Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 53
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
J Biol Chem ; 299(7): 104853, 2023 07.
Artigo em Inglês | MEDLINE | ID: mdl-37220854

RESUMO

We have investigated the equilibrium properties and rapid-reaction kinetics of the isolated butyryl-CoA dehydrogenase (bcd) component of the electron-bifurcating crotonyl-CoA-dependent NADH:ferredoxin oxidoreductase (EtfAB-bcd) from Megasphaera elsdenii. We find that a neutral FADH• semiquinone accumulates transiently during both reduction with sodium dithionite and with NADH in the presence of catalytic concentrations of EtfAB. In both cases full reduction of bcd to the hydroquinone is eventually observed, but the accumulation of FADH• indicates that a substantial portion of reduction occurs in sequential one-electron processes rather than a single two-electron event. In rapid-reaction experiments following the reaction of reduced bcd with crotonyl-CoA and oxidized bcd with butyryl-CoA, long-wavelength-absorbing intermediates are observed that are assigned to bcdred:crotonyl-CoA and bcdox:butyryl-CoA charge-transfer complexes, demonstrating their kinetic competence in the course of the reaction. In the presence of crotonyl-CoA there is an accumulation of semiquinone that is unequivocally the anionic FAD•- rather than the neutral FADH• seen in the absence of substrate, indicating that binding of substrate/product results in ionization of the bcd semiquinone. In addition to fully characterizing the rapid-reaction kinetics of both the oxidative and reductive half-reactions, our results demonstrate that one-electron processes play an important role in the reduction of bcd in EtfAB-bcd.


Assuntos
Butiril-CoA Desidrogenase , Megasphaera elsdenii , Oxirredutases , Butiril-CoA Desidrogenase/química , Butiril-CoA Desidrogenase/metabolismo , Elétrons , Ferredoxinas/metabolismo , Cinética , Megasphaera elsdenii/enzimologia , NAD/metabolismo , Oxirredução , Oxirredutases/química , Oxirredutases/metabolismo , Espectroscopia de Ressonância de Spin Eletrônica , Estrutura Terciária de Proteína , Modelos Moleculares
2.
Int J Med Sci ; 18(16): 3631-3643, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-34790035

RESUMO

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.


Assuntos
Butiril-CoA Desidrogenase/genética , Butiril-CoA Desidrogenase/metabolismo , Neoplasias Colorretais/diagnóstico , Adenocarcinoma/diagnóstico , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Adenocarcinoma/mortalidade , Biomarcadores Tumorais/genética , Biomarcadores Tumorais/metabolismo , Carcinoma/diagnóstico , Carcinoma/genética , Carcinoma/metabolismo , Carcinoma/mortalidade , Linhagem Celular Tumoral , Quimiotaxia de Leucócito/genética , Quimiotaxia de Leucócito/imunologia , Neoplasias Colorretais/genética , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/mortalidade , Ácidos Graxos/metabolismo , Perfilação da Expressão Gênica , Regulação Enzimológica da Expressão Gênica , Regulação Neoplásica da Expressão Gênica , Humanos , Metabolismo dos Lipídeos/genética , Valor Preditivo dos Testes , Prognóstico , Proteômica , Análise de Sobrevida , Microambiente Tumoral/genética , Microambiente Tumoral/imunologia
3.
Br J Nutr ; 122(3): 241-251, 2019 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-31475655

RESUMO

For the same BMI, South Asians have a higher body fat percentage than Caucasians. There might be differences in the fatty acid (FA) handling in adipose tissue when both ethnicities are exposed to high-fat overfeeding. The objective of the present study was to investigate the molecular adaptation in relation to FA metabolism in response to overfeeding with a high-fat diet (OHFD) in South Asian and Caucasian men. Ten South Asian men (BMI 18-29 kg/m2) and ten Caucasian men (BMI 22-33 kg/m2), matched for body fat percentage, aged 20-40 years were included. A weight-maintenance diet (30 % fat, 55 % carbohydrate and 15 % protein) was given for 3 d followed by 3 d of overfeeding (150 % energy requirement) with a high-fat diet (60 % fat, 25 % carbohydrate and 15 % protein) while staying in a respiration chamber. Before and after overfeeding, abdominal subcutaneous fat biopsies were taken. Proteins were isolated, analysed and quantified for short-chain 3-hydroxyacyl-CoA dehydrogenase (HADH), carnitine palmitoyl-transferase 1α (CPT1a), adipose TAG lipase, perilipin A (PLINA), perilipin B, lipoprotein lipase and fatty acid binding protein 4 using Western blotting. OHFD decreased the HADH level (P < 0·05) in Caucasians more than in Asians (P < 0·05), but the baseline and after intervention HADH level was relatively higher in Caucasians. The level of CPT1a decreased in South Asians and increased in Caucasians (P < 0·05). PLINA did not change with diet but the level was higher in South Asians (P < 0·05). The observed differences in HADH and PLINA levels as well as in CPT1a response may be important for differences in the long-term regulation of energy (fat) metabolism in these populations.


Assuntos
Tecido Adiposo/metabolismo , Adiposidade , Dieta Hiperlipídica , Ingestão de Energia , Adaptação Fisiológica , Adulto , Povo Asiático , Biópsia , Composição Corporal , Índice de Massa Corporal , Peso Corporal , Butiril-CoA Desidrogenase/metabolismo , Carnitina O-Palmitoiltransferase/metabolismo , Carboidratos da Dieta , Gorduras na Dieta , Proteínas Alimentares , Metabolismo Energético , Exercício Físico , Ácidos Graxos/metabolismo , Humanos , Lipase/metabolismo , Masculino , Mitocôndrias/metabolismo , Nutrientes , Perilipina-1/metabolismo , População Branca , Adulto Jovem
4.
J Biol Chem ; 291(23): 11993-2002, 2016 Jun 03.
Artigo em Inglês | MEDLINE | ID: mdl-27048649

RESUMO

Electron-transferring flavoprotein (Etf) and butyryl-CoA dehydrogenase (Bcd) from Acidaminococcus fermentans catalyze the endergonic reduction of ferredoxin by NADH, which is also driven by the concomitant reduction of crotonyl-CoA by NADH, a process called electron bifurcation. Here we show that recombinant flavodoxin from A. fermentans produced in Escherichia coli can replace ferredoxin with almost equal efficiency. After complete reduction of the yellow quinone to the blue semiquinone, a second 1.4 times faster electron transfer affords the colorless hydroquinone. Mediated by a hydrogenase, protons reoxidize the fully reduced flavodoxin or ferredoxin to the semi-reduced species. In this hydrogen-generating system, both electron carriers act catalytically with apparent Km = 0.26 µm ferredoxin or 0.42 µm flavodoxin. Membrane preparations of A. fermentans contain a highly active ferredoxin/flavodoxin-NAD(+) reductase (Rnf) that catalyzes the irreversible reduction of flavodoxin by NADH to the blue semiquinone. Using flavodoxin hydroquinone or reduced ferredoxin obtained by electron bifurcation, Rnf can be measured in the forward direction, whereby one NADH is recycled, resulting in the simple equation: crotonyl-CoA + NADH + H(+) = butyryl-CoA + NAD(+) with Km = 1.4 µm ferredoxin or 2.0 µm flavodoxin. This reaction requires Na(+) (Km = 0.12 mm) or Li(+) (Km = 0.25 mm) for activity, indicating that Rnf acts as a Na(+) pump. The redox potential of the quinone/semiquinone couple of flavodoxin (Fld) is much higher than that of the semiquinone/hydroquinone couple. With free riboflavin, the opposite is the case. Based on this behavior, we refine our previous mechanism of electron bifurcation.


