23 research outputs found

    Estratègies de modulació de l'oxidació d'àcids grassos com a tractament per combatre l'obesitat

    Get PDF
    L'estil de vida actual, amb dietes d'alt contingut calòric i falta d'exercici físic, fa que la incidència d'obesitat s'incrementi notablement. Augmentar la degradació de greixos o bé reduir la ingesta calòrica poden ser potencials estratègies terapèutiques. L'enzim carnitina palmitoïltransferasa I (CPT1) és el pas limitant de l'oxidació dels àcids grassos. En aquest article, es mostra com la modulació de la seva activitat en diferents teixits, com el fetge, el teixit adipós o l'hipotàlem, pot ser clau a l'hora d'augmentar la despesa energètica i controlar la ingesta d'aliments

    CPT1C promotes human mesenchymal stem cells survival under glucose deprivation through the modulation of autophagy.

    Get PDF
    Human mesenchymal stem cells (hMSCs) are widely used in regenerative medicine. In some applications, they must survive under low nutrient conditions engendered by avascularity. Strategies to improve hMSCs survival may be of high relevance in tissue engineering. Carnitine palmitoyltransferase 1 C (CPT1C) is a pseudoenzyme exclusively expressed in neurons and cancer cells. In the present study, we show that CPT1C is also expressed in hMSCs and protects them against glucose starvation, glycolysis inhibition, and oxygen/glucose deprivation. CPT1C overexpression in hMSCs did not increase fatty acid oxidation capacity, indicating that the role of CPT1C in these cells is different from that described in tumor cells. The increased survival of CPT1C-overexpressing hMSCs observed during glucose deficiency was found to be the result of autophagy enhancement, leading to a greater number of lipid droplets and increased intracellular ATP levels. In fact, inhibition of autophagy or lipolysis was observed to completely block the protective effects of CPT1C. Our results indicate that CPT1C-mediated autophagy enhancement in glucose deprivation conditions allows a greater availability of lipids to be used as fuel substrate for ATP generation, revealing a new role of CPT1C in stem cell adaptation to low nutrient environments

    Fatty acid metabolism and the basis of brown adipose tissue function

    Get PDF
    Obesity has reached epidemic proportions, leading to severe associated pathologies such as insulin resistance, cardiovascular disease, cancer and type 2 diabetes. Adipose tissue has become crucial due to its involvement in the pathogenesis of obesity-induced insulin resistance, and traditionally white adipose tissue has captured the most attention. However in the last decade the presence and activity of heat-generating brown adipose tissue (BAT) in adult humans has been rediscovered. BAT decreases with age and in obese and diabetic patients. It has thus attracted strong scientific interest, and any strategy to increase its mass or activity might lead to new therapeutic approaches to obesity and associated metabolic diseases. In this review we highlight the mechanisms of fatty acid uptake, trafficking and oxidation in brown fat thermogenesis. We focus on BAT's morphological and functional characteristics and fatty acid synthesis, storage, oxidation and use as a source of energy

    (−)-UB006: A new fatty acid synthase inhibitor and cytotoxic agent without anorexic side effects

    Get PDF
    C75 is a synthetic anticancer drug that inhibits fatty acid synthase (FAS) and shows a potent anorexigenic side effect. In order to find new cytotoxic compounds that do not impact food intake, we synthesized a new family of C75 derivatives. The most promising anticancer compound among them was UB006 ((4SR,5SR)-4-(hydroxymethyl)-3-methylene-5-octyldihydrofuran-2(3H)-one). The effects of this compound on cytotoxicity, food intake and body weight were studied in UB006 racemic mixture and in both its enantiomers separately. The results showed that both enantiomers inhibit FAS activity and have potent cytotoxic effects in several tumour cell lines, such as the ovarian cell cancer line OVCAR-3. The (−)-UB006 enantiomer's cytotoxic effect on OVCAR-3 was 40-fold higher than that of racemic C75, and 2- and 38-fold higher than that of the racemic mixture and its opposite enantiomer, respectively. This cytotoxic effect on the OVCAR-3 cell line involves mechanisms that reduce mitochondrial respiratory capacity and ATP production, DDIT4/REDD1 upregulation, mTOR activity inhibition, and caspase-3 activation, resulting in apoptosis. In addition, central and peripheral administration of (+)-UB006 or (−)-UB006 into rats and mice did not affect food intake or body weight. Altogether, our data support the discovery of a new potential anticancer compound (−)-UB006 that has no anorexigenic side effects

    Ghrelin causes a decline in GABA release by reducing fatty acid oxidation in cortex

    Get PDF
    Lipid metabolism, specifically fatty acid oxidation (FAO) mediated by carnitine palmitoyltransferase (CPT) 1A, has been described to be an important actor of ghrelin action in hypothalamus. However, it is not known whether CPT1A and FAO mediate the effect of ghrelin on the cortex. Here, we show that ghrelin produces a differential effect on CPT1 activity and γ-aminobutyric acid (GABA) metabolism in the hypothalamus and cortex of mice. In the hypothalamus, ghrelin enhances CPT1A activity while GABA transaminase (GABAT) activity, a key enzyme in GABA shunt metabolism, is unaltered. However, in cortex CPT1A activity and GABAT activity are reduced after ghrelin treatment. Furthermore, in primary cortical neurons, ghrelin reduces GABA release through a CPT1A reduction. By using CPT1A floxed mice, we have observed that genetic ablation of CPT1A recapitulates the effect of ghrelin on GABA release in cortical neurons, inducing reductions in mitochondrial oxygen consumption, cell content of citrate and α-ketoglutarate, and GABA shunt enzyme activity. Taken together, these observations indicate that ghrelin-induced changes in CPT1A activity modulate mitochondrial function, yielding changes in GABA metabolism. This evidence suggests that the action of ghrelin on GABA release is region specific within the brain, providing a basis for differential effects of ghrelin in the central nervous system. Keywords: Ghrelin, GABA, Fatty acid oxidation, CPT1A, Cortical neuron

    Long-Term Increased Carnitine Palmitoyltransferase 1A Expression in Ventromedial Hypotalamus Causes Hyperphagia and Alters the Hypothalamic Lipidomic Profile

    Get PDF
    Lipid metabolism in the ventromedial hypothalamus (VMH) has emerged as a crucial pathway in the regulation of feeding and energy homeostasis. Carnitine palmitoyltransferase (CPT) 1A is the rate-limiting enzyme in mitochondrial fatty acid boxidation and it has been proposed as a crucial mediator of fasting and ghrelin orexigenic signalling. However, the relationship between changes in CPT1A activity and the intracellular downstream effectors in the VMH that contribute to appetite modulation is not fully understood. To this end, we examined the effect of long-term expression of a permanently activated CPT1A isoform by using an adeno-associated viral vector injected into the VMH of rats. Peripherally, this procedure provoked hyperghrelinemia and hyperphagia, which led to overweight, hyperglycemia and insulin resistance. In the mediobasal hypothalamus (MBH), long-term CPT1AM expression in the VMH did not modify acyl-CoA or malonyl-CoA levels. However, it altered the MBH lipidomic profile since ceramides and sphingolipids increased and phospholipids decreased. Furthermore, we detected increased vesicular c-aminobutyric acid transporter (VGAT) and reduced vesicular glutamate transporter 2 (VGLUT2) expressions, both transporters involved in this orexigenic signal. Taken together, these observations indicate that CPT1A contributes to the regulation of feeding by modulating the expression of neurotransmitter transporters and lipid components that influence the orexigenic pathways in VMH

    Role of carnitine palmitoyltransferase 1A as a downstream effector of ghrelin in cortical neurons and hypothalamus

    Get PDF
    [eng] Previous studies have reported the importance of carnitine palmitoyltransferase (CPT) 1A as an essential part of downstream ghrelin signaling in the central nervous system (CNS) for the control of food intake. Lipid metabolism in the ventromedial hypothalamus (VMH) has emerged as a crucial pathway in the regulation of feeding and energy homeostasis. However, the relationship between changes in CPT1A activity and the intracellular downstream effectors in the VMH that contribute to appetite modulation is not fully understood, nor its possible involvement in central extra-hypothalamic functions of ghrelin. In this work, we examine the effect of long-term expression of a permanently activated CPT1A isoform (CPT1AM) by using an adeno-associated viral vector, injected into the VMH of rats. CPT1AM overexpression produces a sustained increase in food intake which leads to overweight. Mechanistically, CPT1AM alters the lipidomic profile of mediobasal hypothalamus, provoking an increase in ceramides and sphingolipids and a decrease in phospholipids. Furthermore, we detect increased vesicular y-aminobutyric acid transporter (VGAT) and reduced vesicular glutamate transporter 2 (VGLUT2) expressions. These changes are noteworthy, since both GABA and glutamate have been proposed to be mediators in food intake control. This signature led us to assess the effect of ghrelin in GABAergic neurons. Ghrelin reduces fatty acid oxidation, mitochondrial respiration and mitochondrial reactive oxygen species formation in GT1-7 cells. Moreover, ghrelin produces a reduction in GABA release from primary cortical neurons which is blocked by both pharmacological and genetic inhibition of CPT1A. In addition, ghrelin produces a reduction in: (1) mitochondrial oxygen consumption, (2) citrate and a-ketoglutarate cellular content and (3) GABA shunt connecting TCA cycle and releasable GABA pool. Taken together, these observations indicate that CPT1A contributes to the regulation of feeding by modulating the expression of neurotransmitter transporters and lipid components that influence the orexigenic pathways in VMH. Moreover, it seems that ghrelin and changes in CPT1A activity modulate mitochondrial function, yielding changes in GABA metabolism, which affect eventually to GABAergic neurotransmission.[cat] Estudis previs havien destacat la importància de la carnitina palmitoïltransferasa (CPT) 1A com a part essencial de la senyalització de la grelina al sistema nerviós central (SNC) per al control de la ingesta alimentària. El metabolisme lipídic en el nucli ventromedial de l'hipotàlem (VMH) ha emergit com una via crucial per a la regulació de la ingesta i l'homeòstasi energètica. Això no obstant, la relació entre els canvis en l'activitat de la CPT1A i els efectors intrace•ulars subjacents en el VMH que contribueixen a la modulació de la gana no són totalment compresos, com tampoc la seva involucració en les funcions centrals extrahipotalàmiques de la grelina. En aquest treball, hem examinat l'efecte a llarg termini de l'expressió d'una isoforma mutada permanentment activa de la CPT1A (CPT1AM), fent ús d'un vector víric adeno-associat, injectat en el VMH de rates. La sobreexpressió de la CPT1AM produeix un augment sostingut de la ingesta alimentària que hi promou sobrepès. En la descripció mecanística, la CPT1AM altera el perfil lipidòmic del hipotàlem mediobasal, induint-hi un augment de ceramides i esfingolípids i alhora una reducció en fosfolípids. A més a més, hem detectat un augment en les expressions del transportador vesicular de l'àcid y-aminobutíric (GABA) (VGAT, en les seves sigles en anglès) i una reducció del transportador vesicular de glutamat 2 (VGLUT2, en les seves sigles en anglès). Aquests canvis són destacables, ja que tant GABA como glutamat han sigut proposats com a mediadors en el control de la ingesta alimentària. Aquestes observacions ens dugueren a estudiar l'efecte de la grelina en neurones GABAèrgiques. La grelina redueix l'oxidació d'àcids grassos, la respiració mitocondrial i la formació d'espècies reactives d'oxigen en cèl•lules GT1-7. D'afegitó, la grelina produeix una reducció en l'alliberament de GABA de neurones corticals primàries, la qual cosa és blocada tant per inhibició genètica i farmacològica de la CPT1A. A més a més, la grelina hi produeix una reducció: (1) del consum mitocondrial d'oxigen, (2) del contingut cel•lular de citrat i d'a-cetoglutarat i (3) de la via de desviació de GABA que connecta el cicle dels àcids tricarboxílics i el contingut alliberable de GABA. En conjunt, aquestes observacions indiquen que la CPT1A contribueix a la regulació de la ingesta amb la modulació de l'expressió de transportadors involucrats en la neurotransmissió i la modulació de components lipídics que influencien les vies orexigèniques del VMH. A més a més, sembla que la grelina i els canvis en l'activitat CPT1A modulen la funció mitocondrial, obtenint-ne canvis al metabolisme del GABA, que afecten en darrera instància a la neurotransmissió GABAèrgica

    Role of carnitine palmitoyltransferase 1A as a downstream effector of ghrelin in cortical neurons and hypothalamus

    No full text
    Previous studies have reported the importance of carnitine palmitoyltransferase (CPT) 1A as an essential part of downstream ghrelin signaling in the central nervous system (CNS) for the control of food intake. Lipid metabolism in the ventromedial hypothalamus (VMH) has emerged as a crucial pathway in the regulation of feeding and energy homeostasis. However, the relationship between changes in CPT1A activity and the intracellular downstream effectors in the VMH that contribute to appetite modulation is not fully understood, nor its possible involvement in central extra-hypothalamic functions of ghrelin. In this work, we examine the effect of long-term expression of a permanently activated CPT1A isoform (CPT1AM) by using an adeno-associated viral vector, injected into the VMH of rats. CPT1AM overexpression produces a sustained increase in food intake which leads to overweight. Mechanistically, CPT1AM alters the lipidomic profile of mediobasal hypothalamus, provoking an increase in ceramides and sphingolipids and a decrease in phospholipids. Furthermore, we detect increased vesicular y-aminobutyric acid transporter (VGAT) and reduced vesicular glutamate transporter 2 (VGLUT2) expressions. These changes are noteworthy, since both GABA and glutamate have been proposed to be mediators in food intake control. This signature led us to assess the effect of ghrelin in GABAergic neurons. Ghrelin reduces fatty acid oxidation, mitochondrial respiration and mitochondrial reactive oxygen species formation in GT1-7 cells. Moreover, ghrelin produces a reduction in GABA release from primary cortical neurons which is blocked by both pharmacological and genetic inhibition of CPT1A. In addition, ghrelin produces a reduction in: (1) mitochondrial oxygen consumption, (2) citrate and a-ketoglutarate cellular content and (3) GABA shunt connecting TCA cycle and releasable GABA pool. Taken together, these observations indicate that CPT1A contributes to the regulation of feeding by modulating the expression of neurotransmitter transporters and lipid components that influence the orexigenic pathways in VMH. Moreover, it seems that ghrelin and changes in CPT1A activity modulate mitochondrial function, yielding changes in GABA metabolism, which affect eventually to GABAergic neurotransmission.Estudis previs havien destacat la importància de la carnitina palmitoïltransferasa (CPT) 1A com a part essencial de la senyalització de la grelina al sistema nerviós central (SNC) per al control de la ingesta alimentària. El metabolisme lipídic en el nucli ventromedial de l'hipotàlem (VMH) ha emergit com una via crucial per a la regulació de la ingesta i l'homeòstasi energètica. Això no obstant, la relació entre els canvis en l'activitat de la CPT1A i els efectors intrace•ulars subjacents en el VMH que contribueixen a la modulació de la gana no són totalment compresos, com tampoc la seva involucració en les funcions centrals extrahipotalàmiques de la grelina. En aquest treball, hem examinat l'efecte a llarg termini de l'expressió d'una isoforma mutada permanentment activa de la CPT1A (CPT1AM), fent ús d'un vector víric adeno-associat, injectat en el VMH de rates. La sobreexpressió de la CPT1AM produeix un augment sostingut de la ingesta alimentària que hi promou sobrepès. En la descripció mecanística, la CPT1AM altera el perfil lipidòmic del hipotàlem mediobasal, induint-hi un augment de ceramides i esfingolípids i alhora una reducció en fosfolípids. A més a més, hem detectat un augment en les expressions del transportador vesicular de l'àcid y-aminobutíric (GABA) (VGAT, en les seves sigles en anglès) i una reducció del transportador vesicular de glutamat 2 (VGLUT2, en les seves sigles en anglès). Aquests canvis són destacables, ja que tant GABA como glutamat han sigut proposats com a mediadors en el control de la ingesta alimentària. Aquestes observacions ens dugueren a estudiar l'efecte de la grelina en neurones GABAèrgiques. La grelina redueix l'oxidació d'àcids grassos, la respiració mitocondrial i la formació d'espècies reactives d'oxigen en cèl•lules GT1-7. D'afegitó, la grelina produeix una reducció en l'alliberament de GABA de neurones corticals primàries, la qual cosa és blocada tant per inhibició genètica i farmacològica de la CPT1A. A més a més, la grelina hi produeix una reducció: (1) del consum mitocondrial d'oxigen, (2) del contingut cel•lular de citrat i d'a-cetoglutarat i (3) de la via de desviació de GABA que connecta el cicle dels àcids tricarboxílics i el contingut alliberable de GABA. En conjunt, aquestes observacions indiquen que la CPT1A contribueix a la regulació de la ingesta amb la modulació de l'expressió de transportadors involucrats en la neurotransmissió i la modulació de components lipídics que influencien les vies orexigèniques del VMH. A més a més, sembla que la grelina i els canvis en l'activitat CPT1A modulen la funció mitocondrial, obtenint-ne canvis al metabolisme del GABA, que afecten en darrera instància a la neurotransmissió GABAèrgica

    Mitochondrial fatty acid oxidation in obesity

    Get PDF
    Significance: Current lifestyles with high-energy diets and little exercise are triggering an alarming growth in obesity. Excess of adiposity is leading to severe increases in associated pathologies, such as insulin resistance, type 2 diabetes, atherosclerosis, cancer, arthritis, asthma, and hypertension. This, together with the lack of efficient obesity drugs, is the driving force behind much research. Recent Advances: Traditional anti-obesity strategies focused on reducing food intake and increasing physical activity. However, recent results suggest that enhancing cellular energy expenditure may be an attractive alternative therapy. Critical Issues: This review evaluates recent discoveries regarding mitochondrial fatty acid oxidation (FAO) and its potential as a therapy for obesity. We focus on the still controversial beneficial effects of increased FAO in liver and muscle, recent studies on how to potentiate adipose tissue energy expenditure, and the different hypotheses involving FAO and the reactive oxygen species production in the hypothalamic control of food intake. Future Directions: The present review aims to provide an overview of novel anti-obesity strategies that target mitochondrial FAO and that will definitively be of high interest in the future research to fight against obesity-related disorders

    Mitochondrial fatty acid oxidation in obesity

    No full text
    Significance: Current lifestyles with high-energy diets and little exercise are triggering an alarming growth in obesity. Excess of adiposity is leading to severe increases in associated pathologies, such as insulin resistance, type 2 diabetes, atherosclerosis, cancer, arthritis, asthma, and hypertension. This, together with the lack of efficient obesity drugs, is the driving force behind much research. Recent Advances: Traditional anti-obesity strategies focused on reducing food intake and increasing physical activity. However, recent results suggest that enhancing cellular energy expenditure may be an attractive alternative therapy. Critical Issues: This review evaluates recent discoveries regarding mitochondrial fatty acid oxidation (FAO) and its potential as a therapy for obesity. We focus on the still controversial beneficial effects of increased FAO in liver and muscle, recent studies on how to potentiate adipose tissue energy expenditure, and the different hypotheses involving FAO and the reactive oxygen species production in the hypothalamic control of food intake. Future Directions: The present review aims to provide an overview of novel anti-obesity strategies that target mitochondrial FAO and that will definitively be of high interest in the future research to fight against obesity-related disorders
    corecore