Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
FASEB J ; 35(10): e21881, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34478587

RESUMEN

Group IIA secreted phospholipase A2 (PLA2G2A) hydrolyzes glycerophospholipids at the sn-2 position resulting in the release of fatty acids and lysophospholipids. C57BL/6 mice do not express Pla2g2a due to a frameshift mutation (wild-type [WT] mice). We previously reported that transgenic expression of human PLA2G2A in C57BL/6 mice (IIA+ mice) protects against weight gain and insulin resistance, in part by increasing total energy expenditure. Additionally, we found that brown and white adipocytes from IIA+ mice have increased expression of mitochondrial uncoupling markers, such as uncoupling protein 1 (UCP1), peroxisome proliferator-activated receptor-gamma coactivator, and PR domain containing 16, suggesting that the energy expenditure phenotype might be due to an increased thermogenic capacity in adipose tissue. Here, we further characterize the impact of PLA2G2A on thermogenic mechanisms in adipose tissue. Metabolic analysis of WT and IIA+ mice revealed that even when housed within their thermoneutral zone, IIA+ mice have elevated energy expenditure compared to WT littermates. Increased energy expenditure in IIA+ mice is associated with increased citrate synthase activity in brown adipose tissue (BAT) and increased mitochondrial respiration in both brown and white adipocytes. We also observed that direct addition of recombinant PLA2G2A enzyme to in vitro cultured adipocytes results in the marked induction of UCP1 protein expression. Finally, we report that PLA2G2A induces the expression of numerous transcripts related to energy substrate transport and metabolism in BAT, suggestive of an increase in substrate flux to fuel BAT activity. These data demonstrate that PLA2G2A enhances adipose tissue thermogenesis, in part, through elevated substrate delivery and increased mitochondrial content in BAT.


Asunto(s)
Tejido Adiposo Pardo/fisiopatología , Metabolismo Energético , Fosfolipasas A2 Grupo II/fisiología , Mitocondrias/patología , Termogénesis , Proteína Desacopladora 1/metabolismo , Tejido Adiposo Blanco/fisiopatología , Animales , Transporte Biológico , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mitocondrias/metabolismo
2.
FASEB J ; 33(1): 738-749, 2019 01.
Artículo en Inglés | MEDLINE | ID: mdl-30020829

RESUMEN

Secretory phospholipase A2 group IIA (PLA2G2A) is a phospholipase which has a role in inflammation, atherogenesis, and host defense. Previously, we found that PLA2G2A protects mice on high-fat diets from weight gain and insulin resistance. Here, we examined the regulation of PLA2G2A and the metabolic changes that occur in response to variations in thyroid status. In particular, the impact of PLA2G2A on the brown adipose tissue (BAT) thermogenic gene expression was explored. We induced hypothyroidism in C57BL/6 and PLA2G2A-overexpressing (IIA+) mice over a 10 wk period or treated them with thyroid hormone (T3) for 5 wk. There were no significant changes in PLA2G2A abundance in response to thyroid status. The energy expenditure of hypothyroid IIA+ mice did not increase; however, the energy expenditure, substrate utilization, insulin sensitivity, and glucose tolerance were all elevated in the IIA+ mice given T3. Moreover, white adipocytes from IIA+ mice were much more prone to "beiging," including increased expression of brown adipose thermogenic markers such as uncoupling protein 1 (UCP1), PR domain containing 16, and early B cell factor 2. Finally, the BAT of IIA+ mice had increased UCP1 and other proteins indicative of mitochondrial uncoupling and nonshivering adaptive thermogenesis. These data reveal a novel role for PLA2G2A on adipose tissue thermogenesis depending on thyroid status.-Kuefner, M. S., Deng, X., Stephenson, E. J., Pham, K., Park, E. A. Secretory phospholipase A2 group IIA enhances the metabolic rate and increases glucose utilization in response to thyroid hormone.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Metabolismo Energético/efectos de los fármacos , Glucosa/metabolismo , Fosfolipasas A2 Grupo II/metabolismo , Hipotiroidismo/tratamiento farmacológico , Triyodotironina/farmacología , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Blanco/efectos de los fármacos , Animales , Peso Corporal/efectos de los fármacos , Femenino , Fosfolipasas A2 Grupo II/genética , Hipotiroidismo/metabolismo , Hipotiroidismo/patología , Insulina/metabolismo , Resistencia a la Insulina , Masculino , Ratones , Ratones Endogámicos C57BL , Termogénesis
3.
J Lipid Res ; 58(9): 1822-1833, 2017 09.
Artículo en Inglés | MEDLINE | ID: mdl-28663239

RESUMEN

Secretory phospholipase A2 group IIA (PLA2G2A) is a member of a family of secretory phospholipases that have been implicated in inflammation, atherogenesis, and antibacterial actions. Here, we evaluated the role of PLA2G2A in the metabolic response to a high fat diet. C57BL/6 (BL/6) mice do not express PLA2g2a due to a frameshift mutation. We fed BL/6 mice expressing the human PLA2G2A gene (IIA+ mice) a fat diet and assessed the physiologic response. After 10 weeks on the high fat diet, the BL/6 mice were obese, but the IIA+ mice did not gain weight or accumulate lipid. The lean mass in chow- and high fat-fed IIA+ mice was constant and similar to the BL/6 mice on a chow diet. Surprisingly, the IIA+ mice had an elevated metabolic rate, which was not due to differences in physical activity. The IIA+ mice were more insulin sensitive and glucose tolerant than the BL/6 mice, even when the IIA+ mice were provided the high fat diet. The IIA+ mice had increased expression of uncoupling protein 1 (UCP1), sirtuin 1 (SIRT1), and PPARγ coactivator 1α (PGC-1α) in brown adipose tissue (BAT), suggesting that PLA2G2A activates mitochondrial uncoupling in BAT. Our data indicate that PLA2G2A has a previously undiscovered impact on insulin sensitivity and metabolism.


Asunto(s)
Fosfolipasas A2 Grupo II/metabolismo , Resistencia a la Insulina , Insulina/metabolismo , Animales , Peso Corporal , Metabolismo Energético , Femenino , Fosfolipasas A2 Grupo II/genética , Humanos , Hígado/metabolismo , Masculino , Ratones
4.
Am J Physiol Heart Circ Physiol ; 313(3): H479-H490, 2017 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-28687587

RESUMEN

Pyruvate dehydrogenase (PDH) is the rate-limiting enzyme for glucose oxidation and a critical regulator of metabolic flexibility during the fasting to feeding transition. PDH is regulated via both PDH kinases (PDHK) and PDH phosphatases, which phosphorylate/inactivate and dephosphorylate/activate PDH, respectively. Our goal was to determine whether the transcription factor forkhead box O1 (FoxO1) regulates PDH activity and glucose oxidation in the heart via increasing the expression of Pdk4, the gene encoding PDHK4. To address this question, we differentiated H9c2 myoblasts into cardiac myocytes and modulated FoxO1 activity, after which Pdk4/PDHK4 expression and PDH phosphorylation/activity were assessed. We assessed binding of FoxO1 to the Pdk4 promoter in cardiac myocytes in conjunction with measuring the role of FoxO1 on glucose oxidation in the isolated working heart. Both pharmacological (1 µM AS1842856) and genetic (siRNA mediated) inhibition of FoxO1 decreased Pdk4/PDHK4 expression and subsequent PDH phosphorylation in H9c2 cardiac myocytes, whereas 10 µM dexamethasone-induced Pdk4/PDHK4 expression was abolished via pretreatment with 1 µM AS1842856. Furthermore, transfection of H9c2 cardiac myocytes with a vector expressing FoxO1 increased luciferase activity driven by a Pdk4 promoter construct containing the FoxO1 DNA-binding element region, but not in a Pdk4 promoter construct lacking this region. Finally, AS1842856 treatment in fasted mice enhanced glucose oxidation rates during aerobic isolated working heart perfusions. Taken together, FoxO1 directly regulates Pdk4 transcription in the heart, thereby controlling PDH activity and subsequent glucose oxidation rates.NEW & NOTEWORTHY Although studies have shown an association between FoxO1 activity and pyruvate dehydrogenase kinase 4 expression, our study demonstrated that pyruvate dehydrogenase kinase 4 is a direct transcriptional target of FoxO1 (but not FoxO3/FoxO4) in the heart. Furthermore, we report here, for the first time, that FoxO1 inhibition increases glucose oxidation in the isolated working mouse heart.


Asunto(s)
Metabolismo Energético , Proteína Forkhead Box O1/metabolismo , Regulación Enzimológica de la Expresión Génica , Glucosa/metabolismo , Miocitos Cardíacos/enzimología , Proteínas Serina-Treonina Quinasas/metabolismo , Transcripción Genética , Angiotensina II/toxicidad , Animales , Apoptosis/efectos de los fármacos , Sitios de Unión , Línea Celular , Dexametasona/farmacología , Metabolismo Energético/efectos de los fármacos , Femenino , Proteína Forkhead Box O1/antagonistas & inhibidores , Proteína Forkhead Box O1/genética , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Cinética , Ratones Endogámicos C57BL , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/patología , Oxidación-Reducción , Fosforilación , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Serina-Treonina Quinasas/genética , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Quinolonas/farmacología , Interferencia de ARN , Transducción de Señal , Transcripción Genética/efectos de los fármacos , Transfección
5.
Biochim Biophys Acta Gen Subj ; 1861(2): 307-312, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27845231

RESUMEN

BACKGROUND: Diabetic cardiomyopathy develops in insulin-dependent diabetic patients who have no hypertension, cardiac hypertrophy or vascular disease. Diabetes increases cardiac fatty acid oxidation, but cardiac hypertrophy limits fatty acid oxidation. Here we examined effects of diabetes on gene expression in rat hearts. METHODS: We used oligonucleotide microarrays to examine effects of insulindependent diabetes in the rat heart. RTQ PCR confirmed results of microarrays. Specific antibodies were used to examine changes in the mitochondrial 3-hydroxy-3-methylglutaryl-CoA synthase 2 (HMGCS2). RESULTS: A surprising result of diabetes was increased mRNA encoding all enzymes of the ketone body synthesis pathway. Increased mRNA expression for these enzymes was confirmed by RTQ PCR. The mRNA encoding HMGCS2, the rate-controlling enzyme, was 27 times greater in diabetic hearts. Total HMGCS2 protein increased 8-fold in diabetic hearts, but no difference was found in HMGCS2 protein in control vs. diabetic liver. CONCLUSIONS: Insulin-dependent diabetes induced the enzymes of ketone body synthesis in the heart, including HMGCS2, as well as increasing enzymes of fatty acid oxidation. GENERAL SIGNIFICANCE: The mammalian heart does not export ketone bodies to other tissues, but rather is a major consumer of ketone bodies. Induction of HMGCS2, which is normally expressed only in the fetal and newborn heart, may indicate an adaptation by the heart to combat "metabolic inflexibility" by shifting the flux of excess intramitochondrial acetyl-CoA derived from elevated fatty acid oxidation into ketone bodies, liberating free CoA to balance the acetyl-CoA/CoA ratio in favor of increased glucose oxidation through the pyruvate dehydrogenase complex.


Asunto(s)
Acilcoenzima A/genética , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/genética , Hidroximetilglutaril-CoA Sintasa/genética , Miocardio/metabolismo , ARN Mensajero/genética , Estreptozocina/farmacología , Animales , Cardiomiopatías Diabéticas/genética , Cardiomiopatías Diabéticas/metabolismo , Ácidos Grasos/metabolismo , Expresión Génica/genética , Corazón/fisiopatología , Insulina/metabolismo , Cuerpos Cetónicos/metabolismo , Mitocondrias/metabolismo , Oxidación-Reducción , Ratas , Ratas Sprague-Dawley
6.
J Biol Chem ; 290(37): 22532-42, 2015 Sep 11.
Artículo en Inglés | MEDLINE | ID: mdl-26240147

RESUMEN

Insulin resistance and neuroinflammation have emerged as two likely key contributors in the pathogenesis of Alzheimer disease (AD), especially in those sporadic AD cases compromised by diabetes or cardiovascular disease. Amyloid-ß (Aß) deposition and its associated inflammatory response are hallmarks in sporadic AD brains. Elevated expression and activity of ß-secretase 1 (BACE1), the rate-limiting enzyme responsible for the ß-cleavage of amyloid precursor proteins to Aß peptides, are also observed in sporadic AD brains. Previous studies have suggested that there is therapeutic potential for retinoic acid in treating neurodegeneration based on decreased Aß. Here we discovered that BACE1 expression is elevated in the brains of both Tg2576 transgenic mice and mice on high fat diets. These conditions are associated with a neuroinflammatory response. We found that administration of all-trans-retinoic acid (atRA) down-regulated the expression of BACE1 in the brains of Tg2576 mice and in mice fed a high fat diet. Moreover, in LPS-treated mice and cultured neurons, BACE1 expression was repressed by the addition of atRA, correlating with the anti-inflammatory efficacy of atRA. Mutations of the NFκB binding site in BACE1 promoter abolished the suppressive effect of atRA. Furthermore, atRA disrupted LPS-induced nuclear translocation of NFκB and its binding to BACE1 promoter as well as promoting the recruitment of the corepressor NCoR. Our findings indicate that atRA represses BACE1 gene expression under inflammatory conditions via the modulation of NFκB signaling.


Asunto(s)
Enfermedad de Alzheimer/metabolismo , Secretasas de la Proteína Precursora del Amiloide/biosíntesis , Ácido Aspártico Endopeptidasas/biosíntesis , Encéfalo/metabolismo , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , FN-kappa B/metabolismo , Transducción de Señal/efectos de los fármacos , Tretinoina/farmacología , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/patología , Secretasas de la Proteína Precursora del Amiloide/genética , Animales , Ácido Aspártico Endopeptidasas/genética , Encéfalo/patología , Grasas de la Dieta/farmacología , Modelos Animales de Enfermedad , Ratones , Ratones Transgénicos , FN-kappa B/genética , Transducción de Señal/genética
7.
Biochim Biophys Acta ; 1851(12): 1521-9, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26327595

RESUMEN

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.


Asunto(s)
Proteínas Quinasas Activadas por AMP/metabolismo , Ácidos Docosahexaenoicos/farmacología , Hiperlipidemias/metabolismo , Hígado/metabolismo , Obesidad/metabolismo , Proteolisis/efectos de los fármacos , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Proteínas Quinasas Activadas por AMP/genética , Animales , Línea Celular Tumoral , Hiperlipidemias/tratamiento farmacológico , Hiperlipidemias/genética , Hiperlipidemias/patología , Insulina/genética , Insulina/metabolismo , Hígado/patología , Diana Mecanicista del Complejo 1 de la Rapamicina , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Obesidad/dietoterapia , Obesidad/genética , Obesidad/patología , Fosforilación/efectos de los fármacos , Fosforilación/genética , Ratas , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/metabolismo
8.
J Biol Chem ; 288(23): 16321-16333, 2013 Jun 07.
Artículo en Inglés | MEDLINE | ID: mdl-23629656

RESUMEN

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.


Asunto(s)
Regulación Enzimológica de la Expresión Génica/fisiología , Fosfolipasas A2 Grupo II/biosíntesis , Hepatocitos/metabolismo , Proteínas Represoras/metabolismo , Elementos de Respuesta/fisiología , Receptores beta de Hormona Tiroidea/metabolismo , Transcripción Genética/fisiología , Triyodotironina/metabolismo , Animales , Fosfolipasas A2 Grupo II/genética , Células Hep G2 , Hepatocitos/citología , Humanos , Hígado/citología , Hígado/metabolismo , Masculino , Ratas , Ratas Sprague-Dawley , Proteínas Represoras/genética , Receptores X Retinoide/genética , Receptores X Retinoide/metabolismo , Receptores beta de Hormona Tiroidea/genética , Triyodotironina/genética
9.
J Biol Chem ; 288(2): 807-18, 2013 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-23209300

RESUMEN

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.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Hígado/metabolismo , Sirtuina 1/fisiología , Triyodotironina/fisiología , Secuencia de Bases , Línea Celular , Cartilla de ADN , Regulación de la Expresión Génica/efectos de los fármacos , Técnicas de Silenciamiento del Gen , Humanos , Reacción en Cadena en Tiempo Real de la Polimerasa , Resveratrol , Sirtuina 1/genética , Estilbenos/farmacología
10.
Biochem Biophys Res Commun ; 444(1): 56-62, 2014 Jan 31.
Artículo en Inglés | MEDLINE | ID: mdl-24440706

RESUMEN

Thyroid hormone (T3) stimulates various metabolic pathways and the hepatic actions of T3 are mediated primarily through the thyroid hormone receptor beta (TRß). Hypothyroidism has been linked with low grade inflammation, elevated risk of hepatic steatosis and atherosclerosis. Secretory phospholipases (sPLA2) are associated with inflammation, hyperlipidemia and atherosclerosis. Due to potential linkage between thyroid hormone and sPLA2, we investigated the effect of thyroid hormone status on the regulation of secretory phospholipases in mice, rats and human liver. T3 suppressed the expression of the sPLA2 group IIa (PLA2g2a) gene in the liver of BALB/c mice and C57BL/6 transgenic mice expressing the human PLA2g2a. PLA2g2a was elevated with hypothyroidism and high fat diets which may contribute to the low grade inflammation associated with hypothyroidism and diet induced obesity. We also examined the effects of the TRß agonist eprotirome on hepatic gene regulation. We observed that eprotirome inhibited the expression of selected sPLA2 genes and furthermore the cytokine mediated induction PLA2g2a was suppressed. In addition, eprotirome induced genes involved in fatty acid oxidation and cholesterol clearance while inhibiting lipogenic genes. Our results indicate that in vivo thyroid hormone status regulates the abundance of sPLA2 and the inhibition of PLA2g2a by T3 is conserved across species. By regulating sPLA2 genes, T3 may impact processes associated with atherosclerosis and inflammation and TRß agonists may ameliorate inflammation and hyperlipidemia.


Asunto(s)
Fosfolipasas A2 Secretoras/genética , Triyodotironina/metabolismo , Anilidas/farmacología , Animales , Regulación Enzimológica de la Expresión Génica , Fosfolipasas A2 Grupo II/genética , Fosfolipasas A2 Grupo II/metabolismo , Células Hep G2 , Hepatocitos/efectos de los fármacos , Hepatocitos/metabolismo , Humanos , Hiperlipidemias/genética , Hiperlipidemias/metabolismo , Hipertiroidismo/genética , Hipertiroidismo/metabolismo , Hipotiroidismo/genética , Hipotiroidismo/metabolismo , Inflamación/genética , Inflamación/metabolismo , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Transgénicos , Fosfolipasas A2 Secretoras/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Receptores beta de Hormona Tiroidea/agonistas , Receptores beta de Hormona Tiroidea/metabolismo
11.
Biochem Biophys Res Commun ; 449(4): 449-54, 2014 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-24853806

RESUMEN

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.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Activación Transcripcional , Animales , Glucagón/farmacología , Células HEK293 , Humanos , Espectrometría de Masas , Fosforilación , Alineación de Secuencia , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética
12.
Amino Acids ; 46(7): 1589-604, 2014 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-24633358

RESUMEN

While the ribosome constitution is similar in all biota, there is a considerable increase in size of both ribosomal proteins (RPs) and RNAs in eukaryotes as compared to archaea and bacteria. This is pronounced in the large (60S) ribosomal subunit (LSU). In addition to enlargement (apparently maximized already in lower eukarya), the RP changes include increases in fraction, segregation and clustering of basic residues, and decrease in hydrophobicity. The acidic fraction is lower in eukaryote as compared to prokaryote RPs. In all eukaryote groups tested, the LSU RPs have significantly higher content of basic residues and homobasic segments than the SSU RPs. The vertebrate LSU RPs have much higher sequestration of basic residues than those of bacteria, archaea and even of the lower eukarya. The basic clusters are highly aligned in the vertebrate, but less in the lower eukarya, and only within families in archaea and bacteria. Increase in the basicity of RPs, besides helping transport to the nucleus, should promote stability of the assembled ribosome as well as the association with translocons and other intracellular matrix proteins. The size and GC nucleotide bias of the expansion segments of large LSU rRNAs also culminate in the vertebrate, and should support ribosome association with the endoplasmic reticulum and other intracellular networks. However, the expansion and nucleotide bias of eukaryote LSU rRNAs do not clearly correlate with changes in ionic parameters of LSU ribosomal proteins.


Asunto(s)
Eucariontes/fisiología , Evolución Molecular , ARN Ribosómico/fisiología , Proteínas Ribosómicas/fisiología , Animales , Proteínas Arqueales/genética , Proteínas Arqueales/metabolismo , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Secuencia de Bases , Secuencia Conservada , Células Eucariotas , Interacciones Hidrofóbicas e Hidrofílicas , Mamíferos/genética , Células Procariotas , ARN Bacteriano/química , ARN Bacteriano/fisiología
13.
Int J Mol Sci ; 15(3): 4856-77, 2014 Mar 19.
Artículo en Inglés | MEDLINE | ID: mdl-24651459

RESUMEN

The status and use of transmembrane, extracellular and intracellular domains in oligomerization of heptahelical G-protein coupled receptors (GPCRs) are reviewed and for transmembrane assemblies also supplemented by new experimental evidence. The transmembrane-linked GPCR oligomers typically have as the minimal unit an asymmetric ~180 kDa pentamer consisting of receptor homodimer or heterodimer and a G-protein αßγ subunit heterotrimer. With neuropeptide Y (NPY) receptors, this assembly is converted to ~90 kDa receptor monomer-Gα complex by receptor and Gα agonists, and dimers/heteropentamers are depleted by neutralization of Gαi subunits by pertussis toxin. Employing gradient centrifugation, quantification and other characterization of GPCR dimers at the level of physically isolated and identified heteropentamers is feasible with labeled agonists that do not dissociate upon solubilization. This is demonstrated with three neuropeptide Y (NPY) receptors and could apply to many receptors that use large peptidic agonists.


Asunto(s)
Multimerización de Proteína , Receptores Acoplados a Proteínas G/química , Receptores de Neuropéptido Y/química , Secuencias de Aminoácidos , Secuencia de Aminoácidos , Animales , Arrestina/química , Arrestina/metabolismo , Sitios de Unión , Unión Competitiva/efectos de los fármacos , Células CHO , Cricetinae , Cricetulus , Humanos , Cinética , Neuropéptido Y/metabolismo , Neuropéptido Y/farmacología , Fragmentos de Péptidos/metabolismo , Fragmentos de Péptidos/farmacología , Péptido YY/metabolismo , Péptido YY/farmacología , Unión Proteica/efectos de los fármacos , Subunidades de Proteína/química , Subunidades de Proteína/metabolismo , Conejos , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Neuropéptido Y/agonistas , Receptores de Neuropéptido Y/metabolismo
14.
J Biol Chem ; 287(24): 20132-43, 2012 Jun 08.
Artículo en Inglés | MEDLINE | ID: mdl-22511764

RESUMEN

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.


Asunto(s)
Factores de Transcripción Forkhead/metabolismo , Regulación de la Expresión Génica/fisiología , Hígado/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Factor de Transcripción Sp1/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Animales , Células Cultivadas , Proteína Forkhead Box O1 , Factores de Transcripción Forkhead/genética , Hepatocitos/metabolismo , Insulina/genética , Insulina/metabolismo , Receptores X del Hígado , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Receptores Nucleares Huérfanos/genética , Receptores Nucleares Huérfanos/metabolismo , Ratas , Elementos de Respuesta/fisiología , Factor de Transcripción Sp1/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Transcripción Genética/fisiología
15.
PLoS One ; 18(12): e0294498, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-38100464

RESUMEN

BACKGROUND: Between 5-10% of patients discontinue statin therapy due to statin-associated adverse reactions, primarily statin associated muscle symptoms (SAMS). The absence of a clear clinical phenotype or of biomarkers poses a challenge for diagnosis and management of SAMS. Similarly, our incomplete understanding of the pathogenesis of SAMS hinders the identification of treatments for SAMS. Metabolomics, the profiling of metabolites in biofluids, cells and tissues is an important tool for biomarker discovery and provides important insight into the origins of symptomatology. In order to better understand the pathophysiology of this common disorder and to identify biomarkers, we undertook comprehensive metabolomic and lipidomic profiling of plasma samples from patients with SAMS who were undergoing statin rechallenge as part of their clinical care. METHODS AND FINDINGS: We report our findings in 67 patients, 28 with SAMS (cases) and 39 statin-tolerant controls. SAMS patients were studied during statin rechallenge and statin tolerant controls were studied while on statin. Plasma samples were analyzed using untargeted LC-MS metabolomics and lipidomics to detect differences between cases and controls. Differences in lipid species in plasma were observed between cases and controls. These included higher levels of linoleic acid containing phospholipids and lower ether lipids and sphingolipids. Reduced levels of acylcarnitines and altered amino acid profile (tryptophan, tyrosine, proline, arginine, and taurine) were observed in cases relative to controls. Pathway analysis identified significant increase of urea cycle metabolites and arginine and proline metabolites among cases along with downregulation of pathways mediating oxidation of branched chain fatty acids, carnitine synthesis, and transfer of acetyl groups into mitochondria. CONCLUSIONS: The plasma metabolome of patients with SAMS exhibited reduced content of long chain fatty acids and increased levels of linoleic acid (18:2) in phospholipids, altered energy production pathways (ß-oxidation, citric acid cycle and urea cycles) as well as reduced levels of carnitine, an essential mediator of mitochondrial energy production. Our findings support the hypothesis that alterations in pro-inflammatory lipids (arachidonic acid pathway) and impaired mitochondrial energy metabolism underlie the muscle symptoms of patients with statin associated muscle symptoms (SAMS).


Asunto(s)
Inhibidores de Hidroximetilglutaril-CoA Reductasas , Humanos , Inhibidores de Hidroximetilglutaril-CoA Reductasas/efectos adversos , Prostaglandinas , Músculos/metabolismo , Carnitina , Ácidos Grasos/metabolismo , Metabolómica/métodos , Prolina , Arginina , Biomarcadores , Ácidos Linoleicos , Urea
16.
J Biol Chem ; 286(27): 23799-807, 2011 Jul 08.
Artículo en Inglés | MEDLINE | ID: mdl-21586575

RESUMEN

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.


Asunto(s)
Proteína beta Potenciadora de Unión a CCAAT/metabolismo , Regulación Enzimológica de la Expresión Génica/fisiología , Hepatocitos/metabolismo , Proteínas Serina-Treonina Quinasas/biosíntesis , Elementos de Respuesta/fisiología , Animales , Proteína beta Potenciadora de Unión a CCAAT/genética , Carnitina O-Palmitoiltransferasa/biosíntesis , Carnitina O-Palmitoiltransferasa/genética , Gluconeogénesis/fisiología , Células Hep G2 , Hepatocitos/citología , Humanos , Proteínas Serina-Treonina Quinasas/genética , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Complejo Piruvato Deshidrogenasa/biosíntesis , Complejo Piruvato Deshidrogenasa/genética , Ratas , Triyodotironina/genética , Triyodotironina/metabolismo
17.
J Biol Chem ; 285(4): 2375-85, 2010 Jan 22.
Artículo en Inglés | MEDLINE | ID: mdl-19948729

RESUMEN

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.


Asunto(s)
Hepatocitos/enzimología , Hipertiroidismo/fisiopatología , Hipotiroidismo/fisiopatología , Proteínas Serina-Treonina Quinasas/metabolismo , Proteínas de Unión al ARN/metabolismo , Factores de Transcripción/metabolismo , Triyodotironina/metabolismo , Animales , Secuencia de Bases , Carcinoma Hepatocelular , Línea Celular Tumoral , Hepatocitos/citología , Humanos , Hipertiroidismo/metabolismo , Hipofisectomía , Hipotiroidismo/metabolismo , Neoplasias Hepáticas , Masculino , Datos de Secuencia Molecular , PPAR gamma/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Regiones Promotoras Genéticas/fisiología , Proteínas Serina-Treonina Quinasas/genética , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora , Complejo Piruvato Deshidrogenasa/metabolismo , ARN Mensajero/metabolismo , Proteínas de Unión al ARN/genética , Ratas , Ratas Sprague-Dawley , Receptores beta de Hormona Tiroidea/metabolismo , Factores de Transcripción/genética , Transcripción Genética/fisiología , Transfección
18.
Biochem Soc Trans ; 39(3): 833-7, 2011 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-21599656

RESUMEN

CPT (carnitine palmitoyltransferase) 1 and CPT2 regulate fatty acid oxidation. Recombinant rat CPT2 was isolated from the soluble fractions of bacterial extracts and expressed in Escherichia coli. The acyl-CoA chain-length-specificity of the recombinant CPT2 was identical with that of the purified enzyme from rat liver mitochondrial inner membranes. The Km for carnitine for both the mitochondrial preparation and the recombinant enzyme was identical. In isolated mitochondrial outer membranes, cardiolipin (diphosphatidylglycerol) increased CPT1 activity 4-fold and the Km for carnitine 6-fold. It decreased the Ki for malonyl-CoA inhibition 60-fold, but had no effect on the apparent Km for myristoyl-CoA. Cardiolipin also activated recombinant CPT2 almost 4-fold, whereas phosphatidylglycerol, phosphatidylserine and phosphatidylcholine activated the enzyme 3-, 2- and 2-fold respectively. Most of the recombinant CPT2 was found to have substantial interaction with cardiolipin. A model is proposed whereby cardiolipin may hold the fatty-acid-oxidizing enzymes in the active functional conformation between the mitochondrial inner and outer membranes in conjunction with the translocase and the acyl-CoA synthetase, thus combining all four enzymes into a functional unit.


Asunto(s)
Carnitina O-Palmitoiltransferasa/metabolismo , Membranas Intracelulares/enzimología , Microdominios de Membrana/metabolismo , Animales , Cardiolipinas/metabolismo , Carnitina O-Palmitoiltransferasa/genética , Ácidos Grasos/metabolismo , Isoenzimas/genética , Isoenzimas/metabolismo , Microdominios de Membrana/química , Mitocondrias Hepáticas/enzimología , Fosfolípidos/química , Fosfolípidos/metabolismo , Ratas , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo
19.
Amino Acids ; 40(2): 261-8, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20571842

RESUMEN

For many G-protein coupling receptors (GPCRs), the upkeep of receptor dimers could depend on association with functional Gi α subunits. This is known for Y1, Y2 and Y4 neuropeptide Y receptors [presented in the companion paper (Estes et al., Amino Acids, doi: 10.1007/s00726-010-0642-z , 2010)]. Interactions with transducers use mainly intracellular domains of the receptors. Intracellular loops 1 and 2 in GPCRs are short and lack extensive helicity that could support transducer anchoring. Interaction with G-proteins is known to use the juxtamembrane Helix 8 in the fourth intracellular domain, for which we document a helix-stabilizing n/(n + 4) pattern of large hydrophobic sidechains. Another intracellular helix located in the C-terminal portion of the third intracellular loop does not display a strong stabilizing pattern, and is found in many studies to serve dynamically in association and activation of transducers and effectors. We show that these tracts share features across metazoan phyla not only in opsins and opsin-like receptors (including the Y receptors), but also in Taste-2 and Frizzled receptors. Similarities of these helices across GPCR groups could have both phylogenetic and functional roots.


Asunto(s)
Proteínas de Unión al GTP/metabolismo , Receptores Acoplados a Proteínas G/química , Receptores Acoplados a Proteínas G/metabolismo , Transducción de Señal , Animales , Humanos , Multimerización de Proteína , Estructura Secundaria de Proteína , Estructura Terciaria de Proteína
20.
Amino Acids ; 40(2): 371-80, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20577889

RESUMEN

Treatment of CHO cells expressing human Y receptors (Y(1), Y(2) or Y4 subtype) with pertussis toxin results in a large decrease in functional receptors, with a preferential loss of heteropentameric assemblies of receptor dimers and G-protein trimers. This occurs in parallel to inactivation of the nucleotide site of Gi α subunits, with a half period of about 4 h. The loss could be mainly due to proteolysis at the level of recycling/perinuclear endosomes, and of receptor completion in the ER, since it is reduced by co-treatment with ammonium chloride, an inhibitor of particulate proteinases. Antagonists do not strongly decrease the heteropentameric fraction. These findings indicate that the upkeep of Y receptor dimers in epithelial cell lines depends on the association of receptor oligomers with functional Gi α subunits. This interaction could use the juxtamembrane helix 8 in the fourth intracellular domain, and could also be supported by the C-terminal helix of the third intracellular loop, as outlined in the companion review (Parker et al., Amino Acids, doi: 10.1007/s00726-010-0616-1 , 2010).


Asunto(s)
Células Epiteliales/metabolismo , Proteínas de Unión al GTP Heterotriméricas/metabolismo , Receptores de Neuropéptido Y/metabolismo , Animales , Células CHO , Bovinos , Cricetinae , Cricetulus , Dimerización , Proteínas de Unión al GTP Heterotriméricas/antagonistas & inhibidores , Proteínas de Unión al GTP Heterotriméricas/genética , Humanos , Toxina del Pertussis/metabolismo , Unión Proteica , Ratas , Receptores de Neuropéptido Y/antagonistas & inhibidores , Receptores de Neuropéptido Y/química , Receptores de Neuropéptido Y/genética , Porcinos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA