Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
J Exp Bot ; 72(4): 1349-1369, 2021 02 24.
Artículo en Inglés | MEDLINE | ID: mdl-33130852

RESUMEN

Malonyl-CoA:flavonoid acyltransferases (MaTs) modify isoflavones, but only a few have been characterized for activity and assigned to specific physiological processes. Legume roots exude isoflavone malonates into the rhizosphere, where they are hydrolyzed into isoflavone aglycones. Soybean GmMaT2 was highly expressed in seeds, root hairs, and nodules. GmMaT2 and GmMaT4 recombinant enzymes used isoflavone 7-O-glucosides as acceptors and malonyl-CoA as an acyl donor to generate isoflavone glucoside malonates. GmMaT2 had higher activity towards isoflavone glucosides than GmMaT4. Overexpression in hairy roots of GmMaT2 and GmMaT4 produced more malonyldaidzin, malonylgenistin, and malonylglycitin, and resulted in more nodules than control. However, only GmMaT2 knockdown (KD) hairy roots showed reduced levels of malonyldaidzin, malonylgenistin, and malonylglycitin, and, likewise, reduced nodule numbers. These were consistent with the up-regulation of only GmMaT2 by rhizobial infection, and higher expression levels of early nodulation genes in GmMaT2- and GmMaT4-overexpressing roots, but lower only in GmMaT2-KD roots compared with control roots. Higher malonyl isoflavonoid levels in transgenic hairy roots were associated with higher levels of isoflavones in root exudates and more nodules, and vice versa. We suggest that GmMaT2 participates in soybean nodulation by catalyzing isoflavone malonylation and affecting malonyl isoflavone secretion for activation of Nod factor and nodulation.


Asunto(s)
Aciltransferasas/fisiología , Glycine max , Isoflavonas , Malonil Coenzima A/fisiología , Nodulación de la Raíz de la Planta , Aciltransferasas/genética , Malonil Coenzima A/genética , Glycine max/enzimología , Glycine max/genética
2.
Am J Physiol Endocrinol Metab ; 305(3): E336-47, 2013 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-23736540

RESUMEN

Carnitine palmitoyltransferase-1 (CPT-1) liver isoform, or CPT-1a, is implicated in CNS control of food intake. However, the exact brain nucleus site(s) in mediating this action of CPT-1a has not been identified. In this report, we assess the role of CPT-1a in hypothalamic ventromedial nucleus (VMN). We stereotaxically injected an adenoviral vector containing CPT-1a coding sequence into the VMN of rats to induce overexpression and activation of CPT-1a. The VMN-selective activation of CPT-1a induced an orexigenic effect, suggesting CPT-1a in the VMN is involved in the central control of feeding. Intracerebroventricular administration of etomoxir, a CPT-1 inhibitor, decreases food intake. Importantly, in the animals with VMN overexpression of a CPT-1a mutant that antagonizes the CPT-1 inhibition by etomoxir, the anorectic response to etomoxir was attenuated. This suggests that VMN is involved in mediating the anorectic effect of central inhibition of CPT-1a. In contrast, arcuate nucleus (Arc) overexpression of the mutant did not alter etomoxir-induced inhibition of food intake, suggesting that Arc CPT-1a does not play significant roles in this anorectic action. Furthermore, in the VMN, CPT-1a appears to act downstream of hypothalamic malonyl-CoA action of feeding. Finally, we show that in the VMN CPT-1 activity was altered in concert with fasting and refeeding states, supporting a physiological role of CPT-1a in mediating the control of feeding. All together, CPT-1a in the hypothalamic VMN appears to play an important role in central control of food intake. VMN-selective modulation of CPT-1a activity may therefore be a promising strategy in controlling food intake and maintaining normal body weight.


Asunto(s)
Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/fisiología , Ingestión de Alimentos/fisiología , Núcleo Hipotalámico Ventromedial/enzimología , Núcleo Hipotalámico Ventromedial/fisiología , Acilcoenzima A/metabolismo , Animales , Depresores del Apetito/farmacología , Núcleo Arqueado del Hipotálamo/metabolismo , Western Blotting , Peso Corporal/fisiología , Carnitina/análogos & derivados , Carnitina/metabolismo , Dependovirus , Activación Enzimática/efectos de los fármacos , Compuestos Epoxi/farmacología , Ayuno/fisiología , Vectores Genéticos , Hipoglucemiantes/farmacología , Inyecciones Intraventriculares , Masculino , Malonil Coenzima A/fisiología , Ratas , Ratas Sprague-Dawley
3.
Am J Physiol Regul Integr Comp Physiol ; 301(1): R209-17, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21508288

RESUMEN

Hypothalamic fatty acid metabolism is involved in central nervous system controls of feeding and energy balance. Malonyl-CoA, an intermediate of fatty acid biosynthesis, is emerging as a significant player in these processes. Notably, hypothalamic malonyl-CoA has been implicated in leptin's feeding effect. Leptin treatment increases malonyl-CoA level in the hypothalamic arcuate nucleus (Arc), and this increase is required for leptin-induced decrease in food intake. However, the intracellular downstream mediators of malonyl-CoA's feeding effect have not been identified. A primary biochemical action of malonyl-CoA is the inhibition of the acyltransferase activity of carnitine palmitoyltransferase-1 (CPT-1). In the hypothalamus, the predominant isoform of CPT-1 that possesses the acyltransferase activity is CPT-1 liver type (CPT-1a). To address the role of CPT-1a in malonyl-CoA's anorectic action, we used a recombinant adenovirus expressing a mutant CPT-1a that is insensitive to malonyl-CoA inhibition. We show that Arc overexpression of the mutant CPT-1a blocked the malonyl-CoA-mediated inhibition of CPT-1 activity. However, the overexpression of this mutant did not affect the anorectic actions of leptin or central cerulenin for which an increase in Arc malonyl-CoA level is also required. Thus, CPT-1a does not appear to be involved in the malonyl-CoA's anorectic actions induced by leptin. Furthermore, long-chain fatty acyl-CoAs, substrates of CPT-1a, dissociate from malonyl-CoA's actions in the Arc under different feeding states. Together, our results suggest that Arc intracellular mechanisms of malonyl-CoA's anorectic actions induced by leptin are independent of CPT-1a. The data suggest that target(s), rather than CPT-1a, mediates malonyl-CoA action on feeding.


Asunto(s)
Regulación del Apetito/fisiología , Carnitina O-Palmitoiltransferasa/antagonistas & inhibidores , Hipotálamo/fisiología , Leptina/fisiología , Malonil Coenzima A/fisiología , Aciltransferasas/fisiología , Animales , Carnitina O-Palmitoiltransferasa/genética , Carnitina O-Palmitoiltransferasa/fisiología , Cerulenina/metabolismo , Metabolismo Energético/fisiología , Masculino , Modelos Animales , Mutación/genética , Ratas , Ratas Sprague-Dawley
4.
mBio ; 12(1)2021 02 02.
Artículo en Inglés | MEDLINE | ID: mdl-33531402

RESUMEN

Fatty acid biosynthesis (FASII) enzymes are considered valid targets for antimicrobial drug development against the human pathogen Staphylococcus aureus However, incorporation of host fatty acids confers FASII antibiotic adaptation that compromises prospective treatments. S. aureus adapts to FASII inhibitors by first entering a nonreplicative latency period, followed by outgrowth. Here, we used transcriptional fusions and direct metabolite measurements to investigate the factors that dictate the duration of latency prior to outgrowth. We show that stringent response induction leads to repression of FASII and phospholipid synthesis genes. (p)ppGpp induction inhibits synthesis of malonyl-CoA, a molecule that derepresses FapR, a key regulator of FASII and phospholipid synthesis. Anti-FASII treatment also triggers transient expression of (p)ppGpp-regulated genes during the anti-FASII latency phase, with concomitant repression of FapR regulon expression. These effects are reversed upon outgrowth. GTP depletion, a known consequence of the stringent response, also occurs during FASII latency, and is proposed as the common signal linking these responses. We next showed that anti-FASII treatment shifts malonyl-CoA distribution between its interactants FapR and FabD, toward FapR, increasing expression of the phospholipid synthesis genes plsX and plsC during outgrowth. We conclude that components of the stringent response dictate malonyl-CoA availability in S. aureus FASII regulation, and contribute to latency prior to anti-FASII-adapted outgrowth. A combinatory approach, coupling a (p)ppGpp inducer and an anti-FASII, blocks S. aureus outgrowth, opening perspectives for bi-therapy treatment.IMPORTANCEStaphylococcus aureus is a major human bacterial pathogen for which new inhibitors are urgently needed. Antibiotic development has centered on the fatty acid synthesis (FASII) pathway, which provides the building blocks for bacterial membrane phospholipids. However, S. aureus overcomes FASII inhibition and adapts to anti-FASII by using exogenous fatty acids that are abundant in host environments. This adaptation mechanism comprises a transient latency period followed by bacterial outgrowth. Here, we use metabolite sensors and promoter reporters to show that responses to stringent conditions and to FASII inhibition intersect, in that both involve GTP and malonyl-CoA. These two signaling molecules contribute to modulating the duration of latency prior to S. aureus adaptation outgrowth. We exploit these novel findings to propose a bi-therapy treatment against staphylococcal infections.


Asunto(s)
Antibacterianos/farmacología , Ácidos Grasos/antagonistas & inhibidores , Guanosina Pentafosfato/fisiología , Guanosina Trifosfato/fisiología , Malonil Coenzima A/fisiología , Staphylococcus aureus/efectos de los fármacos , Adaptación Fisiológica/efectos de los fármacos , Ácidos Grasos/biosíntesis , Humanos , Malonil Coenzima A/análisis , Mupirocina/farmacología , Fosfolípidos/biosíntesis , Infecciones Estafilocócicas/tratamiento farmacológico , Staphylococcus aureus/fisiología
5.
Int J Obes (Lond) ; 32 Suppl 4: S49-54, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18719599

RESUMEN

Energy balance is monitored by the hypothalamus. Malonyl-CoA, an intermediate in fatty acid synthesis, serves as an indicator of energy status in the hypothalamic neurons. The cellular malonyl-CoA level is determined by its rate of synthesis, catalyzed by acetyl-CoA carboxylase (ACC), and rate of removal, by fatty acid synthase (FAS). Malonyl-CoA functions in the hypothalamic neurons that express orexigenic and anorexigenic neuropeptides. Inhibitors of FAS, administered systemically or intracerebroventricularly to mice, increase hypothalamic malony-CoA and suppress food intake. Recent evidence suggests that the changes of hypothalamic malonyl-CoA during feeding and fasting cycles are caused by changes in the phosphorylation state and activity of ACC mediated via 5'-AMP-activated protein kinase (AMPK). Stereotactic delivery of a viral malonyl-CoA decarboxylase (MCD) vector into the ventral hypothalamus lowers malonyl-CoA and increases food intake. Fasting decreases hypothalamic malonyl-CoA and refeeding increases hypothalamic malonyl-CoA, to alter feeding behavior in the predicted manner. Malonyl-CoA level is under the control of AMP kinase which phosphorylates/inactivates ACC. Malonyl-CoA is an inhibitor of carnitine palmitoyl-CoA transferase-1 (CPT1), an outer mitochondrial membrane enzyme that regulates entry into, and oxidation of fatty acids, by mitochondria. CPT1c, a recently discovered, brain-specific enzyme expressed in the hypothalamus, has high sequence similarity to liver/muscle CPT1a/b and binds malonyl-CoA, but does not catalyze the prototypical reaction. This suggests that CPT1c has a unique function or activation mechanism. CPT1c knockout (KO) mice have lower food intake, weigh less and have less body fat, consistent with the role as an energy-sensing malonyl-CoA target. Paradoxically, CPT1c protects against the effects of a high-fat diet. CPT1cKO mice exhibit decreased rates of fatty acid oxidation, consistent with their increased susceptibility to diet-induced obesity. We suggest that CPT1c may be a downstream target of malonyl-CoA that regulates energy homeostasis.


Asunto(s)
Ingestión de Alimentos/fisiología , Metabolismo Energético/fisiología , Hipotálamo/enzimología , Malonil Coenzima A/metabolismo , Acetil-CoA Carboxilasa/metabolismo , Animales , Carboxiliasas/metabolismo , Carnitina O-Palmitoiltransferasa/metabolismo , Ácido Graso Sintasas/metabolismo , Hipotálamo/fisiología , Malonil Coenzima A/fisiología , Ratones
6.
Cardiovasc Res ; 73(2): 278-87, 2007 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-17126822

RESUMEN

Obesity is an important contributor to the risk of developing insulin resistance, diabetes, and heart disease. Alterations in tissue levels of malonyl-CoA have the potential to impact on the severity of a number of these disorders. This review will focus on the emerging role of malonyl-CoA as a key "metabolic effector" of both obesity and cardiac fatty acid oxidation. In addition to being a substrate for fatty acid biosynthesis, malonyl-CoA is a potent inhibitor of mitochondrial carnitine palmitoyltransferase (CPT) 1, a key enzyme involved in mitochondrial fatty acid uptake. A decrease in myocardial malonyl-CoA levels and an increase in CPT1 activity contribute to an increase in cardiac fatty acid oxidation. An increase in malonyl-CoA degradation due to increased malonyl-CoA decarboxylase (MCD) activity may be one mechanism responsible for this decrease in malonyl-CoA. Another mechanism involves the inhibition of acetyl-CoA carboxylase (ACC) synthesis of malonyl-CoA, due to AMP-activated protein kinase (AMPK) phosphorylation of ACC. Recent studies have demonstrated a role of malonyl-CoA in the hypothalamus as a regulator of food intake. Increases in hypothalamic malonyl-CoA and inhibition of CPT1 are associated with a decrease in food intake in mice and rats, while a decrease in hypothalamic malonyl-CoA increases food intake and weight gain. The exact mechanism(s) responsible for these effects of malonyl-CoA are not clear, but have been proposed to be due to an increase in the levels of long chain acyl CoA, which occurs as a result of malonyl-CoA inhibition of CPT1. Both hypothalamic and cardiac studies have demonstrated that control of malonyl-CoA levels has an important impact on obesity and heart disease. Targeting enzymes that control malonyl-CoA levels may be an important therapeutic approach to treating heart disease and obesity.


Asunto(s)
Regulación del Apetito/fisiología , Cardiopatías/metabolismo , Hipotálamo/fisiología , Malonil Coenzima A/fisiología , Obesidad/metabolismo , Proteínas Quinasas Activadas por AMP , Acetil-CoA Carboxilasa/metabolismo , Acilcoenzima A/metabolismo , Animales , Carnitina O-Palmitoiltransferasa/metabolismo , Metabolismo Energético , Humanos , Complejos Multienzimáticos/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo
7.
Diabetes ; 55 Suppl 2: S16-23, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17130640

RESUMEN

Fatty acids (FAs) and other lipid molecules are important for many cellular functions, including vesicle exocytosis. For the pancreatic beta-cell, while the presence of some FAs is essential for glucose-stimulated insulin secretion, FAs have enormous capacity to amplify glucose-stimulated insulin secretion, which is particularly operative in situations of beta-cell compensation for insulin resistance. In this review, we propose that FAs do this via three interdependent processes, which we have assigned to a "trident model" of beta-cell lipid signaling. The first two arms of the model implicate intracellular metabolism of FAs, whereas the third is related to membrane free fatty acid receptor (FFAR) activation. The first arm involves the AMP-activated protein kinase/malonyl-CoA/long-chain acyl-CoA (LC-CoA) signaling network in which glucose, together with other anaplerotic fuels, increases cytosolic malonyl-CoA, which inhibits FA partitioning into oxidation, thus increasing the availability of LC-CoA for signaling purposes. The second involves glucose-responsive triglyceride (TG)/free fatty acid (FFA) cycling. In this pathway, glucose promotes LC-CoA esterification to complex lipids such as TG and diacylglycerol, concomitant with glucose stimulation of lipolysis of the esterification products, with renewal of the intracellular FFA pool for reactivation to LC-CoA. The third arm involves FFA stimulation of the G-protein-coupled receptor GPR40/FFAR1, which results in enhancement of glucose-stimulated accumulation of cytosolic Ca2+ and consequently insulin secretion. It is possible that FFA released by the lipolysis arm of TG/FFA cycling is partly "secreted" and, via an autocrine/paracrine mechanism, is additive to exogenous FFAs in activating the FFAR1 pathway. Glucose-stimulated release of arachidonic acid from phospholipids by calcium-independent phospholipase A2 and/or from TG/FFA cycling may also be involved. Improved knowledge of lipid signaling in the beta-cell will allow a better understanding of the mechanisms of beta-cell compensation and failure in diabetes.


Asunto(s)
Ácidos Grasos no Esterificados/fisiología , Células Secretoras de Insulina/metabolismo , Insulina/metabolismo , Acilcoenzima A/fisiología , Animales , Humanos , Secreción de Insulina , Malonil Coenzima A/fisiología , Modelos Biológicos , Fosfolipasas A/fisiología , Fosfolipasas A2 , Transducción de Señal/fisiología
8.
Arch Physiol Biochem ; 113(1): 13-24, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17522981

RESUMEN

There seems to be an association between increased concentrations of malonyl coenzyme A (malonyl CoA) in skeletal muscle and diabetes and/or insulin resistance. The purpose of the current study was to test the hypothesis that treatments designed to manipulate malonyl CoA concentrations would affect insulin-stimulated glucose transport in cultured C2C12 myotubes. We assessed glucose transport after polyamine-mediated delivery of malonyl CoA to myotubes, after incubation with dichloroacetate (which reportedly increases malonyl CoA levels), or after exposure of myotubes to 2-bromopalmitate, a carnitine palmitoyl transferase I inhibitor. All three of these treatments prevented stimulation of glucose transport by insulin. We also assayed glucose transport after 30 min of inhibition of acetyl coenzyme A carboxylase (ACC), the enzyme which catalyzes the production of malonyl CoA. Three unrelated ACC inhibitors (diclofop, clethodim, and Pfizer CP-640186) all enhanced insulin-stimulated glucose transport. However, none of the treatments designed to manipulate malonyl CoA concentrations altered markers of proximal insulin signaling through Akt. The findings support the hypothesis that acute changes in malonyl CoA concentrations affect insulin action in muscle cells but suggest that the effects do not involve alterations in proximal insulin signaling.


Asunto(s)
Glucosa/metabolismo , Insulina/fisiología , Malonil Coenzima A/fisiología , Fibras Musculares Esqueléticas/enzimología , Animales , Transporte Biológico Activo/efectos de los fármacos , Transporte Biológico Activo/fisiología , Bovinos , Línea Celular , Ácido Dicloroacético/farmacología , Ratones , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo
9.
Nutr Rev ; 64(8): 379-83, 2006 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16958315

RESUMEN

In animals, food intake and therefore energy balance is regulated by a center in the hypothalamus of the brain. Neurons there release appetite-inhibiting (anorexigenic) or appetite-stimulating (orexigenic) peptide hormones according to whether energy intake exceeds or is less than expenditure, respectively. Recent evidence for the "malonyl coenzyme A hypothesis" showed that the level of malonyl coenzyme A (MalCoA) in the arcuate nucleus of the hypothalamus determines the stimulation or inhibition of food intake. A high level of MalCoA, indicative of energy surplus, signals the release of anorexigenic neuropeptides, resulting in decreased food intake; a low level of MalCoA, due to an energy deficit such as during fasting, signals the release of orexigenic neuropeptides, stimulating food intake.


Asunto(s)
Ingestión de Energía/fisiología , Conducta Alimentaria/fisiología , Hipotálamo/fisiología , Malonil Coenzima A/fisiología , Animales , Metabolismo Energético/fisiología , Humanos , Ratones , Modelos Animales
10.
Cancer Res ; 60(2): 213-8, 2000 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-10667561

RESUMEN

A biologically aggressive subset of human breast cancers and other malignancies is characterized by elevated fatty-acid synthase (FAS) enzyme expression, elevated fatty acid (FA) synthesis, and selective sensitivity to pharmacological inhibition of FAS activity by cerulenin or the novel compound C75. In this study, inhibition of FA synthesis at the physiologically regulated step of carboxylation of acetyl-CoA to malonyl-CoA by 5-(tetradecyloxy)-2-furoic acid (TOFA) was not cytotoxic to breast cancer cells in clonogenic assays. FAS inhibitors induced a rapid increase in intracellular malonyl-CoA to several fold above control levels, whereas TOFA reduced intracellular malonyl-CoA by 60%. Simultaneous exposure of breast cancer cells to TOFA and an FAS inhibitor resulted in significantly reduced cytotoxicity and apoptosis. Subcutaneous xenografts of MCF7 breast cancer cells in nude mice treated with C75 showed FA synthesis inhibition, apoptosis, and inhibition of tumor growth to less than 1/8 of control volumes, without comparable toxicity in normal tissues. The data suggest that differences in intermediary metabolism render tumor cells susceptible to toxic fluxes in malonyl-CoA, both in vitro and in vivo.


Asunto(s)
Antineoplásicos/toxicidad , Neoplasias de la Mama/patología , Cerulenina/toxicidad , Ácido Graso Sintasas/antagonistas & inhibidores , Furanos/farmacología , Malonil Coenzima A/fisiología , Animales , Antineoplásicos/uso terapéutico , Neoplasias de la Mama/tratamiento farmacológico , Neoplasias de la Mama/enzimología , Supervivencia Celular/efectos de los fármacos , Cerulenina/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Inhibidores Enzimáticos/toxicidad , Femenino , Humanos , Hipolipemiantes/farmacología , Ratones , Ratones Desnudos , Células Tumorales Cultivadas , Ensayo de Tumor de Célula Madre
11.
Diabetes ; 51 Suppl 3: S405-13, 2002 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-12475783

RESUMEN

Beta-cells possess inherent mechanisms to adapt to overnutrition and the prevailing concentrations of glucose, fatty acids, and other fuels to maintain glucose homeostasis. However, this is balanced by potentially harmful actions of the same nutrients. Both glucose and fatty acids may cause good/adaptive or evil/toxic actions on the beta-cell, depending on their concentrations and the time during which they are elevated. Chronic high glucose dramatically influences beta-cell lipid metabolism via substrate availability, changes in the activity and expression of enzymes of glucose and lipid metabolism, and modifications in the expression level of key transcription factors. We discuss here the emerging view that beta-cell "glucotoxicity" is in part indirectly caused by "lipotoxicity," and that beta-cell abnormalities will become particularly apparent when both glucose and circulating fatty acids are high. We support the concept that elevated glucose and fatty acids synergize in causing toxicity in islets and other organs, a process that may be instrumental in the pleiotropic defects associated with the metabolic syndrome and type 1 and type 2 diabetes. The mechanisms by which hyperglycemia and hyperlipidemia alter insulin secretion are discussed and a model of beta-cell "glucolipotoxicity" that implicates alterations in beta-cell malonyl-CoA concentrations; peroxisome proliferator-activated receptor-alpha and -gamma and sterol regulatory element binding protein-1c expression; and lipid partitioning is proposed.


Asunto(s)
Diabetes Mellitus/etiología , Glucosa/metabolismo , Islotes Pancreáticos/fisiología , Metabolismo de los Lípidos , Malonil Coenzima A/fisiología , Transducción de Señal/fisiología , Adaptación Fisiológica/fisiología , Animales , Humanos
12.
Trends Endocrinol Metab ; 11(2): 49-53, 2000 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-10675890

RESUMEN

For two decades it has been assumed that inhibition of carnitine palmitoyltransferase I (CPT-I) by malonyl-CoA represents the main regulatory mechanism of liver ketogenesis. However, recent evidence indicates that CPT-I activity is also controlled by interactions between mitochondria and cytoskeletal components. This newly recognized mechanism emphasizes the emerging role of the cytoskeleton in the regulation of metabolic pathways.


Asunto(s)
Citoesqueleto/fisiología , Ácidos Grasos/metabolismo , Mitocondrias Hepáticas/metabolismo , Animales , Transporte Biológico/fisiología , Carnitina O-Palmitoiltransferasa/fisiología , Humanos , Malonil Coenzima A/fisiología
13.
Anim Sci J ; 86(2): 181-8, 2015 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-25040023

RESUMEN

Depression induces anorexia, leading to suppressed feeding behaviors and energy intake. Previously, we revealed that chronic social defeat induced a mild suppression of feeding in rats with elevated levels of hypothalamic malonyl-CoA which regulates feeding. Therefore, we attempted to elucidate the effects of chronic mild food restriction on behavior and on hypothalamic malonyl-CoA. The chronic mild food restricted rats were fed a restricted diet approximately 80% to 90% amount of diet compared to the control for 5 weeks. Ratios of restriction were adjusted with feed consumption in the chronic social defeat stressed rats. Chronic mild food restricted rats exhibited a suppression of body weight gain similar to that of the chronic social defeat stressed rats. Also these rats showed increased time spent in the center area of an open field (OF), prolonged immobility time in forced swim, increased phosphorylation of hypothalamic adenosine monophosphate-activated protein kinase (AMPK) and acetyl-CoA carboxylase and a decreased concentration of hypothalamic malonyl-CoA. Weight of the adrenal glands, locomotion in an OF, mitogen-activated protein kinase cascade and calcium/calmodulin-dependent protein kinases II in the hippocampus were not affected by chronic mild food restriction. Our findings suggest that chronic mild food restriction activates AMPK following a decreased hypothalamic malonyl-CoA.


Asunto(s)
Conducta Animal/fisiología , Ingestión de Alimentos/fisiología , Privación de Alimentos/fisiología , Hipotálamo/metabolismo , Malonil Coenzima A/metabolismo , Transducción de Señal/fisiología , Proteínas Quinasas Activadas por AMP/metabolismo , Acetil-CoA Carboxilasa/metabolismo , Animales , Masculino , Malonil Coenzima A/fisiología , Fosforilación , Ratas Wistar , Estrés Psicológico/fisiopatología
14.
J Basic Clin Physiol Pharmacol ; 9(2-4): 295-308, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-10212840

RESUMEN

Malonyl CoA is an inhibitor of carnitine palmitoyl transferase 1 (CPT1), the enzyme that regulates the transfer of long chain fatty acyl CoA into mitochondria. By virtue of this effect, it is thought to play a key role in regulating fatty acid oxidation. Thus, when the supply of glucose to muscle is increased, malonyl CoA levels increase in keeping with a decreased need for fatty acid oxidation, and fatty acids are preferentially esterified to form diaglycerol and triglycerides. In contrast, during exercise, when the need for fatty acid oxidation is increased, malonyl CoA levels fall. Changes in glucose supply regulate malonyl CoA by modulating the concentration of cytosolic citrate, an allosteric activator of acetyl CoA carboxylase (ACC), the rate-limiting enzyme for malonyl CoA formation and a precursor of its substrate cytosolic acetyl CoA. Conversely, exercise lowers the concentration of malonyl CoA, by activating an AMP-activated protein kinase, which phosphorylates and inhibits ACC. A number of reports have linked sustained increases in the concentration of malonyl CoA in muscle to insulin resistance. In this paper, we review these reports, as well as the notion that changes in malonyl CoA contribute to the increases in long chain fatty acyl CoA, (LCFA CoA), diacylglycerol and triglyceride content and changes in protein kinase C activity and distribution observed in insulin-resistant muscle. We also review the implications of the malonyl CoA/LCFA CoA hypothesis to two other proposed mechanisms for insulin resistance, the glucose-fatty acid cycle and the hexosamine theory.


Asunto(s)
Acilcoenzima A/biosíntesis , Resistencia a la Insulina/fisiología , Malonil Coenzima A/metabolismo , Músculo Esquelético/metabolismo , Animales , Glucemia/metabolismo , Ácidos Grasos/metabolismo , Glucólisis/fisiología , Hexosaminas/biosíntesis , Insulina/farmacología , Malonil Coenzima A/fisiología , Ratones , Músculo Esquelético/efectos de los fármacos , Ratas
SELECCIÓN DE REFERENCIAS
Detalles de la búsqueda