Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 64
Filtrar
Mais filtros










Intervalo de ano de publicação
1.
Biomed Pharmacother ; 137: 111378, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33601148

RESUMO

Hydroxy-3-methylglutaryl-CoA synthase 1 (HMGCS1) is a key enzyme in the mevalonate pathway of cholesterol synthesis. Dysregulation of HMGCS1 expression is a common occurrence in many solid tumors. It was also found to be overexpressed in newly diagnosed (ND) and relapsed/refractory (RR) acute myeloid leukemia (AML) patients. Previous study proved that HMGCS1 could induce drug-resistance in AML cells. However, the underlying mechanism how HMGCS1 contributed to chemoresistance remains elusive. Here, we confirmed that HMGCS1 inhibitor Hymeglusin enhanced cytarabine/Adriamycin (Ara-c/ADR) chemo-sensitivity in AML cells lines. Moreover, Ara-c-resistant HL-60 cells (HL-60/Ara-c) and ADR-resistant HL-60 cells (HL-60/ADR) were more sensitive to HMGCS1 inhibition than HL-60 cells. In addition, we demonstrated that the transcription factor GATA1 was the upstream regulator of HMGCS1 and could directly bind to the HMGCS1 promoter. After treatment of Tunicamycin (Tm), the number of mitochondria was increased and the damage of endoplasmic reticulum (ER) was reduced in bone marrow cells from AML-RR patients, compared to cells from AML-CR group. HMGCS1 protected mitochondria and ER under ER stress and up-regulated unfold protein response (UPR) downstream molecules in AML cells. In summary, we proved that HMGCS1 could upregulate UPR downstream components, protect mitochondria and ER from damage in AML cells under stress, therefore conferring drug resistance. Therefore, HMGCS1 could serve as a novel target for treatment of patients with intolerant chemotherapy and AML-RR patients.


Assuntos
Resistencia a Medicamentos Antineoplásicos/genética , Retículo Endoplasmático/efeitos dos fármacos , Hidroximetilglutaril-CoA Sintase/genética , Leucemia Mieloide Aguda/genética , Mitocôndrias/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Resposta a Proteínas não Dobradas/efeitos dos fármacos , Apoptose/efeitos dos fármacos , Células da Medula Óssea/efeitos dos fármacos , Linhagem Celular Tumoral , Resistencia a Medicamentos Antineoplásicos/efeitos dos fármacos , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Fator de Transcrição GATA1/genética , Células HL-60 , Humanos , Hidroximetilglutaril-CoA Sintase/antagonistas & inibidores , Tunicamicina/farmacologia
2.
Plant Cell Physiol ; 62(1): 205-218, 2021 Mar 25.
Artigo em Inglês | MEDLINE | ID: mdl-33340324

RESUMO

Little has been established on the relationship between the mevalonate (MVA) pathway and other metabolic pathways except for the sterol and glucosinolate biosynthesis pathways. In the MVA pathway, 3-hydroxy-3-methylglutaryl-CoA synthase (HMGS) catalyzes the condensation of acetoacetyl-CoA and acetyl-CoA to form 3-hydroxy-3-methylglutaryl-coenzyme A. Our previous studies had shown that, while the recombinant Brassica juncea HMGS1 (BjHMGS1) mutant S359A displayed 10-fold higher enzyme activity than wild-type (wt) BjHMGS1, transgenic tobacco overexpressing S359A (OE-S359A) exhibited higher sterol content, growth rate and seed yield than OE-wtBjHMGS1. Herein, untargeted proteomics and targeted metabolomics were employed to understand the phenotypic effects of HMGS overexpression in tobacco by examining which other metabolic pathways were affected. Sequential window acquisition of all theoretical mass spectra quantitative proteomics analysis on OE-wtBjHMGS1 and OE-S359A identified the misregulation of proteins in primary metabolism and cell wall modification, while some proteins related to photosynthesis and the tricarboxylic acid cycle were upregulated in OE-S359A. Metabolomic analysis indicated corresponding changes in carbohydrate, amino acid and fatty acid contents in HMGS-OEs, and F-244, a specific inhibitor of HMGS, was applied successfully on tobacco to confirm these observations. Finally, the crystal structure of acetyl-CoA-liganded S359A revealed that improved activity of S359A likely resulted from a loss in hydrogen bonding between Ser359 and acyl-CoA, which is evident in wtBjHMGS1. This work suggests that regulation of plant growth by HMGS can influence the central metabolic pathways. Furthermore, this study demonstrates that the application of the HMGS-specific inhibitor (F-244) in tobacco represents an effective approach for studying the HMGS/MVA pathway.


Assuntos
Hidroximetilglutaril-CoA Sintase/metabolismo , Redes e Vias Metabólicas , Nicotiana/metabolismo , Proteínas de Plantas/metabolismo , Dimetil Sulfóxido/farmacologia , Ácidos Graxos/metabolismo , Ácidos Graxos Insaturados/farmacologia , Regulação da Expressão Gênica de Plantas/efeitos dos fármacos , Ligação de Hidrogênio , Hidroximetilglutaril-CoA Sintase/antagonistas & inibidores , Hidroximetilglutaril-CoA Sintase/química , Lactonas/farmacologia , Espectrometria de Massas , Redes e Vias Metabólicas/efeitos dos fármacos , Estrutura Terciária de Proteína , Nicotiana/enzimologia
3.
IUBMB Life ; 72(9): 1997-2009, 2020 09.
Artigo em Inglês | MEDLINE | ID: mdl-32734614

RESUMO

Diabetic cardiomyopathy (DCM) is a cardiac disorder, which affects around 12% diabetic patients, resulting in overt heart death. Our initial bioinformatic analysis identified the differentially expressed gene 3-hydroxy-3-methylglutaryl-coenzyme A synthase 2 (HMGCS2) in DCM, which may be activated by peroxisome proliferator-activated receptor-alpha (PPARα) based on previous evidence. Therefore, the present study aims to explore the effect of PPARα on the development of DCM through regulating HMGCS2. The expression of PPARα and HMGCS2 was detected by reverse transcription quantitative polymerase chain reaction in cardiomyocytes and high-glucose-cultured cardiomyocytes. The proliferation and apoptosis of cardiomyocytes were examined by 5-ethynyl-2'-deoxyuridine assay and flow cytometry, separately. Mitoehondrial membrane potential (MMP) and intracellular reactive oxygen species (ROS) levels were determined. Then, the protein levels of B-cell lymphoma 2, Bcl-2-associated X protein, and cleaved Caspase-3 were detected by Western blot analysis. The myocardial apoptosis index, heart weight, and serum lipids of rats were examined. At last, the expressions of atrial natriuretic peptide (ANP), brain natriuretic peptide (BNP), transforming growth factor ß1 (TGFß1), peroxisome proliferator activator receptor gamma coactivator-1 alpha (PGC1α), nuclear respiratory factor (NRF)-1, NRF-2, NAD(P)H oxidase 1, and superoxide dismutase-1 were examined. HMGCS2 was the most differentially expressed gene in DCM. The levels of HMGCS2 and PPARα were upregulated in patients with DCM. HMGCS2 silencing was shown to inhibit HMGCS2 expression to suppress the apoptosis of high-glucose-induced cardiomyocytes and the loss of MMP, reduce the accumulation of ROS, and promote cardiomyocyte proliferation. Silencing of HMGCS2 and PPARα alleviated myocardial injury, decreased blood glucose, and lipid in DCM rats, downregulated the expression of ANP, BNP, and TGFß1 to reduce myocardial injury, and elevated PGC1α, NRF-1, and NRF-2 levels to enhance oxidative stress levels. Our results demonstrated that silencing of PPARα could alleviate cardiomyocyte injury and oxidative stress via a mechanism related to the downregulation of HMGCS2, which could provide a novel target for DCM treatment.


Assuntos
Apoptose , Diabetes Mellitus Experimental/complicações , Cardiomiopatias Diabéticas/prevenção & controle , Hidroximetilglutaril-CoA Sintase/antagonistas & inibidores , Miócitos Cardíacos/metabolismo , Estresse Oxidativo , PPAR alfa/antagonistas & inibidores , Animais , Cardiomiopatias Diabéticas/etiologia , Cardiomiopatias Diabéticas/metabolismo , Cardiomiopatias Diabéticas/patologia , Humanos , Masculino , Ratos , Espécies Reativas de Oxigênio
4.
J Antibiot (Tokyo) ; 73(7): 475-479, 2020 07.
Artigo em Inglês | MEDLINE | ID: mdl-32139880

RESUMO

We found that the protein synthesis inhibitor hygromycin B induced the production of secondary metabolites, including lucilactaene, NG-391, fusarubin, 1233A, and 1233B, in the filamentous fungus, Fusarium sp. RK97-94. We identified the biosynthetic gene cluster for 1233A, an HMG-CoA synthase inhibitor. The biosynthetic gene cluster consisted of four genes, one of which was involved in conferring self-resistance to 1233A.


Assuntos
Ácidos Graxos Insaturados/genética , Higromicina B/metabolismo , Família Multigênica/genética , Fungos/genética , Fungos/metabolismo , Furanos/metabolismo , Fusarium/genética , Fusarium/metabolismo , Hidroximetilglutaril-CoA Sintase/antagonistas & inibidores , Lactonas , Naftoquinonas/metabolismo , Pirróis/metabolismo
5.
Neuropharmacology ; 148: 377-393, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-28987936

RESUMO

The brain is a high energy-consuming organ that typically utilizes glucose as the main energy source for cerebral activity. When glucose becomes scarce under conditions of stress, ketone bodies, such as ß-hydroxybutyrate, acetoacetate and acetone, become extremely important. Alterations in brain energy metabolism have been observed in psychostimulant abusers; however, the mode of brain metabolic programming in cocaine dependence remains largely unknown. Here, we profiled the metabolites and metabolic enzymes from brain nucleus accumbens (NAc) of mice exposed to cocaine. We found that cocaine modified energy metabolism and markedly activated ketogenesis pathway in the NAc. The expression of HMG-CoA synthase 2 (HMGCS2), a critical rate-limiting ketogenesis enzyme, was markedly up-regulated. After switching metabolic pathways from ketogenesis to glycolysis through activation of glucokinase, cocaine-evoked metabolic reprogramming regained homeostasis, and the cocaine effect was attenuated. Importantly, both the pharmacological and genetic inhibition of HMGCS2 significantly suppressed cocaine-induced ketogenesis and behavior. In conclusion, cocaine induces a remarkable energy reprogramming in the NAc, which is characterized by HMGCS2-driven ketogenesis. Such effect may facilitate adaptations to cocaine-induced energy stress in the brain. Our findings establish an important link between drug-induced energy reprogramming and cocaine effect, and may have implication in the treatment of cocaine addiction.


Assuntos
Cocaína/farmacologia , Metabolismo Energético/efeitos dos fármacos , Hidroximetilglutaril-CoA Sintase/biossíntese , Corpos Cetônicos/metabolismo , Animais , Homeostase , Hidroximetilglutaril-CoA Sintase/antagonistas & inibidores , Masculino , Camundongos , Núcleo Accumbens/metabolismo , Regulação para Cima/efeitos dos fármacos
6.
Endocrine ; 63(3): 615-631, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30460485

RESUMO

PURPOSE: Diabetic mellitus-induced erectile dysfunction (DMED) represents a significant complication associated with diabetes mellitus (DM) that greatly affects human life quality. Various reports have highlighted the involvement of mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase 2 (HMGCS2) in the regulation of mitochondrial fatty acid oxidation, which has also been linked with DM. Through bioinformatics analysis, HMGCS2 was determined to be a novel target among DM patients suffering from erectile dysfunction (ED), and enriched in the Ras/ERK/PPAR signaling axis. Owing to the fact that the key mechanism HMGCS2 involved in DM remains largely unknown, we set out to investigate the role of the Ras/MAPK/PPARγ signaling axis and HMGCS2 in the corpus cavernosal endothelial cells (CCECs) of rats with DMED. METHODS: Firstly, bioinformatics analysis was used to screen out differentially expressed genes in DMED. Then, to investigate the influence of the Ras/MAPK/PPARγ signaling axis and HMGCS2 on DMED, a rat model of DMED was established and injected with Simvastatin and si-Hmgcs2. The individual expression patterns of Ras, MAPK, PPARγ and HMGCS2 were determined by RT-qPCR, immunohistochemistry and western blot analysis methods. Afterwards, to investigate the mechanism of Ras/MAPK/PPARγ signaling axis and HMGCS2, CCECs were isolated from DMED rats and transfected with agonists and inhibitors of the Ras/MAPK/PPARγ signaling axis and siRNA of HMGCS2, with their respective functions in apoptosis and impairment of CCECs evaluated using TUNEL staining and flow cytometry. RESULTS: Microarray analysis and KEGG pathway enrichment analysis revealed that Ras/ERK/PPAR signaling axis mediated HMGCS2 in DMED. Among the DMED rats, the Ras/MAPK/PPAR signaling axis was also activated while the expression of HMGCS2 was upregulated. The activation of Ras was determined to be capable of upregulating ERK expression which resulted in the inhibition of the transcription of PPARγ and subsequent upregulation of HMGCS2 expression. The inhibited activation of the Ras/ERK/PPAR signaling axis and silencing HMGCS2 were observed to provide an alleviatory effect on the injury of DMED while acting to inhibit the apoptosis of CCECs. CONCLUSION: Collectively, the key findings suggested that suppression of the Ras/MAPK/PPARγ signaling axis could downregulate expression of HMGCS2, so as to alleviate DMED. This study defines the potential treatment for DMED through inhibition of the Ras/MAPK/PPARγ signaling axis and silencing HMGCS2.


Assuntos
Diabetes Mellitus Experimental/complicações , Disfunção Erétil/tratamento farmacológico , Hidroximetilglutaril-CoA Sintase/antagonistas & inibidores , Sistema de Sinalização das MAP Quinases , Sinvastatina/uso terapêutico , Animais , Ensaios de Seleção de Medicamentos Antitumorais , Células Endoteliais/metabolismo , Disfunção Erétil/enzimologia , Disfunção Erétil/etiologia , Hidroximetilglutaril-CoA Sintase/metabolismo , Masculino , PPAR gama/metabolismo , Pênis/metabolismo , RNA Interferente Pequeno/uso terapêutico , Ratos Sprague-Dawley , Sinvastatina/farmacologia , Proteínas ras/metabolismo
7.
J Biol Chem ; 292(24): 10142-10152, 2017 06 16.
Artigo em Inglês | MEDLINE | ID: mdl-28468827

RESUMO

Contributions of metabolic changes to cancer development and maintenance have received increasing attention in recent years. Although many human cancers share similar metabolic alterations, it remains unclear whether oncogene-specific metabolic alterations are required for tumor development. Using an RNAi-based screen targeting the majority of the known metabolic proteins, we recently found that oncogenic BRAFV600E up-regulates HMG-CoA lyase (HMGCL), which converts HMG-CoA to acetyl-CoA and a ketone body, acetoacetate, that selectively enhances BRAFV600E-dependent MEK1 activation in human cancer. Here, we identified HMG-CoA synthase 1 (HMGCS1), the upstream ketogenic enzyme of HMGCL, as an additional "synthetic lethal" partner of BRAFV600E Although HMGCS1 expression did not correlate with BRAFV600E mutation in human melanoma cells, HMGCS1 was selectively important for proliferation of BRAFV600E-positive melanoma and colon cancer cells but not control cells harboring active N/KRAS mutants, and stable knockdown of HMGCS1 only attenuated colony formation and tumor growth potential of BRAFV600E melanoma cells. Moreover, cytosolic HMGCS1 that co-localized with HMGCL and BRAFV600E was more important than the mitochondrial HMGCS2 isoform in BRAFV600E-expressing cancer cells in terms of acetoacetate production. Interestingly, HMGCL knockdown did not affect HMGCS1 expression levels, whereas HMGCS1 knockdown caused a compensating increase in HMGCL protein level because of attenuated protein degradation. However, this increase did not reverse the reduced ketogenesis in HMGCS1 knockdown cells. Mechanistically, HMGCS1 inhibition decreased intracellular acetoacetate levels, leading to reduced BRAFV600E-MEK1 binding and consequent MEK1 activation. We conclude that the ketogenic HMGCS1-HMGCL-acetoacetate axis may represent a promising therapeutic target for managing BRAFV600E-positive human cancers.


Assuntos
Neoplasias do Colo/enzimologia , Hidroximetilglutaril-CoA Sintase/metabolismo , MAP Quinase Quinase 1/metabolismo , Melanoma/enzimologia , Proteínas de Neoplasias/metabolismo , Oxo-Ácido-Liases/metabolismo , Proteínas Proto-Oncogênicas B-raf/metabolismo , Acetoacetatos/metabolismo , Substituição de Aminoácidos , Animais , Linhagem Celular Tumoral , Proliferação de Células , Neoplasias do Colo/metabolismo , Neoplasias do Colo/patologia , Citosol/enzimologia , Citosol/metabolismo , Ativação Enzimática , Estabilidade Enzimática , Feminino , Humanos , Hidroximetilglutaril-CoA Sintase/antagonistas & inibidores , Hidroximetilglutaril-CoA Sintase/genética , Isoenzimas/antagonistas & inibidores , Isoenzimas/genética , Isoenzimas/metabolismo , MAP Quinase Quinase 1/química , Melanoma/metabolismo , Melanoma/patologia , Camundongos Nus , Mutação , Proteínas de Neoplasias/antagonistas & inibidores , Proteínas de Neoplasias/química , Proteínas de Neoplasias/genética , Transplante de Neoplasias , Oxo-Ácido-Liases/antagonistas & inibidores , Oxo-Ácido-Liases/química , Oxo-Ácido-Liases/genética , Proteólise , Proteínas Proto-Oncogênicas B-raf/genética , Interferência de RNA , Carga Tumoral
8.
Biochem Biophys Res Commun ; 486(2): 492-498, 2017 04 29.
Artigo em Inglês | MEDLINE | ID: mdl-28320515

RESUMO

HMGCS2 (mitochondrial 3-hydroxy-3-methylglutaryl-COA synthase 2) is a control enzyme in ketogenesis. The mitochondrial localization and interaction with APP (ß-amyloid precursor protein) suggest that HMGCS2 may play a role in the pathophysiology of AD (Alzheimer's disease). Here we report that overexpression of HMGCS2 decreased levels of APP and related CTFs (carboxy-terminal fragments), which was largely prevented by an autophagic inhibitor chloroquine. In addition, HMGCS2 enhancement of autophagic marker LC3II was diminished by rapamycin, an inhibitor of mechanistic target of rapamycin. Moreover, deprivation of EBSS (Earle's Balanced Salt Solution) significantly augmented the effect of HMGCS2 on LC3II, while acetoacetate reversed the reduction of LC3II, APP and CTFs which was induced by HMGCS2 knockdown. In the presence of acetoacetate, rapamycin failed to induce further increase of LC3II, which mimicked the effect of HMGCS2 overexpression. Finally, HMGCS2 enhanced the antioxidant response. Collectively, HMGCS2 shares with ketone bodies common features in autophagic clearance of APP and CTFs, suggesting that ketone bodies play an important role in HMGCS2 regulation of the autophagy.


Assuntos
Precursor de Proteína beta-Amiloide/metabolismo , Autofagia/genética , Hidroximetilglutaril-CoA Sintase/genética , Corpos Cetônicos/metabolismo , Proteínas Associadas aos Microtúbulos/genética , Serina-Treonina Quinases TOR/genética , Acetoacetatos/farmacologia , Animais , Linhagem Celular , Cloroquina/farmacologia , Regulação da Expressão Gênica , Células HEK293 , Hipocampo/citologia , Hipocampo/efeitos dos fármacos , Hipocampo/metabolismo , Humanos , Hidroximetilglutaril-CoA Sintase/antagonistas & inibidores , Hidroximetilglutaril-CoA Sintase/metabolismo , Camundongos , Proteínas Associadas aos Microtúbulos/metabolismo , Neurônios/citologia , Neurônios/efeitos dos fármacos , Neurônios/metabolismo , Proteólise/efeitos dos fármacos , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transdução de Sinais , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Transgenes
9.
Cell Death Differ ; 24(3): 458-468, 2017 03.
Artigo em Inglês | MEDLINE | ID: mdl-27935584

RESUMO

The intestinal epithelium undergoes a continual process of proliferation, differentiation and apoptosis. Previously, we have shown that the PI3K/Akt/mTOR pathway has a critical role in intestinal homeostasis. However, the downstream targets mediating the effects of mTOR in intestinal cells are not known. Here, we show that the ketone body ß-hydroxybutyrate (ßHB), an endogenous inhibitor of histone deacetylases (HDACs) induces intestinal cell differentiation as noted by the increased expression of differentiation markers (Mucin2 (MUC2), lysozyme, IAP, sucrase-isomaltase, KRT20, villin, Caudal-related homeobox transcription factor 2 (CDX2) and p21Waf1). Conversely, knockdown of the ketogenic mitochondrial enzyme hydroxymethylglutaryl CoA synthase 2 (HMGCS2) attenuated spontaneous differentiation in the human colon cancer cell line Caco-2. Overexpression of HMGCS2, which we found is localized specifically in the more differentiated portions of the intestinal mucosa, increased the expression of CDX2, thus further suggesting the contributory role of HMGCS2 in intestinal differentiation. In addition, mice fed a ketogenic diet demonstrated increased differentiation of intestinal cells as noted by an increase in the enterocyte, goblet and Paneth cell lineages. Moreover, we showed that either knockdown of mTOR or inhibition of mTORC1 with rapamycin increases the expression of HMGCS2 in intestinal cells in vitro and in vivo, suggesting a possible cross-talk between mTOR and HMGCS2/ßHB signaling in intestinal cells. In contrast, treatment of intestinal cells with ßHB or feeding mice with a ketogenic diet inhibits mTOR signaling in intestinal cells. Together, we provide evidence showing that HMGCS2/ßHB contributes to intestinal cell differentiation. Our results suggest that mTOR acts cooperatively with HMGCS2/ßHB to maintain intestinal homeostasis.


Assuntos
Diferenciação Celular , Hidroximetilglutaril-CoA Sintase/metabolismo , Ácido 3-Hidroxibutírico/farmacologia , Fosfatase Alcalina/metabolismo , Animais , Fator de Transcrição CDX2/metabolismo , Células CACO-2 , Diferenciação Celular/efeitos dos fármacos , Inibidor de Quinase Dependente de Ciclina p21/metabolismo , Dieta Cetogênica , Células HT29 , Humanos , Hidroximetilglutaril-CoA Sintase/antagonistas & inibidores , Hidroximetilglutaril-CoA Sintase/genética , Queratina-20/metabolismo , Masculino , Alvo Mecanístico do Complexo 1 de Rapamicina/antagonistas & inibidores , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Mucina-2/metabolismo , Transdução de Sinais/efeitos dos fármacos , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
10.
Anim Biotechnol ; 28(3): 189-197, 2017 Jul 03.
Artigo em Inglês | MEDLINE | ID: mdl-27874312

RESUMO

Ketogenesis is the production of ketone bodies, which provide energy when the body lacks glucose. Under ketogenic conditions, the body switches from primarily carbohydrate to fat metabolism to maintain energy balance. However, accumulation of high levels of ketone bodies in the blood results in ketosis. Treating ketosis with natural substances is preferable, because they are unlikely to cause side-effects. Momilactone B is an active compound isolated from Korean rice. Based on previous studies, we hypothesized that momilactone B could inhibit ketosis. We constructed an in vitro ketosis model by glucose starvation. We used this model to test the anti-ketosis effects of momilactone B. A primary target for treating ketosis is angiopoietin-like-3 (ANGPTL3), which modulates lipoprotein metabolism by inhibiting lipoprotein lipase (LPL), a multifunctional enzyme that breaks down stored fat to produce triglycerides. We showed that momilactone B could regulate the ANGPTL3-LPL pathway. However, a strong anti-ketosis candidate drug should also inhibit ketogenesis. Ketogenesis can be suppressed by inhibiting the expression of 3-hydroxy-3-methylglutaryl-CoA synthase-2 (HMGCS2), a mitochondrial enzyme that converts acetyl-CoA to ketone bodies. We found that momilactone B suppressed the expression of HMGCS2 through the increased expression of STAT5b. We also elucidated the relationship of STAT5b to ANGPTL3 and LPL expression.


Assuntos
Angiopoietinas/metabolismo , Diterpenos/farmacologia , Hidroximetilglutaril-CoA Sintase/antagonistas & inibidores , Cetose/metabolismo , Lactonas/farmacologia , Lipase Lipoproteica/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Linhagem Celular , Proliferação de Células/efeitos dos fármacos , Sobrevivência Celular/efeitos dos fármacos , Hidroximetilglutaril-CoA Sintase/metabolismo , Corpos Cetônicos/metabolismo , Camundongos , Modelos Biológicos , Fator de Transcrição STAT5/metabolismo
11.
Arch Toxicol ; 90(12): 3061-3071, 2016 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26838045

RESUMO

It was reported that 2,4-dichlorophenoxyacetic acid (2,4-D), a commonly used herbicide and a possible endocrine disruptor, can disturb spermatogenesis, but the precise mechanism is not understood. Since 2,4-D is a weak peroxisome proliferator in hepatocytes and peroxisome proliferator-activated receptor α (PPARα) is also expressed in Leydig cells, this study aimed to investigate the link between PPARα and 2,4-D-mediated testicular dysfunction. 2,4-D (130 mg/kg/day) was administered to wild-type and Ppara-null mice for 2 weeks, and the alterations in testis and testosterone/cholesterol metabolism in Leydig cells were examined. Treatment with 2,4-D markedly decreased testicular testosterone in wild-type mice, leading to degeneration of spermatocytes and Sertoli cells. The 2,4-D decreased cholesterol levels in Leydig cells of wild-type mice through down-regulating the expression of 3-hydroxy-3-methylglutaryl coenzyme A synthase 1 and reductase, involved in de novo cholesterogenesis. However, the mRNAs encoding the important proteins involved in testosterone synthesis were unchanged by 2,4-D except for CYP17A1, indicating that exhausted cholesterol levels in the cells is a main reason for reduced testicular testosterone. Additionally, pregnancy rate and the number of pups between 2,4-D-treated wild-type male mice and untreated female mice were significantly lower compared with those between untreated couples. These phenomena were not observed in 2,4-D-treated Ppara-null males. Collectively, these results suggest a critical role for PPARα in 2,4-D-induced testicular toxicity due to disruption of cholesterol/testosterone homeostasis in Leydig cells. This study yields novel insights into the possible mechanism of testicular dysfunction and male infertility caused by 2,4-D.


Assuntos
Ácido 2,4-Diclorofenoxiacético/toxicidade , Disruptores Endócrinos/toxicidade , Herbicidas/toxicidade , Infertilidade Masculina/induzido quimicamente , Células Intersticiais do Testículo/efeitos dos fármacos , PPAR alfa/metabolismo , Testosterona/metabolismo , Ácido 2,4-Diclorofenoxiacético/administração & dosagem , Animais , Colesterol/química , Relação Dose-Resposta a Droga , Disruptores Endócrinos/administração & dosagem , Repressão Enzimática/efeitos dos fármacos , Herbicidas/administração & dosagem , Hidroximetilglutaril-CoA Redutases/química , Hidroximetilglutaril-CoA Redutases/genética , Hidroximetilglutaril-CoA Redutases/metabolismo , Hidroximetilglutaril-CoA Sintase/antagonistas & inibidores , Hidroximetilglutaril-CoA Sintase/genética , Hidroximetilglutaril-CoA Sintase/metabolismo , Infertilidade Masculina/metabolismo , Infertilidade Masculina/patologia , Infertilidade Masculina/fisiopatologia , Células Intersticiais do Testículo/metabolismo , Células Intersticiais do Testículo/patologia , Gotículas Lipídicas/efeitos dos fármacos , Gotículas Lipídicas/metabolismo , Gotículas Lipídicas/patologia , Masculino , Camundongos da Linhagem 129 , Camundongos Knockout , PPAR alfa/genética , Proliferadores de Peroxissomos/administração & dosagem , Proliferadores de Peroxissomos/toxicidade , Distribuição Aleatória , Epitélio Seminífero/efeitos dos fármacos , Epitélio Seminífero/metabolismo , Epitélio Seminífero/patologia , Epitélio Seminífero/fisiopatologia , Espermatogênese/efeitos dos fármacos
12.
Biochemistry ; 51(23): 4713-22, 2012 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-22510038

RESUMO

Hymeglusin (1233A, F244, L-659-699) is established as a specific ß-lactone inhibitor of eukaryotic hydroxymethylglutaryl-CoA synthase (HMGCS). Inhibition results from formation of a thioester adduct to the active site cysteine. In contrast, the effects of hymeglusin on bacterial HMG-CoA synthase, mvaS, have been minimally characterized. Hymeglusin blocks growth of Enterococcus faecalis. After removal of the inhibitor from culture media, a growth curve inflection point at 3.1 h is observed (vs 0.7 h for the uninhibited control). Upon hymeglusin inactivation of purified E. faecalis mvaS, the thioester adduct is more stable than that measured for human HMGCS. Hydroxylamine cleaves the thioester adduct; substantial enzyme activity is restored at a rate that is 8-fold faster for human HMGCS than for mvaS. Structural results explain these differences in enzyme-inhibitor thioester adduct stability and solvent accessibility. The E. faecalis mvaS-hymeglusin cocrystal structure (1.95 Å) reveals virtually complete occlusion of the bound inhibitor in a narrow tunnel that is largely sequestered from bulk solvent. In contrast, eukaryotic (Brassica juncea) HMGCS binds hymeglusin in a more solvent-exposed cavity.


Assuntos
Enterococcus faecalis/enzimologia , Inibidores Enzimáticos/farmacologia , Ácidos Graxos Insaturados/farmacologia , Hidroximetilglutaril-CoA Sintase/antagonistas & inibidores , Lactonas/farmacologia , Clonagem Molecular , Cristalografia por Raios X , Inibidores Enzimáticos/química , Ácidos Graxos Insaturados/química , Regulação Bacteriana da Expressão Gênica/fisiologia , Regulação Enzimológica da Expressão Gênica , Humanos , Hidroxilamina/química , Hidroxilamina/farmacologia , Hidroximetilglutaril-CoA Sintase/genética , Hidroximetilglutaril-CoA Sintase/metabolismo , Cinética , Lactonas/química , Modelos Moleculares , Estrutura Molecular , Ligação Proteica , Difração de Raios X
13.
J Infect Dis ; 204(4): 609-16, 2011 Aug 15.
Artigo em Inglês | MEDLINE | ID: mdl-21791663

RESUMO

BACKGROUND: Hepatitis C virus (HCV) chronically infects >170 million persons worldwide and is a leading cause of cirrhosis and hepatocellular carcinoma. The identification of more effective and better-tolerated agents for treating HCV is a high priority. We have reported elsewhere the discovery of the anti-HCV compound ceestatin using a high-throughput screen of a small molecule library. METHODS: To identify host or viral protein targets in an unbiased fashion, we performed affinity chromatography, using tandem liquid chromatography/mass spectrometry to identify specific potential targets. RESULTS. Ceestatin binds to 3-hydroxy-3-methylglutaryl-coenzyme A (HMG-CoA) synthase and irreversibly inhibits HMG-CoA synthase in a dose-dependent manner. Ceestatin's anti-HCV effects are reversed by addition of HMG-CoA, mevalonic acid, or geranylgeraniol. Treatment with small interfering RNA against HMG-CoA synthase led to a substantial reduction in HCV replication, further validating HMG-CoA synthase as an enzyme essential for HCV replication. CONCLUSIONS: Ceestatin therefore exerts its anti-HCV effects through inhibition of HMG-CoA synthase. It may prove useful as an antiviral agent, as a probe to study HCV replication, and as a cholesterol-lowering agent. The logical stepwise process employed to discover the mechanism of action of ceestatin can serve as a general experimental strategy to uncover the targets on which novel uncharacterized anti-HCV compounds act.


Assuntos
Antivirais/farmacologia , Compostos Bicíclicos Heterocíclicos com Pontes/farmacologia , Hepacivirus/efeitos dos fármacos , Hidroximetilglutaril-CoA Sintase/antagonistas & inibidores , Lactonas/farmacologia , Replicação Viral/efeitos dos fármacos , Linhagem Celular , Cromatografia de Afinidade , Hepacivirus/fisiologia , Humanos , Espectrometria de Massas , Ligação Proteica , Interferência de RNA , RNA Interferente Pequeno
14.
J Biomol Screen ; 16(6): 637-46, 2011 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-21593486

RESUMO

A simple, optical density-based assay for inhibitors of the mevalonate-dependent pathway for isoprenoid biosynthesis was developed. The assay uses pathway-sensitized Staphylococcus aureus strains and is fully compatible with high-density screening in a 1536-well format. S. aureus strains were constructed in which genes required for mevalonate-dependent isopentenyl pyrophosphate (IPP) synthesis were regulated by an isopropyl-ß-D-thiogalactopyranoside (IPTG)-inducible promoter. Inhibitors of the target enzymes displayed greater antibacterial potency in media containing low concentrations of IPTG, and therefore less induction of mevalonate pathway genes, than in media with high IPTG conditions. This differential growth phenotype was exploited to bias the cell-based screening hits toward specific inhibitors of mevalonate-dependent IPP biosynthesis. Screens were run against strains engineered for regulation of the enzymes HMG-CoA synthase (MvaS) and mevalonate kinase (mvaK1), mevalonate diphosphate decarboxylase (mvaD), and phosphomevalonate kinase (mvaK2). The latter three enzymes are regulated as an operon. These assays resulted in the discovery of potent antibacterial hits that were progressed to an active hit-to-lead program. The example presented here demonstrates that a cell sensitization strategy can be successfully applied to a 1.3-million compound high-throughput screen in a high-density 1536-well format.


Assuntos
Antibacterianos/farmacologia , Inibidores Enzimáticos/farmacologia , Ensaios de Triagem em Larga Escala , Ácido Mevalônico/metabolismo , Staphylococcus aureus/efeitos dos fármacos , Vias Biossintéticas/efeitos dos fármacos , Vias Biossintéticas/genética , Hidroximetilglutaril-CoA Sintase/antagonistas & inibidores , Hidroximetilglutaril-CoA Sintase/genética , Hidroximetilglutaril-CoA Sintase/metabolismo , Ácido Mevalônico/química , Testes de Sensibilidade Microbiana , Fosfotransferases (Aceptor do Grupo Álcool)/antagonistas & inibidores , Fosfotransferases (Aceptor do Grupo Álcool)/genética , Fosfotransferases (Aceptor do Grupo Álcool)/metabolismo , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo
15.
J Mol Biol ; 398(4): 497-506, 2010 May 14.
Artigo em Inglês | MEDLINE | ID: mdl-20346956

RESUMO

3-Hydroxy-3-methylglutaryl coenzyme A (CoA) synthase (HMGCS) catalyzes the condensation of acetyl-CoA and acetoacetyl-CoA into 3-hydroxy-3-methylglutaryl CoA. It is ubiquitous across the phylogenetic tree and is broadly classified into three classes. The prokaryotic isoform is essential in Gram-positive bacteria for isoprenoid synthesis via the mevalonate pathway. The eukaryotic cytosolic isoform also participates in the mevalonate pathway but its end product is cholesterol. Mammals also contain a mitochondrial isoform; its deficiency results in an inherited disorder of ketone body formation. Here, we report high-resolution crystal structures of the human cytosolic (hHMGCS1) and mitochondrial (hHMGCS2) isoforms in binary product complexes. Our data represent the first structures solved for human HMGCS and the mitochondrial isoform, allowing for the first time structural comparison among the three isoforms. This serves as a starting point for the development of isoform-specific inhibitors that have potential cholesterol-reducing and antibiotic applications. In addition, missense mutations that cause mitochondrial HMGCS deficiency have been mapped onto the hHMGCS2 structure to rationalize the structural basis for the disease pathology.


Assuntos
Hidroximetilglutaril-CoA Sintase/antagonistas & inibidores , Hidroximetilglutaril-CoA Sintase/química , Cetonas/metabolismo , Cristalografia por Raios X , Dimerização , Humanos , Hidroximetilglutaril-CoA Sintase/deficiência , Hidroximetilglutaril-CoA Sintase/genética , Modelos Moleculares , Mutação de Sentido Incorreto , Isoformas de Proteínas/antagonistas & inibidores , Isoformas de Proteínas/química , Isoformas de Proteínas/deficiência , Isoformas de Proteínas/genética , Estrutura Quaternária de Proteína
16.
Anal Biochem ; 396(1): 96-102, 2010 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-19706283

RESUMO

3-Hydroxy-3-methylglutaryl-CoA (HMG-CoA) synthase catalyzes the first physiologically irreversible step in biosynthesis of isoprenoids and sterols from acetyl-CoA. Inhibition of enzyme activity by beta-lactone-containing natural products correlates with substantial diminution of sterol synthesis, identifying HMG-CoA synthase as a potential drug target and suggesting that identification of effective inhibitors would be valuable. A visible wavelength spectrophotometric assay for HMG-CoA synthase has been developed. The assay uses dithiobisnitrobenzoic acid (DTNB) to detect coenzyme A (CoASH) release on acetylation of enzyme by the substrate acetyl-CoA, which precedes condensation with acetoacetyl-CoA to form the HMG-CoA product. The assay method takes advantage of the stability of recombinant enzyme in the absence of a reducing agent. It can be scaled down to a 60 microl volume to allow the use of 384-well microplates, facilitating high-throughput screening of compound libraries. Enzyme activity measured in the microplate assay is comparable to values measured by using conventional scale spectrophotometric assays with the DTNB method (412 nm) for CoASH production or by monitoring the use of a second substrate, acetoacetyl-CoA (300 nm). The high-throughput assay method has been successfully used to screen a library of more than 100,000 drug-like compounds and has identified both reversible and irreversible inhibitors of the human enzyme.


Assuntos
Ensaios Enzimáticos/métodos , Ensaios de Triagem em Larga Escala/métodos , Hidroximetilglutaril-CoA Sintase/análise , Luz , Espectrofotometria/métodos , Acetilcoenzima A/metabolismo , Ácido Ditionitrobenzoico/metabolismo , Ativação Enzimática/efeitos dos fármacos , Inibidores Enzimáticos/análise , Inibidores Enzimáticos/química , Inibidores Enzimáticos/farmacologia , Humanos , Hidroximetilglutaril-CoA Sintase/antagonistas & inibidores , Hidroximetilglutaril-CoA Sintase/metabolismo , Cinética , Reprodutibilidade dos Testes , Bibliotecas de Moléculas Pequenas/análise , Bibliotecas de Moléculas Pequenas/química , Bibliotecas de Moléculas Pequenas/farmacologia , Especificidade por Substrato/efeitos dos fármacos , Terpenos/metabolismo , Fatores de Tempo
17.
Mol Endocrinol ; 21(10): 2361-77, 2007 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-17595321

RESUMO

The peroxisome proliferator-activated receptors (PPARalpha, PPARdelta, and PPARgamma) constitute a family of nuclear receptors that regulates metabolic processes involved in lipid and glucose homeostasis. Although generally considered to function as ligand-regulated receptors, all three PPARs exhibit a high level of constitutive activity that may result from their stimulation by intracellularly produced endogenous ligands. Consequently, complete inhibition of PPAR signaling requires the development of inverse agonists. However, the currently available small molecule antagonists for the PPARs function only as partial agonists, or their efficacy is not sufficient to inhibit the constitutive activity of these receptors. Due to the lack of efficacious antagonists that interact with the ligand-binding domain of the PPARs, we decided to target an interaction that is central to nuclear receptor-mediated gene transcription: the nuclear receptor-coactivator interaction. We utilized phage display technology to identify short LXXLL-containing peptides that bind to the PPARs. Analysis of these peptides revealed a consensus binding motif consisting of HPLLXXLL. Cross-screening of these peptides for binding to other nuclear receptors enabled the identification of a high-affinity PPAR-selective peptide that has the ability to repress PPARgamma1-dependent transcription of transfected reporter genes. Most importantly, when introduced into HepG2 cells, the peptide inhibited the expression of endogenous PPARgamma1 target genes, adipose differentiation-related protein and mitochondrial 3-hydroxy-3-methylglutaryl coenzyme A synthase 2. This work lends support for the rational development of peptidomimetics that block receptor-mediated transcription by targeting the nuclear receptor-coactivator interaction surface.


Assuntos
PPAR gama/antagonistas & inibidores , Biblioteca de Peptídeos , Peptídeos/farmacologia , Motivos de Aminoácidos , Sequência de Aminoácidos , Linhagem Celular , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Expressão Gênica/efeitos dos fármacos , Humanos , Hidroximetilglutaril-CoA Sintase/antagonistas & inibidores , Hidroximetilglutaril-CoA Sintase/genética , Proteínas de Membrana/antagonistas & inibidores , Proteínas de Membrana/genética , Mitocôndrias/enzimologia , Dados de Sequência Molecular , PPAR gama/química , PPAR gama/genética , Peptídeos/química , Peptídeos/genética , Perilipina-2 , Conformação Proteica
18.
Proc Natl Acad Sci U S A ; 103(31): 11491-6, 2006 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-16864776

RESUMO

3-Hydroxy-3-methylglutaryl CoA synthase (HMGS) catalyzes the first committed step in the mevalonate metabolic pathway for isoprenoid biosynthesis and serves as an alternative target for cholesterol-lowering and antibiotic drugs. We have determined a previously undescribed crystal structure of a eukaryotic HMGS bound covalently to a potent and specific inhibitor F-244 [(E,E)-11-[3-(hydroxymethyl)-4-oxo-2-oxytanyl]-3,5,7-trimethyl-2,4-undecadienenoic acid]. Given the accessibility of synthetic analogs of the F-244 natural product, this inhibited eukaryotic HMGS structure serves as a necessary starting point for structure-based methods that may improve the potency and species-specific selectivity of the next generation of F-244 analogs designed to target particular eukaryotic and prokaryotic HMGS.


Assuntos
Inibidores Enzimáticos , Ácidos Graxos Insaturados , Hidroximetilglutaril-CoA Sintase/antagonistas & inibidores , Hidroximetilglutaril-CoA Sintase/química , Lactonas , Mostardeira/enzimologia , Estrutura Terciária de Proteína , Sítios de Ligação , Cristalografia por Raios X , Inibidores Enzimáticos/química , Inibidores Enzimáticos/metabolismo , Ácidos Graxos Insaturados/química , Ácidos Graxos Insaturados/metabolismo , Humanos , Hidroximetilglutaril-CoA Sintase/metabolismo , Lactonas/química , Lactonas/metabolismo , Modelos Moleculares , Dados de Sequência Molecular , Proteínas de Plantas/antagonistas & inibidores , Proteínas de Plantas/química , Proteínas de Plantas/metabolismo , Relação Estrutura-Atividade
19.
J Biol Chem ; 276(15): 11674-82, 2001 Apr 13.
Artigo em Inglês | MEDLINE | ID: mdl-11148203

RESUMO

The amino acid leucine is efficiently used by the trypanosomatid Leishmania mexicana for sterol biosynthesis. The incubation of [2-(13)C]leucine with L. mexicana promastigotes in the presence of ketoconazole gave 14alpha-methylergosta-8,24(24(1))-3beta-ol as the major sterol, which was shown by mass spectrometry to contain up to six atoms of (13)C per molecule. (13)C NMR analysis of the 14alpha-methylergosta-8,24(24(1))-3beta-ol revealed that it was labeled in only six positions: C-2, C-6, C-11, C-12, C-16, and C-23. This established that the leucine skeleton is incorporated intact into the isoprenoid pathway leading to sterol; it is not converted first to acetyl-CoA, as in animals and plants, with utilization of the acetyl-CoA to regenerate 3-hydroxy-3-methylglutaryl-CoA (HMG-CoA). An inhibitor of HMG-CoA synthase (L-659,699) blocked the incorporation of [1-(14)C]acetate into sterol but had no inhibitory effect on [U-(14)C]leucine incorporation. The HMG-CoA reductase inhibitor lovastatin inhibited promastigote growth and [U-(14)C]leucine incorporation into sterol. The addition of unlabeled mevalonic acid (MVA) overcame the lovastatin inhibition of growth and also diluted the incorporation of [1-(14)C]leucine into sterol. These results are compatible with two routes by which the leucine skeleton may enter intact into the isoprenoid pathway. The catabolism of leucine could generate HMG-CoA that is then directly reduced to MVA for incorporation into sterol. Alternatively, a compound produced as an intermediate in leucine breakdown to HMG-CoA (e.g. dimethylcrotonyl-CoA) could be directly reduced to produce an isoprene alcohol followed by phosphorylation to enter the isoprenoid pathway post-MVA.


Assuntos
Leishmania mexicana/metabolismo , Leucina/metabolismo , Esteróis/biossíntese , Animais , Antiprotozoários/farmacologia , Isótopos de Carbono , Inibidores Enzimáticos/farmacologia , Ácidos Graxos Insaturados/farmacologia , Hidroximetilglutaril-CoA Sintase/antagonistas & inibidores , Lactonas/farmacologia , Leishmania mexicana/efeitos dos fármacos , Leishmania mexicana/enzimologia , Espectroscopia de Ressonância Magnética , Espectrometria de Massas , Ressonância Magnética Nuclear Biomolecular , Esteróis/química
20.
Zhonghua Xue Ye Xue Za Zhi ; 22(2): 72-5, 2001 Feb.
Artigo em Chinês | MEDLINE | ID: mdl-11877052

RESUMO

OBJECTIVE: To investigate the anti-apoptotic mechanism and explore approach to inhibiting proliferation and inducing apoptosis of chronic myclogenous leukemia (CML) cells. METHODS: K562 cell line was used to evaluate the effects of simvastatin, an inhibitor of 3-hydroxy-3-methylglutaryl coenzyme A (HMG-CoA) reductase, and the combination of simvastatin with chemotherapeutic agents on the proliferation and apoptosis of CML cells. RESULTS: Simvastatin could significantly inhibit proliferation and induce apoptosis of K562 cells, and could increase the sensitivity of K562 cells to chemotherapeutic agents. Addition of mevalonate, the immediate product of HMG-CoA, could completely reverse this effect. CONCLUSION: Simvastatin inhibited proliferation and induced apoptosis of K562 cells through inhibiting the metabolic pathway of mevalonate. It is promising that HMG-CoA reductase inhibitors may be an effective chemotherapeutic approach to the treatment of CML.


Assuntos
Apoptose/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Inibidores Enzimáticos/farmacologia , Leucemia/fisiopatologia , Sinvastatina/farmacologia , Antineoplásicos/farmacologia , Humanos , Hidroximetilglutaril-CoA Sintase/antagonistas & inibidores , Hidroximetilglutaril-CoA Sintase/metabolismo , Células K562 , Leucemia/tratamento farmacológico , Leucemia/enzimologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...