Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
J Clin Lipidol ; 16(1): 23-27, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34887219

RESUMO

We report the case of an individual with severe hypercholesterolemia who experienced rhabdomyolysis with high dose atorvastatin. Genetic testing was undertaken to evaluate for suspected familial hypercholesterolemia (FH) and for the presence of gene variants associated with susceptibility to statin associated muscle disease. Genetic testing identified the presence of a potentially damaging variant of the hepatic xenobiotic transporter pump SLCO1B1, a single nucleotide variant (SNV) (rs77271279, c.481+1G>T) that disrupts the canonical donor splice motif. Although this variant has not previously been reported as associated with rhabdomyolysis and thus requires validation in population studies, it likely played a role in this patient's susceptibility to rhabdomyolysis based on functional assessment of the effect of this variant on SLCO1B1 protein function and given the known role of this transporter in statin uptake by the liver. The presence of this gene variant reinforced our decision to treat the patient's hypercholesterolemia with non-statin alternatives (PCSK9 inhibitor and ezetimibe). Genetic testing also identified the presence of a second SLCO1B1 gene variant, c.1200C>G (p.Phe400Leu, rs59113707) and homozygosity for an intron variant of the apolipoprotein(a) (LPA) gene (c.2604.138G>A intron variant, rs9457951) associated with increased Lp(a), a risk factor for atherosclerotic cardiovascular disease. Notably, all three variants are rare in persons of European descent but more frequent in African-Americans. These findings underscore the role of disabling mutations of the SLCO1B1 gene in statin myopathy and the need to validate these and other gene variants associated with statin myopathy in a population of patients with statin-associated muscle disease.


Assuntos
Inibidores de Hidroximetilglutaril-CoA Redutases , Hipercolesterolemia , Doenças Musculares , Rabdomiólise , Atorvastatina/efeitos adversos , Humanos , Inibidores de Hidroximetilglutaril-CoA Redutases/efeitos adversos , Hipercolesterolemia/tratamento farmacológico , Transportador 1 de Ânion Orgânico Específico do Fígado/genética , Doenças Musculares/genética , Polimorfismo de Nucleotídeo Único , Pró-Proteína Convertase 9/genética , Rabdomiólise/tratamento farmacológico , Rabdomiólise/genética
2.
Mol Cell Biochem ; 463(1-2): 13-31, 2020 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-31541353

RESUMO

Insulin stimulates de novo lipid synthesis in the liver and in cultured hepatocytes via its ability to activate sterol regulatory element-binding protein 1c (SREBP-1c). Although PI3K-AKT-mTORC1-p70S6K-signaling kinases are known to drive feed-forward expression of SREBP-1c, the identity of the phosphorylated amino acid residue(s) putatively involved in insulin-stimulated de novo lipogenesis remains elusive. We obtained in silico and mass spectrometry evidence, that was combined with siRNA strategies, to discover that insulin-induced phosphorylation of serine 418, serine 419, and serine 422 in rat SREBP-1c was most likely mediated by p70S6 kinase. Here, for the first time, we show that insulin-induced phosphorylation of these 3 serine residues mainly impinged on the mechanisms of proteostasis of both full-length and mature SREBP-1c in the McArdle-RH7777 hepatoma cells. Consistent with this conclusion, nascent SREBP-1c, substituted with phosphomimetic aspartic acid residues at these 3 sites, was resistant to proteasomal degradation. As a consequence, endoplasmic reticulum to Golgi migration and proteolytic maturation of pSREBP-1c was significantly enhanced which led to increased accumulation of mature nSREBP-1c, even in the absence of insulin. Remarkably, aspartic acid substitutions at S418, S419 and S422 also protected the nascent SREBP-1c from ubiquitin-mediated proteasome degradation thus increasing its steady-state levels and transactivation potential in the nucleus. These complementary effects of p70S6K-mediated phosphorylation on proteostasis of pSREBP-1c were necessary and sufficient to account for insulin's ability to enhance transcription of genes controlling de novo lipogenesis in hepatocytes.


Assuntos
Hepatócitos/metabolismo , Lipídeos/biossíntese , Lipogênese , Proteostase , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Serina-Treonina Quinases TOR/metabolismo , Linhagem Celular Tumoral , Hepatócitos/citologia , Humanos , Lipídeos/genética , Proteínas Quinases S6 Ribossômicas 70-kDa/genética , Proteínas Quinases S6 Ribossômicas 70-kDa/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Serina-Treonina Quinases TOR/genética , Transcrição Gênica
3.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1864(8): 1145-1156, 2019 08.
Artigo em Inglês | MEDLINE | ID: mdl-31067497

RESUMO

Lipid homeostasis is critically dependent on the liver. Hepatic genes involved in lipid biosynthesis are controlled by combinatorial actions of multiple transcription factors that include three sterol regulatory element binding proteins (SREBPs), carbohydrate responsive element binding protein, liver X receptors, and others. SREBP-1c, a seminal regulator of de novo lipogenesis, resides in the endoplasmic reticulum as a transcriptionally inert precursor and must undergo a regulated intramembrane proteolysis (RIP) prior to its nuclear translocation as a bone fide transcription factor. The regulation of biosynthesis, turnover and actions of SREBP-1c and lipogenesis are mechanistically linked to signaling kinases, canonically induced by macronutrients and insulin. Here, we briefly review the evidence showing that phosphorylation of SREBP-1c and its interacting partners, catalyzed by phosphatidyl inositol-3-kinase, protein kinase B, mechanistic target of rapamycin complex 1 and 2, mitogen activated protein kinases, glycogen synthase kinase-3ß, protein kinase A and 5' adenosine monophosphate-activated protein kinase regulates the mechanisms of RIP and stability of SREBP-1c and de novo lipogenesis.


Assuntos
Proteostase , Proteínas de Ligação a Elemento Regulador de Esterol/metabolismo , Animais , Humanos , Lipogênese , Fosforilação , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo
4.
Biol Sex Differ ; 9(1): 40, 2018 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-30201044

RESUMO

BACKGROUND: Patients with metabolic syndrome, who are characterized by co-existence of insulin resistance, hypertension, hyperlipidemia, and obesity, are also prone to develop non-alcoholic fatty liver disease (NAFLD). Although the prevalence and severity of NAFLD is significantly greater in men than women, the mechanisms by which gender modulates the pathogenesis of hepatic steatosis are poorly defined. The obese spontaneously hypertensive (SHROB) rats represent an attractive model of metabolic syndrome without overt type 2 diabetes. Although pathological manifestation caused by the absence of a functional leptin receptor has been extensively studied in SHROB rats, it is unknown whether these animals elicited sex-specific differences in the development of hepatic steatosis. METHODS: We compared hepatic pathology in male and female SHROB rats. Additionally, we examined key biochemical and molecular parameters of signaling pathways linked with hyperinsulinemia and hyperlipidemia. Finally, using methods of quantitative polymerase chain reaction (qPCR) and western blot analysis, we quantified expression of 45 genes related to lipid biosynthesis and metabolism in the livers of male and female SHROB rats. RESULTS: We show that all SHROB rats developed hepatic steatosis that was accompanied by enhanced expression of SREBP1, SREBP2, ACC1, and FASN proteins. The livers of male rats also elicited higher induction of Pparg, Ppara, Slc2a4, Atox1, Skp1, Angptl3, and Pnpla3 mRNAs. In contrast, the livers of female SHROB rats elicited constitutively higher levels of phosphorylated JNK and AMPK and enhanced expression of Cd36. CONCLUSION: Based on these data, we conclude that the severity of hepatic steatosis in male and female SHROB rats was mainly driven by increased de novo lipogenesis. Moreover, male and female SHROB rats also elicited differential severity of hepatic steatosis that was coupled with sex-specific differences in fatty acid transport and esterification.


Assuntos
Hipertensão , Hepatopatia Gordurosa não Alcoólica , Obesidade , Caracteres Sexuais , Animais , Antígenos CD36/metabolismo , Ácidos Graxos/metabolismo , Feminino , Hipertensão/metabolismo , Lipogênese , Fígado/metabolismo , Masculino , Proteínas de Membrana/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Obesidade/metabolismo , Fosfolipases A2/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos
5.
Biosci Rep ; 36(1): e00284, 2015 Nov 20.
Artigo em Inglês | MEDLINE | ID: mdl-26589965

RESUMO

Sterol regulatory element binding protein-1c (SREBP-1c) is a key transcription factor that regulates genes involved in the de novo lipid synthesis and glycolysis pathways. The structure, turnover and transactivation potential of SREBP-1c are regulated by macronutrients and hormones via a cascade of signalling kinases. Using MS, we have identified serine 73 as a novel glycogen synthase kinase-3 (GSK-3) phosphorylation site in the rat SREBP-1c purified from McA-RH7777 hepatoma cells. Our site-specific mutagenesis strategy revealed that the turnover of SREBP-1c, containing wild type, phospho-null (serine to alanine) or phospho-mimetic (serine to aspartic acid) substitutions, was differentially regulated. We show that the S73D mutant of pSREBP-1c, that mimicked a state of constitutive phosphorylation, dissociated from the SREBP-1c-SCAP complex more readily and underwent GSK-3-dependent proteasomal degradation via SCF(Fbw7) ubiquitin ligase pathway. Pharmacologic inhibition of GSK-3 or knockdown of GSK-3 by siRNA prevented accelerated degradation of SREBP-1c. As demonstrated by MS, SREBP-1c was phosphorylated in vitro by GSK-3ß at serine 73. Phosphorylation of serine 73 also occurs in the intact liver. We propose that GSK-3-mediated phosphorylation of serine 73 in the rat SREBP-1c and its concomitant destabilization represents a novel mechanism involved in the inhibition of de novo lipid synthesis in the liver.


Assuntos
Quinase 3 da Glicogênio Sintase/metabolismo , Lipídeos/biossíntese , Fígado/metabolismo , Complexo de Endopeptidases do Proteassoma/metabolismo , Proteólise , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Substituição de Aminoácidos , Animais , Linhagem Celular Tumoral , Quinase 3 da Glicogênio Sintase/genética , Células HEK293 , Humanos , Lipídeos/genética , Mutação de Sentido Incorreto , Fosforilação/fisiologia , Complexo de Endopeptidases do Proteassoma/genética , Estabilidade Proteica , Ratos , Proteína de Ligação a Elemento Regulador de Esterol 1/genética
6.
Biochim Biophys Acta ; 1851(12): 1521-9, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26327595

RESUMO

In hyperinsulinemic states including obesity and T2DM, overproduction of fatty acid and triglyceride contributes to steatosis of the liver, hyperlipidemia and hepatic insulin resistance. This effect is mediated in part by the transcriptional regulator sterol responsive element binding protein-1c (SREBP-1c), which stimulates the expression of genes involved in hepatic fatty acid and triglyceride synthesis. SREBP-1c is up regulated by insulin both via increased transcription of nascent full-length SREBP-1c and by enhanced proteolytic processing of the endoplasmic reticulum (ER)-bound precursor to yield the transcriptionally active n-terminal form, nSREBP-1c. Polyunsaturated fatty acids of marine origin (n-3 PUFA) prevent induction of SREBP-1c by insulin thereby reducing plasma and hepatic triglycerides. Despite widespread use of n-3 PUFA supplements to reduce triglycerides in clinical practice, the exact mechanisms underlying their hypotriglyceridemic effect remain elusive. Here we demonstrate that the n-3 PUFA docosahexaenoic acid (DHA; 22:5 n-3) reduces nSREBP-1c by inhibiting regulated intramembrane proteolysis (RIP) of the nascent SREBP-1c. We further show that this effect of DHA is mediated both via activation of AMP-activated protein kinase (AMPK) and by inhibition of mechanistic target of rapamycin complex 1 (mTORC1). The inhibitory effect of AMPK on SREBP-1c processing is linked to phosphorylation of serine 365 of SREBP-1c in the rat. We have defined a novel regulatory mechanism by which n-3 PUFA inhibit induction of SREBP-1c by insulin. These findings identify AMPK as an important negative regulator of hepatic lipid synthesis and as a potential therapeutic target for hyperlipidemia in obesity and T2DM.


Assuntos
Proteínas Quinases Ativadas por AMP/metabolismo , Ácidos Docosa-Hexaenoicos/farmacologia , Hiperlipidemias/metabolismo , Fígado/metabolismo , Obesidade/metabolismo , Proteólise/efeitos dos fármacos , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Proteínas Quinases Ativadas por AMP/genética , Animais , Linhagem Celular Tumoral , Hiperlipidemias/tratamento farmacológico , Hiperlipidemias/genética , Hiperlipidemias/patologia , Insulina/genética , Insulina/metabolismo , Fígado/patologia , Alvo Mecanístico do Complexo 1 de Rapamicina , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Obesidade/dietoterapia , Obesidade/genética , Obesidade/patologia , Fosforilação/efeitos dos fármacos , Fosforilação/genética , Ratos , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Serina-Treonina Quinases TOR/genética , Serina-Treonina Quinases TOR/metabolismo
7.
Anal Biochem ; 474: 25-7, 2015 Apr 01.
Artigo em Inglês | MEDLINE | ID: mdl-25579785

RESUMO

We combined the TRIzol method of nucleic acid extraction with QIAamp columns to achieve coextraction of RNA and genomic DNA from peripheral blood mononuclear cells (PBMCs) and biopsied skeletal muscle, both stored at -80 °C for many months. Total RNA was recovered from the upper aqueous phase of TRIzol. The interphase and organic phases were precipitated with ethanol, digested with proteinase K, and filtered through QIAamp MinElute columns to recover DNA. The combined protocol yielded excellent quality and quantity of nucleic acids from archived human PBMCs and muscle and may be easily adapted for other tissues.


Assuntos
DNA/sangue , DNA/isolamento & purificação , Genoma Humano , Músculos/metabolismo , RNA/sangue , RNA/isolamento & purificação , Bancos de Tecidos , Humanos , Leucócitos Mononucleares/metabolismo
8.
Biochem Biophys Res Commun ; 449(4): 449-54, 2014 Jul 11.
Artigo em Inglês | MEDLINE | ID: mdl-24853806

RESUMO

The counter-regulatory hormone glucagon inhibits lipogenesis via downregulation of sterol regulatory element binding protein 1 (SREBP-1). The effect of glucagon is mediated via protein kinase A (PKA). To determine if SREBP-1 is a direct phosphorylation target of PKA, we conducted mass spectrometry analysis of recombinant n-terminal SREBP-1a following PKA treatment in vitro. This analysis identified serines 331/332 as bona-fide phosphorylation targets of PKA. To determine the functional consequences of phosphorylation at these sites, we constructed mammalian expression vector for both nSREBP-1a and 1c isoforms in which the candidate PKA phosphorylation sites were mutated to active phosphomimetic or non-phosphorylatable amino acids. The transcriptional activity of SREBP was reduced by the phosphomimetic mutation of S332 of nSREBP-1a and the corresponding serine (S308) of nSREBP-1c. This site is a strong candidate for mediating the negative regulatory effect of glucagon on SREBP-1 and lipogenesis.


Assuntos
Proteínas Quinases Dependentes de AMP Cíclico/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Ativação Transcricional , Animais , Glucagon/farmacologia , Células HEK293 , Humanos , Espectrometria de Massas , Fosforilação , Alinhamento de Sequência , Proteína de Ligação a Elemento Regulador de Esterol 1/genética
9.
J Biol Chem ; 288(23): 16321-16333, 2013 Jun 07.
Artigo em Inglês | MEDLINE | ID: mdl-23629656

RESUMO

Secretory phospholipase A2 group IIa (PLA2g2a) is associated with inflammation, hyperlipidemia, and atherogenesis. Transcription of the PLA2g2a gene is induced by multiple cytokines. Here, we report the surprising observation that thyroid hormone (T3) inhibited PLA2g2a gene expression in human and rat hepatocytes as well as in rat liver. Moreover, T3 reduced the cytokine-mediated induction of PLA2g2a, suggesting that the thyroid status may modulate aspects of the inflammatory response. In an effort to dissect the mechanism of repression by T3, we cloned the PLA2g2a gene and identified a negative T3 response element in the promoter. This T3 receptor (TRß)-binding site differed considerably from consensus T3 stimulatory elements. Using in vitro and in vivo binding assays, we found that TRß bound directly to the PLA2g2a promoter as a heterodimer with the retinoid X receptor. Knockdown of nuclear corepressor or silencing mediator for retinoid and thyroid receptors by siRNA blocked the T3 inhibition of PLA2g2a. Using chromatin immunoprecipitation assays, we showed that nuclear corepressor and silencing mediator for retinoid and thyroid receptors were associated with the PLA2g2a gene in the presence of T3. In contrast with the established role of T3 to promote coactivator association with TRß, our experiments demonstrate a novel inverse recruitment mechanism in which liganded TRß recruits corepressors to inhibit PLA2g2a expression.


Assuntos
Regulação Enzimológica da Expressão Gênica/fisiologia , Fosfolipases A2 do Grupo II/biossíntese , Hepatócitos/metabolismo , Proteínas Repressoras/metabolismo , Elementos de Resposta/fisiologia , Receptores beta dos Hormônios Tireóideos/metabolismo , Transcrição Gênica/fisiologia , Tri-Iodotironina/metabolismo , Animais , Fosfolipases A2 do Grupo II/genética , Células Hep G2 , Hepatócitos/citologia , Humanos , Fígado/citologia , Fígado/metabolismo , Masculino , Ratos , Ratos Sprague-Dawley , Proteínas Repressoras/genética , Receptores X de Retinoides/genética , Receptores X de Retinoides/metabolismo , Receptores beta dos Hormônios Tireóideos/genética , Tri-Iodotironina/genética
10.
J Biol Chem ; 288(2): 807-18, 2013 Jan 11.
Artigo em Inglês | MEDLINE | ID: mdl-23209300

RESUMO

Sirtuin 1 (SIRT1) is a nuclear deacetylase that modulates lipid metabolism and enhances mitochondrial activity. SIRT1 targets multiple transcription factors and coactivators. Thyroid hormone (T(3)) stimulates the expression of hepatic genes involved in mitochondrial fatty acid oxidation and gluconeogenesis. We reported that T(3) induces genes for carnitine palmitoyltransferase (cpt1a), pyruvate dehydrogenase kinase 4 (pdk4), and phosphoenolpyruvate carboxykinase (pepck). SIRT1 increases the expression of these genes via the activation of several factors, including peroxisome proliferator-activated receptor α, estrogen-related receptor α, and peroxisome proliferator-activated receptor γ coactivator (PGC-1α). Previously, we reported that PGC-1α participates in the T(3) induction of cpt1a and pdk4 in the liver. Given the overlapping targets of T(3) and SIRT1, we investigated whether SIRT1 participated in the T(3) regulation of these genes. Resveratrol is a small phenolic compound whose actions include the activation of SIRT1. Addition of resveratrol increased the T(3) induction of the pdk4 and cpt1a genes in hepatocytes. Furthermore, expression of SIRT1 in hepatocytes mimicked resveratrol in the regulation of gene expression by T(3). The deacetylase activity of SIRT1 was required and PGC-1α was deacetylated following addition of T(3). We found that SIRT1 interacted directly with T(3) receptor (TRß). Knockdown of SIRT1 decreased the T(3) induction of cpt1a and pdk4 and reduced the T(3) inhibition of sterol response element binding protein (srebp-1c) both in isolated hepatocytes and in rat liver. Our results indicate that SIRT1 contributes to the T(3) regulation of hepatic genes.


Assuntos
Regulação da Expressão Gênica/fisiologia , Fígado/metabolismo , Sirtuína 1/fisiologia , Tri-Iodotironina/fisiologia , Sequência de Bases , Linhagem Celular , Primers do DNA , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Reação em Cadeia da Polimerase em Tempo Real , Resveratrol , Sirtuína 1/genética , Estilbenos/farmacologia
11.
J Biol Chem ; 287(24): 20132-43, 2012 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-22511764

RESUMO

Induction of lipogenesis in response to insulin is critically dependent on the transcription factor, sterol regulatory element-binding protein-1c (SREBP-1c). FoxO1, a forkhead box class-O transcription factor, is an important mediator of insulin action, but its role in the regulation of lipid metabolism has not been clearly defined. We examined the effects of FoxO1 on srebp1 gene expression in vivo and in vitro. In vivo studies showed that constitutively active (CA) FoxO1 (CA-FoxO1) reduced basal expression of SREBP-1c mRNA in liver by ∼60% and blunted induction of SREBP-1c in response to feeding. In liver-specific FoxO knock-out mice, SREBP-1c expression was increased ∼2-fold. Similarly, in primary hepatocytes, CA-FoxO1 suppressed SREBP1-c expression and inhibited basal and insulin-induced SREBP-1c promoter activity. SREBP-1c gene expression is induced by the liver X receptor (LXR), but CA-FoxO1 did not block the activation of SREBP-1c by the LXR agonist TO9. Insulin stimulates SREBP-1c transcription through Sp1 and via "feed forward" regulation by newly synthesized SREBP-1c. CA-FoxO1 inhibited SREBP-1c by reducing the transactivational capacity of both Sp1 and SREBP-1c. In addition, chromatin immunoprecipitation assays indicate that FoxO1 can associate with the proximal promoter region of the srebp1 gene and disrupt the assembly of key components of the transcriptional complex of the SREBP-1c promoter. We conclude that FoxO1 inhibits SREBP-1c transcription via combined actions on multiple transcription factors and that this effect is exerted at least in part through reduced transcriptional activity of Sp1 and SREBP-1c and disrupted assembly of the transcriptional initiation complex on the SREBP-1c promoter.


Assuntos
Fatores de Transcrição Forkhead/metabolismo , Regulação da Expressão Gênica/fisiologia , Fígado/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Fator de Transcrição Sp1/metabolismo , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Animais , Células Cultivadas , Proteína Forkhead Box O1 , Fatores de Transcrição Forkhead/genética , Hepatócitos/metabolismo , Insulina/genética , Insulina/metabolismo , Receptores X do Fígado , Camundongos , Camundongos Knockout , Proteínas do Tecido Nervoso/genética , Receptores Nucleares Órfãos/genética , Receptores Nucleares Órfãos/metabolismo , Ratos , Elementos de Resposta/fisiologia , Fator de Transcrição Sp1/genética , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Transcrição Gênica/fisiologia
12.
Diabetes Care ; 35(5): 1008-14, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22432114

RESUMO

OBJECTIVE: To assess the reversibility of the elevation of serum creatinine levels in patients with diabetes after 5 years of continuous on-trial fenofibrate therapy. RESEARCH DESIGN AND METHODS: An on-drug/off-drug ancillary study to the Action to Control Cardiovascular Risk in Diabetes (ACCORD) Lipid Trial to investigate posttrial changes in serum creatinine and cystatin C. Eligible participants were recruited into a prospective, nested, three-group study based on retrospective on-trial serum creatinine levels: fenofibrate case subjects (n = 321, ≥ 20% increase after 3 months of therapy); fenofibrate control subjects (n = 175, ≤ 2% increase); and placebo control subjects (n = 565). Serum creatinine and cystatin C were measured at trial end and 6-8 weeks after discontinuation of trial therapy. RESULTS At trial end, case subjects had the highest adjusted serum creatinine (± SE) mg/dL (1.11 ± 0.02) and the lowest adjusted estimated glomerular filtration rate (eGFR) (± SE) mL/min/1.73 m(2) (68.4 ± 1.0) versus control subjects (1.01 ± 0.02; 74.8 ± 1.3) and placebo subjects (0.98 ± 0.01; 77.8 ± 0.7). After 51 days off-drug, serum creatinine in case subjects was still higher (0.97 ± 0.02) and eGFR still lower (77.8 ± 1.0) than control subjects (0.90 ± 0.02; 81.8 ± 1.3) but not different from placebo subjects (0.99 ± 0.01; 76.6 ± 0.7). Changes in serum cystatin C recapitulated the serum creatinine changes. CONCLUSIONS: Participants with significant initial on-trial increases in serum creatinine (≥ 20%) returned to the same level of renal function as participants receiving placebo while participants who had ≤ 2% increase in serum creatinine had net preservation of renal function compared with the same unselected placebo reference group. The fenofibrate-associated on-trial increases in serum creatinine were reversible, and the reversal was complete after 51 days off-drug. The similarity of the cystatin C results suggests that the mechanism of this change is not specific for serum creatinine.


Assuntos
Fenofibrato/efeitos adversos , Fenofibrato/uso terapêutico , Hipolipemiantes/efeitos adversos , Insuficiência Renal/induzido quimicamente , Idoso , Creatinina/sangue , Taxa de Filtração Glomerular/efeitos dos fármacos , Humanos , Hipolipemiantes/uso terapêutico , Pessoa de Meia-Idade , Insuficiência Renal/sangue
13.
J Biol Chem ; 286(27): 23799-807, 2011 Jul 08.
Artigo em Inglês | MEDLINE | ID: mdl-21586575

RESUMO

The conversion of pyruvate to acetyl-CoA in mitochondria is catalyzed by the pyruvate dehydrogenase complex (PDC). Activity of PDC is inhibited by phosphorylation via the pyruvate dehydrogenase kinases (PDKs). Here, we examined the regulation of Pdk4 gene expression by the CCAAT/enhancer-binding protein ß (C/EBPß). C/EBPß modulates the expression of multiple hepatic genes including those involved in metabolism, development, and inflammation. We found that C/EBPß induced Pdk4 gene expression and decreased PDC activity. This transcriptional induction was mediated through two C/EBPß binding sites in the Pdk4 promoter. C/EBPß participates in the hormonal regulation of gluconeogenic genes. Previously, we reported that Pdk4 was induced by thyroid hormone (T(3)). Therefore, we investigated the role of C/EBPß in the T(3) regulation of Pdk4. T(3) increased C/EBPß abundance in primary rat hepatocytes. Knockdown of C/EBPß with siRNA diminished the T(3) induction of the Pdk4 and carnitine palmitoyltransferase (Cpt1a) genes. CPT1a is an initiating step in the mitochondrial oxidation of long chain fatty acids. Our results indicate that C/EBPß stimulates Pdk4 expression and participates in the T(3) induction of the Cpt1a and Pdk4 genes.


Assuntos
Proteína beta Intensificadora de Ligação a CCAAT/metabolismo , Regulação Enzimológica da Expressão Gênica/fisiologia , Hepatócitos/metabolismo , Proteínas Serina-Treonina Quinases/biossíntese , Elementos de Resposta/fisiologia , Animais , Proteína beta Intensificadora de Ligação a CCAAT/genética , Carnitina O-Palmitoiltransferase/biossíntese , Carnitina O-Palmitoiltransferase/genética , Gluconeogênese/fisiologia , Células Hep G2 , Hepatócitos/citologia , Humanos , Proteínas Serina-Treonina Quinases/genética , Piruvato Desidrogenase Quinase de Transferência de Acetil , Complexo Piruvato Desidrogenase/biossíntese , Complexo Piruvato Desidrogenase/genética , Ratos , Tri-Iodotironina/genética , Tri-Iodotironina/metabolismo
14.
J Biol Chem ; 285(4): 2375-85, 2010 Jan 22.
Artigo em Inglês | MEDLINE | ID: mdl-19948729

RESUMO

PDK4 (pyruvate dehydrogenase kinase 4) regulates pyruvate oxidation through the phosphorylation and inhibition of the pyruvate dehydrogenase complex (PDC). PDC catalyzes the conversion of pyruvate to acetyl-CoA and is an important control point in glucose and pyruvate metabolism. PDK4 gene expression is stimulated by thyroid hormone (T(3)), glucocorticoids, and long chain fatty acids. The effects of T(3) on gene expression in the liver are mediated via the thyroid hormone receptor. Here, we have identified two binding sites for thyroid hormone receptor beta in the promoter of the rat PDK4 (rPDK4) gene. In addition, we have investigated the role of transcriptional coactivators and found that the PGC-1 alpha (peroxisome proliferator-activated receptor gamma coactivator) enhances the T(3) induction of rPDK4. Following T(3) administration, there is an increase in the association of PGC-1 alpha with the rPDK4 promoter. Interestingly, this increased association is with the proximal rPDK4 promoter rather than the distal region of the gene that contains the T(3) response elements. Administration of T(3) to hypothyroid rats elevated the abundance of PGC-1 alpha mRNA and protein in the liver. In addition, we observed greater association of PGC-1 alpha not only with the rPDK4 gene but also with phosphoenolpyruvate carboxykinase and CPT-1a (carnitine palmitoyltransferase 1a) genes. Knockdown of PGC-1 alpha in rat hepatocytes reduced the T(3) induction of PDK4, PEPCK, and CPT-1a genes. Our results indicate that T(3) regulates PGC-1 alpha abundance and association with hepatic genes, and in turn PGC-1 alpha is an important participant in the T(3) induction of selected genes.


Assuntos
Hepatócitos/enzimologia , Hipertireoidismo/fisiopatologia , Hipotireoidismo/fisiopatologia , Proteínas Serina-Treonina Quinases/metabolismo , Proteínas de Ligação a RNA/metabolismo , Fatores de Transcrição/metabolismo , Tri-Iodotironina/metabolismo , Animais , Sequência de Bases , Carcinoma Hepatocelular , Linhagem Celular Tumoral , Hepatócitos/citologia , Humanos , Hipertireoidismo/metabolismo , Hipofisectomia , Hipotireoidismo/metabolismo , Neoplasias Hepáticas , Masculino , Dados de Sequência Molecular , PPAR gama/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Regiões Promotoras Genéticas/fisiologia , Proteínas Serina-Treonina Quinases/genética , Piruvato Desidrogenase Quinase de Transferência de Acetil , Complexo Piruvato Desidrogenase/metabolismo , RNA Mensageiro/metabolismo , Proteínas de Ligação a RNA/genética , Ratos , Ratos Sprague-Dawley , Receptores beta dos Hormônios Tireóideos/metabolismo , Fatores de Transcrição/genética , Transcrição Gênica/fisiologia , Transfecção
15.
Metabolism ; 59(4): 587-98, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-19913854

RESUMO

We compared hepatic expression of genes that regulate lipid biosynthesis and metabolic signaling in liver biopsy specimens from women who were undergoing gastric bypass surgery (GBP) for morbid obesity with that in women undergoing ventral hernia repair who had experienced massive weight loss (MWL) after prior GBP. Comprehensive metabolic profiles of morbidly obese (MO) (22 subjects) and MWL (9 subjects) were also compared. Analyses of gene expression in liver biopsies from MO and MWL were accomplished by Affymetrix microarray, real-time polymerase chain reaction, and Western blotting techniques. After GBP, MWL subjects had lost on average 102 lb as compared with MO subjects. This was accompanied by effective reversal of the dyslipidemia and insulin resistance that were present in MO. As compared with MWL, livers of MO subjects exhibited increased expression of sterol regulatory element binding protein (SREBP)-1c and its downstream lipogenic targets, fatty acid synthase and acetyl-coenzyme A-carboxylase-1. Livers of MO subjects also exhibited enhanced expression of suppressor of cytokine signaling-3 protein and attenuated Janus kinase signal transducer and activator of transcription (JAK/STAT) signaling. Consistent with these findings, we found that the human SREBP-1c promoter was positively regulated by insulin and negatively regulated by STAT3. These data support the hypothesis that suppressor of cytokine signaling-3-mediated attenuation of the STAT signaling pathway and resulting enhanced expression of SREBP-1c, a key regulator of de novo lipid biosynthesis, are mechanistically related to the development of hepatic insulin resistance and dyslipidemia in MO women.


Assuntos
Regulação da Expressão Gênica , Fígado/metabolismo , Obesidade Mórbida/metabolismo , Fator de Transcrição STAT3/fisiologia , Proteína de Ligação a Elemento Regulador de Esterol 1/genética , Proteínas Supressoras da Sinalização de Citocina/fisiologia , Adulto , Ácidos Graxos/metabolismo , Feminino , Derivação Gástrica , Humanos , Hidrocarbonetos Fluorados/farmacologia , Insulina/farmacologia , Resistência à Insulina , Lipoproteínas VLDL/biossíntese , Reação em Cadeia da Polimerase , Regiões Promotoras Genéticas , Fator de Transcrição STAT1/fisiologia , Estearoil-CoA Dessaturase/fisiologia , Sulfonamidas/farmacologia , Proteína 3 Supressora da Sinalização de Citocinas , Triglicerídeos/biossíntese , Redução de Peso
16.
Mol Cell Endocrinol ; 315(1-2): 159-67, 2010 Feb 05.
Artigo em Inglês | MEDLINE | ID: mdl-19703515

RESUMO

The pyruvate dehydrogenase complex (PDC) catalyzes the conversion of pyruvate to acetyl-CoA in mitochondria and is a key regulatory enzyme in the oxidation of glucose to acetyl-CoA. Phosphorylation of PDC by the pyruvate dehydrogenase kinases (PDK) inhibits its activity. The expression of the pyruvate dehydrogenase kinase 4 (PDK4) gene is increased in fasting and other conditions associated with the switch from the utilization of glucose to fatty acids as an energy source. Transcription of the PDK4 gene is elevated by glucocorticoids and inhibited by insulin. In this study, we have investigated the factors involved in the regulation of the PDK4 gene by these hormones. Glucocorticoids stimulate PDK4 through two glucocorticoid receptor (GR) binding sites located more than 6000 base pairs upstream of the transcriptional start site. Insulin inhibits the glucocorticoid induction in part by causing dissociation of the GR from the promoter. Previously, we found that the estrogen related receptor alpha (ERRalpha) stimulates the expression of PDK4. Here, we determined that one of the ERRalpha binding sites contributes to the insulin inhibition of PDK4. A binding site for the forkhead transcription factor (FoxO1) is adjacent to the ERRalpha binding sites. FoxO1 participates in the glucocorticoid induction of PDK4 and the regulation of this gene by insulin. Our data demonstrate that glucocorticoids and insulin each modulate PDK4 gene expression through complex hormone response units that contain multiple factors.


Assuntos
Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/farmacologia , Insulina/farmacologia , Isoenzimas/metabolismo , Proteínas Quinases/metabolismo , Animais , Células Cultivadas , Fatores de Transcrição Forkhead/genética , Fatores de Transcrição Forkhead/metabolismo , Genes Reporter , Glucocorticoides/metabolismo , Humanos , Insulina/metabolismo , Isoenzimas/genética , Mutagênese Sítio-Dirigida , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , Coativador 1-alfa do Receptor gama Ativado por Proliferador de Peroxissomo , Proteínas Quinases/genética , Proteínas de Ligação a RNA/genética , Proteínas de Ligação a RNA/metabolismo , Ratos , Receptores de Estrogênio/genética , Receptores de Estrogênio/metabolismo , Receptores de Glucocorticoides/metabolismo , Elementos de Resposta , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Receptor ERRalfa Relacionado ao Estrogênio
17.
Obesity (Silver Spring) ; 17(8): 1563-73, 2009 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-19265796

RESUMO

The objective of this study was to determine the molecular bases of disordered hepatic function and disease susceptibility in obesity. We compared global gene expression in liver biopsies from morbidly obese (MO) women undergoing gastric bypass (GBP) surgery with that of women undergoing ventral hernia repair who had experienced massive weight loss (MWL) following prior GBP. Metabolic and hormonal profiles were examined in MO vs. MWL groups. Additionally, we analyzed individual profiles of hepatic gene expression in liver biopsy specimens obtained from MO and MWL subjects. All patients underwent preoperative metabolic profiling. RNAs were extracted from wedge biopsies of livers from MO and MWL subjects, and analysis of mRNA expression was carried out using Affymetrix HG-U133A microarray gene chips. Genes exhibiting greater than twofold differential expression between MO and MWL subjects were organized according to gene ontology and hierarchical clustering, and expression of key genes exhibiting differential regulation was quantified by real-time-polymerase chain reaction (RT-PCR). We discovered 154 genes to be differentially expressed in livers of MWL and MO subjects. A total of 28 candidate disease susceptibility genes were identified that encoded proteins regulating lipid and energy homeostasis (PLIN, ENO3, ELOVL2, APOF, LEPR, IGFBP1, DDIT4), signal transduction (MAP2K6, SOCS-2), postinflammatory tissue repair (HLA-DQB1, SPP1, P4HA1, LUM), bile acid transport (SULT2A, ABCB11), and metabolism of xenobiotics (GSTT2, CYP1A1). Using gene expression profiling, we have identified novel candidate disease susceptibility genes whose expression is altered in livers of MO subjects. The significance of altered expression of these genes to obesity-related disease is discussed.


Assuntos
Regulação da Expressão Gênica , Predisposição Genética para Doença , Fígado/metabolismo , Obesidade Mórbida/genética , Adulto , Biópsia , Proliferação de Células , Feminino , Perfilação da Expressão Gênica , Humanos , Inflamação , Metabolismo dos Lipídeos , Fígado/patologia , Obesidade Mórbida/patologia , Análise de Sequência com Séries de Oligonucleotídeos , RNA/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
18.
J Biol Chem ; 284(12): 7518-32, 2009 Mar 20.
Artigo em Inglês | MEDLINE | ID: mdl-19158095

RESUMO

The regulation of lipid homeostasis by insulin is mediated in part by the enhanced transcription of the gene encoding SREBP-1c (sterol regulatory element-binding protein-1c). Nascent SREBP-1c is synthesized and embedded in the endoplasmic reticulum (ER) and must be transported to the Golgi in coatomer protein II (COPII) vesicles where two sequential cleavages generate the transcriptionally active NH(2)-terminal fragment, nSREBP-1c. There is limited indirect evidence to suggest that insulin may also regulate the posttranslational processing of the nascent SREBP-1c protein. Therefore, we designed experiments to directly assess the action of insulin on the post-translational processing of epitope-tagged full-length SREBP-1c and SREBP-2 proteins expressed in cultured hepatocytes. We demonstrate that insulin treatment led to enhanced post-translational processing of SREBP-1c, which was associated with phosphorylation of ER-bound nascent SREBP-1c protein that increased affinity of the SREBP-1c cleavage-activating protein (SCAP)-SREBP-1c complex for the Sec23/24 proteins of the COPII vesicles. Furthermore, chemical and molecular inhibitors of the phosphoinositide 3-kinase pathway and its downstream kinase protein kinase B (PKB)/Akt prevented both insulin-mediated phosphorylation of nascent SREBP-1c protein and its posttranslational processing. Insulin had no effect on the proteolysis of nascent SREBP-2 under identical conditions. We also show that in vitro incubation of an active PKB/Akt enzyme with recombinant full-length SREBP-1c led to its phosphorylation. Thus, insulin selectively stimulates the processing of SREBP-1c in rat hepatocytes by enhancing the association between the SCAP-SREBP-1c complex and COPII proteins and subsequent ER to Golgi transport and proteolytic cleavage. This effect of insulin is tightly linked to phosphoinositide 3-kinase and PKB/Akt-dependent serine phosphorylation of the precursor SREBP-1c protein.


Assuntos
Vesículas Revestidas pelo Complexo de Proteína do Envoltório/metabolismo , Hepatócitos/metabolismo , Hipoglicemiantes/farmacologia , Insulina/farmacologia , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteína de Ligação a Elemento Regulador de Esterol 1/metabolismo , Animais , Vesículas Revestidas pelo Complexo de Proteína do Envoltório/genética , Linhagem Celular , Retículo Endoplasmático/genética , Retículo Endoplasmático/metabolismo , Complexo de Golgi/genética , Complexo de Golgi/metabolismo , Hipoglicemiantes/metabolismo , Insulina/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Fosfatidilinositol 3-Quinases/genética , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação/efeitos dos fármacos , Fosforilação/fisiologia , Processamento de Proteína Pós-Traducional/fisiologia , Transporte Proteico/efeitos dos fármacos , Transporte Proteico/fisiologia , Proteínas Proto-Oncogênicas c-akt/genética , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Sprague-Dawley , Proteína de Ligação a Elemento Regulador de Esterol 1/genética
19.
Trends Endocrinol Metab ; 19(2): 65-73, 2008 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-18291668

RESUMO

The uptake, biosynthesis and metabolism of cholesterol and other lipids are exquisitely regulated by feedback and feed-forward pathways in organisms ranging from Caenorhabditis elegans to humans. As endoplasmic reticulum (ER) membrane-embedded transcription factors that are activated in the Golgi apparatus, sterol regulatory element-binding proteins (SREBPs) are central to the intracellular surveillance of lipid catabolism and de novo biogenesis. The biosynthesis of SREBP proteins, their migration from the ER to the Golgi compartment, intra-membrane proteolysis, nuclear translocation and trans-activation potential are tightly controlled in vivo. Here we summarize recent studies elucidating the transcriptional and post-transcriptional regulation of SREBP-1c through nutrition and the action of hormones, particularly insulin, and the resulting implications for dyslipidemia of obesity, metabolic syndrome and type 2 diabetes.


Assuntos
Transtornos do Metabolismo dos Lipídeos/genética , Metabolismo dos Lipídeos/genética , Proteínas de Ligação a Elemento Regulador de Esterol/fisiologia , Animais , Sequência de Bases , AMP Cíclico/farmacologia , Retículo Endoplasmático/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Glucagon/farmacologia , Complexo de Golgi/metabolismo , Homeostase/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/fisiologia , Lipídeos/farmacologia , Proteínas de Membrana/fisiologia , Modelos Biológicos , Dados de Sequência Molecular , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Processamento de Proteína Pós-Traducional/fisiologia , Transporte Proteico/efeitos dos fármacos , Homologia de Sequência do Ácido Nucleico , Proteínas de Ligação a Elemento Regulador de Esterol/biossíntese , Proteínas de Ligação a Elemento Regulador de Esterol/genética , Proteínas de Ligação a Elemento Regulador de Esterol/metabolismo
20.
Am J Med Sci ; 333(3): 168-72, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17496735

RESUMO

BACKGROUND: Marine fish oil supplements are frequently administered with other lipid medications for treatment of hypertriglyceridemia. The efficacy of fish oil may be reduced in the presence of other lipid agents, particularly fibrates that also act as PPARalpha agonists. We therefore sought to determine the efficacy of fish-oil supplements when coadministered with other lipid-modifying agents. METHODS: Patients receiving fish oil supplements were identified from the computer database of a large governmental HMO. Change in plasma lipoprotein levels after administration of fish oil was compared between patients receiving fish oil as their only treatment and those for whom fish oil was added to other drugs. RESULTS: A total of 166 evaluable records were identified, 66 from patients treated with fish oil alone and 100 from patients for whom fish oil was added to another agent or other agents. Fish oil effectively reduced triglyceride levels to an equal extent in the fish oil only and fish oil added groups (-30% versus -27% respectively; P = 0.84). CONCLUSION: Fish oil effectively reduces plasma triglyceride levels when administered with concomitant lipid medications. These findings suggest the presence of additional and even complementary mechanisms of action of fish oil to lower triglyceride when added to other lipid drugs. These findings validate the common clinical practice of combining fish oil supplements with other lipid-lowering medications in patients with hypertriglyceridemia.


Assuntos
Óleos de Peixe/uso terapêutico , Hipertrigliceridemia/tratamento farmacológico , Hipolipemiantes/uso terapêutico , Lipoproteínas/sangue , Análise de Variância , Quimioterapia Combinada , Óleos de Peixe/farmacologia , Humanos , Hipolipemiantes/farmacologia , Masculino , Pessoa de Meia-Idade , Estudos Retrospectivos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA