondriales/metabolismo , Animales , Complejo IV de Transporte de Electrones/genética , Femenino , Fibroblastos/patología , Humanos , Enfermedad de Leigh/genética , Enfermedad de Leigh/patología , Masculino , Proteínas de la Membrana/genética , Ratones , Ratones Noqueados , Proteínas Mitocondriales/genética , Especificidad de Órganos , Especificidad de la Especie
12.
Hum Mol Genet ; 25(21): 4674-4685, 2016 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-28173120

RESUMEN

TMEM70, a 21-kDa protein localized in the inner mitochondrial membrane, has been shown to facilitate the biogenesis of mammalian F1Fo ATP synthase. Mutations of the TMEM70 gene represent the most frequent cause of isolated ATP synthase deficiency resulting in a severe mitochondrial disease presenting as neonatal encephalo-cardiomyopathy (OMIM 604273). To better understand the biological role of this factor, we generated Tmem70-deficient mice and found that the homozygous Tmem70-/- knockouts exhibited profound growth retardation and embryonic lethality at ∼9.5 days post coitum. Blue-Native electrophoresis demonstrated an isolated deficiency in fully assembled ATP synthase in the Tmem70-/- embryos (80% decrease) and a marked accumulation of F1 complexes indicative of impairment in ATP synthase biogenesis that was stalled at the early stage, following the formation of F1 oligomer. Consequently, a decrease in ADP-stimulated State 3 respiration, respiratory control ratio and ATP/ADP ratios, indicated compromised mitochondrial ATP production. Tmem70-/- embryos exhibited delayed development of the cardiovascular system and a disturbed heart mitochondrial ultrastructure, with concentric or irregular cristae structures. Tmem70+/- heterozygous mice were fully viable and displayed normal postnatal growth and development of the mitochondrial oxidative phosphorylation system. Nevertheless, they presented with mild deterioration of heart function. Our results demonstrated that Tmem70 knockout in the mouse results in embryonic lethality due to the lack of ATP synthase and impairment of mitochondrial energy provision. This is analogous to TMEM70 dysfunction in humans and verifies the crucial role of this factor in the biosynthesis and assembly of mammalian ATP synthase.


Asunto(s)
Proteínas de la Membrana/genética , Proteínas Mitocondriales/genética , ATPasas de Translocación de Protón Mitocondriales/genética , Adenosina Trifosfato/metabolismo , Animales , Cardiomiopatías/metabolismo , Femenino , Homocigoto , Proteínas de la Membrana/deficiencia , Proteínas de la Membrana/metabolismo , Errores Innatos del Metabolismo/metabolismo , Ratones , Ratones Noqueados , Mitocondrias/metabolismo , Enfermedades Mitocondriales/genética , Enfermedades Mitocondriales/metabolismo , Membranas Mitocondriales/metabolismo , Proteínas Mitocondriales/deficiencia , Proteínas Mitocondriales/metabolismo , ATPasas de Translocación de Protón Mitocondriales/biosíntesis , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Mutación , Fosforilación Oxidativa , Embarazo
13.
Biochem Biophys Res Commun ; 464(3): 787-93, 2015 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-26168732

RESUMEN

Mitochondrial ATP synthase, ADP/ATP translocase (ANT), and inorganic phosphate carrier (PiC) are supposed to form a supercomplex called ATP synthasome. Our protein and transcript analysis of rat tissues indicates that the expression of ANT and PiC is transcriptionally controlled in accordance with the biogenesis of ATP synthase. In contrast, the content of ANT and PiC is increased in ATP synthase deficient patients' fibroblasts, likely due to a post-transcriptional adaptive mechanism. A structural analysis of rat heart mitochondria by immunoprecipitation, blue native/SDS electrophoresis, immunodetection and MS analysis revealed the presence of ATP synthasome. However, the majority of PiC and especially ANT did not associate with ATP synthase, suggesting that most of PiC, ANT and ATP synthase exist as separate entities.


Asunto(s)
Adenosina Trifosfato/biosíntesis , Mitocondrias/metabolismo , ATPasas de Translocación de Protón Mitocondriales/química , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Animales , Animales Recién Nacidos , Células Cultivadas , Fibroblastos/metabolismo , Humanos , Sustancias Macromoleculares/química , Sustancias Macromoleculares/metabolismo , Mitocondrias Cardíacas/metabolismo , Translocasas Mitocondriales de ADP y ATP/química , Translocasas Mitocondriales de ADP y ATP/genética , Translocasas Mitocondriales de ADP y ATP/metabolismo , ATPasas de Translocación de Protón Mitocondriales/genética , Fosfatos/química , Fosfatos/metabolismo , Ratas , Ratas Wistar
14.
Biochem J ; 466(3): 601-11, 2015 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-25588698

RESUMEN

Mutations in the MT-ATP6 gene are frequent causes of severe mitochondrial disorders. Typically, these are missense mutations, but another type is represented by the 9205delTA microdeletion, which removes the stop codon of the MT-ATP6 gene and affects the cleavage site in the MT-ATP8/MT-ATP6/MT-CO3 polycistronic transcript. This interferes with the processing of mRNAs for the Atp6 (Fo-a) subunit of ATP synthase and the Cox3 subunit of cytochrome c oxidase (COX). Two cases described so far presented with strikingly different clinical phenotypes-mild transient lactic acidosis or fatal encephalopathy. To gain more insight into the pathogenic mechanism, we prepared 9205delTA cybrids with mutation load ranging between 52 and 99% and investigated changes in the structure and function of ATP synthase and the COX. We found that 9205delTA mutation strongly reduces the levels of both Fo-a and Cox3 proteins. Lack of Fo-a alters the structure but not the content of ATP synthase, which assembles into a labile, ∼60 kDa smaller, complex retaining ATP hydrolytic activity but which is unable to synthesize ATP. In contrast, lack of Cox3 limits the biosynthesis of COX but does not alter the structure of the enzyme. Consequently, the diminished mitochondrial content of COX and non-functional ATP synthase prevent most mitochondrial ATP production. The biochemical effects caused by the 9205delTA microdeletion displayed a pronounced threshold effect above ∼90% mutation heteroplasmy. We observed a linear relationship between the decrease in subunit Fo-a or Cox3 content and the functional presentation of the defect. Therefore we conclude that the threshold effect originated from a gene-protein level.


Asunto(s)
ADN Mitocondrial/genética , Complejo IV de Transporte de Electrones/genética , ATPasas de Translocación de Protón Mitocondriales/fisiología , Mutación/genética , Línea Celular , Complejo IV de Transporte de Electrones/metabolismo , Eliminación de Gen , Humanos , ATPasas de Translocación de Protón Mitocondriales/química , ATPasas de Translocación de Protón Mitocondriales/deficiencia , ATPasas de Translocación de Protón Mitocondriales/genética , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Subunidades de Proteína/deficiencia , Subunidades de Proteína/genética , Subunidades de Proteína/metabolismo
15.
BBA Clin ; 2: 62-71, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26675066

RESUMEN

BACKGROUND: Mitochondrial diseases belong to the most severe inherited metabolic disorders affecting pediatric population. Despite detailed knowledge of mtDNA mutations and progress in identification of affected nuclear genes, diagnostics of a substantial part of mitochondrial diseases relies on clinical symptoms and biochemical data from muscle biopsies and cultured fibroblasts. METHODS: To investigate manifestation of oxidative phosphorylation defects in isolated lymphocytes, digitonin-permeabilized cells from 48 children were analyzed by high resolution respirometry, cytofluorometric detection of mitochondrial membrane potential and immunodetection of respiratory chain proteins with SDS and Blue Native electrophoreses. RESULTS: Evaluation of individual respiratory complex activities, ATP synthesis, kinetic parameters of mitochondrial respiratory chain and the content and subunit composition of respiratory chain complexes enabled detection of inborn defects of respiratory complexes I, IV and V within 2 days. Low respiration with NADH-dependent substrates and increased respiration with glycerol-3-phosphate revealed complex I defects; changes in p 50 for oxygen and elevated uncoupling control ratio pointed to complex IV deficiency due to SURF1 or SCO2 mutation; high oligomycin sensitivity of state 3-ADP respiration, upregulated mitochondrial membrane potential and low content of complex V were found in lymphocytes with ATP synthase deficiency due to TMEM70 mutations. CONCLUSION: Based on our results, we propose the best biochemical parameters predictive for defects of respiratory complexes I, IV and V manifesting in peripheral blood lymphocytes. GENERAL SIGNIFICANCE: The noninvasiveness, reliability and speed of an approach utilizing novel biochemical criteria demonstrate the high potential of isolated lymphocytes for diagnostics of oxidative phosphorylation disorders in pediatric patients.

16.
PLoS One ; 8(8): e71869, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23967256

RESUMEN

Mitochondrial respiratory chain is organised into supramolecular structures that can be preserved in mild detergent solubilisates and resolved by native electrophoretic systems. Supercomplexes of respiratory complexes I, III and IV as well as multimeric forms of ATP synthase are well established. However, the involvement of complex II, linking respiratory chain with tricarboxylic acid cycle, in mitochondrial supercomplexes is questionable. Here we show that digitonin-solubilised complex II quantitatively forms high molecular weight structures (CIIhmw) that can be resolved by clear native electrophoresis. CIIhmw structures are enzymatically active and differ in electrophoretic mobility between tissues (500 - over 1000 kDa) and cultured cells (400-670 kDa). While their formation is unaffected by isolated defects in other respiratory chain complexes, they are destabilised in mtDNA-depleted, rho0 cells. Molecular interactions responsible for the assembly of CIIhmw are rather weak with the complexes being more stable in tissues than in cultured cells. While electrophoretic studies and immunoprecipitation experiments of CIIhmw do not indicate specific interactions with the respiratory chain complexes I, III or IV or enzymes of the tricarboxylic acid cycle, they point out to a specific interaction between CII and ATP synthase.


Asunto(s)
Complejo II de Transporte de Electrones/química , Animales , Línea Celular , Transporte de Electrón , Proteínas del Complejo de Cadena de Transporte de Electrón/química , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Complejo II de Transporte de Electrones/metabolismo , Humanos , Redes y Vías Metabólicas , Mitocondrias/genética , Mitocondrias/metabolismo , Peso Molecular , Especificidad de Órganos , Fosforilación Oxidativa , Unión Proteica
17.
Int J Dev Neurosci ; 31(2): 123-30, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23238024

RESUMEN

We have recently demonstrated the evidence of oxidative stress in brain of immature rats during seizures induced by DL-homocysteic acid (DL-HCA). The aim of the present study was to investigate the antioxidant defense mechanisms under these conditions. Seizures were induced in immature 12-day-old rats by bilateral icv infusion of DL-HCA (600 nmol/side), and the activities of the main antioxidant enzymes were examined in supernatants of the cerebral cortex during the acute phase of seizures and at several periods of survival, up to 5 weeks, following these seizures. In control animals individual antioxidant enzymes revealed different changes during the studied postnatal period (PD 12 till PD 47). Total superoxide dismutase (SOD), CuZn SOD (SOD1), Mn SOD (SOD2) and glutathione peroxidase (GPX) activities were increasing while, catalase activity decreased and the activity of glutathione reductase (GR) remained unchanged. In HCA-treated animals, the activity of total SOD, SOD1 and particularly SOD2 significantly increased at 20 h and 6 days of survival. Importantly, upregulation of SOD2 was also confirmed in mitochondria at the protein level by immunoblotting. The activities of other antioxidant enzymes including catalase and GPX did not significantly differ upon HCA treatment from the appropriate controls at any of the studied time intervals. The pronounced and selective upregulation of SOD2 points to enhanced ROS levels in the mitochondrial matrix. This may be associated with inhibition of respiratory chain complex I that we have demonstrated in our previous studies. The present findings suggest that oxidative stress occurring in the brain of immature rats during and following the seizures induced by DL-HCA is apparently due to both the increased free radical production and the limited antioxidant defense.


Asunto(s)
Envejecimiento/metabolismo , Antioxidantes/metabolismo , Corteza Cerebral/enzimología , Especies Reactivas de Oxígeno/metabolismo , Convulsiones/enzimología , Superóxido Dismutasa/metabolismo , Animales , Estrés Oxidativo , Ratas , Ratas Wistar , Distribución Tisular , Regulación hacia Arriba
18.
Biochim Biophys Acta ; 1817(7): 1037-43, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-22433607

RESUMEN

Early onset mitochondrial encephalo-cardiomyopathy due to isolated deficiency of ATP synthase is frequently caused by mutations in TMEM70 gene encoding enzyme-specific ancillary factor. Diminished ATP synthase results in low ATP production, elevated mitochondrial membrane potential and increased ROS production. To test whether the patient cells may react to metabolic disbalance by changes in oxidative phosphorylation system, we performed a quantitative analysis of respiratory chain complexes and intramitochondrial proteases involved in their turnover. SDS- and BN-PAGE Western blot analysis of fibroblasts from 10 patients with TMEM70 317-2A>G homozygous mutation showed a significant 82-89% decrease of ATP synthase and 50-162% increase of respiratory chain complex IV and 22-53% increase of complex III. The content of Lon protease, paraplegin and prohibitins 1 and 2 was not significantly changed. Whole genome expression profiling revealed a generalized upregulation of transcriptional activity, but did not show any consistent changes in mRNA levels of structural subunits, specific assembly factors of respiratory chain complexes, or in regulatory genes of mitochondrial biogenesis which would parallel the protein data. The mtDNA content in patient cells was also not changed. The results indicate involvement of posttranscriptional events in the adaptive regulation of mitochondrial biogenesis that allows for the compensatory increase of respiratory chain complexes III and IV in response to deficiency of ATP synthase.


Asunto(s)
Complejo III de Transporte de Electrones/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Proteínas de la Membrana/genética , Proteínas Mitocondriales/genética , ATPasas de Translocación de Protón Mitocondriales/deficiencia , Mutación/genética , Regulación hacia Arriba , ADN Mitocondrial/metabolismo , Transporte de Electrón/genética , Fibroblastos/metabolismo , Fibroblastos/patología , Perfilación de la Expresión Génica , Humanos , Mitocondrias/enzimología , Mitocondrias/genética , ATPasas de Translocación de Protón Mitocondriales/metabolismo , Fosforilación Oxidativa , Péptido Hidrolasas/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo
19.
Mitochondrion ; 11(5): 722-8, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21664301

RESUMEN

The primary attempt in diagnostic and experimental studies of numerous pathological states associated with mitochondrial dysfunction is a precise evaluation of changes in function, content and structure of mitochondrial OXPHOS system. The analysis of rat heart, liver, brain and kidney by oxygraphy, enzyme activities, membrane potential, and BN/SDS-PAGE western blotting demonstrated that tissue homogenates can substitute for isolated mitochondria, providing comparable qualitative mitochondrial parameters. The use of homogenate avoids the loss of the majority of mitochondria during their isolation. Only 50-100mg of the tissue is required for the complex OXPHOS analysis, i.e. five times less as compared with isolated mitochondria.


Asunto(s)
Encéfalo/metabolismo , Riñón/metabolismo , Hígado/metabolismo , Mitocondrias/metabolismo , Miocardio/metabolismo , Animales , Encéfalo/enzimología , Citrato (si)-Sintasa/metabolismo , Proteínas del Complejo de Cadena de Transporte de Electrón/metabolismo , Complejo IV de Transporte de Electrones/metabolismo , Pruebas de Enzimas , Riñón/enzimología , Hígado/enzimología , Masculino , Potencial de la Membrana Mitocondrial , Mitocondrias/enzimología , Proteínas Mitocondriales/metabolismo , Miocardio/enzimología , Fosforilación Oxidativa , Consumo de Oxígeno , Ratas , Ratas Wistar
20.
J Bioenerg Biomembr ; 42(5): 395-403, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20725851

RESUMEN

The mechanism of cyanide's inhibitory effect on the mitochondrial cytochrome c oxidase (COX) as well as the conditions for its recovery have not yet been fully explained. We investigated three parameters of COX function, namely electron transport (oxygen consumption), proton transport (mitochondrial membrane potential Δψ(m)) and the enzyme affinity to oxygen (p50 value) with regard to the inhibition by KCN and its reversal by pyruvate. 250 µM KCN completely inhibited both the electron and proton transport function of COX. The inhibition was reversible as demonstrated by washing of mitochondria. The addition of 60 mM pyruvate induced the maximal recovery of both parameters to 60-80% of the original values. When using low KCN concentrations of up to 5 µM, we observed a profound, 30-fold decrease of COX affinity for oxygen. Again, this decrease was completely reversed by washing mitochondria while pyruvate induced only a partial, yet significant recovery of oxygen affinity. Our results demonstrate that the inhibition of COX by cyanide is reversible and that the potential of pyruvate as a cyanide poisoning antidote is limited. Importantly, we also showed that the COX affinity for oxygen is the most sensitive indicator of cyanide toxic effects.


Asunto(s)
Complejo IV de Transporte de Electrones/antagonistas & inhibidores , Complejo IV de Transporte de Electrones/metabolismo , Transporte de Electrón/fisiología , Mitocondrias/metabolismo , Cianuro de Potasio/farmacología , Protones , Ácido Pirúvico/metabolismo , Animales , Hígado/metabolismo , Masculino , Potencial de la Membrana Mitocondrial/fisiología , Oxígeno/metabolismo , Consumo de Oxígeno/fisiología , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...