Assuntos
Proteínas de Bactérias/metabolismo , Flavoproteínas Transferidoras de Elétrons/metabolismo , NAD/metabolismo , Oxirredutases/metabolismo , Sódio/metabolismo , Acidaminococcus/enzimologia , Acidaminococcus/genética , Acidaminococcus/metabolismo , Acil Coenzima A/metabolismo , Benzoquinonas/metabolismo , Butiril-CoA Desidrogenase/metabolismo , Catálise , Transporte de Elétrons , Flavoproteínas Transferidoras de Elétrons/genética , Elétrons , Hidrogênio/metabolismo , Hidroquinonas/metabolismo , Cinética , Oxirredução , Proteínas Recombinantes/metabolismo , Riboflavina/metabolismo , Espectrofotometria
5.
J Cell Mol Med ; 20(7): 1381-91, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-26989860

RESUMO

Short-chain acyl-CoA dehydrogenase (SCAD), a key enzyme of fatty acid ß-oxidation, plays an important role in cardiac hypertrophy. However, its effect on the cardiomyocyte apoptosis remains unknown. We aimed to determine the role of SCAD in tert-butyl hydroperoxide (tBHP)-induced cardiomyocyte apoptosis. The mRNA and protein expression of SCAD were significantly down-regulated in the cardiomyocyte apoptosis model. Inhibition of SCAD with siRNA-1186 significantly decreased SCAD expression, enzyme activity and ATP content, but obviously increased the content of free fatty acids. Meanwhile, SCAD siRNA treatment triggered the same apoptosis as cardiomyocytes treated with tBHP, such as the increase in cell apoptotic rate, the activation of caspase3 and the decrease in the Bcl-2/Bax ratio, which showed that SCAD may play an important role in primary cardiomyocyte apoptosis. The changes of phosphonate AMP-activated protein kinase α (p-AMPKα) and Peroxisome proliferator-activated receptor α (PPARα) in cardiomyocyte apoptosis were consistent with that of SCAD. Furthermore, PPARα activator fenofibrate and AMPKα activator AICAR treatment significantly increased the expression of SCAD and inhibited cardiomyocyte apoptosis. In conclusion, for the first time our findings directly demonstrated that SCAD may be as a new target to prevent cardiomyocyte apoptosis through the AMPK/PPARα/SCAD signal pathways.


Assuntos
Apoptose , Butiril-CoA Desidrogenase/metabolismo , Miócitos Cardíacos/citologia , Miócitos Cardíacos/enzimologia , Proteínas Quinases Ativadas por AMP/metabolismo , Aminoimidazol Carboxamida/análogos & derivados , Aminoimidazol Carboxamida/farmacologia , Animais , Animais Recém-Nascidos , Apoptose/efeitos dos fármacos , Cardiotônicos/farmacologia , Morte Celular/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Fenofibrato/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , PPAR alfa/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos Sprague-Dawley , Reação em Cadeia da Polimerase em Tempo Real , Ribonucleotídeos/farmacologia , terc-Butil Hidroperóxido/farmacologia
6.
J Cell Mol Med ; 19(7): 1672-88, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25753319

RESUMO

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.


Assuntos
Butiril-CoA Desidrogenase/metabolismo , Cardiomegalia/enzimologia , Coração/crescimento & desenvolvimento , Animais , Animais Recém-Nascidos , Pressão Sanguínea/efeitos dos fármacos , Butiril-CoA Desidrogenase/genética , Cardiomegalia/diagnóstico por imagem , Cardiomegalia/patologia , Cardiomegalia/fisiopatologia , Modelos Animais de Doenças , Ácidos Graxos/metabolismo , Fenofibrato/farmacologia , Coração/efeitos dos fármacos , Coração/fisiopatologia , Ventrículos do Coração/patologia , Ventrículos do Coração/fisiopatologia , Fator de Crescimento Insulin-Like I/farmacologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/patologia , Tamanho do Órgão/efeitos dos fármacos , Oxirredução/efeitos dos fármacos , PPAR alfa/metabolismo , Fenilefrina/farmacologia , Interferência de RNA/efeitos dos fármacos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Ratos Endogâmicos SHR , Ratos Wistar , Especificidade por Substrato/efeitos dos fármacos , Sístole/efeitos dos fármacos , Fatores de Tempo , Ultrassonografia
7.
J Biol Chem ; 289(8): 5145-57, 2014 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-24379410

RESUMO

Electron bifurcation is a fundamental strategy of energy coupling originally discovered in the Q-cycle of many organisms. Recently a flavin-based electron bifurcation has been detected in anaerobes, first in clostridia and later in acetogens and methanogens. It enables anaerobic bacteria and archaea to reduce the low-potential [4Fe-4S] clusters of ferredoxin, which increases the efficiency of the substrate level and electron transport phosphorylations. Here we characterize the bifurcating electron transferring flavoprotein (EtfAf) and butyryl-CoA dehydrogenase (BcdAf) of Acidaminococcus fermentans, which couple the exergonic reduction of crotonyl-CoA to butyryl-CoA to the endergonic reduction of ferredoxin both with NADH. EtfAf contains one FAD (α-FAD) in subunit α and a second FAD (ß-FAD) in subunit ß. The distance between the two isoalloxazine rings is 18 Å. The EtfAf-NAD(+) complex structure revealed ß-FAD as acceptor of the hydride of NADH. The formed ß-FADH(-) is considered as the bifurcating electron donor. As a result of a domain movement, α-FAD is able to approach ß-FADH(-) by about 4 Å and to take up one electron yielding a stable anionic semiquinone, α-FAD, which donates this electron further to Dh-FAD of BcdAf after a second domain movement. The remaining non-stabilized neutral semiquinone, ß-FADH(•), immediately reduces ferredoxin. Repetition of this process affords a second reduced ferredoxin and Dh-FADH(-) that converts crotonyl-CoA to butyryl-CoA.


Assuntos
Acidaminococcus/enzimologia , Biocatálise , Butiril-CoA Desidrogenase/metabolismo , Flavoproteínas Transferidoras de Elétrons/metabolismo , Elétrons , Butiril-CoA Desidrogenase/química , Cristalografia por Raios X , Transporte de Elétrons , Flavoproteínas Transferidoras de Elétrons/química , Eletroforese em Gel de Poliacrilamida , Ferredoxinas/química , Ferredoxinas/metabolismo , Flavina-Adenina Dinucleotídeo/química , Flavina-Adenina Dinucleotídeo/metabolismo , Flavinas/química , Flavinas/metabolismo , Cinética , Modelos Biológicos , Simulação de Acoplamento Molecular , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Espectrofotometria Ultravioleta
8.
Appl Environ Microbiol ; 81(14): 4782-90, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25956767

RESUMO

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.


Assuntos
Butiratos/metabolismo , Monóxido de Carbono/metabolismo , Eubacterium/metabolismo , Acil Coenzima A/metabolismo , Trifosfato de Adenosina/metabolismo , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Butiril-CoA Desidrogenase/genética , Butiril-CoA Desidrogenase/metabolismo , Metabolismo Energético , Eubacterium/enzimologia , Eubacterium/genética , Flavinas/metabolismo , Genômica , Oxirredução
9.
Mol Genet Metab ; 111(3): 360-368, 2014 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-24485985

RESUMO

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.


Assuntos
Acil-CoA Desidrogenase/deficiência , Butiril-CoA Desidrogenase/genética , Erros Inatos do Metabolismo Lipídico/genética , Mitocôndrias/genética , Proteínas Mitocondriais/biossíntese , Acil-CoA Desidrogenase/genética , Acil-CoA Desidrogenase/metabolismo , Butiril-CoA Desidrogenase/metabolismo , Células Cultivadas , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Regulação da Expressão Gênica , Humanos , Erros Inatos do Metabolismo Lipídico/metabolismo , Erros Inatos do Metabolismo Lipídico/patologia , Masculino , Mitocôndrias/patologia , Estresse Oxidativo/genética , Proteômica , Espectrometria de Massas em Tandem
10.
J Ind Microbiol Biotechnol ; 41(9): 1425-33, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25037723

RESUMO

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.


Assuntos
Proteínas de Bactérias/química , Proteínas de Bactérias/metabolismo , Butileno Glicóis/metabolismo , Butiril-CoA Desidrogenase/química , Butiril-CoA Desidrogenase/metabolismo , Klebsiella pneumoniae/enzimologia , Acetoína/metabolismo , Sequência de Aminoácidos , Proteínas de Bactérias/genética , Butiril-CoA Desidrogenase/genética , Estabilidade Enzimática , Klebsiella pneumoniae/química , Klebsiella pneumoniae/genética , Klebsiella pneumoniae/metabolismo , Dados de Sequência Molecular , Alinhamento de Sequência
11.
J Bacteriol ; 195(16): 3704-13, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23772070

RESUMO

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.


Assuntos
Butiratos/metabolismo , Butiril-CoA Desidrogenase/metabolismo , Clostridioides difficile/metabolismo , Flavoproteínas Transferidoras de Elétrons/metabolismo , Escherichia coli/metabolismo , Oxigênio , Proteínas de Bactérias/genética , Proteínas de Bactérias/metabolismo , Butiril-CoA Desidrogenase/genética , Clonagem Molecular , Clostridioides difficile/enzimologia , Clostridioides difficile/genética , Flavoproteínas Transferidoras de Elétrons/genética , Escherichia coli/genética , Regulação Bacteriana da Expressão Gênica/fisiologia , Proteínas Recombinantes
12.
Plant Physiol ; 160(4): 2202-18, 2012 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-23027666

RESUMO

The related proteins D1 and D2 together build up the photosystem II reaction center. Synthesis of D1 (PsbA) is highly regulated in all photosynthetic organisms. The mechanisms and specific protein factors involved in controlled expression of the psbA gene in higher plants are highly elusive. Here, we report on the identification of a chloroplast-located protein, HCF244 (for high chlorophyll fluorescence244), which is essentially required for translational initiation of the psbA messenger RNA in Arabidopsis (Arabidopsis thaliana). The factor is highly conserved between land plants, algae, and cyanobacteria. HCF244 was identified by coexpression analysis of HCF173, which encodes a protein that is also necessary for psbA translational initiation and in addition for stabilization of this messenger RNA. Phenotypic characterization of the mutants hcf244 and hcf173 suggests that the corresponding proteins operate cooperatively during psbA translation. Immunolocalization studies detected the majority of the two proteins at the thylakoid membrane. Both HCF244 and HCF173 are members of the atypical short-chain dehydrogenase/reductase superfamily, a modified group, which has lost enzyme activity but acquires new functions in the metabolism of the cell.


Assuntos
Proteínas de Arabidopsis/metabolismo , Arabidopsis/enzimologia , Arabidopsis/genética , Butiril-CoA Desidrogenase/metabolismo , Fatores de Iniciação em Eucariotos/metabolismo , Iniciação Traducional da Cadeia Peptídica , Sequência de Aminoácidos , Proteínas de Arabidopsis/química , Proteínas de Arabidopsis/genética , Butiril-CoA Desidrogenase/química , Centrifugação com Gradiente de Concentração , Fatores de Iniciação em Eucariotos/química , Regulação da Expressão Gênica de Plantas , Genes de Plantas/genética , Dados de Sequência Molecular , Mutação/genética , Fotossíntese/genética , Complexo de Proteína do Fotossistema II/metabolismo , Filogenia , Ligação Proteica/genética , Estrutura Terciária de Proteína , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA de Plantas/metabolismo , Análise Espectral , Tilacoides/metabolismo
13.
J Hypertens ; 41(5): 775-793, 2023 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-36883465

RESUMO

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.


Assuntos
Butiril-CoA Desidrogenase , Hipertensão , Ratos , Animais , Camundongos , Humanos , Recém-Nascido , Butiril-CoA Desidrogenase/genética , Butiril-CoA Desidrogenase/metabolismo , Remodelação Vascular , Ratos Endogâmicos SHR , Ratos Wistar , Células Endoteliais da Veia Umbilical Humana/metabolismo , RNA Interferente Pequeno/metabolismo , Camundongos Knockout
14.
Eur J Pharmacol ; 954: 175849, 2023 Sep 05.
Artigo em Inglês | MEDLINE | ID: mdl-37331684

RESUMO

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.


Assuntos
Butiril-CoA Desidrogenase , Flavina-Adenina Dinucleotídeo , Animais , Camundongos , Butiril-CoA Desidrogenase/genética , Butiril-CoA Desidrogenase/metabolismo , Flavina-Adenina Dinucleotídeo/farmacologia , Riboflavina/farmacologia , Cardiomegalia/patologia , Ácidos Graxos não Esterificados , Fibrose
15.
Metabolism ; 146: 155661, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-37454871

RESUMO

Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease worldwide; however, the underlying mechanisms remain poorly understood. FAM3D is a member of the FAM3 family; however, its role in hepatic glycolipid metabolism remains unknown. Serum FAM3D levels are positively correlated with fasting blood glucose levels in patients with diabetes. Hepatocytes express and secrete FAM3D, and its expression is increased in steatotic human and mouse livers. Hepatic FAM3D overexpression ameliorated hyperglycemia and steatosis in obese mice, whereas FAM3D-deficient mice exhibited exaggerated hyperglycemia and steatosis after high-fat diet (HFD)-feeding. In cultured hepatocytes, FAM3D overexpression or recombinant FAM3D protein (rFAM3D) treatment reduced gluconeogenesis and lipid deposition, which were blocked by anti-FAM3D antibodies or inhibition of its receptor, formyl peptide receptor 1 (FPR1). FPR1 overexpression suppressed gluconeogenesis and reduced lipid deposition in wild hepatocytes but not in FAM3D-deficient hepatocytes. The addition of rFAM3D restored FPR1's inhibitory effects on gluconeogenesis and lipid deposition in FAM3D-deficient hepatocytes. Hepatic FPR1 overexpression ameliorated hyperglycemia and steatosis in obese mice. RNA sequencing and DNA pull-down revealed that the FAM3D-FPR1 axis upregulated the expression of heterogeneous nuclear ribonucleoprotein U (hnRNP U), which recruits the glucocorticoid receptor (GR) to the promoter region of the short-chain acyl-CoA dehydrogenase (SCAD) gene, promoting its transcription to enhance lipid oxidation. Moreover, FAM3D-FPR1 axis also activates calmodulin-Akt pathway to suppress gluconeogenesis in hepatocytes. In conclusion, hepatocyte-secreted FAM3D activated the FPR1-hnRNP U-GR-SCAD pathway to enhance lipid oxidation in hepatocytes. Under obesity conditions, increased hepatic FAM3D expression is a compensatory mechanism against dysregulated glucose and lipid metabolism.


Assuntos
Hiperglicemia , Hepatopatia Gordurosa não Alcoólica , Animais , Humanos , Camundongos , Butiril-CoA Desidrogenase/metabolismo , Dieta Hiperlipídica , Hepatócitos/metabolismo , Ribonucleoproteínas Nucleares Heterogêneas Grupo U/metabolismo , Hiperglicemia/metabolismo , Metabolismo dos Lipídeos , Lipídeos , Fígado/metabolismo , Camundongos Endogâmicos C57BL , Camundongos Obesos , Hepatopatia Gordurosa não Alcoólica/metabolismo , Receptores de Formil Peptídeo/metabolismo , Receptores de Glucocorticoides/metabolismo
16.
Br J Pharmacol ; 180(23): 3024-3044, 2023 12.
Artigo em Inglês | MEDLINE | ID: mdl-37377111

RESUMO

BACKGROUND AND PURPOSE: Our recent studies have shown that flavin adenine dinucleotide (FAD) exerts cardiovascular protective effects by supplementing short-chain acyl-CoA dehydrogenase (SCAD). The current study aimed to elucidate whether riboflavin (the precursor of FAD) could improve heart failure via activating SCAD and the DJ-1-Keap1-Nrf2 signalling pathway. EXPERIMENTAL APPROACH: Riboflavin treatment was given to the mouse transverse aortic constriction (TAC)-induced heart failure model. Cardiac structure and function, energy metabolism and apoptosis index were assessed, and relevant signalling proteins were analysed. The mechanisms underlying the cardioprotection by riboflavin were analysed in the cell apoptosis model induced by tert-butyl hydroperoxide (tBHP). KEY RESULTS: In vivo, riboflavin ameliorated myocardial fibrosis and energy metabolism, improved cardiac dysfunction and inhibited oxidative stress and cardiomyocyte apoptosis in TAC-induced heart failure. In vitro, riboflavin ameliorated cell apoptosis in H9C2 cardiomyocytes by decreasing reactive oxygen species (ROS). At the molecular level, riboflavin significantly restored FAD content, SCAD expression and enzymatic activity, activated DJ-1 and inhibited the Keap1-Nrf2/HO1 signalling pathway in vivo and in vitro. SCAD knockdown exaggerated the tBHP-induced DJ-1 decrease and Keap1-Nrf2/HO1 signalling pathway activation in H9C2 cardiomyocytes. The knockdown of SCAD abolished the anti-apoptotic effects of riboflavin on H9C2 cardiomyocytes. DJ-1 knockdown hindered SCAD overexpression anti-apoptotic effects and regulation on Keap1-Nrf2/HO1 signalling pathway in H9C2 cardiomyocytes. CONCLUSIONS AND IMPLICATIONS: Riboflavin exerts cardioprotective effects on heart failure by improving oxidative stress and cardiomyocyte apoptosis via FAD to stimulate SCAD and then activates the DJ-1-Keap1-Nrf2 signalling pathway.


Assuntos
Butiril-CoA Desidrogenase , Insuficiência Cardíaca , Animais , Camundongos , Butiril-CoA Desidrogenase/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Flavina-Adenina Dinucleotídeo/metabolismo , Flavina-Adenina Dinucleotídeo/farmacologia , Proteína 1 Associada a ECH Semelhante a Kelch/metabolismo , Insuficiência Cardíaca/tratamento farmacológico , Insuficiência Cardíaca/metabolismo , Estresse Oxidativo , Apoptose , Miócitos Cardíacos/metabolismo
17.
J Ind Microbiol Biotechnol ; 39(8): 1101-7, 2012 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-22776992

RESUMO

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.


Assuntos
1-Butanol/metabolismo , Reatores Biológicos , Butanóis/metabolismo , Escherichia coli/metabolismo , Fermentação , Engenharia Metabólica , 3-Hidroxiacil-CoA Desidrogenases/genética , 3-Hidroxiacil-CoA Desidrogenases/metabolismo , Acetil-CoA C-Acetiltransferase/genética , Acetil-CoA C-Acetiltransferase/metabolismo , Álcool Desidrogenase/genética , Álcool Desidrogenase/metabolismo , Biocombustíveis , Butiril-CoA Desidrogenase/genética , Butiril-CoA Desidrogenase/metabolismo , Clostridium acetobutylicum/enzimologia , Clostridium acetobutylicum/genética , Enoil-CoA Hidratase/genética , Enoil-CoA Hidratase/metabolismo , Escherichia coli/classificação , Escherichia coli/genética , Glucose/metabolismo , Glicólise , NAD/metabolismo , Oxirredução , Complexo Piruvato Desidrogenase/genética , Complexo Piruvato Desidrogenase/metabolismo
19.
Drug Metab Dispos ; 39(7): 1155-60, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21430231

RESUMO

Many biological systems including the oxidative catabolic pathway for branched-chain amino acids (BCAAs) are affected in vivo by valproate therapy. In this study, we investigated the potential effect of valproic acid (VPA) and some of its metabolites on the metabolism of BCAAs. In vitro studies were performed using isovaleryl-CoA dehydrogenase (IVD), isobutyryl-CoA dehydrogenase (IBD), and short branched-chain acyl-CoA dehydrogenase (SBCAD), enzymes involved in the degradation pathway of leucine, valine, and isoleucine. The enzymatic activities of the three purified human enzymes were measured using optimized high-performance liquid chromatography procedures, and the respective kinetic parameters were determined in the absence and presence of VPA and the corresponding CoA and dephosphoCoA conjugates. Valproyl-CoA and valproyl-dephosphoCoA inhibited IVD activity significantly by a purely competitive mechanism with K(i) values of 74 ± 4 and 170 ± 12 µM, respectively. IBD activity was not affected by any of the tested VPA esters. However, valproyl-CoA did inhibit SBCAD activity by a purely competitive mechanism with a K(i) of 249 ± 29 µM. In addition, valproyl-dephosphoCoA inhibited SBCAD activity via a distinct mechanism (K(i) = 511 ± 96 µM) that appeared to be of the mixed type. Furthermore, we show that both SBCAD and IVD are active, using valproyl-CoA as a substrate. The catalytic efficiency of SBCAD turned out to be much higher than that of IVD, demonstrating that SBCAD is the most probable candidate for the first dehydrogenation step of VPA ß-oxidation. Our data explain some of the effects of valproate on the branched-chain amino acid metabolism and shed new light on the biotransformation pathway of valproate.


Assuntos
Aminoácidos de Cadeia Ramificada/metabolismo , Butiril-CoA Desidrogenase/metabolismo , Isovaleril-CoA Desidrogenase/metabolismo , Ácido Valproico/metabolismo , Cromatografia Líquida de Alta Pressão , Oxirredução , Espectrometria de Massas por Ionização por Electrospray , Espectrometria de Massas em Tandem
20.
J Inherit Metab Dis ; 34(2): 465-75, 2011 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-21170680

RESUMO

BACKGROUND: Variations in the gene ACADS, encoding the mitochondrial protein short-chain acyl CoA-dehydrogenase (SCAD), have been observed in individuals with clinical symptoms. The phenotype of SCAD deficiency (SCADD) is very heterogeneous, ranging from asymptomatic to severe, without a clear genotype-phenotype correlation, which suggests a multifactorial disorder. The pathophysiological relevance of the genetic variations in the SCAD gene is therefore disputed, and has not yet been elucidated, which is an important step in the investigation of SCADD etiology. AIM: To determine whether the disease-associated misfolding variant of SCAD protein, p.Arg107Cys, disturbs mitochondrial function. METHODS: We have developed a cell model system, stably expressing either the SCAD wild-type protein or the misfolding SCAD variant protein, p.Arg107Cys (c.319 C > T). The model system was used for investigation of SCAD with respect to expression, degree of misfolding, and enzymatic SCAD activity. Furthermore, cell proliferation and expression of selected stress response genes were investigated as well as proteomic analysis of mitochondria-enriched extracts in order to study the consequences of p.Arg107Cys protein expression using a global approach. CONCLUSIONS: We found that expression of the p.Arg107Cys variant SCAD protein gives rise to inactive misfolded protein species, eliciting a mild toxic response manifested though a decreased proliferation rate and oxidative stress, as shown by an increased demand for the mitochondrial antioxidant SOD2. In addition, we found markers of apoptotic activity in the p.Arg107Cys expressing cells, which points to a possible pathophysiological role of this variant protein.


Assuntos
Butiril-CoA Desidrogenase/metabolismo , Doenças Mitocondriais/metabolismo , Proteínas Mitocondriais/química , Animais , Antioxidantes/química , Butiril-CoA Desidrogenase/química , Butiril-CoA Desidrogenase/toxicidade , Proliferação de Células , Variação Genética , Genótipo , Humanos , Camundongos , Estresse Oxidativo , Fenótipo , Desnaturação Proteica , Dobramento de Proteína , Proteômica/métodos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA