Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 113
Filtrar
1.
Mol Med ; 30(1): 69, 2024 May 23.
Artigo em Inglês | MEDLINE | ID: mdl-38783226

RESUMO

BACKGROUND: The Enoyl-CoA hydratase/isomerase family plays a crucial role in the metabolism of tumors, being crucial for maintaining the energy balance and biosynthetic needs of cancer cells. However, the enzymes within this family that are pivotal in gastric cancer (GC) remain unclear. METHODS: We employed bioinformatics techniques to identify key Enoyl-CoA hydratase/isomerase in GC. The expression of ECHDC2 and its clinical significance were validated through tissue microarray analysis. The role of ECHDC2 in GC was further assessed using colony formation assays, CCK8 assay, EDU assay, Glucose and lactic acid assay, and subcutaneous tumor experiments in nude mice. The mechanism of action of ECHDC2 was validated through Western blotting, Co-immunoprecipitation, and immunofluorescence experiments. RESULTS: Our analysis of multiple datasets indicates that low expression of ECHDC2 in GC is significantly associated with poor prognosis. Overexpression of ECHDC2 notably inhibits aerobic glycolysis and proliferation of GC cells both in vivo and in vitro. Further experiments revealed that overexpression of ECHDC2 suppresses the P38 MAPK pathway by inhibiting the protein level of MCCC2, thereby restraining glycolysis and proliferation in GC cells. Ultimately, it was discovered that ECHDC2 promotes the ubiquitination and subsequent degradation of MCCC2 protein by binding with NEDD4. CONCLUSIONS: These findings underscore the pivotal role of the ECHDC2 in regulating aerobic glycolysis and proliferation in GC cells, suggesting ECHDC2 as a potential therapeutic target in GC.


Assuntos
Proliferação de Células , Ubiquitina-Proteína Ligases Nedd4 , Neoplasias Gástricas , Animais , Feminino , Humanos , Masculino , Camundongos , Linhagem Celular Tumoral , Enoil-CoA Hidratase/metabolismo , Enoil-CoA Hidratase/genética , Regulação Neoplásica da Expressão Gênica , Glicólise , Camundongos Nus , Ubiquitina-Proteína Ligases Nedd4/metabolismo , Ubiquitina-Proteína Ligases Nedd4/genética , Ligação Proteica , Proteólise , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , Neoplasias Gástricas/genética , Ubiquitinação , Efeito Warburg em Oncologia
2.
Int J Biochem Cell Biol ; 172: 106585, 2024 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-38734232

RESUMO

Tamoxifen is an estrogen receptor modulator that has been reported to alleviate hepatic lipid accumulation in mice, but the mechanism is still unclear. Peroxisome fatty acid ß-oxidation is the main metabolic pathway for the overload of long-chain fatty acids. As long-chain fatty acids are a cause of hepatic lipid accumulation, the activation of peroxisome fatty acid ß-oxidation might be a novel therapeutic strategy for metabolic associated fatty liver disease. In this study, we investigated the mechanism of tamoxifen against hepatic lipid accumulation based on the activation of peroxisome fatty acid ß-oxidation. Tamoxifen reduced liver long-chain fatty acids and relieved hepatic lipid accumulation in high fat diet mice without sex difference. In vitro, tamoxifen protected primary hepatocytes against palmitic acid-induced lipotoxicity. Mechanistically, the RNA-sequence of hepatocytes isolated from the liver revealed that peroxisome fatty acid ß-oxidation was activated by tamoxifen. Protein and mRNA expression of enoyl CoA hydratase and 3-hydroxyacyl CoA hydratase were significantly increased in vivo and in vitro. Small interfering RNA enoyl CoA hydratase and 3-hydroxyacyl CoA hydratase in primary hepatocytes abolished the therapeutic effects of tamoxifen in lipid accumulation. In conclusion, our results indicated that tamoxifen could relieve hepatic lipid accumulation in high fat diet mice based on the activation of enoyl CoA hydratase and 3-hydroxyacyl CoA hydratase-mediated peroxisome fatty acids ß-oxidation.


Assuntos
Enoil-CoA Hidratase , Hepatócitos , Metabolismo dos Lipídeos , Fígado , Camundongos Endogâmicos C57BL , Oxirredução , Peroxissomos , Tamoxifeno , Animais , Tamoxifeno/farmacologia , Camundongos , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/metabolismo , Fígado/efeitos dos fármacos , Hepatócitos/metabolismo , Hepatócitos/efeitos dos fármacos , Oxirredução/efeitos dos fármacos , Masculino , Peroxissomos/metabolismo , Peroxissomos/efeitos dos fármacos , Enoil-CoA Hidratase/metabolismo , Enoil-CoA Hidratase/genética , Regulação para Cima/efeitos dos fármacos , Dieta Hiperlipídica/efeitos adversos , Feminino , Ácidos Graxos/metabolismo
3.
Int J Mol Sci ; 23(20)2022 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-36293464

RESUMO

The lack of effective treatments for mitochondrial disease has seen the development of new approaches, including those that stimulate mitochondrial biogenesis to boost ATP production. Here, we examined the effects of deoxyribonucleosides (dNs) on mitochondrial biogenesis and function in Short chain enoyl-CoA hydratase 1 (ECHS1) 'knockout' (KO) cells, which exhibit combined defects in both oxidative phosphorylation (OXPHOS) and mitochondrial fatty acid ß-oxidation (FAO). DNs treatment increased mitochondrial DNA (mtDNA) copy number and the expression of mtDNA-encoded transcripts in both CONTROL (CON) and ECHS1 KO cells. DNs treatment also altered global nuclear gene expression, with key gene sets including 'respiratory electron transport' and 'formation of ATP by chemiosmotic coupling' increased in both CON and ECHS1 KO cells. Genes involved in OXPHOS complex I biogenesis were also upregulated in both CON and ECHS1 KO cells following dNs treatment, with a corresponding increase in the steady-state levels of holocomplex I in ECHS1 KO cells. Steady-state levels of OXPHOS complex V, and the CIII2/CIV and CI/CIII2/CIV supercomplexes, were also increased by dNs treatment in ECHS1 KO cells. Importantly, treatment with dNs increased both basal and maximal mitochondrial oxygen consumption in ECHS1 KO cells when metabolizing either glucose or the fatty acid palmitoyl-L-carnitine. These findings highlight the ability of dNs to improve overall mitochondrial respiratory function, via the stimulation mitochondrial biogenesis, in the face of combined defects in OXPHOS and FAO due to ECHS1 deficiency.


Assuntos
Enoil-CoA Hidratase , Biogênese de Organelas , Enoil-CoA Hidratase/genética , Enoil-CoA Hidratase/metabolismo , DNA Mitocondrial/genética , Ácidos Graxos/metabolismo , Glucose , Carnitina , Desoxirribonucleosídeos , Trifosfato de Adenosina
4.
Cell Death Dis ; 12(10): 911, 2021 10 06.
Artigo em Inglês | MEDLINE | ID: mdl-34615856

RESUMO

Sphingolipid metabolic dysregulation has increasingly been considered to be a drug-resistance mechanism for a variety of tumors. In this study, through an LC-MS assay, LIM and SH3 protein 1 (LASP1) was identified as a sphingolipid-metabolism-involved protein, and short-chain enoyl-CoA hydratase (ECHS1) was identified as a new LASP1-interacting protein through a protein assay in colorectal cancer (CRC). Gain- and loss-of-function analyses demonstrated the stimulatory role played by ECHS1 in CRC cell proliferation, migration, and invasion in vitro and in vivo. Mechanistic studies of the underlying tumor-supportive oncometabolism indicate that ECHS1 enables altering ceramide (Cer) metabolism that increases glycosphingolipid synthesis (HexCer) by promoting UDP-glucose ceramide glycosyltransferase (UGCG). Further analysis showed that ECHS1 promotes CRC progression and drug resistance by releasing reactive oxygen species (ROS) and interfering mitochondrial membrane potential via the PI3K/Akt/mTOR-dependent signaling pathway. Meanwhile, the phenomenon of promoting the survival and drug resistance of CRC cells caused by ECHS1 could be reversed by Eliglustat, a specific inhibitor of UCCG, in vitro and in vivo. IHC assay showed that ECHS1 was overexpressed in CRC tissues, which was related to the differentiation and poor prognosis of CRC patients. This study provides new insight into the mechanism by which phospholipids promote drug resistance in CRC and identifies potential targets for future therapies.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Ceramidas/metabolismo , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Proteínas do Citoesqueleto/metabolismo , Progressão da Doença , Enoil-CoA Hidratase/metabolismo , Proteínas com Domínio LIM/metabolismo , Esfingolipídeos/metabolismo , Animais , Apoptose , Autofagia , Linhagem Celular Tumoral , Neoplasias Colorretais/genética , Resistencia a Medicamentos Antineoplásicos , Feminino , Regulação Neoplásica da Expressão Gênica , Glicosilação , Humanos , Camundongos Endogâmicos BALB C , Camundongos Nus , Proteínas de Transporte de Monossacarídeos , Invasividade Neoplásica , Fenótipo , Fosfatidilinositol 3-Quinases/metabolismo , Prognóstico , Ligação Proteica , Proteínas Proto-Oncogênicas c-akt/metabolismo , Esfingomielinas/metabolismo , Regulação para Cima/genética , Domínios de Homologia de src
5.
Clin Sci (Lond) ; 135(19): 2243-2263, 2021 10 14.
Artigo em Inglês | MEDLINE | ID: mdl-34569605

RESUMO

The protein tyrosine kinase inhibitor imatinib is used in the treatment of various malignancies but may also promote beneficial effects in the treatment of diabetes. The aim of the present investigation was to characterize the mechanisms by which imatinib protects insulin producing cells. Treatment of non-obese diabetic (NOD) mice with imatinib resulted in increased beta-cell AMP-activated kinase (AMPK) phosphorylation. Imatinib activated AMPK also in vitro, resulting in decreased ribosomal protein S6 phosphorylation and protection against islet amyloid polypeptide (IAPP)-aggregation, thioredoxin interacting protein (TXNIP) up-regulation and beta-cell death. 5-Aminoimidazole-4-carboxamide ribonucleotide (AICAR) mimicked and compound C counteracted the effect of imatinib on beta-cell survival. Imatinib-induced AMPK activation was preceded by reduced glucose/pyruvate-dependent respiration, increased glycolysis rates, and a lowered ATP/AMP ratio. Imatinib augmented the fractional oxidation of fatty acids/malate, possibly via a direct interaction with the beta-oxidation enzyme enoyl coenzyme A hydratase, short chain, 1, mitochondrial (ECHS1). In non-beta cells, imatinib reduced respiratory chain complex I and II-mediated respiration and acyl-CoA carboxylase (ACC) phosphorylation, suggesting that mitochondrial effects of imatinib are not beta-cell specific. In conclusion, tyrosine kinase inhibitors modestly inhibit mitochondrial respiration, leading to AMPK activation and TXNIP down-regulation, which in turn protects against beta-cell death.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Diabetes Mellitus/tratamento farmacológico , Metabolismo Energético/efeitos dos fármacos , Hipoglicemiantes/farmacologia , Mesilato de Imatinib/farmacologia , Células Secretoras de Insulina/efeitos dos fármacos , Mitocôndrias/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , Animais , Proteínas de Transporte/metabolismo , Morte Celular/efeitos dos fármacos , Linhagem Celular , Respiração Celular/efeitos dos fármacos , Diabetes Mellitus/enzimologia , Diabetes Mellitus/patologia , Modelos Animais de Doenças , Enoil-CoA Hidratase/metabolismo , Ativação Enzimática , Humanos , Células Secretoras de Insulina/enzimologia , Células Secretoras de Insulina/patologia , Polipeptídeo Amiloide das Ilhotas Pancreáticas/metabolismo , Masculino , Camundongos Endogâmicos NOD , Mitocôndrias/enzimologia , Mitocôndrias/patologia , Fosforilação , Ratos Sprague-Dawley , Proteína S6 Ribossômica/metabolismo
6.
Brain Dev ; 43(2): 308-313, 2021 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-33139125

RESUMO

BACKGROUND: ECHS1 is a key enzyme of the valine catabolic pathway and oxidation of fatty acids. In ECHS1 deficiency (ECHS1D), accumulation of toxic intermediates from the valine induces neurodegeneration, which presents Leigh syndrome (LS). Therefore, valine restriction is suggested as an effective therapy. Further, cysteamine may detoxify the toxic metabolites themselves and N-acetylcysteine (NAC) is a potent antioxidant preventing neurological affect. Herein, we report the therapeutic effects of dietary therapy, cysteamine, and NAC in two siblings with ECHS1D, including their clinical, neuroradiological, and chemical aspects. CASE REPORT: The elder sister was the proband and was diagnosed as LS at 13 months of age. Gene analysis identified compound heterozygous ECHS1 mutations. Her psychomotor development was regressed, and she became bedridden. At 4 years old she started a low protein diet (LPD), but with no obvious neurological change. The younger brother was confirmed early with ECHS1D and received cysteamine and NAC treatment from 5 months of age, which could not prevent him developing LS at 7 months of age. Thus, we started a LPD at 14 months of age, with which he regained his ability to roll over, then we proceeded to a valine-restricted diet. The brain magnetic resonance image hyperintensity was diminished, and the lactate peak on magnetic resonance spectroscopy decreased. His neurological outcome is better than his elder sister. In both cases, excretion of valine metabolites decreased after dietary therapy without obvious adverse effects. CONCLUSION: Early initiation of dietary therapy may reduce neurological sequelae in patients with ECHS1D.


Assuntos
Enoil-CoA Hidratase/deficiência , Valina/metabolismo , Acetilcisteína/farmacologia , Cisteamina/farmacologia , Dietoterapia/métodos , Enoil-CoA Hidratase/genética , Enoil-CoA Hidratase/metabolismo , Enoil-CoA Hidratase/fisiologia , Família , Feminino , Testes Genéticos/métodos , Humanos , Lactente , Japão , Doença de Leigh/genética , Doença de Leigh/prevenção & controle , Imageamento por Ressonância Magnética/métodos , Masculino , Mutação/genética , Linhagem , Irmãos , Resultado do Tratamento , Valina/deficiência , Valina/genética
7.
Cell Rep ; 33(8): 108421, 2020 11 24.
Artigo em Inglês | MEDLINE | ID: mdl-33238129

RESUMO

Emerging evidence indicates that non-mutational drug tolerance mechanisms underlie the survival of residual cancer "persister" cells. Here, we find that BRAF(V600E) mutant melanoma persister cells tolerant to BRAF/MEK inhibitors switch their metabolism from glycolysis to oxidative respiration supported by peroxisomal fatty acid ß-oxidation (FAO) that is transcriptionally regulated by peroxisome proliferator-activated receptor alpha (PPARα). Knockdown of the key peroxisomal FAO enzyme, acyl-CoA oxidase 1 (ACOX1), as well as treatment with the peroxisomal FAO inhibitor thioridazine, specifically suppresses the oxidative respiration of persister cells and significantly decreases their emergence. Consistently, a combination treatment of BRAF/MEK inhibitors with thioridazine in human-melanoma-bearing mice results in a durable anti-tumor response. In BRAF(V600E) melanoma samples from patients treated with BRAF/MEK inhibitors, higher baseline expression of FAO-related genes and PPARα correlates with patients' outcomes. These results pave the way for a metabolic strategy to overcome drug resistance.


Assuntos
3-Hidroxiacil-CoA Desidrogenases/metabolismo , Acetil-CoA C-Aciltransferase/metabolismo , Acil-CoA Oxidase/metabolismo , Isomerases de Ligação Dupla Carbono-Carbono/metabolismo , Enoil-CoA Hidratase/metabolismo , Melanoma/genética , Inibidores de Proteínas Quinases/uso terapêutico , Proteínas Proto-Oncogênicas B-raf/antagonistas & inibidores , Racemases e Epimerases/metabolismo , Animais , Humanos , Melanoma/patologia , Camundongos , Inibidores de Proteínas Quinases/farmacologia
8.
Mol Genet Metab ; 131(1-2): 90-97, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32928639

RESUMO

BACKGROUND: The plasma acylcarnitine profile is frequently used as a biochemical assessment for follow-up in diagnosed patients with fatty acid oxidation disorders (FAODs). Disease specific acylcarnitine species are elevated during metabolic decompensation but there is clinical and biochemical heterogeneity among patients and limited data on the utility of an acylcarnitine profile for routine clinical monitoring. METHODS: We evaluated plasma acylcarnitine profiles from 30 diagnosed patients with long-chain FAODs (carnitine palmitoyltransferase-2 (CPT2), very long-chain acyl-CoA dehydrogenase (VLCAD), and long-chain 3-hydroxy acyl-CoA dehydrogenase or mitochondrial trifunctional protein (LCHAD/TFP) deficiencies) collected after an overnight fast, after feeding a controlled low-fat diet, and before and after moderate exercise. Our purpose was to describe the variability in this biomarker and how various physiologic states effect the acylcarnitine concentrations in circulation. RESULTS: Disease specific acylcarnitine species were higher after an overnight fast and decreased by approximately 60% two hours after a controlled breakfast meal. Moderate-intensity exercise increased the acylcarnitine species but it varied by diagnosis. When analyzed for a genotype/phenotype correlation, the presence of the common LCHADD mutation (c.1528G > C) was associated with higher levels of 3-hydroxyacylcarnitines than in patients with other mutations. CONCLUSIONS: We found that feeding consistently suppressed and that moderate intensity exercise increased disease specific acylcarnitine species, but the response to exercise was highly variable across subjects and diagnoses. The clinical utility of routine plasma acylcarnitine analysis for outpatient treatment monitoring remains questionable; however, if acylcarnitine profiles are measured in the clinical setting, standardized procedures are required for sample collection to be of value.


Assuntos
Cardiomiopatias/sangue , Carnitina O-Palmitoiltransferase/deficiência , Carnitina/análogos & derivados , Síndrome Congênita de Insuficiência da Medula Óssea/sangue , Erros Inatos do Metabolismo Lipídico/sangue , Erros Inatos do Metabolismo/sangue , Doenças Mitocondriais/sangue , Miopatias Mitocondriais/sangue , Proteína Mitocondrial Trifuncional/deficiência , Doenças Musculares/sangue , Doenças do Sistema Nervoso/sangue , Rabdomiólise/sangue , 3-Hidroxiacil-CoA Desidrogenases/genética , 3-Hidroxiacil-CoA Desidrogenases/metabolismo , Acetil-CoA C-Aciltransferase/genética , Acetil-CoA C-Aciltransferase/metabolismo , Acil-CoA Desidrogenase de Cadeia Longa/sangue , Isomerases de Ligação Dupla Carbono-Carbono/genética , Isomerases de Ligação Dupla Carbono-Carbono/metabolismo , Cardiomiopatias/dietoterapia , Cardiomiopatias/patologia , Cardiomiopatias/terapia , Carnitina/sangue , Carnitina/genética , Carnitina/metabolismo , Carnitina O-Palmitoiltransferase/sangue , Síndrome Congênita de Insuficiência da Medula Óssea/dietoterapia , Síndrome Congênita de Insuficiência da Medula Óssea/patologia , Síndrome Congênita de Insuficiência da Medula Óssea/terapia , Enoil-CoA Hidratase/genética , Enoil-CoA Hidratase/metabolismo , Terapia por Exercício , Jejum , Feminino , Humanos , Erros Inatos do Metabolismo Lipídico/dietoterapia , Erros Inatos do Metabolismo Lipídico/patologia , Erros Inatos do Metabolismo Lipídico/terapia , 3-Hidroxiacil-CoA Desidrogenase de Cadeia Longa/sangue , Masculino , Erros Inatos do Metabolismo/dietoterapia , Erros Inatos do Metabolismo/patologia , Erros Inatos do Metabolismo/terapia , Doenças Mitocondriais/dietoterapia , Doenças Mitocondriais/patologia , Doenças Mitocondriais/terapia , Miopatias Mitocondriais/dietoterapia , Miopatias Mitocondriais/patologia , Miopatias Mitocondriais/terapia , Proteína Mitocondrial Trifuncional/sangue , Doenças Musculares/dietoterapia , Doenças Musculares/patologia , Doenças Musculares/terapia , Doenças do Sistema Nervoso/dietoterapia , Doenças do Sistema Nervoso/patologia , Doenças do Sistema Nervoso/terapia , Racemases e Epimerases/genética , Racemases e Epimerases/metabolismo , Rabdomiólise/dietoterapia , Rabdomiólise/patologia , Rabdomiólise/terapia
9.
Biol Pharm Bull ; 43(9): 1382-1392, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32879213

RESUMO

The effects of different dietary fats on hepatic fatty acid oxidation were compared in male ICR mice and Sprague-Dawley rats. Animals were fed diets containing 100 g/kg of either palm oil (saturated fat), safflower oil (rich in linoleic acid), an oil of evening primrose origin (γ-linolenic acid, GLA oil), perilla oil (α-linolenic acid) or fish oil (eicosapentaenoic and doxosahexaenoic acids) for 21 d. GLA, perilla and fish oils, compared with palm and safflower oils, increased the activity of fatty acid oxidation enzymes in both mice and rats, with some exceptions. In mice, GLA and fish oils greatly increased the peroxisomal palmitoyl-CoA oxidation rate, and the activity of acyl-CoA oxidase and enoyl-CoA hydratase to the same degree. The effects were much smaller with perilla oil. In rats, enhancing effects were more notable with fish oil than with GLA and perilla oils, excluding the activity of enoyl-CoA hydratase, and were comparable between GLA and perilla oils. In mice, strong enhancing effects of GLA oil, which were greater than with perilla oil and comparable to those of fish oil, were confirmed on mRNA levels of peroxisomal but not mitochondrial fatty acid oxidation enzymes. In rats, the effects of GLA and perilla oils on mRNA levels of peroxisomal and mitochondrial enzymes were indistinguishable, and lower than those observed with fish oil. Therefore, considerable diversity in the response to dietary polyunsaturated fats, especially the oil rich in γ-linolenic acid and fish oil, of hepatic fatty acid oxidation pathway exists between mice and rats.


Assuntos
Gorduras na Dieta/administração & dosagem , Metabolismo dos Lipídeos/efeitos dos fármacos , Fígado/efeitos dos fármacos , Ácido gama-Linolênico/administração & dosagem , Acil-CoA Oxidase/metabolismo , Ração Animal , Animais , Enoil-CoA Hidratase/metabolismo , Óleos de Peixe/administração & dosagem , Óleos de Peixe/química , Fígado/citologia , Fígado/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Mitocôndrias/efeitos dos fármacos , Mitocôndrias/enzimologia , Oxirredução/efeitos dos fármacos , Peroxissomos/efeitos dos fármacos , Peroxissomos/enzimologia , Óleos de Plantas/administração & dosagem , Óleos de Plantas/química , Ratos , Ratos Sprague-Dawley , Especificidade da Espécie
10.
Int J Biol Macromol ; 164: 1600-1607, 2020 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-32768477

RESUMO

The acyl-CoA dehydrogenase (FadE) and (R)-specific enoyl-CoA hydratase (PhaJ) are functionally related to the degradation of fatty acids and the synthesis of polyhydroxyalkanoates (PHAs). To verify this, a recombinant Cupriavidus necator H16 harboring the plasmid -pMPJAS03- with fadE from Escherichia coli strain K12 and phaJ1 from Pseudomonas putida strain KT2440 under the arabinose promoter (araC-PBAD) was constructed. The impact of co-expressing fadE and phaJ genes on C. necator H16/pMPJAS03 maintaining the wild-type synthase on short-chain-length/medium-chain-length PHA formation from canola or avocado oil at different arabinose concentrations was investigated. The functional activity of fadEE.c led to obtaining higher biomass and PHA concentrations compared to the cultures without expressing the gene. While high transcriptional levels of phaJ1P.p, at 0.1% of arabinose, aid the wild-type synthase to polymerize larger-side chain monomers, such as 3-Hydroxyoctanoate (3HO) and 3-Hydroxydecanoate (3HD). The presence of even small amounts of 3HO and 3HD in the co-polymers significantly depresses the melting temperature of the polymers, compared to those composed of pure 3-hydroxybutyrate (3HB). Our data presents supporting evidence that the synthesis of larger-side chain monomers by the recombinant strain relies not only upon the affinity of the wild-type synthase but also on the functionality of the intermediate supplying enzymes.


Assuntos
Acil-CoA Desidrogenase/genética , Cupriavidus necator/genética , Enoil-CoA Hidratase/genética , Óleos de Plantas/metabolismo , Poli-Hidroxialcanoatos/biossíntese , Poli-Hidroxialcanoatos/genética , Acil-CoA Desidrogenase/metabolismo , Arabinose/genética , Arabinose/metabolismo , Caprilatos/metabolismo , Cupriavidus necator/metabolismo , Ácidos Decanoicos/metabolismo , Enoil-CoA Hidratase/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Ácidos Graxos/genética , Ácidos Graxos/metabolismo , Hidroxibutiratos/metabolismo , Plasmídeos/genética , Poli-Hidroxialcanoatos/metabolismo , Regiões Promotoras Genéticas/genética , Pseudomonas putida/genética , Pseudomonas putida/metabolismo , Transcrição Gênica/genética
11.
Cell Death Dis ; 11(4): 233, 2020 04 16.
Artigo em Inglês | MEDLINE | ID: mdl-32300102

RESUMO

Chemotherapy is the first-tier treatment regime for gastric cancer (GC) patients at advance stages. Mesenchymal stem cell (MSC) cam affect drug-resistance of GC cells in tumor microenvironment, but the detailed mechanism remains poorly understood. Present study aimed to investigate the regulation of MSC-induced long non-coding RNA (lncRNA) in GC. Dysregulated lncRNAs in GC were analyzed based on GEO data. Stemness and drug-resistance of GC cells were detected by sphere formation, colony formation, CCK-8, and flow cytometry analyses. MicroRNA (miRNA)-related pathways were analyzed by online KEGG analysis tool DAVID6.8. Molecular interactions were determined by luciferase reporter assay, pulldown, RNA immunoprecipitation (RIP), chromatin immunoprecipitation (ChIP), and co-immunoprecipitation (CoIP). Results revealed that MSC co-culture improved stemness and drug-resistance of GC cells. LncRNA histocompatibility leukocyte antigen complex P5 (HCP5) was induced in GC cells by MSC co-culture, contributing to stemness and drug-resistance. Mechanistically, HCP5 sequestered miR-3619-5p and upregulated PPARG coactivator 1 alpha (PPARGC1A), increasing transcription complex Peroxisome proliferator activated receptor (PPAR) coactivator-1α (PGC1α)/CEBPB and transcriptionally inducing carnitine palmitoyltransferase 1 (CPT1), which prompted the fatty acid oxidation (FAO) in GC cells. In conclusion, MSC-induced lncRNA HCP5 drove FAO through miR-3619-5p/AMPK/PGC1α/CEBPB axis to promote stemness and chemo-resistance of GC, indicating that targeting HCP5 was a novel approach to enhancing the efficacy of chemotherapy in GC.


Assuntos
3-Hidroxiacil-CoA Desidrogenases/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Acetil-CoA C-Aciltransferase/metabolismo , Isomerases de Ligação Dupla Carbono-Carbono/metabolismo , Enoil-CoA Hidratase/metabolismo , Ácidos Graxos/metabolismo , Células-Tronco Neoplásicas/metabolismo , RNA Longo não Codificante/metabolismo , Racemases e Epimerases/metabolismo , Neoplasias Gástricas/metabolismo , Animais , Resistencia a Medicamentos Antineoplásicos , Humanos , Camundongos , Camundongos Nus , Células-Tronco Neoplásicas/efeitos dos fármacos , Células-Tronco Neoplásicas/patologia , Oxirredução , RNA Longo não Codificante/genética , Neoplasias Gástricas/tratamento farmacológico , Neoplasias Gástricas/genética , Neoplasias Gástricas/patologia , Transfecção
12.
Aquat Toxicol ; 222: 105470, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-32199138

RESUMO

Cadmium (Cd) a highly toxic metal to human and wildlife health and it is hazardous to both terrestrial and aquatic life. In this study, we used RNA sequencing analysis to examine the effects of chronic cadmium exposure on liver lipid metabolism of Bufo gargarizans larvae. Tadpoles were exposed to cadmium concentrations at 0, 5, 10, 50, 100 and 200 µg L-1 from Gosner stage 26-42 of metamorphic climax. The results showed high dose cadmium (50, 100 and 200 µg L-1) caused obvious histological changes characterized by hepatocytes deformation, nuclear pyknosis, increasing melanomacrophage centers (MMCs) and aggregated lipid droplets. Moreover, transcriptome analysis showed that liver function was seriously affected by cadmium exposure. Furthermore, high dose cadmium significantly upregulated the mRNA expression of elongation of very-long-chain fatty acids 1 (ELOVL1), Mitochondrial trans-2-enoyl-CoA reductase (MECR), Trans-2, 3-enoyl-CoA reductase (TER) and Hydroxysteroid (17ß) dehydrogenase type 12 (HSD17B12) which are related with fatty acid synthesis. Meanwhile, mRNA levels of genes related with fat acid oxidation such as acetyl-CoA acyltransferase 2 (ACAA2) and enoyl-coenzyme A (CoA) hydratase short chain 1 (ECHS1) were significantly upregulated while the expression of Acyl-coA thioesterase 1 (ACOT1), 3-hydroxyacyl-CoA dehydrogenase (HADH), Palmitoyl-protein thioesterase 1(PPT1) and Acetyl-CoA acyltransferase 1(ACAA1) was significantly downregulated by high dose cadmium exposure. Furthermore, the mRNA level of ATP-binding cassette subfamily B member 11 (ABCB11) related with bile secretion was significantly decreased exposed to high dose cadmium. Our results suggested cadmium can cause liver dysfunction by inducing histopathological damages, genetic expression alterations and fatty acid metabolism disorder.


Assuntos
Cádmio/toxicidade , Ácidos Graxos/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Larva/efeitos dos fármacos , Fígado/efeitos dos fármacos , Poluentes Químicos da Água/toxicidade , Animais , Bufonidae , Enoil-CoA Hidratase/metabolismo , Larva/genética , Larva/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , Metabolismo dos Lipídeos/genética , Fígado/metabolismo , Oxirredução , RNA Mensageiro/metabolismo
13.
Biomed Pharmacother ; 123: 109750, 2020 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-31891870

RESUMO

Although the management of patients with renal cell carcinoma (RCC) has changed drastically in recent years, it is still faced with the evolving challenge. Elucidation of the mechanisms underlying RCC will help the development of therapies, as well as biomarkers for early diagnosis. In this study, ccRCC tissues from patients in different stages were subject to iTRAQ-based proteomics analysis. 130 common differentially expressed proteins (DEPs) in different stages were found and lipid metabolism pathway was obviously dysregulated in all stages. These 130 common DEPs were enriched in four highly connected subnetworks including metabolic pathway, the TCA cycle, oxidative phosphorylation and fatty acid metabolism. ECHS1, a key enzyme in fatty acid metabolism, was further investigated. ECHS1 expression was significantly downregulated in ccRCC tissues and ECHS1 level discriminated ccRCC tissues in general and in stage I from adjacent normal tissues well and with the area under the receiver operating characteristic curve (AUC) of more than 0.7. ECHS1 overexpression suppressed RCC cell proliferation and migration through inhibiting mTOR pathway activation. ECHS1 may be a novel target for ccRCC therapeutic interventions and diagnostic biomarker for ccRCC.


Assuntos
Carcinoma de Células Renais/metabolismo , Enoil-CoA Hidratase/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Biomarcadores Tumorais , Linhagem Celular Tumoral , Movimento Celular , Proliferação de Células , Ácidos Graxos , Regulação Neoplásica da Expressão Gênica , Redes Reguladoras de Genes , Humanos , Metabolismo dos Lipídeos , Mapas de Interação de Proteínas , Proteômica , Proteínas Quinases S6 Ribossômicas/metabolismo , Transdução de Sinais
14.
Cancer Res ; 80(2): 319-333, 2020 01 15.
Artigo em Inglês | MEDLINE | ID: mdl-31690668

RESUMO

The tumorigenic role and underlying mechanisms of lipid accumulation, commonly observed in many cancers, remain insufficiently understood. In this study, we identified an AMP-activated protein kinase (AMPK)-GATA-binding protein 3 (GATA3)-enoyl-CoA hydratase short-chain 1 (ECHS1) pathway that induces lipid accumulation and promotes cell proliferation in clear cell renal cell carcinoma (ccRCC). Decreased expression of ECHS1, which is responsible for inactivation of fatty acid (FA) oxidation and activation of de novo FA synthesis, positively associated with ccRCC progression and predicted poor patient survival. Mechanistically, ECHS1 downregulation induced FA and branched-chain amino acid (BCAA) accumulation, which inhibited AMPK-promoted expression of GATA3, a transcriptional activator of ECHS1. BCAA accumulation induced activation of mTORC1 and de novo FA synthesis, and promoted cell proliferation. Furthermore, GATA3 expression phenocopied ECHS1 in predicting ccRCC progression and patient survival. The AMPK-GATA3-ECHS1 pathway may offer new therapeutic approaches and prognostic assessment for ccRCC in the clinic. SIGNIFICANCE: These findings uncover molecular mechanisms underlying lipid accumulation in ccRCC, suggesting the AMPK-GATA3-ECHS1 pathway as a potential therapeutic target and prognostic biomarker.


Assuntos
Carcinoma de Células Renais/genética , Regulação Neoplásica da Expressão Gênica , Neoplasias Renais/genética , Lipogênese/genética , Transdução de Sinais/genética , Proteínas Quinases Ativadas por AMP/metabolismo , Adulto , Idoso , Idoso de 80 Anos ou mais , Aminoácidos de Cadeia Ramificada/análise , Aminoácidos de Cadeia Ramificada/biossíntese , Animais , Carcinogênese/genética , Carcinoma de Células Renais/metabolismo , Carcinoma de Células Renais/mortalidade , Carcinoma de Células Renais/patologia , Linhagem Celular Tumoral , Movimento Celular/genética , Proliferação de Células , Regulação para Baixo , Enoil-CoA Hidratase/metabolismo , Ácidos Graxos/análise , Ácidos Graxos/biossíntese , Feminino , Fator de Transcrição GATA3/metabolismo , Células HEK293 , Humanos , Rim/patologia , Rim/cirurgia , Neoplasias Renais/metabolismo , Neoplasias Renais/mortalidade , Neoplasias Renais/patologia , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos Knockout , Pessoa de Meia-Idade , Nefrectomia , Prognóstico , Intervalo Livre de Progressão , Adulto Jovem
15.
NPJ Biofilms Microbiomes ; 5(1): 20, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31396394

RESUMO

Achromobacter xylosoxidans has attracted increasing attention as an emerging pathogen in patients with cystic fibrosis. Intrinsic resistance to several classes of antimicrobials and the ability to form robust biofilms in vivo contribute to the clinical manifestations of persistent A. xylosoxidans infection. Still, much of A. xylosoxidans biofilm formation remains uncharacterized due to the scarcity of existing genetic tools. Here we demonstrate a promising genetic system for use in A. xylosoxidans; generating a transposon mutant library which was then used to identify genes involved in biofilm development in vitro. We further described the effects of one of the genes found in the mutagenesis screen, encoding a putative enoyl-CoA hydratase, on biofilm structure and tolerance to antimicrobials. Through additional analysis, we find that a fatty acid signaling compound is essential to A. xylosoxidans biofilm ultrastructure and maintenance. This work describes methods for the genetic manipulation of A. xylosoxidans and demonstrated their use to improve our understanding of A. xylosoxidans pathophysiology.


Assuntos
Achromobacter denitrificans/efeitos dos fármacos , Achromobacter denitrificans/enzimologia , Antibacterianos/metabolismo , Biofilmes/efeitos dos fármacos , Tolerância a Medicamentos , Enoil-CoA Hidratase/metabolismo , Mutagênese Insercional/métodos , Achromobacter denitrificans/genética , Achromobacter denitrificans/crescimento & desenvolvimento , Biofilmes/crescimento & desenvolvimento , Elementos de DNA Transponíveis , Enoil-CoA Hidratase/genética , Deleção de Genes
16.
Cell Biol Toxicol ; 35(5): 457-470, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30721374

RESUMO

Silent information regulator 1 (SIRT1) is a nicotinamide adenine dinucleotide (NAD+)-dependent deacetylase, and the function is linked to cellular metabolism including mitochondrial biogenesis. Hepatic L-serine concentration is decreased significantly in fatty liver disease. We reported that the supplementation of the amino acid ameliorated the alcoholic fatty liver by enhancing L-serine-dependent homocysteine metabolism. In this study, we hypothesized that the metabolic production of NAD+ from L-serine and thus activation of SIRT1 contribute to the action of L-serine. To this end, we evaluated the effects of L-serine on SIRT1 activity and mitochondria biogenesis in C2C12 myotubes. L-Serine increased intracellular NAD+ content and led to the activation of SIRT1 as determined by p53 luciferase assay and western blot analysis of peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α) acetylation. L-Serine treatment increased the expression of the genes associated with mitochondrial biogenesis and enhanced mitochondrial mass and function. In addition, L-serine reversed cellular insulin resistance determined by insulin-induced phosphorylation of Akt and GLUT4 expression and membrane translocation. L-Serine-induced mitochondrial gene expression, fatty acid oxidation, and insulin sensitization were mediated by enhanced SIRT1 activity, which was verified by selective SIRT1 inhibitor (Ex-527) and siRNA directed to SIRT1. L-Serine effect on cellular NAD+ level is dependent on the L-serine metabolism to pyruvate that is subsequently converted to lactate by lactate dehydrogenase. In summary, these data suggest that L-serine increases cellular NAD+ level and thus SIRT1 activity in C2C12 myotubes.


Assuntos
Ácidos Graxos/metabolismo , Resistência à Insulina/fisiologia , Fibras Musculares Esqueléticas/efeitos dos fármacos , Fibras Musculares Esqueléticas/metabolismo , Serina/farmacologia , Sirtuína 1/metabolismo , 3-Hidroxiacil-CoA Desidrogenases/metabolismo , Proteínas Quinases Ativadas por AMP/metabolismo , Acetil-CoA C-Aciltransferase/metabolismo , Acetilação , Animais , Isomerases de Ligação Dupla Carbono-Carbono/metabolismo , Linhagem Celular , Enoil-CoA Hidratase/metabolismo , Células Hep G2 , Humanos , Insulina/farmacologia , Metabolismo dos Lipídeos , Camundongos , Mitocôndrias/metabolismo , Fibras Musculares Esqueléticas/citologia , Músculo Esquelético/metabolismo , Oxirredução , Fosforilação , Racemases e Epimerases/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transativadores/metabolismo , Fatores de Transcrição/metabolismo
17.
Methods Enzymol ; 604: 207-236, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-29779653

RESUMO

ß-Branching is an expansion upon canonical polyketide synthase extension that allows for the installation of diverse chemical moieties in several natural products. Several of these moieties are unique among natural products, including the two vinyl methylesters found in the core structure of bryostatins. This family of molecules is derived from an obligate bacterial symbiont of a sessile marine bryozoan, Bugula neritina. Within this family, bryostatin 1 has been investigated as an anticancer, neuroprotective, and immunomodulatory compound. We have turned to the biosynthetic gene cluster within the bacterial symbiont to investigate the biosynthesis of bryostatins. Recent sequencing efforts resulted in the annotation of two missing genes: bryT and bryU. Using novel chemoenzymatic techniques, we have validated these as the missing enoyl-CoA hydratase and donor acyl carrier protein, essential components of the ß-branching cassette of the bryostatin pathway. Together, this cassette installs the vinyl methylester moieties essential to the activity of bryostatins.


Assuntos
Bioquímica/métodos , Briostatinas/metabolismo , Enzimas/metabolismo , Proteína de Transporte de Acila/genética , Proteína de Transporte de Acila/metabolismo , Animais , Briostatinas/biossíntese , Briozoários/genética , Briozoários/metabolismo , Enoil-CoA Hidratase/genética , Enoil-CoA Hidratase/metabolismo , Enzimas/genética , Espectroscopia de Ressonância Magnética , Redes e Vias Metabólicas , Metilação , Família Multigênica , Policetídeos/metabolismo
18.
Nat Commun ; 8(1): 464, 2017 09 06.
Artigo em Inglês | MEDLINE | ID: mdl-28878358

RESUMO

The oncogenic mechanisms of overnutrition, a confirmed independent cancer risk factor, remain poorly understood. Herein, we report that enoyl-CoA hydratase-1 (ECHS1), the enzyme involved in the oxidation of fatty acids (FAs) and branched-chain amino acids (BCAAs), senses nutrients and promotes mTOR activation and apoptotic resistance. Nutrients-promoted acetylation of lys101 of ECHS1 impedes ECHS1 activity by impairing enoyl-CoA binding, promoting ECHS1 degradation and blocking its mitochondrial translocation through inducing ubiquitination. As a result, nutrients induce the accumulation of BCAAs and FAs that activate mTOR signaling and stimulate apoptosis, respectively. The latter was overcome by selection of BCL-2 overexpressing cells under overnutrition conditions. The oncogenic effects of nutrients were reversed by SIRT3, which deacetylates lys101 acetylation. Severely decreased ECHS1, accumulation of BCAAs and FAs, activation of mTOR and overexpression of BCL-2 were observed in cancer tissues from metabolic organs. Our results identified ECHS1, a nutrients-sensing protein that transforms nutrient signals into oncogenic signals.Overnutrition has been linked to increased risk of cancer. Here, the authors show that exceeding nutrients suppress Enoyl-CoA hydratase-1 (ECHS1) activity by inducing its acetylation resulting in accumulation of fatty acids and branched-chain amino acids and oncogenic mTOR activation.


Assuntos
Apoptose , Enoil-CoA Hidratase/metabolismo , Transdução de Sinais , Serina-Treonina Quinases TOR/metabolismo , Acetilação , Aminoácidos de Cadeia Ramificada/metabolismo , Animais , Carcinogênese , Linhagem Celular Tumoral , Regulação Neoplásica da Expressão Gênica , Células HCT116 , Células HEK293 , Células Hep G2 , Humanos , Lisina/química , Masculino , Camundongos , Camundongos Knockout , Transplante de Neoplasias , Proteínas Recombinantes/química , Sirtuína 3/metabolismo , Ubiquitina/química
19.
Cancer Lett ; 409: 104-115, 2017 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-28923398

RESUMO

The class III deacetylase sirtuin 1 (SIRT1), a member of the sirtuin family proteins, plays a key role in many types of cancers including colorectal cancer (CRC). Here we report that SIRT1 suppressed CRC metastasis in vitro and in vivo as a negative regulator for miR-15b-5p transcription. Mechanistically, SIRT1 impaired regulatory effects of activator protein (AP-1) on miR-15b-5p trans-activation through deacetylation of AP-1. Importantly, acyl-CoA oxidase 1 (ACOX1), a key enzyme of the fatty acid oxidation (FAO) pathway, was found as a direct target for miR-15b-5p. SIRT1 expression was positively correlated with ACOX1 expression in CRC cells and in xenografts. Moreover, ACOX1 overexpression attenuated the augmentation of migration and invasion of CRC cells by miR-15b-5p overexpression. In conclusion, our study demonstrated a functional role of the SIRT1/miR-15b-5p/ACOX1 axis in CRC metastasis and suggested a potential target for metastatic CRC therapy.


Assuntos
Neoplasias Colorretais/genética , MicroRNAs/genética , Sirtuína 1/genética , 3-Hidroxiacil-CoA Desidrogenases/metabolismo , Acetil-CoA C-Aciltransferase/metabolismo , Animais , Células CACO-2 , Isomerases de Ligação Dupla Carbono-Carbono/metabolismo , Linhagem Celular Tumoral , Neoplasias Colorretais/metabolismo , Neoplasias Colorretais/patologia , Enoil-CoA Hidratase/metabolismo , Células HCT116 , Células HT29 , Xenoenxertos , Humanos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , MicroRNAs/metabolismo , Metástase Neoplásica , Racemases e Epimerases/metabolismo , Transdução de Sinais , Sirtuína 1/metabolismo , Transcrição Gênica , Transfecção
20.
Mol Nutr Food Res ; 61(10)2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28608394

RESUMO

SCOPE: The aim of this work was to study the urinary metabolomics changes of participants that consumed beer, nonalcoholic beer (na-beer), and gin. METHODS AND RESULTS: Thirty-three males at high cardiovascular risk between 55 and 75 years old participated in an open, randomized, crossover, controlled trial with three nutritional interventions consisting of beer, na-beer, and gin for 4 wk. Diet and physical activity was monitored throughout the study and compliance was assessed by measurement of urinary isoxanthohumol. Metabolomic analysis was performed in urine samples by LC coupled to an LTQ-Orbitrap mass spectrometer combined with univariate and multivariate statistical analysis. Ten metabolites were identified. Eight were exogenous metabolites related to beer, na-beer, or gin consumption, but two of them were related to endogenic changes: hydroxyadipic acid linked to fatty acid oxidation, and 4-guanidinobutanoic acid, which correlated with a decrease in urinary creatinine. Plasmatic acylcarnitines were quantified by targeted MS. A regular and moderate consumption of beer and na-beer decreased stearoylcarnitine concentrations. CONCLUSION: Humulinone and 2,3-dihydroxy-3-methylvaleric acid showed to be potential biomarkers of beer and na-beer consumption. Moreover, the results of this trial provide new evidence that the nonalcoholic fraction of beer may increase fatty oxidation.


Assuntos
Cerveja/efeitos adversos , Biomarcadores/urina , Doenças Cardiovasculares/urina , Metaboloma , Metabolômica , 3-Hidroxiacil-CoA Desidrogenases/metabolismo , Acetil-CoA C-Aciltransferase/metabolismo , Adipatos/sangue , Idoso , Consumo de Bebidas Alcoólicas , Bebidas , Isomerases de Ligação Dupla Carbono-Carbono/metabolismo , Carnitina/análogos & derivados , Carnitina/sangue , Creatinina/urina , Estudos Cross-Over , Dieta , Enoil-CoA Hidratase/metabolismo , Exercício Físico , Humanos , Masculino , Pessoa de Meia-Idade , Cooperação do Paciente , Ácidos Pentanoicos/urina , Racemases e Epimerases/metabolismo , Fatores de Risco , Xantonas/urina
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA