Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
1.
Biochemistry ; 63(18): 2352-2368, 2024 Sep 17.
Artículo en Inglés | MEDLINE | ID: mdl-39206807

RESUMEN

Fusobacterium nucleatum, a Gram-negative obligate anaerobe, is common to the oral microbiota, but the species is known to infect other sites of the body where it is associated with a range of pathologies. At present, little is known about the mechanisms by which F. nucleatum mitigates against oxidative and nitrosative stress. Inspection of the F. nucleatum subsp. polymorphum ATCC 10953 genome reveals that it encodes a flavodiiron protein (FDP; FNP2073) that is known in other organisms to reduce NO to N2O and/or O2 to H2O. FNP2073 is dicistronic with a gene encoding a multicomponent enzyme termed BCR for butyryl-CoA reductase. BCR is composed of a butyryl-CoA dehydrogenase domain (BCD), the C-terminal domain of the α-subunit of the electron-transfer flavoprotein (Etfα), and a rubredoxin domain. We show that BCR and the FDP form an α4ß4 heterotetramic complex and use butyryl-CoA to selectively reduce O2 to H2O. The FAD associated with the Etfα domain (α-FAD) forms red anionic semiquinone (FAD•-), whereas the FAD present in the BCD domain (δ-FAD) forms the blue-neutral semiquinone (FADH•), indicating that both cofactors participate in one-electron transfers. This was confirmed in stopped-flow studies where the reduction of oxidized BCR with an excess of butyryl-CoA resulted in rapid (<1.6 ms) interflavin electron transfer evidenced by the formation of the FAD•-. Analysis of bacterial genomes revealed that the dicistron is present in obligate anaerobic gut bacteria considered to be beneficial by virtue of their ability to produce butyrate. Thus, BCR-FDP may help to maintain anaerobiosis in the colon.


Asunto(s)
Proteínas Bacterianas , Fusobacterium nucleatum , Oxidación-Reducción , Oxígeno , Fusobacterium nucleatum/metabolismo , Fusobacterium nucleatum/genética , Fusobacterium nucleatum/enzimología , Oxígeno/metabolismo , Oxígeno/química , Proteínas Bacterianas/metabolismo , Proteínas Bacterianas/química , Proteínas Bacterianas/genética , Flavoproteínas Transportadoras de Electrones/metabolismo , Flavoproteínas Transportadoras de Electrones/genética , Flavoproteínas Transportadoras de Electrones/química , Transporte de Electrón , Acilcoenzima A/metabolismo , Butiril-CoA Deshidrogenasa/metabolismo , Butiril-CoA Deshidrogenasa/genética , Butiril-CoA Deshidrogenasa/química
2.
Int J Med Sci ; 18(16): 3631-3643, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34790035

RESUMEN

Background: Acyl-CoA dehydrogenase short-chain (ACADS) is a crucial enzyme in the fatty acid metabolism pathway located in mitochondria. However, the expression level and prognostic value of ACADS in colorectal cancer (CRC) remain unclear. Methods: The mRNA and protein expression data of ACADS was obtained from The Cancer Genome Atlas (TCGA), Clinical Proteomic Tumor Analysis Consortium (CPTAC), and Oncomine. Prognostic values of ACADS were calculated using Kaplan-Meier survival analysis. Correlations between ACADS and immune infiltration were estimated using TIMER, CIBERSORT, EPIC, quanTIseq, and xCell. The UALCAN and MEXPRESS databases were utilized for Methylation analysis. The co-expression analysis based on mRNA expression and interaction network of ACADS were performed via several online tools. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis on ACADS co-expressed genes were performed using the Metascape. Results: The expression analysis demonstrated that ACADS was down-regulated in CRC tissues compared with paired normal tissue. Expression of ACADS was found to be significantly associated with clinical cancer stages and the consensus molecular subgroups (CMS) constituent ratio in CRC patients. Besides, lower ACADS expression was found to predict poor prognosis and be significantly associated with common immune checkpoint genes and MMR genes in CRC. ACADS expression levels were positively related to B cells, CD4+ T cells, CD8+ T cells, M1 macrophages, neutrophils, and Tregs, while negatively correlated with M0 macrophages, M2 macrophages. The methylation level of ACADS in normal tissues was significantly higher than that in tumor tissues, and several methylation sites were identified. The enrichment analysis suggested the co-expressed genes mainly enriched in cell mitochondrial metabolism. Conclusions: The present study provided multilevel evidences for expression of ACADS in CRC and the function of ACADS in prognostic prediction, immune infiltration, and methylation. ACADS might have the potential as the novel biomarker and therapeutic target in CRC patients.


Asunto(s)
Butiril-CoA Deshidrogenasa/genética , Butiril-CoA Deshidrogenasa/metabolismo , Neoplasias Colorrectales/diagnóstico , Adenocarcinoma/diagnóstico , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/mortalidad , Biomarcadores de Tumor/genética , Biomarcadores de Tumor/metabolismo , Carcinoma/diagnóstico , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma/mortalidad , Línea Celular Tumoral , Quimiotaxis de Leucocito/genética , Quimiotaxis de Leucocito/inmunología , Neoplasias Colorrectales/genética , Neoplasias Colorrectales/metabolismo , Neoplasias Colorrectales/mortalidad , Ácidos Grasos/metabolismo , Perfilación de la Expresión Génica , Regulación Enzimológica de la Expresión Génica , Regulación Neoplásica de la Expresión Génica , Humanos , Metabolismo de los Lípidos/genética , Valor Predictivo de las Pruebas , Pronóstico , Proteómica , Análisis de Supervivencia , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
3.
Fungal Genet Biol ; 127: 23-34, 2019 06.
Artículo en Inglés | MEDLINE | ID: mdl-30822500

RESUMEN

Short-chain acyl-CoA dehydrogenase (Scad) mediated ß-oxidation serves as the fastest route for generating essential energies required to support the survival of organisms under stress or starvation. In this study, we identified three putative SCAD genes in the genome of the globally destructive rice blast pathogen Magnaporthe oryzae, named as MoSCAD1, MoSCAD2, and MoSCAD3. To elucidate their function, we deployed targeted gene deletion strategy to investigate individual and the combined influence of MoSCAD genes on growth, stress tolerance, conidiation and pathogenicity of the rice blast fungus. First, localization and co-localization results obtained from this study showed that MoScad1 localizes to the endoplasmic reticulum (ER), MoScad2 localizes exclusively to the mitochondria while MoScad3 partially localizes to the mitochondria and peroxisome at all developmental stages of M. oryzae. Results obtained from this investigation showed that the deletion of MoSCAD1 and MoSCAD2 caused a minimal but significant reduction in the growth of ΔMoscad1 and ΔMoscad2 strains, while, growth characteristics exhibited by the ΔMoscad3 strain was similar to the wild-type strain. Furthermore, we observed that deletion of MoSCAD2 resulted in drastic reduction in conidiation, delayed conidia germination, triggered the development of abnormal appressorium and suppressed host penetration and colonization efficiencies of the ΔMoscad1 strain. This study provides first material evidence confirming the possible existence of ER ß-oxidation pathway in M. oryzae. We also infer that mitochondria ß-oxidation rather than peroxisomal and ER ß-oxidation play an essential role in the vegetative growth, conidiation, appressorial morphogenesis and progression of pathogenesis in M. oryzae.


Asunto(s)
Butiril-CoA Deshidrogenasa/genética , Proteínas Fúngicas/genética , Magnaporthe/genética , Magnaporthe/patogenicidad , Esporas Fúngicas/crecimiento & desarrollo , Retículo Endoplásmico , Radicales Libres/metabolismo , Eliminación de Gen , Regulación Fúngica de la Expresión Génica , Magnaporthe/enzimología , Mitocondrias/metabolismo , Oryza/microbiología , Oxidación-Reducción , Peroxisomas/metabolismo , Enfermedades de las Plantas/microbiología , Esporas Fúngicas/genética
4.
BMC Med Genet ; 19(1): 64, 2018 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-29678161

RESUMEN

BACKGROUND: Short-chain acyl-CoA dehydrogenase deficiency (SCADD) represents a rare autosomal recessive inborn metabolic disorder of mitochondrial ß-oxidation of monocarboxylic acids. Clinical symptoms can vary from a severe life-threatening condition to an asymptomatic state, reported in the majority of cases. Since the expansion of newborn screenings, more than three hundred probands were admitted for molecular-genetic analysis, most selected because of elevated values of C4-acylcarnitine detected in newborn screenings in Slovakia. Searching for the principal genomic changes led us to the selection of sixty-two patients in whom the presence of sequence variants in the ACADS gene was analysed and correlated with the available biochemical and clinical data. METHODS: Biochemical and molecular genetic tests were performed. Acylcarnitine profiles focused on an elevated level of C4-acylcarnitine, which was analysed via tandem mass spectrometry. Urinary organic acids, specifically a quantity of ethylmalonic acid, were determined by gas chromatography/mass spectrometry. The entire coding region of the ACADS gene was sequenced. A low-cost restriction fragment length polymorphism of PCR amplified fragments analysis (PCR-RFLP) of pathogenic variants was introduced and implemented for the molecular-genetic algorithm appropriate for the Slovak population. RESULTS: Our molecular genetic study was performed on sixty-two patients with a pathological biochemical pattern related to short-chain acyl-CoA dehydrogenase deficiency. In this cohort, we discovered a high occurrence of two rare pathogenic variants-the deletion c.310_312delGAG and the substitution c.1138C>T, with allelic frequencies of 64% and 31%, respectively. Up to 86% of investigated individuals belong to the Roma ethnic group. CONCLUSIONS: Analogous to other countries, SCADD is not included in the newborn screening programme. Based on the exceeded levels of the specific biomarker C4-acylcarnitine as well as ethylmalonic acid, we revealed a high prevalence of short-chain acyl-CoA dehydrogenase deficiency cases, confirmed by the findings of two rare pathogenic variants. A deletion c.310_312delGAG and c.1138C > T substitution in the ACADS gene appear with a high frequency in the Roma ethnic group of Slovakia. Due to the uncertainty of the pathogenicity and clinical consequences, it is important to follow up the morbidity and mortality in these patients over time and evaluate SCADD in relation to clinical outcomes and preventive healthcare recommendations.


Asunto(s)
Acil-CoA Deshidrogenasa/deficiencia , Butiril-CoA Deshidrogenasa/genética , Carnitina/análogos & derivados , Etnicidad/genética , Errores Innatos del Metabolismo Lipídico/genética , Mutación , Acil-CoA Deshidrogenasa/genética , Acil-CoA Deshidrogenasa/metabolismo , Carnitina/metabolismo , Femenino , Frecuencia de los Genes , Pruebas Genéticas , Humanos , Recién Nacido , Errores Innatos del Metabolismo Lipídico/etnología , Errores Innatos del Metabolismo Lipídico/metabolismo , Masculino , Tamizaje Neonatal/métodos , Polimorfismo de Longitud del Fragmento de Restricción , Análisis de Secuencia de ADN , Eslovaquia/etnología
5.
J Cell Mol Med ; 19(7): 1672-88, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25753319

RESUMEN

This study was designed to investigate the expression of short-chain acyl-CoA dehydrogenase (SCAD), a key enzyme of fatty acid ß-oxidation, during rat heart development and the difference of SCAD between pathological and physiological cardiac hypertrophy. The expression of SCAD was lowest in the foetal and neonatal heart, which had time-dependent increase during normal heart development. In contrast, a significant decrease in SCAD expression was observed in different ages of spontaneously hypertensive rats (SHR). On the other hand, swim-trained rats developed physiological cardiac hypertrophy, whereas SHR developed pathological cardiac hypertrophy. The two kinds of cardiac hypertrophy exhibited divergent SCAD changes in myocardial fatty acids utilization. In addition, the expression of SCAD was significantly decreased in pathological cardiomyocyte hypertrophy, however, increased in physiological cardiomyocyte hypertrophy. SCAD siRNA treatment triggered the pathological cardiomyocyte hypertrophy, which showed that the down-regulation of SCAD expression may play an important role in pathological cardiac hypertrophy. The changes in peroxisome proliferator-activated receptor α (PPARα) was accordant with that of SCAD. Moreover, the specific PPARα ligand fenofibrate treatment increased the expression of SCAD and inhibited pathological cardiac hypertrophy. Therefore, we speculate that the down-regulated expression of SCAD in pathological cardiac hypertrophy may be responsible for 'the recapitulation of foetal energy metabolism'. The deactivation of PPARα may result in the decrease in SCAD expression in pathological cardiac hypertrophy. Changes in SCAD are different in pathological and physiological cardiac hypertrophy, which may be used as the molecular markers of pathological and physiological cardiac hypertrophy.


Asunto(s)
Butiril-CoA Deshidrogenasa/metabolismo , Cardiomegalia/enzimología , Corazón/crecimiento & desarrollo , Animales , Animales Recién Nacidos , Presión Sanguínea/efectos de los fármacos , Butiril-CoA Deshidrogenasa/genética , Cardiomegalia/diagnóstico por imagen , Cardiomegalia/patología , Cardiomegalia/fisiopatología , Modelos Animales de Enfermedad , Ácidos Grasos/metabolismo , Fenofibrato/farmacología , Corazón/efectos de los fármacos , Corazón/fisiopatología , Ventrículos Cardíacos/patología , Ventrículos Cardíacos/fisiopatología , Factor I del Crecimiento Similar a la Insulina/farmacología , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Miocitos Cardíacos/patología , Tamaño de los Órganos/efectos de los fármacos , Oxidación-Reducción/efectos de los fármacos , PPAR alfa/metabolismo , Fenilefrina/farmacología , Interferencia de ARN/efectos de los fármacos , ARN Mensajero/genética , ARN Mensajero/metabolismo , ARN Interferente Pequeño/metabolismo , Ratas Endogámicas SHR , Ratas Wistar , Especificidad por Sustrato/efectos de los fármacos , Sístole/efectos de los fármacos , Factores de Tiempo , Ultrasonografía
6.
Appl Environ Microbiol ; 81(14): 4782-90, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25956767

RESUMEN

Eubacterium limosum KIST612 is one of the few acetogens that can produce butyrate from carbon monoxide. We have used a genome-guided analysis to delineate the path of butyrate formation, the enzymes involved, and the potential coupling to ATP synthesis. Oxidation of CO is catalyzed by the acetyl-coenzyme A (CoA) synthase/CO dehydrogenase and coupled to the reduction of ferredoxin. Oxidation of reduced ferredoxin is catalyzed by the Rnf complex and Na(+) dependent. Consistent with the finding of a Na(+)-dependent Rnf complex is the presence of a conserved Na(+)-binding motif in the c subunit of the ATP synthase. Butyrate formation is from acetyl-CoA via acetoacetyl-CoA, hydroxybutyryl-CoA, crotonyl-CoA, and butyryl-CoA and is consistent with the finding of a gene cluster that encodes the enzymes for this pathway. The activity of the butyryl-CoA dehydrogenase was demonstrated. Reduction of crotonyl-CoA to butyryl-CoA with NADH as the reductant was coupled to reduction of ferredoxin. We postulate that the butyryl-CoA dehydrogenase uses flavin-based electron bifurcation to reduce ferredoxin, which is consistent with the finding of etfA and etfB genes next to it. The overall ATP yield was calculated and is significantly higher than the one obtained with H2 + CO2. The energetic benefit may be one reason that butyrate is formed only from CO but not from H2 + CO2.


Asunto(s)
Butiratos/metabolismo , Monóxido de Carbono/metabolismo , Eubacterium/metabolismo , Acilcoenzima A/metabolismo , Adenosina Trifosfato/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Butiril-CoA Deshidrogenasa/genética , Butiril-CoA Deshidrogenasa/metabolismo , Metabolismo Energético , Eubacterium/enzimología , Eubacterium/genética , Flavinas/metabolismo , Genómica , Oxidación-Reducción
7.
Mol Genet Metab ; 111(3): 360-368, 2014 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-24485985

RESUMEN

Short-chain acyl-CoA dehydrogenase (SCAD) deficiency is a rare inherited autosomal recessive disorder with not yet well established mechanisms of disease. In the present study, the mitochondrial proteome of five symptomatic patients homozygous for missense variations in the SCAD gene ACADS was investigated in an extensive large-scale proteomic study to map protein perturbations linked to the disease. Fibroblast cultures of patient cells homozygous for either c.319C>T/p.Arg107Cys (n=2) or c.1138C>T/p.Arg380Trp (n=3) in ACADS, and healthy controls (normal human dermal fibroblasts), were studied. The mitochondrial proteome derived from these cultures was analyzed by label free proteomics using high mass accuracy nanoliquid chromatography tandem mass spectrometry (nanoLC-MS/MS). More than 300 mitochondrial proteins were identified and quantified. Thirteen proteins had significant alteration in protein levels in patients carrying variation c.319C>T in ACADS compared to controls and they belonged to various pathways, such as the antioxidant system and amino acid metabolism. Twenty-two proteins were found significantly altered in patients carrying variation c.1138C>T which included proteins associated with fatty acid ß-oxidation, amino acid metabolism and protein quality control system. Three proteins were found significantly regulated in both patient groups: adenylate kinase 4 (AK4), nucleoside diphosphate kinase A (NME1) and aldehyde dehydrogenase family 4 member A1 (ALDH4A1). Proteins AK4 and NME1 deserve further investigation because of their involvement in energy reprogramming, cell survival and proliferation with relevance for SCAD deficiency and related metabolic disorders.


Asunto(s)
Acil-CoA Deshidrogenasa/deficiencia , Butiril-CoA Deshidrogenasa/genética , Errores Innatos del Metabolismo Lipídico/genética , Mitocondrias/genética , Proteínas Mitocondriales/biosíntesis , Acil-CoA Deshidrogenasa/genética , Acil-CoA Deshidrogenasa/metabolismo , Butiril-CoA Deshidrogenasa/metabolismo , Células Cultivadas , Femenino , Fibroblastos/citología , Fibroblastos/metabolismo , Regulación de la Expresión Génica , Humanos , Errores Innatos del Metabolismo Lipídico/metabolismo , Errores Innatos del Metabolismo Lipídico/patología , Masculino , Mitocondrias/patología , Estrés Oxidativo/genética , Proteómica , Espectrometría de Masas en Tándem
8.
J Ind Microbiol Biotechnol ; 41(9): 1425-33, 2014 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25037723

RESUMEN

Klebsiella pneumoniae synthesize large amounts of L-2,3-butanediol (L-2,3-BD), but the underlying mechanism has been unknown. In this study, we provide the first identification and characterization of an L-2,3-BD dehydrogenase from K. pneumoniae, demonstrating its reductive activities toward diacetyl and acetoin, and oxidative activity toward L-2,3-BD. Optimum pH, temperature, and kinetics determined for reductive and oxidative reactions support the preferential production of 2,3-BD during cell growth. Synthesis of L-2,3-BD was remarkably enhanced by increasing gene dosage, reaching levels that, to the best of our knowledge, are the highest achieved to date.


Asunto(s)
Proteínas Bacterianas/química , Proteínas Bacterianas/metabolismo , Butileno Glicoles/metabolismo , Butiril-CoA Deshidrogenasa/química , Butiril-CoA Deshidrogenasa/metabolismo , Klebsiella pneumoniae/enzimología , Acetoína/metabolismo , Secuencia de Aminoácidos , Proteínas Bacterianas/genética , Butiril-CoA Deshidrogenasa/genética , Estabilidad de Enzimas , Klebsiella pneumoniae/química , Klebsiella pneumoniae/genética , Klebsiella pneumoniae/metabolismo , Datos de Secuencia Molecular , Alineación de Secuencia
9.
Aging Cell ; 23(10): e14256, 2024 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-38898632

RESUMEN

Hepatic steatosis, the first step in the development of nonalcoholic fatty liver disease (NAFLD), is frequently observed in the aging population. However, the underlying molecular mechanism remains largely unknown. In this study, we first employed GSEA enrichment analysis to identify short-chain acyl-CoA dehydrogenase (SCAD), which participates in the mitochondrial ß-oxidation of fatty acids and may be associated with hepatic steatosis in elderly individuals. Subsequently, we examined SCAD expression and hepatic triglyceride content in various aged humans and mice and found that triglycerides were markedly increased and that SCAD was upregulated in aged livers. Our further evidence in SCAD-ablated mice suggested that SCAD deletion was able to slow liver aging and ameliorate aging-associated fatty liver. Examination of the molecular pathways by which the deletion of SCAD attenuates steatosis revealed that the autophagic degradation of lipid droplets, which was not detected in elderly wild-type mice, was maintained in SCAD-deficient old mice. This was due to the decrease in the production of acetyl-coenzyme A (acetyl-CoA), which is abundant in the livers of old wild-type mice. In conclusion, our findings demonstrate that the suppression of SCAD may prevent age-associated hepatic steatosis by promoting lipophagy and that SCAD could be a promising therapeutic target for liver aging and associated steatosis.


Asunto(s)
Envejecimiento , Autofagia , Butiril-CoA Deshidrogenasa , Hígado Graso , Animales , Humanos , Masculino , Ratones , Envejecimiento/metabolismo , Autofagia/genética , Butiril-CoA Deshidrogenasa/metabolismo , Butiril-CoA Deshidrogenasa/genética , Hígado Graso/genética , Hígado Graso/metabolismo , Hígado Graso/patología , Hígado/metabolismo , Hígado/patología , Ratones Endogámicos C57BL
10.
J Bacteriol ; 195(16): 3704-13, 2013 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-23772070

RESUMEN

The butyrogenic genes from Clostridium difficile DSM 1296(T) have been cloned and expressed in Escherichia coli. The enzymes acetyl-coenzyme A (CoA) C-acetyltransferase, 3-hydroxybutyryl-CoA dehydrogenase, crotonase, phosphate butyryltransferase, and butyrate kinase and the butyryl-CoA dehydrogenase complex composed of the dehydrogenase and two electron-transferring flavoprotein subunits were individually produced in E. coli and kinetically characterized in vitro. While most of these enzymes were measured using well-established test systems, novel methods to determine butyrate kinase and butyryl-CoA dehydrogenase activities with respect to physiological function were developed. Subsequently, the individual genes were combined to form a single plasmid-encoded operon in a plasmid vector, which was successfully used to confer butyrate-forming capability to the host. In vitro and in vivo studies demonstrated that C. difficile possesses a bifurcating butyryl-CoA dehydrogenase which catalyzes the NADH-dependent reduction of ferredoxin coupled to the reduction of crotonyl-CoA also by NADH. Since the reoxidation of ferredoxin by a membrane-bound ferredoxin:NAD(+)-oxidoreductase enables electron transport phosphorylation, additional ATP is formed. The butyryl-CoA dehydrogenase from C. difficile is oxygen stable and apparently uses oxygen as a co-oxidant of NADH in the presence of air. These properties suggest that this enzyme complex might be well suited to provide butyryl-CoA for solventogenesis in recombinant strains. The central role of bifurcating butyryl-CoA dehydrogenases and membrane-bound ferredoxin:NAD oxidoreductases (Rhodobacter nitrogen fixation [RNF]), which affect the energy yield of butyrate fermentation in the clostridial metabolism, is discussed.


Asunto(s)
Butiratos/metabolismo , Butiril-CoA Deshidrogenasa/metabolismo , Clostridioides difficile/metabolismo , Flavoproteínas Transportadoras de Electrones/metabolismo , Escherichia coli/metabolismo , Oxígeno , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Butiril-CoA Deshidrogenasa/genética , Clonación Molecular , Clostridioides difficile/enzimología , Clostridioides difficile/genética , Flavoproteínas Transportadoras de Electrones/genética , Escherichia coli/genética , Regulación Bacteriana de la Expresión Génica/fisiología , Proteínas Recombinantes
11.
Eur J Pharmacol ; 954: 175849, 2023 Sep 05.
Artículo en Inglés | MEDLINE | ID: mdl-37331684

RESUMEN

Short-chain acyl-CoA dehydrogenase (SCAD), the rate-limiting enzyme for fatty acid ß-oxidation, has a negative regulatory effect on pathological cardiac hypertrophy and fibrosis. FAD, a coenzyme of SCAD, participates in the electron transfer of SCAD-catalyzed fatty acid ß-oxidation, which plays a crucial role in maintaining the balance of myocardial energy metabolism. Insufficient riboflavin intake can lead to symptoms similar to short-chain acyl-CoA dehydrogenase (SCAD) deficiency or flavin adenine dinucleotide (FAD) gene abnormality, which can be alleviated by riboflavin supplementation. However, whether riboflavin can inhibit pathological cardiac hypertrophy and fibrosis remains unclear. Therefore, we observed the effect of riboflavin on pathological cardiac hypertrophy and fibrosis. In vitro experiments, riboflavin increased SCAD expression and the content of ATP, decreased the free fatty acids content and improved PE-induced cardiomyocytes hypertrophy and AngⅡ-induced cardiac fibroblasts proliferation by increasing the content of FAD, which were attenuated by knocking down the expression of SCAD using small interfering RNA. In vivo experiments, riboflavin significantly increased the expression of SCAD and the energy metabolism of the heart to improve TAC induced pathological myocardial hypertrophy and fibrosis in mice. The results demonstrate that riboflavin improves pathological cardiac hypertrophy and fibrosis by increasing the content of FAD to activate SCAD, which may be a new strategy for treating pathological cardiac hypertrophy and fibrosis.


Asunto(s)
Butiril-CoA Deshidrogenasa , Flavina-Adenina Dinucleótido , Animales , Ratones , Butiril-CoA Deshidrogenasa/genética , Butiril-CoA Deshidrogenasa/metabolismo , Flavina-Adenina Dinucleótido/farmacología , Riboflavina/farmacología , Cardiomegalia/patología , Ácidos Grasos no Esterificados , Fibrosis
12.
J Hypertens ; 41(5): 775-793, 2023 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-36883465

RESUMEN

OBJECTIVES: Short-chain acyl-CoA dehydrogenase (SCAD), a key enzyme in the fatty acid oxidation process, is not only involved in ATP synthesis but also regulates the production of mitochondrial reactive oxygen species (ROS) and nitric oxide synthesis. The purpose of this study was to investigate the possible role of SCAD in hypertension-associated vascular remodelling. METHODS: In-vivo experiments were performed on spontaneously hypertensive rats (SHRs, ages of 4 weeks to 20 months) and SCAD knockout mice. The aorta sections of hypertensive patients were used for measurement of SCAD expression. In-vitro experiments with t-butylhydroperoxide (tBHP), SCAD siRNA, adenovirus-SCAD (MOI 90) or shear stress (4, 15 dynes/cm 2 ) were performed using human umbilical vein endothelial cells (HUVECs). RESULTS: Compared with age-matched Wistar rats, aortic SCAD expression decreased gradually in SHRs with age. In addition, aerobic exercise training for 8 weeks could significantly increase SCAD expression and enzyme activity in the aortas of SHRs while decreasing vascular remodelling in SHRs. SCAD knockout mice also exhibited aggravated vascular remodelling and cardiovascular dysfunction. Likewise, SCAD expression was also decreased in tBHP-induced endothelial cell apoptosis models and the aortas of hypertensive patients. SCAD siRNA caused HUVEC apoptosis in vitro , whereas adenovirus-mediated SCAD overexpression (Ad-SCAD) protected against HUVEC apoptosis. Furthermore, SCAD expression was decreased in HUVECs exposed to low shear stress (4 dynes/cm 2 ) and increased in HUVECs exposed to 15 dynes/cm 2 compared with those under static conditions. CONCLUSION: SCAD is a negative regulator of vascular remodelling and may represent a novel therapeutic target for vascular remodelling.


Asunto(s)
Butiril-CoA Deshidrogenasa , Hipertensión , Ratas , Animales , Ratones , Humanos , Recién Nacido , Butiril-CoA Deshidrogenasa/genética , Butiril-CoA Deshidrogenasa/metabolismo , Remodelación Vascular , Ratas Endogámicas SHR , Ratas Wistar , Células Endoteliales de la Vena Umbilical Humana/metabolismo , ARN Interferente Pequeño/metabolismo , Ratones Noqueados
13.
J Ind Microbiol Biotechnol ; 39(8): 1101-7, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22776992

RESUMEN

A homobutanol fermentation pathway was engineered in a derivative of Escherichia coli B (glucose [glycolysis] => 2 pyruvate + 2 NADH; pyruvate [pyruvate dehydrogenase] => acetyl-CoA + NADH; 2 acetyl-CoA [butanol pathway enzymes] + 4 NADH => butanol; summary stoichiometry: glucose => butanol). Initially, the native fermentation pathways were eliminated from E. coli B by deleting the genes encoding for lactate dehydrogenase (ldhA), acetate kinase (ackA), fumarate reductase (frdABCD), pyruvate formate lyase (pflB), and alcohol dehydrogenase (adhE), and the pyruvate dehydrogenase complex (aceEF-lpd) was anaerobically expressed through promoter replacement. The resulting strain, E. coli EG03 (ΔfrdABCD ΔldhA ΔackA ΔpflB Δ adhE ΔpdhR ::pflBp6-aceEF-lpd ΔmgsA), could generate 4 NADH for every glucose oxidized to two acetyl-CoA through glycolysis and the pyruvate dehydrogenase complex. However, EG03 lost its ability for anaerobic growth due to the lack of NADH oxidation pathways. When the butanol pathway genes that encode for acetyl-CoA acetyltransferase (thiL), 3-hydroxybutyryl-CoA dehydrogenase (hbd), crotonase (crt), butyryl-CoA dehydrogenase (bcd, etfA, etfB), and butyraldehyde dehydrogenase (adheII) were cloned from Clostridium acetobutylicum ATCC 824, and expressed in E. coli EG03, a balanced NADH oxidation pathway was established for homobutanol fermentation (glucose => 4 NADH + 2 acetyl-CoA => butanol). This strain was able to convert glucose to butanol (1,254 mg l(-1)) under anaerobic condition.


Asunto(s)
1-Butanol/metabolismo , Reactores Biológicos , Butanoles/metabolismo , Escherichia coli/metabolismo , Fermentación , Ingeniería Metabólica , 3-Hidroxiacil-CoA Deshidrogenasas/genética , 3-Hidroxiacil-CoA Deshidrogenasas/metabolismo , Acetil-CoA C-Acetiltransferasa/genética , Acetil-CoA C-Acetiltransferasa/metabolismo , Alcohol Deshidrogenasa/genética , Alcohol Deshidrogenasa/metabolismo , Biocombustibles , Butiril-CoA Deshidrogenasa/genética , Butiril-CoA Deshidrogenasa/metabolismo , Clostridium acetobutylicum/enzimología , Clostridium acetobutylicum/genética , Enoil-CoA Hidratasa/genética , Enoil-CoA Hidratasa/metabolismo , Escherichia coli/clasificación , Escherichia coli/genética , Glucosa/metabolismo , Glucólisis , NAD/metabolismo , Oxidación-Reducción , Complejo Piruvato Deshidrogenasa/genética , Complejo Piruvato Deshidrogenasa/metabolismo
14.
J Pediatr Endocrinol Metab ; 25(7-8): 795-7, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23155713

RESUMEN

Short-chain acyl-CoA dehydrogenase deficiency (SCADD) is a rare autosomal recessive inborn error of mitochondrial fatty acid oxidation. It is caused by rare mutations as well as polymorphic susceptibility variants. We describe here the case of a 1-year-old male patient who had growth and mental retardation, seizures, and recurring fever since infancy. Urinary gas chromatography/mass spectrometry (GC/MS) showed elevated levels of ethylmalonic acid. Plasma acylcarnitines on tandem mass spectrometry (MS/MS) and elevations of C4-cartinitine are consistently present. The two polymorphic susceptibility variants of the short-chain acyl-CoA dehydrogenase (SCAD) gene, c.625G>A and c.322G>A, were detected. Because of its highly variable clinical characteristics, there are no related reports in China. This report broadens the phenotype and genotype of SCADD in China and underlines the difficulty of diagnosis.


Asunto(s)
Errores Innatos del Metabolismo Lipídico/diagnóstico , Acil-CoA Deshidrogenasa/deficiencia , Acil-CoA Deshidrogenasa/genética , Butiril-CoA Deshidrogenasa/genética , China , Genotipo , Humanos , Lactante , Discapacidad Intelectual/complicaciones , Discapacidad Intelectual/diagnóstico , Discapacidad Intelectual/genética , Errores Innatos del Metabolismo Lipídico/complicaciones , Errores Innatos del Metabolismo Lipídico/genética , Masculino , Polimorfismo de Nucleótido Simple/fisiología , Convulsiones/complicaciones , Convulsiones/congénito , Convulsiones/diagnóstico , Convulsiones/genética
15.
Metab Eng ; 13(4): 353-63, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21569861

RESUMEN

Production of chemicals and fuels directly from CO(2) is an attractive approach to solving the energy and environmental problems. 1-Butanol, a chemical feedstock and potential fuel, has been produced by fermentation of carbohydrates, both in native Clostridium species and various engineered hosts. To produce 1-butanol from CO(2), we transferred a modified CoA-dependent 1-butanol production pathway into a cyanobacterium, Synechococcus elongatus PCC 7942. We demonstrated the activity of each enzyme in the pathway by chromosomal integration and expression of the genes. In particular, Treponema denticola trans-enoyl-CoA reductase (Ter), which utilizes NADH as the reducing power, was used for the reduction of crotonyl-CoA to butyryl-CoA instead of Clostridium acetobutylicum butyryl-CoA dehydrogenase to by-pass the need of Clostridial ferredoxins. Addition of polyhistidine-tag increased the overall activity of Ter and resulted in higher 1-butanol production. Removal of oxygen is an important factor in the synthesis of 1-butanol in this organism. This result represents the first autotrophic 1-butanol production.


Asunto(s)
1-Butanol/metabolismo , Dióxido de Carbono/metabolismo , Ingeniería Genética , Organismos Modificados Genéticamente , Synechococcus , Proteínas Bacterianas/biosíntesis , Proteínas Bacterianas/genética , Butiril-CoA Deshidrogenasa/biosíntesis , Butiril-CoA Deshidrogenasa/genética , Clostridium acetobutylicum/enzimología , Clostridium acetobutylicum/genética , Ácido Graso Desaturasas/biosíntesis , Ácido Graso Desaturasas/genética , Organismos Modificados Genéticamente/genética , Organismos Modificados Genéticamente/metabolismo , Synechococcus/genética , Synechococcus/metabolismo , Treponema denticola/enzimología , Treponema denticola/genética
16.
BMC Med Genet ; 12: 4, 2011 Jan 06.
Artículo en Inglés | MEDLINE | ID: mdl-21211036

RESUMEN

BACKGROUND: A genome-wide association study (GWAS) using metabolite concentrations as proxies for enzymatic activity, suggested that two variants: rs2014355 in the gene encoding short-chain acyl-coenzyme A dehydrogenase (ACADS) and rs11161510 in the gene encoding medium-chain acyl-coenzyme A dehydrogenase (ACADM) impair fatty acid ß-oxidation. Chronic exposure to fatty acids due to an impaired ß-oxidation may down-regulate the glucose-stimulated insulin release and result in an increased risk of type 2 diabetes (T2D). We aimed to investigate whether the two variants associate with altered insulin release following an oral glucose load or with T2D. METHODS: The variants were genotyped using KASPar® PCR SNP genotyping system and investigated for associations with estimates of insulin release and insulin sensitivity following an oral glucose tolerance test (OGTT) in a random sample of middle-aged Danish individuals (nACADS = 4,324; nACADM = 4,337). The T2D-case-control study involved a total of ~8,300 Danish individuals (nACADS = 8,313; nACADM = 8,344). RESULTS: In glucose-tolerant individuals the minor C-allele of rs2014355 of ACADS associated with reduced measures of serum insulin at 30 min following an oral glucose load (per allele effect (ß) = -3.8% (-6.3%;-1.3%), P = 0.003), reduced incremental area under the insulin curve (ß = -3.6% (-6.3%;-0.9%), P = 0.009), reduced acute insulin response (ß = -2.2% (-4.2%;0.2%), P = 0.03), and with increased insulin sensitivity ISIMatsuda (ß = 2.9% (0.5%;5.2%), P = 0.02). The C-allele did not associate with two other measures of insulin sensitivity or with a derived disposition index. The C-allele was not associated with T2D in the case-control analysis (OR 1.07, 95% CI 0.96-1.18, P = 0.21). rs11161510 of ACADM did not associate with any indices of glucose-stimulated insulin release or with T2D. CONCLUSIONS: In glucose-tolerant individuals the minor C-allele of rs2014355 of ACADS was associated with reduced measures of glucose-stimulated insulin release during an OGTT, a finding which in part may be mediated through an impaired ß-oxidation of fatty acids.


Asunto(s)
Alelos , Glucemia/genética , Butiril-CoA Deshidrogenasa/genética , Insulina/sangre , Insulina/metabolismo , Adulto , Estudios de Casos y Controles , Ensayos Clínicos como Asunto , Estudios de Cohortes , Diabetes Mellitus Tipo 2/epidemiología , Diabetes Mellitus Tipo 2/genética , Femenino , Prueba de Tolerancia a la Glucosa , Humanos , Secreción de Insulina , Masculino , Persona de Mediana Edad , Prevalencia
17.
Neuropediatrics ; 42(1): 13-7, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21500142

RESUMEN

Short-chain acyl-CoA dehydrogenase deficiency (SCADD) is an autosomal recessive inborn error of metabolism, most frequently associated with developmental delay and/or epilepsy. Most SCADD patients carry common SCAD-encoding gene ( ACADS) variants or these variants in combination with a rare ACADS mutation, in the Netherlands predominantly the c.1058C>T. Epilepsy in childhood often remains unexplained and patients with epilepsy related to SCADD may remain undiagnosed because studies for SCADD are often not performed. To test this hypothesis and to further estimate the extent of the Dutch SCADD population, we performed a study on blood spot samples in 131 paediatric patients with epilepsy and 909 anonymous newborns and investigated the presence of the 2 common ACADS variants and the rare c.1058C>T mutation. Overall, the 2 common ACADS variants and the rare c.1058C>T mutation were detected in either homozygous or compound heterozygous forms in 9.2% of the epilepsy and 7.5% of the reference group. A birth prevalence of SCADD with a mutation/variant genotype in the Netherlands as high as >1:1,000 was calculated. This is in contrast with the low number of patients diagnosed clinically and supports the hypothesis that SCADD is clinically irrelevant. Furthermore our study does not support an association between SCADD and epilepsy.


Asunto(s)
Epilepsia/epidemiología , Errores Innatos del Metabolismo Lipídico/epidemiología , Acil-CoA Deshidrogenasa/deficiencia , Acil-CoA Deshidrogenasa/genética , Adolescente , Butiril-CoA Deshidrogenasa/genética , Niño , Preescolar , Análisis Mutacional de ADN , Femenino , Humanos , Incidencia , Lactante , Recién Nacido , Errores Innatos del Metabolismo Lipídico/diagnóstico , Errores Innatos del Metabolismo Lipídico/genética , Masculino , Mutación/genética , Países Bajos/epidemiología , Pediatría
18.
Hum Genet ; 127(6): 619-28, 2010 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-20376488

RESUMEN

Short-chain acyl-CoA dehydrogenase (SCAD) is a mitochondrial enzyme involved in the beta-oxidation of fatty acids. Genetic defect of SCAD was documented to cause clinical symptoms such as progressive psychomotor retardation, muscle hypotonia, and myopathy in early reports. However, clinical significance of SCAD deficiency (SCADD) has been getting ambiguous, for some variants in the ACADS gene, which encodes the SCAD protein, has turned out to be widely prevailed among general populations. Accordingly, the pathophysiology of SCADD has not been clarified thus far. The present report focuses on two suspected cases of SCADD detected through the screening of newborns by tandem mass spectrometry. In both subjects, compound heterozygous mutations in ACADS were detected. The mutated genes were expressed in a transient gene expression system, and the enzymatic activities of the obtained mutant SCAD proteins were measured. The activities of the mutant SCAD proteins were significantly lower than that of the wild-type enzyme, confirming the mechanism underlying the diagnosis of SCADD in both subjects. Moreover, the mutant SCAD proteins gave rise to mitochondrial fragmentation and autophagy, both of which were proportional to the decrease in SCAD activities. The association of autophagy with programmed cell death suggests that the mutant SCAD proteins are toxic to mitochondria and to the cells in which they are expressed. The expression of recombinant ACADS-encoded mutant proteins offers a technique to evaluate both the nature of the defective SCAD proteins and their toxicity. Moreover, our results provide insight into possible molecular pathophysiology of SCADD.


Asunto(s)
Butiril-CoA Deshidrogenasa/deficiencia , Butiril-CoA Deshidrogenasa/genética , Genes , Trastornos del Metabolismo de los Lípidos/genética , Mutación , Errores Innatos del Metabolismo de los Aminoácidos/genética , Errores Innatos del Metabolismo de los Aminoácidos/metabolismo , Secuencia de Bases , Butiril-CoA Deshidrogenasa/metabolismo , Genotipo , Heterocigoto , Humanos , Recién Nacido , Trastornos del Metabolismo de los Lípidos/metabolismo , Mitocondrias/genética , Mitocondrias/metabolismo , Datos de Secuencia Molecular , Oxidación-Reducción , Estructura Secundaria de Proteína/genética , Proteínas Recombinantes/metabolismo
19.
Biochem Biophys Res Commun ; 400(3): 318-22, 2010 Sep 24.
Artículo en Inglés | MEDLINE | ID: mdl-20727852

RESUMEN

Brown adipose tissue is a highly specialized organ that uses mitochondrial fatty acid oxidation to fuel non-shivering thermogenesis. In mice, mutations in the acyl-CoA dehydrogenase family of fatty acid oxidation genes are associated with sensitivity to cold. Brown adipose tissue function has not previously been characterized in these knockout strains. Short-chain acyl-CoA dehydrogenase (SCAD) deficient mice were found to have increased brown adipose tissue mass as well as modest cardiac hypertrophy. Uncoupling protein-1 was reduced by 70% in brown adipose tissue and this was not due to a change in mitochondrial number, nor was it due to decreased signal transduction through protein kinase A which is known to be a major regulator of uncoupling protein-1 expression. PKA activity and in vitro lipolysis were normal in brown adipose tissue, although in white adipose tissue a modest increase in basal lipolysis was seen in SCAD-/- mice. Finally, an in vivo norepinephrine challenge of brown adipose tissue thermogenesis revealed normal heat production in SCAD-/- mice. These results suggest that reduced brown adipose tissue function is not the major factor causing cold sensitivity in acyl-CoA dehydrogenase knockout strains. We speculate that other mechanisms such as shivering capacity, cardiac function, and reduced hepatic glycogen stores are involved.


Asunto(s)
Tejido Adiposo Pardo/fisiología , Butiril-CoA Deshidrogenasa/genética , Frío , Termogénesis/genética , Acil-CoA Deshidrogenasa de Cadena Larga/genética , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/enzimología , Animales , AMP Cíclico/farmacología , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Canales Iónicos/metabolismo , Lipólisis/genética , Ratones , Ratones Noqueados , Proteínas Mitocondriales/metabolismo , Norepinefrina/farmacología , Tiritona/genética , Proteína Desacopladora 1
20.
Biochem Biophys Res Commun ; 396(1): 125-30, 2010 May 21.
Artículo en Inglés | MEDLINE | ID: mdl-20494124

RESUMEN

Two large gene and protein superfamilies, SDR and MDR (short- and medium-chain dehydrogenases/reductases), were originally defined from analysis of alcohol and polyol dehydrogenases. The superfamilies contain minimally 82 and 25 genes, respectively, in humans, minimally 324 and 86 enzyme families when known lines in other organisms are also included, and over 47,000 and 15,000 variants in existing sequence data bank entries. SDR enzymes have one-domain subunits without metal and MDR two-domain subunits without or with zinc, and these three lines appear to have emerged in that order from the universal cellular ancestor. This is compatible with their molecular architectures, present multiplicity, and overall distribution in the kingdoms of life, with SDR also of viral occurrence. An MDR-zinc, when present, is often, but not always, catalytic. It appears also to have a structural role in inter-domain interactions, coenzyme binding and substrate pocket formation, as supported by domain variability ratios and ligand positions. Differences among structural and catalytic zinc ions may be relative and involve several states. Combined, the comparisons trace evolutionary properties of huge superfamilies, with partially redundant enzymes in cellular redox functions.


Asunto(s)
Acil-CoA Deshidrogenasa/clasificación , Butiril-CoA Deshidrogenasa/clasificación , Evolución Molecular , Metaloproteínas/clasificación , Zinc/metabolismo , Acil-CoA Deshidrogenasa/química , Acil-CoA Deshidrogenasa/genética , Butiril-CoA Deshidrogenasa/química , Butiril-CoA Deshidrogenasa/genética , Humanos , Metaloproteínas/química , Metaloproteínas/genética , Filogenia , Conformación Proteica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA