Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 101
Filtrar
1.
J Biol Chem ; 294(6): 2009-2020, 2019 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-30523156

RESUMO

Hepatic insulin resistance in the setting of steatosis is attributable at least in part to the accumulation of bioactive lipids that suppress insulin signaling. The mitochondria-associated glycerol-3-phosphate acyltransferase 1 (GPAT1) catalyzes the first committed step in glycerolipid synthesis, and its activity diverts fatty acids from mitochondrial ß-oxidation. GPAT1 overexpression in mouse liver leads to hepatic steatosis even in the absence of overnutrition. The mice develop insulin resistance owing to the generation of saturated diacylglycerol and phosphatidic acid molecular species that reduce insulin signaling by activating PKCϵ and by suppressing mTORC2, respectively. Them2, a mitochondria-associated acyl-CoA thioesterase, also participates in the trafficking of fatty acids into oxidative versus glycerolipid biosynthetic pathways. Them2-/- mice are protected against diet-induced hepatic steatosis and insulin resistance. To determine whether Them2 contributes to hepatic insulin resistance due to hepatic overexpression of GPAT1, recombinant adenovirus was used to overexpress GPAT1 in livers of chow-fed Them2+/+ and Them2-/- mice. Hepatic GPAT1 overexpression led to steatosis in both genotypes. In the setting of GPAT1 overexpression, glucose tolerance was reduced in Them2+/+ but not Them2-/- mice, without influencing whole-body insulin sensitivity or basal hepatic glucose production. Improved glucose tolerance in Them2-/- mice was associated with reduced PKCϵ translocation. Preserved insulin receptor activity was supported by Thr-308 phosphorylation of Akt following GPAT1 overexpression in Them2-/- hepatocytes. These findings suggest a pathogenic role of Them2 in the biosynthesis of glycerolipid metabolites that promote hepatic insulin resistance.


Assuntos
Fígado Gorduroso/complicações , Glicerol-3-Fosfato O-Aciltransferase/metabolismo , Resistência à Insulina , Tioléster Hidrolases/farmacologia , Animais , Ácidos Graxos/metabolismo , Fígado Gorduroso/induzido quimicamente , Glicerídeos/biossíntese , Hepatócitos/metabolismo , Hepatopatias , Camundongos , Proteína Quinase C-épsilon/metabolismo , Tioléster Hidrolases/genética
2.
J Biol Chem ; 294(22): 8819-8833, 2019 05 31.
Artigo em Inglês | MEDLINE | ID: mdl-30975900

RESUMO

Loss of long-chain acyl-CoA synthetase isoform-1 (ACSL1) in mouse skeletal muscle (Acsl1M-/-) severely reduces acyl-CoA synthetase activity and fatty acid oxidation. However, the effects of decreased fatty acid oxidation on skeletal muscle function, histology, use of alternative fuels, and mitochondrial function and morphology are unclear. We observed that Acsl1M-/- mice have impaired voluntary running capacity and muscle grip strength and that their gastrocnemius muscle contains myocytes with central nuclei, indicating muscle regeneration. We also found that plasma creatine kinase and aspartate aminotransferase levels in Acsl1M-/- mice are 3.4- and 1.5-fold greater, respectively, than in control mice (Acsl1flox/flox ), indicating muscle damage, even without exercise, in the Acsl1M-/- mice. Moreover, caspase-3 protein expression exclusively in Acsl1M-/- skeletal muscle and the presence of cleaved caspase-3 suggested myocyte apoptosis. Mitochondria in Acsl1M-/- skeletal muscle were swollen with abnormal cristae, and mitochondrial biogenesis was increased. Glucose uptake did not increase in Acsl1M-/- skeletal muscle, and pyruvate oxidation was similar in gastrocnemius homogenates from Acsl1M-/- and control mice. The rate of protein synthesis in Acsl1M-/- glycolytic muscle was 2.1-fold greater 30 min after exercise than in the controls, suggesting resynthesis of proteins catabolized for fuel during the exercise. At this time, mTOR complex 1 was activated, and autophagy was blocked. These results suggest that fatty acid oxidation is critical for normal skeletal muscle homeostasis during both rest and exercise. We conclude that ACSL1 deficiency produces an overall defect in muscle fuel metabolism that increases protein catabolism, resulting in exercise intolerance, muscle weakness, and myocyte apoptosis.


Assuntos
Aminoácidos/metabolismo , Coenzima A Ligases/genética , Ácidos Graxos/metabolismo , Músculo Esquelético/metabolismo , Animais , Apoptose , Aspartato Aminotransferases/metabolismo , Caspase 3/metabolismo , Coenzima A Ligases/deficiência , Creatina Quinase/metabolismo , Metabolismo dos Lipídeos , Alvo Mecanístico do Complexo 1 de Rapamicina/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/metabolismo , Músculo Esquelético/patologia , Oxirredução , Condicionamento Físico Animal , Regulação para Cima
3.
Biochem J ; 476(1): 85-99, 2019 01 11.
Artigo em Inglês | MEDLINE | ID: mdl-30523059

RESUMO

Macrophage classical M1 activation via TLR4 triggers a variety of responses to achieve the elimination of foreign pathogens. During this process, there is also an increase in lipid droplets which contain large quantities of triacylglycerol (TAG) and phospholipid (PL). The functional consequences of this increment in lipid mass are poorly understood. Here, we studied the contribution of glycerolipid synthesis to lipid accumulation, focusing specifically on the first and rate-limiting enzyme of the pathway: glycerol-3-phosphate acyltransferase (GPAT). Using bone marrow-derived macrophages (BMDMs) treated with Kdo2-lipid A, we showed that glycerolipid synthesis is induced during macrophage activation. GPAT4 protein level and GPAT3/GPAT4 enzymatic activity increase during this process, and these two isoforms were required for the accumulation of cell TAG and PL. The phagocytic capacity of Gpat3-/- and Gpat4-/- BMDM was impaired. Additionally, inhibiting fatty acid ß-oxidation reduced phagocytosis only partially, suggesting that lipid accumulation is not necessary for the energy requirements for phagocytosis. Finally, Gpat4-/- BMDM expressed and released more pro-inflammatory cytokines and chemokines after macrophage activation, suggesting a role for GPAT4 in suppressing inflammatory responses. Together, these results provide evidence that glycerolipid synthesis directed by GPAT4 is important for the attenuation of the inflammatory response in activated macrophages.


Assuntos
1-Acilglicerol-3-Fosfato O-Aciltransferase/metabolismo , Glicerol-3-Fosfato O-Aciltransferase/metabolismo , Lipogênese , Macrófagos/enzimologia , Fosfolipídeos/biossíntese , Triglicerídeos/biossíntese , 1-Acilglicerol-3-Fosfato O-Aciltransferase/genética , Animais , Glicerol-3-Fosfato O-Aciltransferase/genética , Inflamação/enzimologia , Inflamação/genética , Inflamação/patologia , Ativação de Macrófagos/genética , Macrófagos/patologia , Camundongos , Camundongos Knockout , Fosfolipídeos/genética , Triglicerídeos/genética
4.
J Lipid Res ; 60(3): 490-497, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30683668

RESUMO

Diet, hormones, gene transcription, and posttranslational modifications control the hepatic metabolism of FAs; metabolic dysregulation causes chronic diseases, including cardiovascular disease, and warrants exploration into the mechanisms directing FA and triacylglycerol (TAG) synthesis and degradation. Long-chain FA metabolism begins by formation of an acyl-CoA by a member of the acyl-CoA synthetase (ACSL) family. Subsequently, TAG synthesis begins with acyl-CoA esterification to glycerol-3-phosphate by a member of the glycerol-3-phosphate acyltransferase (GPAT) family. Our studies of the isoforms ACSL1 and GPAT1 strongly suggest that these proteins are members of larger protein assemblies (interactomes). ACSL1 targeted to the ER interacts with peroxisomal, lipid droplet, and tethering proteins, uncovering a dynamic role for ACSL1 in organelle and lipid droplet interactions. On the outer mitochondrial membrane (OMM), PPARα upregulates ACSL1, which interacts with proteins believed to tether lipid droplets to the OMM. In contrast, GPAT1 is upregulated nutritionally by carbohydrate and insulin in a coordinated sequence of enzyme reactions, from saturated FA formation via de novo lipogenesis to FA esterification by GPAT1 and entry into the TAG biosynthesis pathway. We propose that involved enzymes form a dynamic protein interactome that facilitates esterification and that other lipid-metabolizing pathways will exist in similar physiologically regulated interactomes.


Assuntos
Ácidos Graxos/metabolismo , Mapeamento de Interação de Proteínas , Triglicerídeos/metabolismo , Animais , Humanos
5.
J Biol Chem ; 293(43): 16724-16740, 2018 10 26.
Artigo em Inglês | MEDLINE | ID: mdl-30190326

RESUMO

Fatty acid channeling into oxidation or storage modes depends on physiological conditions and hormonal signaling. However, the directionality of this channeling may also depend on the association of each of the five acyl-CoA synthetase isoforms with specific protein partners. Long-chain acyl-CoA synthetases (ACSLs) catalyze the conversion of long-chain fatty acids to fatty acyl-CoAs, which are then either oxidized or used in esterification reactions. In highly oxidative tissues, ACSL1 is located on the outer mitochondrial membrane (OMM) and directs fatty acids into mitochondria for ß-oxidation. In the liver, however, about 50% of ACSL1 is located on the endoplasmic reticulum (ER) where its metabolic function is unclear. Because hepatic fatty acid partitioning is likely to require the interaction of ACSL1 with other specific proteins, we used an unbiased protein interaction technique, BioID, to discover ACSL1-binding partners in hepatocytes. We targeted ACSL1 either to the ER or to the OMM of Hepa 1-6 cells as a fusion protein with the Escherichia coli biotin ligase, BirA*. Proteomic analysis identified 98 proteins that specifically interacted with ACSL1 at the ER, 55 at the OMM, and 43 common to both subcellular locations. We found subsets of peroxisomal and lipid droplet proteins, tethering proteins, and vesicle proteins, uncovering a dynamic role for ACSL1 in organelle and lipid droplet interactions. Proteins involved in lipid metabolism were also identified, including acyl-CoA-binding proteins and ceramide synthase isoforms 2 and 5. Our results provide fundamental and detailed insights into protein interaction networks that control fatty acid metabolism.


Assuntos
Coenzima A Ligases/fisiologia , Retículo Endoplasmático/metabolismo , Ácidos Graxos/metabolismo , Fígado/metabolismo , Mitocôndrias/metabolismo , Domínios e Motivos de Interação entre Proteínas , Animais , Feminino , Fígado/citologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
6.
J Biol Chem ; 293(41): 15933-15946, 2018 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-30158245

RESUMO

Cardiolipin (CL) is an anionic phospholipid mainly located in the inner mitochondrial membrane, where it helps regulate bioenergetics, membrane structure, and apoptosis. Localized, phase-segregated domains of CL are hypothesized to control mitochondrial inner membrane organization. However, the existence and underlying mechanisms regulating these mitochondrial domains are unclear. Here, we first isolated detergent-resistant cardiac mitochondrial membranes that have been reported to be CL-enriched domains. Experiments with different detergents yielded only nonspecific solubilization of mitochondrial phospholipids, suggesting that CL domains are not recoverable with detergents. Next, domain formation was investigated in biomimetic giant unilamellar vesicles (GUVs) and newly synthesized giant mitochondrial vesicles (GMVs) from mouse hearts. Confocal fluorescent imaging revealed that introduction of cytochrome c into membranes promotes macroscopic proteolipid domain formation associated with membrane morphological changes in both GUVs and GMVs. Domain organization was also investigated after lowering tetralinoleoyl-CL concentration and substitution with monolyso-CL, two common modifications observed in cardiac pathologies. Loss of tetralinoleoyl-CL decreased proteolipid domain formation in GUVs, because of a favorable Gibbs-free energy of lipid mixing, whereas addition of monolyso-CL had no effect on lipid mixing. Moreover, murine GMVs generated from cardiac acyl-CoA synthetase-1 knockouts, which have remodeled CL acyl chains, did not perturb proteolipid domains. Finally, lowering the tetralinoleoyl-CL content had a stronger influence on the oxidation status of cytochrome c than did incorporation of monolyso-CL. These results indicate that proteolipid domain formation in the cardiac mitochondrial inner membrane depends on tetralinoleoyl-CL concentration, driven by underlying lipid-mixing properties, but not the presence of monolyso-CL.


Assuntos
Cardiolipinas/metabolismo , Microdomínios da Membrana/metabolismo , Mitocôndrias/metabolismo , Membranas Mitocondriais/metabolismo , Proteolipídeos/metabolismo , Lipossomas Unilamelares/metabolismo , Animais , Materiais Biomiméticos/metabolismo , Coenzima A Ligases/genética , Citocromos c/metabolismo , Técnicas de Silenciamento de Genes , Lisofosfolipídeos/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Miocárdio/metabolismo , Ratos Sprague-Dawley
7.
J Lipid Res ; 58(5): 884-894, 2017 05.
Artigo em Inglês | MEDLINE | ID: mdl-28209804

RESUMO

Because the signaling eicosanoids, epoxyeicosatrienoic acids (EETs) and HETEs, are esterified to membrane phospholipids, we asked which long-chain acyl-CoA synthetase (ACSL) isoforms would activate these molecules and whether the apparent FA substrate preferences of each ACSL isoform might differ depending on whether it was assayed in mammalian cell membranes or as a purified bacterial recombinant protein. We found that all five ACSL isoforms were able to use EETs and HETEs as substrates and showed by LC-MS/MS that ACSLs produce EET-CoAs. We found differences in substrate preference between ACS assays performed in COS7 cell membranes and recombinant purified proteins. Similarly, preferences and Michaelis-Menten kinetics for long-chain FAs were distinctive. Substrate preferences identified for the purified ACSLs did not correspond to those observed in ACSL-deficient mouse models. Taken together, these data support the concept that each ACSL isoform exhibits a distinct substrate preference, but apparent substrate specificities depend upon multiple factors including membrane character, coactivators, inhibitors, protein interactions, and posttranslational modification.


Assuntos
Coenzima A Ligases/metabolismo , Eicosanoides/química , Eicosanoides/metabolismo , Animais , Células COS , Chlorocebus aethiops , Isoenzimas/metabolismo , Ratos , Especificidade por Substrato
8.
Biochim Biophys Acta ; 1861(10): 1425-33, 2016 10.
Artigo em Inglês | MEDLINE | ID: mdl-26993579

RESUMO

The heart's extraordinary metabolic flexibility allows it to adapt to normal changes in physiology in order to preserve its function. Alterations in the metabolic profile of the heart have also been attributed to pathological conditions such as ischemia and hypertrophy; however, research during the past decade has established that cardiac metabolic adaptations can precede the onset of pathologies. It is therefore critical to understand how changes in cardiac substrate availability and use trigger events that ultimately result in heart dysfunction. This review examines the mechanisms by which the heart obtains fuels from the circulation or from mobilization of intracellular stores. We next describe experimental models that exhibit either an increase in glucose use or a decrease in FA oxidation, and how these aberrant conditions affect cardiac metabolism and function. Finally, we highlight the importance of alternative, relatively under-investigated strategies for the treatment of heart failure. This article is part of a Special Issue entitled: Heart Lipid Metabolism edited by G.D. Lopaschuk.


Assuntos
Metabolismo Energético , Ácidos Graxos/metabolismo , Glucose/metabolismo , Miocárdio/metabolismo , Animais , Humanos , Oxirredução , Especificidade por Substrato
9.
Biochim Biophys Acta ; 1861(7): 650-62, 2016 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-27091636

RESUMO

Long-chain acyl-CoA esters are important intermediates in lipid metabolism and are synthesized from fatty acids by long-chain acyl-CoA synthetases (ACSL). The hematophagous insect Rhodnius prolixus, a vector of Chagas' disease, produces glycerolipids in the midgut after a blood meal, which are stored as triacylglycerol in the fat body and eggs. We identified twenty acyl-CoA synthetase genes in R. prolixus, two encoding ACSL isoforms (RhoprAcsl1 and RhoprAcsl2). RhoprAcsl1 transcripts increased in posterior midgut on the second day after feeding, and RhoprAcsl2 was highly transcribed on the tenth day. Both enzymes were expressed in Escherichia coli. Recombinant RhoprACSL1 and RhoprACSL2 had broad pH optima (7.5-9.5 and 6.5-9.5, respectively), were inhibited by triacsin C, and were rosiglitazone-insensitive. Both showed similar apparent Km for palmitic and oleic acid (2-6 µM), but different Km for arachidonic acid (0.5 and 6 µM for RhoprACSL1-Flag and RhoprACSL2-Flag, respectively). The knockdown of RhoprAcsl1 did not result in noticeable phenotypes. However, RhoprACSL2 deficient insects exhibited a 2.5-fold increase in triacylglycerol content in the fat body, and 90% decrease in fatty acid ß-oxidation. RhoprAcsl2 knockdown also resulted in 20% increase in lifespan, delayed digestion, 30% reduced oviposition, and 50% reduction in egg hatching. Laid eggs and hatched nymphs showed remarkable alterations in morphology. In summary, R. prolixus ACSL isoforms have distinct roles on lipid metabolism. Although RhoprACSL1 functions remain unclear, we propose that RhoprACSL2 is the main contributor for the formation of the intracellular acyl-CoA pool channeled for ß-oxidation in the fat body, and is also required for normal reproduction.


Assuntos
Coenzima A Ligases/genética , Corpo Adiposo/metabolismo , Ácidos Graxos/metabolismo , Oogênese/genética , Rhodnius/genética , Triglicerídeos/biossíntese , Sequência de Aminoácidos , Animais , Coenzima A Ligases/metabolismo , Escherichia coli/genética , Escherichia coli/metabolismo , Feminino , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Proteínas de Insetos , Isoenzimas/genética , Isoenzimas/metabolismo , Dados de Sequência Molecular , Oxirredução , Filogenia , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Reprodução/genética , Rhodnius/classificação , Alinhamento de Sequência , Transcrição Gênica , Triazenos , Zigoto/metabolismo
10.
Artigo em Inglês | MEDLINE | ID: mdl-27377347

RESUMO

Our understanding of the synthesis and remodeling of mitochondrial phospholipids remains incomplete. Two isoforms of glycerol-3-phosphate acyltransferase (GPAT1 and 2) and two isoforms of acylglycerol-3-phosphate acyltransferase (AGPAT4 and 5) are located on the outer mitochondrial membrane, suggesting that both lysophosphatidic acid and phosphatidic acid are synthesized in situ for de novo glycerolipid biosynthesis. However, it is believed that the phosphatidic acid substrate for cardiolipin and phosphatidylethanolamine biosynthesis is produced at the endoplasmic reticulum whereas the phosphatidic acid synthesized in the mitochondria must be transferred to the endoplasmic reticulum before it undergoes additional steps to form the mature phospholipids that are trafficked back to the mitochondria. It is unclear whether mitochondrial phospholipids are remodeled by mitochondrial acyltransferases or whether lysophospholipids must return to the endoplasmic reticulum or to the mitochondrial associated membrane for reesterification. In this review we will focus on the few glycerolipid acyltransferases that are known to be mitochondrial. This article is part of a Special Issue entitled: Lipids of Mitochondria edited by Guenther Daum.


Assuntos
Aciltransferases/metabolismo , Glicerofosfolipídeos/metabolismo , Mitocôndrias/metabolismo , Animais , Retículo Endoplasmático/metabolismo , Glicerol-3-Fosfato O-Aciltransferase , Humanos , Metabolismo dos Lipídeos/fisiologia , Transporte Proteico/fisiologia
11.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1862(3): 324-336, 2017 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-27956137

RESUMO

Glycerol-3-phosphate acyltransferases (GPAT) catalyze the initial and rate-limiting step for the de novo synthesis of triacylglycerol (TAG). Four mammalian GPAT isoforms have been identified: the mitochondria-associated GPAT1 and 2, and the endoplasmic reticulum (ER)-associated GPAT3 and 4. In the insect Rhodnius prolixus, a vector of Chagas' disease, we previously predicted a mitochondrial-like isoform (RhoprGPAT1) from genomic data. In the current study, we clone the RhoprGPAT1 coding sequence and identify an ER-associated GPAT (RhoprGPAT4) as the second isoform in the insect. RhoprGPAT1 contributes 15% of the total GPAT activity in anterior midgut, 50% in posterior midgut and fat body, and 70% in the ovary. The RhoprGpat1 gene is the predominant transcript in the midgut and fat body. To evaluate the physiological relevance of RhoprGPAT1, we generate RhoprGPAT1-deficient insects. The knockdown of RhoprGpat1 results in 50% and 65% decrease in TAG content in the posterior midgut and fat body, respectively. RhoprGpat1-deficient insects also exhibits impaired lipid droplet expansion and a 2-fold increase in fatty acid ß-oxidation rates in the fat body. We propose that the RhoprGPAT1 mitochondrial-like isoform is required to channel fatty acyl chains towards TAG synthesis and away from ß-oxidation. Such a process is crucial for the insect lipid homeostasis.


Assuntos
Corpo Adiposo/metabolismo , Ácidos Graxos/metabolismo , Glicerol-3-Fosfato O-Aciltransferase/metabolismo , Insetos/metabolismo , Rhodnius/metabolismo , Triglicerídeos/metabolismo , Animais , Retículo Endoplasmático/metabolismo , Gotículas Lipídicas/metabolismo , Mitocôndrias/metabolismo , Oxirredução
12.
Proc Natl Acad Sci U S A ; 111(23): E2414-22, 2014 Jun 10.
Artigo em Inglês | MEDLINE | ID: mdl-24872453

RESUMO

The tumor suppressor p53 has recently been shown to regulate energy metabolism through multiple mechanisms. However, the in vivo signaling pathways related to p53-mediated metabolic regulation remain largely uncharacterized. By using mice bearing a single amino acid substitution at cysteine residue 305 of mouse double minute 2 (Mdm2(C305F)), which renders Mdm2 deficient in binding ribosomal proteins (RPs) RPL11 and RPL5, we show that the RP-Mdm2-p53 signaling pathway is critical for sensing nutrient deprivation and maintaining liver lipid homeostasis. Although the Mdm2(C305F) mutation does not significantly affect growth and development in mice, this mutation promotes fat accumulation under normal feeding conditions and hepatosteatosis under acute fasting conditions. We show that nutrient deprivation inhibits rRNA biosynthesis, increases RP-Mdm2 interaction, and induces p53-mediated transactivation of malonyl-CoA decarboxylase (MCD), which catalyzes the degradation of malonyl-CoA to acetyl-CoA, thus modulating lipid partitioning. Fasted Mdm2(C305F) mice demonstrate attenuated MCD induction and enhanced malonyl-CoA accumulation in addition to decreased oxidative respiration and increased fatty acid accumulation in the liver. Thus, the RP-Mdm2-p53 pathway appears to function as an endogenous sensor responsible for stimulating fatty acid oxidation in response to nutrient depletion.


Assuntos
Fenômenos Fisiológicos da Nutrição Animal/fisiologia , Carboxiliases/metabolismo , Ácidos Graxos/metabolismo , Metabolismo dos Lipídeos/fisiologia , Proteínas Proto-Oncogênicas c-mdm2/metabolismo , Proteínas Ribossômicas/metabolismo , Proteína Supressora de Tumor p53/metabolismo , Animais , Células Cultivadas , Embrião de Mamíferos/citologia , Jejum , Fígado Gorduroso/genética , Fígado Gorduroso/fisiopatologia , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Immunoblotting , Metabolismo dos Lipídeos/genética , Camundongos , Camundongos Knockout , Análise de Sequência com Séries de Oligonucleotídeos , Oxirredução , Ligação Proteica , Proteínas Proto-Oncogênicas c-mdm2/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estresse Fisiológico/fisiologia , Tamoxifeno/análogos & derivados , Tamoxifeno/farmacologia , Transcriptoma/efeitos dos fármacos , Proteína Supressora de Tumor p53/genética , Redução de Peso/genética , Redução de Peso/fisiologia
13.
J Biol Chem ; 290(33): 20023-31, 2015 Aug 14.
Artigo em Inglês | MEDLINE | ID: mdl-26124277

RESUMO

Meeting the complex physiological demands of mammalian life requires strict control of the metabolism of long-chain fatty acyl-CoAs because of the multiplicity of their cellular functions. Acyl-CoAs are substrates for energy production; stored within lipid droplets as triacylglycerol, cholesterol esters, and retinol esters; esterified to form membrane phospholipids; or used to activate transcriptional and signaling pathways. Indirect evidence suggests that acyl-CoAs do not wander freely within cells, but instead, are channeled into specific pathways. In this review, we will discuss the evidence for acyl-CoA compartmentalization, highlight the key modes of acyl-CoA regulation, and diagram potential mechanisms for controlling acyl-CoA partitioning.


Assuntos
Acil Coenzima A/metabolismo , Compartimento Celular , Acilação , Animais , Meios de Cultura , Homeostase , Camundongos , Camundongos Knockout , Transdução de Sinais , Especificidade por Substrato , Termogênese
14.
J Biol Chem ; 290(24): 15112-20, 2015 Jun 12.
Artigo em Inglês | MEDLINE | ID: mdl-25918168

RESUMO

Glycerol-3-phosphate acyltransferase-4 (GPAT4) null pups grew poorly during the suckling period and, as adults, were protected from high fat diet-induced obesity. To determine why Gpat4(-/-) mice failed to gain weight during these two periods of high fat feeding, we examined energy metabolism. Compared with controls, the metabolic rate of Gpat4(-/-) mice fed a 45% fat diet was 12% higher. Core body temperature was 1 ºC higher after high fat feeding. Food intake, fat absorption, and activity were similar in both genotypes. Impaired weight gain in Gpat4(-/-) mice did not result from increased heat loss, because both cold tolerance and response to a ß3-adrenergic agonist were similar in both genotypes. Because GPAT4 comprises 65% of the total GPAT activity in brown adipose tissue (BAT), we characterized BAT function. A 45% fat diet increased the Gpat4(-/-) BAT expression of peroxisome proliferator-activated receptor α (PPAR) target genes, Cpt1α, Pgc1α, and Ucp1, and BAT mitochondria oxidized oleate and pyruvate at higher rates than controls, suggesting that fatty acid signaling and flux through the TCA cycle were enhanced. To assess the role of GPAT4 directly, neonatal BAT preadipocytes were differentiated to adipocytes. Compared with controls, Gpat4(-/-) brown adipocytes incorporated 33% less fatty acid into triacylglycerol and 46% more into the pathway of ß-oxidation. The increased oxidation rate was due solely to an increase in the oxidation of exogenous fatty acids. These data suggest that in the absence of cold exposure, GPAT4 limits excessive fatty acid oxidation and the detrimental induction of a hypermetabolic state.


Assuntos
Adipócitos/metabolismo , Tecido Adiposo Marrom/metabolismo , Ácidos Graxos/metabolismo , Glicerol-3-Fosfato O-Aciltransferase/metabolismo , Adipócitos/enzimologia , Tecido Adiposo Marrom/citologia , Tecido Adiposo Marrom/enzimologia , Animais , Gorduras na Dieta/administração & dosagem , Glicerol-3-Fosfato O-Aciltransferase/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Oxirredução , Termogênese/genética , Triglicerídeos/metabolismo , Aumento de Peso
15.
J Biol Chem ; 290(6): 3519-28, 2015 Feb 06.
Artigo em Inglês | MEDLINE | ID: mdl-25512376

RESUMO

Although an elevated triacylglycerol content in non-adipose tissues is often associated with insulin resistance, the mechanistic relationship remains unclear. The data support roles for intermediates in the glycerol-3-phosphate pathway of triacylglycerol synthesis: diacylglycerol (DAG), which may cause insulin resistance in liver by activating PKCϵ, and phosphatidic acid (PA), which inhibits insulin action in hepatocytes by disrupting the assembly of mTOR and rictor. To determine whether increases in DAG and PA impair insulin signaling when produced by pathways other than that of de novo synthesis, we examined primary mouse hepatocytes after enzymatically manipulating the cellular content of DAG or PA. Overexpressing phospholipase D1 or phospholipase D2 inhibited insulin signaling and was accompanied by an elevated cellular content of total PA, without a change in total DAG. Overexpression of diacylglycerol kinase-θ inhibited insulin signaling and was accompanied by an elevated cellular content of total PA and a decreased cellular content of total DAG. Overexpressing glycerol-3-phosphate acyltransferase-1 or -4 inhibited insulin signaling and increased the cellular content of both PA and DAG. Insulin signaling impairment caused by overexpression of phospholipase D1/D2 or diacylglycerol kinase-θ was always accompanied by disassociation of mTOR/rictor and reduction of mTORC2 kinase activity. However, although the protein ratio of membrane to cytosolic PKCϵ increased, PKC activity itself was unaltered. These data suggest that PA, but not DAG, is associated with impaired insulin action in mouse hepatocytes.


Assuntos
Diglicerídeos/metabolismo , Hepatócitos/metabolismo , Insulina/metabolismo , Ácidos Fosfatídicos/metabolismo , Transdução de Sinais , Animais , Proteínas de Transporte/metabolismo , Células Cultivadas , Diacilglicerol Quinase/genética , Diacilglicerol Quinase/metabolismo , Glicerol-3-Fosfato O-Aciltransferase/genética , Glicerol-3-Fosfato O-Aciltransferase/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Fosfolipase D/genética , Fosfolipase D/metabolismo , Proteína Quinase C/metabolismo , Proteína Companheira de mTOR Insensível à Rapamicina , Serina-Treonina Quinases TOR/metabolismo
16.
FASEB J ; 29(11): 4641-53, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26220174

RESUMO

Because hearts with a temporally induced knockout of acyl-CoA synthetase 1 (Acsl1(T-/-)) are virtually unable to oxidize fatty acids, glucose use increases 8-fold to compensate. This metabolic switch activates mechanistic target of rapamycin complex 1 (mTORC1), which initiates growth by increasing protein and RNA synthesis and fatty acid metabolism, while decreasing autophagy. Compared with controls, Acsl1(T-/-) hearts contained 3 times more mitochondria with abnormal structure and displayed a 35-43% lower respiratory function. To study the effects of mTORC1 activation on mitochondrial structure and function, mTORC1 was inhibited by treating Acsl1(T-/-) and littermate control mice with rapamycin or vehicle alone for 2 wk. Rapamycin treatment normalized mitochondrial structure, number, and the maximal respiration rate in Acsl1(T-/-) hearts, but did not improve ADP-stimulated oxygen consumption, which was likely caused by the 33-51% lower ATP synthase activity present in both vehicle- and rapamycin-treated Acsl1(T-/-) hearts. The turnover of microtubule associated protein light chain 3b in Acsl1(T-/-) hearts was 88% lower than controls, indicating a diminished rate of autophagy. Rapamycin treatment increased autophagy to a rate that was 3.1-fold higher than in controls, allowing the formation of autophagolysosomes and the clearance of damaged mitochondria. Thus, long-chain acyl-CoA synthetase isoform 1 (ACSL1) deficiency in the heart activated mTORC1, thereby inhibiting autophagy and increasing the number of damaged mitochondria.


Assuntos
Autofagia/efeitos dos fármacos , Coenzima A Ligases/deficiência , Mitocôndrias Cardíacas/metabolismo , Complexos Multiproteicos/metabolismo , Miocárdio/metabolismo , Sirolimo/farmacologia , Serina-Treonina Quinases TOR/metabolismo , Animais , Autofagia/genética , Alvo Mecanístico do Complexo 1 de Rapamicina , Camundongos , Camundongos Knockout , Mitocôndrias Cardíacas/genética , Mitocôndrias Cardíacas/patologia , Complexos Multiproteicos/genética , Miocárdio/patologia , Consumo de Oxigênio/efeitos dos fármacos , Consumo de Oxigênio/genética , ATPases Translocadoras de Prótons/metabolismo , Serina-Treonina Quinases TOR/genética
17.
Biochem J ; 471(2): 211-20, 2015 Oct 15.
Artigo em Inglês | MEDLINE | ID: mdl-26268560

RESUMO

Spermatogenesis is a highly regulated process that involves both mitotic and meiotic divisions, as well as cellular differentiation to yield mature spermatozoa from undifferentiated germinal stem cells. Although Gpat2 was originally annotated as encoding a glycerol-3-phosphate acyltransferase by sequence homology to Gpat1, GPAT2 is highly expressed in testis but not in lipogenic tissues and is not up-regulated during adipocyte differentiation. New data show that GPAT2 is required for the synthesis of piRNAs (piwi-interacting RNAs), a group of small RNAs that protect the germ cell genome from retrotransposable elements. In order to understand the relationship between GPAT2 and its role in the testis, we focused on Gpat2 expression during the first wave of mouse spermatogenesis. Gpat2 expression was analysed by qPCR (quantitative real-time PCR), in situ hybridization, immunohistochemistry and Western blotting. Gpat2 mRNA content and protein expression were maximal at 15 dpp (days post-partum) and were restricted to pachytene spermatocytes. To achieve this transient expression, both epigenetic mechanisms and trans-acting factors are involved. In vitro assays showed that Gpat2 expression correlates with DNA demethylation and histone acetylation and that it is up-regulated by retinoic acid. Epigenetic regulation by DNA methylation was confirmed in vivo in germ cells by bisulfite sequencing of the Gpat2 promoter. Consistent with the initiation of meiosis at 11 dpp, methylation decreased dramatically. Thus, Gpat2 is expressed at a specific stage of spermatogenesis, consistent with piRNA synthesis and meiosis I prophase, and its on-off expression pattern responds predominantly to epigenetic modifications.


Assuntos
Metilação de DNA/fisiologia , Epigênese Genética/fisiologia , Glicerol-3-Fosfato O-Aciltransferase/metabolismo , Prófase Meiótica I/fisiologia , Estágio Paquíteno/fisiologia , Regiões Promotoras Genéticas/fisiologia , Espermatócitos/metabolismo , Espermatogênese/fisiologia , Animais , Glicerol-3-Fosfato O-Aciltransferase/genética , Masculino , Camundongos , Camundongos Endogâmicos BALB C , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Espermatócitos/citologia
18.
J Lipid Res ; 56(8): 1572-82, 2015 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-26136511

RESUMO

Long-chain acyl-CoA synthetase 1 (ACSL1) contributes more than 90% of total cardiac ACSL activity, but its role in phospholipid synthesis has not been determined. Mice with an inducible knockout of ACSL1 (Acsl1(T-/-)) have impaired cardiac fatty acid oxidation and rely on glucose for ATP production. Because ACSL1 exhibited a strong substrate preference for linoleate, we investigated the composition of heart phospholipids. Acsl1(T-/-) hearts contained 83% less tetralinoleoyl-cardiolipin (CL), the major form present in control hearts. A stable knockdown of ACSL1 in H9c2 rat cardiomyocytes resulted in low incorporation of linoleate into CL and in diminished incorporation of palmitate and oleate into other phospholipids. Overexpression of ACSL1 in H9c2 and HEK-293 cells increased incorporation of linoleate into CL and other phospholipids. To determine whether increasing the content of linoleate in CL would improve mitochondrial respiratory function in Acsl1(T-/-) hearts, control and Acsl1(T-/-) mice were fed a high-linoleate diet; this diet normalized the amount of tetralinoleoyl-CL but did not improve respiratory function. Thus, ACSL1 is required for the normal composition of several phospholipid species in heart. Although ACSL1 determines the acyl-chain composition of heart CL, a high tetralinoleoyl-CL content may not be required for normal function.


Assuntos
Cardiolipinas/metabolismo , Coenzima A Ligases/deficiência , Mitocôndrias/metabolismo , Animais , Linhagem Celular , Respiração Celular , Coenzima A Ligases/genética , Coenzima A Ligases/metabolismo , Gorduras na Dieta/farmacologia , Ácidos Graxos/metabolismo , Técnicas de Silenciamento de Genes , Células HEK293 , Humanos , Ácido Linoleico/farmacologia , Masculino , Camundongos , Mitocôndrias/efeitos dos fármacos , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Oxirredução/efeitos dos fármacos , Transporte Proteico , Ratos
19.
Biochim Biophys Acta ; 1841(6): 880-7, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24631848

RESUMO

In mice with temporally-induced cardiac-specific deficiency of acyl-CoA synthetase-1 (Acsl1(H-/-)), the heart is unable to oxidize long-chain fatty acids and relies primarily on glucose for energy. These metabolic changes result in the development of both a spontaneous cardiac hypertrophy and increased phosphorylated S6 kinase (S6K), a substrate of the mechanistic target of rapamycin, mTOR. Doppler echocardiography revealed evidence of significant diastolic dysfunction, indicated by a reduced E/A ratio and increased mean performance index, although the deceleration time and the expression of sarco/endoplasmic reticulum calcium ATPase and phospholamban showed no difference between genotypes. To determine the role of mTOR in the development of cardiac hypertrophy, we treated Acsl1(H-/-) mice with rapamycin. Six to eight week old Acsl1(H-/-) mice and their littermate controls were given i.p. tamoxifen to eliminate cardiac Acsl1, then concomitantly treated for 10weeks with i.p. rapamycin or vehicle alone. Rapamycin completely blocked the enhanced ventricular S6K phosphorylation and cardiac hypertrophy and attenuated the expression of hypertrophy-associated fetal genes, including α-skeletal actin and B-type natriuretic peptide. mTOR activation of the related Acsl3 gene, usually associated with pathologic hypertrophy, was also attenuated in the Acsl1(H-/-) hearts, indicating that alternative pathways of fatty acid activation did not compensate for the loss of Acsl1. Compared to controls, Acsl1(H-/-) hearts exhibited an 8-fold higher uptake of 2-deoxy[1-(14)C]glucose and a 35% lower uptake of the fatty acid analog 2-bromo[1-(14)C]palmitate. These data indicate that Acsl1-deficiency causes diastolic dysfunction and that mTOR activation is linked to the development of cardiac hypertrophy in Acsl1(H-/-) mice.


Assuntos
Cardiomegalia/enzimologia , Coenzima A Ligases/genética , Insuficiência Cardíaca Diastólica/enzimologia , Sirolimo/administração & dosagem , Serina-Treonina Quinases TOR/genética , Animais , Cardiomegalia/tratamento farmacológico , Cardiomegalia/patologia , Coenzima A Ligases/deficiência , Coenzima A Ligases/metabolismo , Retículo Endoplasmático/metabolismo , Insuficiência Cardíaca Diastólica/genética , Insuficiência Cardíaca Diastólica/patologia , Humanos , Metabolismo dos Lipídeos/genética , Camundongos , Oxirredução , Tamoxifeno/farmacologia
20.
Annu Rev Nutr ; 34: 1-30, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24819326

RESUMO

Long-chain fatty acyl-coenzyme As (CoAs) are critical regulatory molecules and metabolic intermediates. The initial step in their synthesis is the activation of fatty acids by one of 13 long-chain acyl-CoA synthetase isoforms. These isoforms are regulated independently and have different tissue expression patterns and subcellular locations. Their acyl-CoA products regulate metabolic enzymes and signaling pathways, become oxidized to provide cellular energy, and are incorporated into acylated proteins and complex lipids such as triacylglycerol, phospholipids, and cholesterol esters. Their differing metabolic fates are determined by a network of proteins that channel the acyl-CoAs toward or away from specific metabolic pathways and serve as the basis for partitioning. This review evaluates the evidence for acyl-CoA partitioning by reviewing experimental data on proteins that are believed to contribute to acyl-CoA channeling, the metabolic consequences of loss of these proteins, and the potential role of maladaptive acyl-CoA partitioning in the pathogenesis of metabolic disease and carcinogenesis.


Assuntos
Acil Coenzima A/metabolismo , Membrana Celular/metabolismo , Coenzima A Ligases/metabolismo , Retículo Endoplasmático/metabolismo , Proteínas de Transporte de Ácido Graxo/metabolismo , Metabolismo dos Lipídeos , Modelos Biológicos , Animais , Membrana Celular/enzimologia , Coenzima A Ligases/genética , Retículo Endoplasmático/enzimologia , Proteínas de Transporte de Ácido Graxo/genética , Regulação da Expressão Gênica , Humanos , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Transporte Proteico
SELEÇÃO DE REFERÊNCIAS
Detalhe da pesquisa