Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 57
Filtrar
Mais filtros

Base de dados
Tipo de documento
Intervalo de ano de publicação
1.
Cell Commun Signal ; 22(1): 297, 2024 May 28.
Artigo em Inglês | MEDLINE | ID: mdl-38807218

RESUMO

BACKGROUND: Endoplasmic reticulum (ER) stress-mediated increases in the hepatic levels of the very low-density lipoprotein (VLDL) receptor (VLDLR) promote hepatic steatosis by increasing the delivery of triglyceride-rich lipoproteins to the liver. Here, we examined whether the NAD(+)-dependent deacetylase sirtuin 1 (SIRT1) regulates hepatic lipid accumulation by modulating VLDLR levels and the subsequent uptake of triglyceride-rich lipoproteins. METHODS: Rats fed with fructose in drinking water, Sirt1-/- mice, mice treated with the ER stressor tunicamycin with or without a SIRT1 activator, and human Huh-7 hepatoma cells transfected with siRNA or exposed to tunicamycin or different inhibitors were used. RESULTS: Hepatic SIRT1 protein levels were reduced, while those of VLDLR were upregulated in the rat model of metabolic dysfunction-associated steatotic liver disease (MASLD) induced by fructose-drinking water. Moreover, Sirt1-/- mice displayed increased hepatic VLDLR levels that were not associated with ER stress, but were accompanied by an increased expression of hypoxia-inducible factor 1α (HIF-1α)-target genes. The pharmacological inhibition or gene knockdown of SIRT1 upregulated VLDLR protein levels in the human Huh-7 hepatoma cell line, with this increase abolished by the pharmacological inhibition of HIF-1α. Finally, SIRT1 activation prevented the increase in hepatic VLDLR protein levels in mice treated with the ER stressor tunicamycin. CONCLUSIONS: Overall, these findings suggest that SIRT1 attenuates fatty liver development by modulating hepatic VLDLR levels.


Assuntos
Fígado , Receptores de LDL , Sirtuína 1 , Animais , Sirtuína 1/metabolismo , Sirtuína 1/genética , Humanos , Fígado/metabolismo , Fígado/efeitos dos fármacos , Receptores de LDL/metabolismo , Receptores de LDL/genética , Camundongos , Masculino , Estresse do Retículo Endoplasmático/efeitos dos fármacos , Ratos , Linhagem Celular Tumoral , Camundongos Knockout , Fígado Gorduroso/metabolismo , Fígado Gorduroso/genética , Fígado Gorduroso/patologia , Camundongos Endogâmicos C57BL , Tunicamicina/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Ratos Sprague-Dawley
2.
Int J Mol Sci ; 24(2)2023 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-36675212

RESUMO

Sensing of long-chain fatty acids (LCFA) in the hypothalamus modulates energy balance, and its disruption leads to obesity. To date, the effects of saturated or unsaturated LCFA on hypothalamic-brown adipose tissue (BAT) axis and the underlying mechanisms have remained largely unclear. Our aim was to characterize the main molecular pathways involved in the hypothalamic regulation of BAT thermogenesis in response to LCFA with different lengths and degrees of saturation. One-week administration of high-fat diet enriched in monounsaturated FA led to higher BAT thermogenesis compared to a saturated FA-enriched diet. Intracerebroventricular infusion of oleic and linoleic acids upregulated thermogenesis markers and temperature in brown fat of mice, and triggered neuronal activation of paraventricular (PaV), ventromedial (VMH) and arcuate (ARC) hypothalamic nuclei, which was not found with saturated FAs. The neuron-specific protein carnitine palmitoyltransferase 1-C (CPT1C) was a crucial effector of oleic acid since the FA action was blunted in CPT1C-KO mice. Moreover, changes in the AMPK/ACC/malonyl-CoA pathway and fatty acid synthase expression were evoked by oleic acid. Altogether, central infusion of unsaturated but not saturated LCFA increases BAT thermogenesis through CPT1C-mediated sensing of FA metabolism shift, which in turn drive melanocortin system activation. These findings add new insight into neuronal circuitries activated by LCFA to drive thermogenesis.


Assuntos
Tecido Adiposo Marrom , Hipotálamo , Termogênese , Animais , Camundongos , Tecido Adiposo Marrom/metabolismo , Ácidos Graxos/metabolismo , Hipotálamo/metabolismo , Ácidos Oleicos/metabolismo , Termogênese/genética , Termogênese/fisiologia
3.
Cell Mol Life Sci ; 78(23): 7469-7490, 2021 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-34718828

RESUMO

The crucial role of the hypothalamus in the pathogenesis of obesity is widely recognized, while the precise molecular and cellular mechanisms involved are the focus of intense research. A disrupted endocannabinoid system, which critically modulates feeding and metabolic functions, through central and peripheral mechanisms, is a landmark indicator of obesity, as corroborated by investigations centered on the cannabinoid receptor CB1, considered to offer promise in terms of pharmacologically targeted treatment for obesity. In recent years, novel insights have been obtained, not only into relation to the mode of action of CB receptors, but also CB ligands, non-CB receptors, and metabolizing enzymes considered to be part of the endocannabinoid system (particularly the hypothalamus). The outcome has been a substantial expansion in knowledge of this complex signaling system and in drug development. Here we review recent literature, providing further evidence on the role of hypothalamic endocannabinoids in regulating energy balance and the implication for the pathophysiology of obesity. We discuss how these lipids are dynamically regulated in obesity onset, by diet and metabolic hormones in specific hypothalamic neurons, the impact of gender, and the role of endocannabinoid metabolizing enzymes as promising targets for tackling obesity and related diseases.


Assuntos
Endocanabinoides/metabolismo , Hipotálamo/patologia , Obesidade/patologia , Receptores de Canabinoides/metabolismo , Animais , Metabolismo Energético , Humanos , Hipotálamo/metabolismo , Obesidade/etiologia , Obesidade/metabolismo
4.
Int J Mol Sci ; 22(24)2021 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-34948254

RESUMO

Obesity has now reached pandemic proportions and represents a major socioeconomic and health problem in our societies [...].


Assuntos
Hipotálamo/metabolismo , Obesidade/fisiopatologia , Metabolismo Energético , Humanos , Hipotálamo/fisiopatologia
5.
Int J Mol Sci ; 22(12)2021 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-34201257

RESUMO

Despite the substantial role played by the hypothalamus in the regulation of energy balance and glucose homeostasis, the exact mechanisms and neuronal circuits underlying this regulation remain poorly understood. In the last 15 years, investigations using transgenic models, optogenetic, and chemogenetic approaches have revealed that SF1 neurons in the ventromedial hypothalamus are a specific lead in the brain's ability to sense glucose levels and conduct insulin and leptin signaling in energy expenditure and glucose homeostasis, with minor feeding control. Deletion of hormonal receptors, nutritional sensors, or synaptic receptors in SF1 neurons triggers metabolic alterations mostly appreciated under high-fat feeding, indicating that SF1 neurons are particularly important for metabolic adaptation in the early stages of obesity. Although these studies have provided exciting insight into the implications of hypothalamic SF1 neurons on whole-body energy homeostasis, new questions have arisen from these results. Particularly, the existence of neuronal sub-populations of SF1 neurons and the intricate neurocircuitry linking these neurons with other nuclei and with the periphery. In this review, we address the most relevant studies carried out in SF1 neurons to date, to provide a global view of the central role played by these neurons in the pathogenesis of obesity and diabetes.


Assuntos
Diabetes Mellitus/patologia , Hipotálamo/patologia , Neurônios/patologia , Obesidade/patologia , Fator Esteroidogênico 1/metabolismo , Animais , Diabetes Mellitus/etiologia , Diabetes Mellitus/metabolismo , Humanos , Hipotálamo/metabolismo , Neurônios/metabolismo , Obesidade/etiologia , Obesidade/metabolismo
6.
Cell Commun Signal ; 18(1): 147, 2020 09 10.
Artigo em Inglês | MEDLINE | ID: mdl-32912335

RESUMO

BACKGROUND: Deficiency of mitochondrial sirtuin 3 (SIRT3), a NAD+-dependent protein deacetylase that maintains redox status and lipid homeostasis, contributes to hepatic steatosis. In this study, we investigated additional mechanisms that might play a role in aggravating hepatic steatosis in Sirt3-deficient mice fed a high-fat diet (HFD). METHODS: Studies were conducted in wild-type (WT) and Sirt3-/- mice fed a standard diet or a HFD and in SIRT3-knockdown human Huh-7 hepatoma cells. RESULTS: Sirt3-/- mice fed a HFD presented exacerbated hepatic steatosis that was accompanied by decreased expression and DNA-binding activity of peroxisome proliferator-activated receptor (PPAR) α and of several of its target genes involved in fatty acid oxidation, compared to WT mice fed the HFD. Interestingly, Sirt3 deficiency in liver and its knockdown in Huh-7 cells resulted in upregulation of the nuclear levels of LIPIN1, a PPARα co-activator, and of the protein that controls its levels and localization, hypoxia-inducible factor 1α (HIF-1α). These changes were prevented by lipid exposure through a mechanism that might involve a decrease in succinate levels. Finally, Sirt3-/- mice fed the HFD showed increased levels of some proteins involved in lipid uptake, such as CD36 and the VLDL receptor. The upregulation in CD36 was confirmed in Huh-7 cells treated with a SIRT3 inhibitor or transfected with SIRT3 siRNA and incubated with palmitate, an effect that was prevented by the Nrf2 inhibitor ML385. CONCLUSION: These findings demonstrate new mechanisms by which Sirt3 deficiency contributes to hepatic steatosis. Video abstract.


Assuntos
Antígenos CD36/metabolismo , Fígado Gorduroso/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Fator 2 Relacionado a NF-E2/metabolismo , Fosfatidato Fosfatase/metabolismo , Sirtuína 3/genética , Animais , Linhagem Celular , Fígado Gorduroso/metabolismo , Fígado Gorduroso/patologia , Deleção de Genes , Humanos , Lipogênese , Masculino , Camundongos Endogâmicos C57BL , Transdução de Sinais , Sirtuína 3/metabolismo
7.
J Lipid Res ; 60(7): 1260-1269, 2019 07.
Artigo em Inglês | MEDLINE | ID: mdl-31138606

RESUMO

The endocannabinoid (eCB) system regulates energy homeostasis and is linked to obesity development. However, the exact dynamic and regulation of eCBs in the hypothalamus during obesity progression remain incompletely described and understood. Our study examined the time course of responses in two hypothalamic eCBs, 2-arachidonoylglycerol (2-AG) and arachidonoylethanolamine (AEA), in male and female mice during diet-induced obesity and explored the association of eCB levels with changes in brown adipose tissue (BAT) thermogenesis and body weight. We fed mice a high-fat diet (HFD), which induced a transient increase (substantial at 7 days) in hypothalamic eCBs, followed by a progressive decrease to basal levels with a long-term HFD. This transient rise at early stages of obesity is considered a physiologic compensatory response to BAT thermogenesis, which is activated by diet surplus. The eCB dynamic was sexually dimorphic: hypothalamic eCBs levels were higher in female mice, who became obese at later time points than males. The hypothalamic eCBs time course positively correlated with thermogenesis activation, but negatively matched body weight, leptinemia, and circulating eCB levels. Increased expression of eCB-synthetizing enzymes accompanied the transient hypothalamic eCB elevation. Icv injection of eCB did not promote BAT thermogenesis; however, administration of thermogenic molecules, such as central leptin or a peripheral ß3-adrenoreceptor agonist, induced a significant increase in hypothalamic eCBs, suggesting a directional link from BAT thermogenesis to hypothalamic eCBs. This study contributes to the understanding of hypothalamic regulation of obesity.


Assuntos
Dieta Hiperlipídica/efeitos adversos , Endocanabinoides/metabolismo , Hipotálamo/metabolismo , Obesidade/etiologia , Obesidade/metabolismo , Tecido Adiposo Marrom/metabolismo , Animais , Ácidos Araquidônicos/metabolismo , Feminino , Glicerídeos/metabolismo , Masculino , Camundongos , Alcamidas Poli-Insaturadas/metabolismo , Caracteres Sexuais
8.
Biochim Biophys Acta ; 1852(5): 1049-58, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25728706

RESUMO

We studied whether PPARß/δ deficiency modifies the effects of high fructose intake (30% fructose in drinking water) on glucose tolerance and adipose tissue dysfunction, focusing on the CD36-dependent pathway that enhances adipose tissue inflammation and impairs insulin signaling. Fructose intake for 8 weeks significantly increased body and liver weight, and hepatic triglyceride accumulation in PPARß/δ-deficient mice but not in wild-type mice. Feeding PPARß/δ-deficient mice with fructose exacerbated glucose intolerance and led to macrophage infiltration, inflammation, enhanced mRNA and protein levels of CD36, and activation of the JNK pathway in white adipose tissue compared to those of water-fed PPARß/δ-deficient mice. Cultured adipocytes exposed to fructose also exhibited increased CD36 protein levels and this increase was prevented by the PPARß/δ activator GW501516. Interestingly, the levels of the nuclear factor E2-related factor 2 (Nrf2), a transcription factor reported to up-regulate Cd36 expression and to impair insulin signaling, were increased in fructose-exposed adipocytes whereas co-incubation with GW501516 abolished this increase. In agreement with Nrf2 playing a role in the fructose-induced CD36 protein level increases, the Nrf2 inhibitor trigonelline prevented the increase and the reduction in insulin-stimulated AKT phosphorylation caused by fructose in adipocytes. Protein levels of the well-known Nrf2 target gene NAD(P)H: quinone oxidoreductase 1 (Nqo1) were increased in water-fed PPARß/δ-null mice, suggesting that PPARß/δ deficiency increases Nrf2 activity; and this increase was exacerbated in fructose-fed PPARß/δ-deficient mice. These findings indicate that the combination of high fructose intake and PPARß/δ deficiency increases CD36 protein levels via Nrf2, a process that promotes chronic inflammation and insulin resistance in adipose tissue.


Assuntos
Adipócitos/efeitos dos fármacos , Frutose/farmacologia , Resistência à Insulina , Fator 2 Relacionado a NF-E2/metabolismo , PPAR delta/metabolismo , PPAR beta/metabolismo , Células 3T3-L1 , Adipócitos/metabolismo , Adipócitos/patologia , Alcaloides/farmacologia , Animais , Antígenos CD36/genética , Antígenos CD36/metabolismo , Linhagem Celular , Citocinas/genética , Citocinas/metabolismo , Intolerância à Glucose/genética , Humanos , Immunoblotting , Proteínas Quinases JNK Ativadas por Mitógeno/metabolismo , Lipoproteínas LDL/metabolismo , Masculino , Camundongos , Camundongos da Linhagem 129 , Camundongos Endogâmicos C57BL , Camundongos Knockout , Fator 2 Relacionado a NF-E2/antagonistas & inibidores , PPAR delta/agonistas , PPAR delta/genética , PPAR beta/agonistas , PPAR beta/genética , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Transdução de Sinais/efeitos dos fármacos , Tiazóis/farmacologia
9.
Nanotechnology ; 27(42): 425603, 2016 Oct 21.
Artigo em Inglês | MEDLINE | ID: mdl-27631422

RESUMO

This paper presents the study of the dynamics of the formation of polymer-assisted highly-orientated polycrystalline cubic structures (CS) by a fractal-mediated mechanism. This mechanism involves the formation of seed Ag@Co nanoparticles by InterMatrix Synthesis and subsequent overgrowth after incubation at a low temperature in chloride and phosphate solutions. These ions promote the dissolution and recrystallization in an ordered configuration of pre-synthetized nanoparticles initially embedded in negatively-charged polymeric matrices. During recrystallization, silver ions aggregate in AgCl@Co fractal-like structures, then evolve into regular polycrystalline solid nanostructures (e.g. CS) in a single crystallization step on specific regions of the ion exchange resin (IER) which maintain the integrity of polycrystalline nanocubes. Here, we study the essential role of the IER in the formation of these CS for the maintenance of their integrity and stability. Thus, this synthesis protocol may be easily expanded to the composition of other nanoparticles providing an interesting, cheap and simple alternative for cubic structure formation and isolation.

10.
Eur J Nutr ; 55(6): 2011-9, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-26266932

RESUMO

BACKGROUND: The inflammatory process associated with obesity mainly arises from white adipose tissue (WAT) alterations. In the last few years, nutritional-based strategies have been positioned as promising alternatives to pharmacological approaches against these pathologies. Our aim was to determine the potential of a rice bran enzymatic extract (RBEE)-supplemented diet in the prevention of metabolic, biochemical and functional adipose tissue and macrophage changes associated with a diet-induced obesity (DIO) in mice. METHODS: C57BL/6J mice were fed high-fat diet (HF), 1 and 5 % RBEE-supplemented high-fat diet (HF1 % and HF5 %, respectively) and standard diet as control. Serum cardiometabolic parameters, adipocytes size and mRNA expression of pro-inflammatory biomarkers and macrophage polarization-related genes from WAT and liver were evaluated. RESULTS: RBEE administration significantly decreased insulin resistance in obese mice. Serum triglycerides, total cholesterol, glucose, insulin, adiponectin and nitrites from treated mice were partially restored, mainly by 1 % RBEE-enriched diet. The incremented adipocytes size observed in HF group was reduced by RBEE treatment, being 1 % more effective than 5 % RBEE. Pro-inflammatory biomarkers in WAT such as IL-6 and IL-1ß were significantly decreased in RBEE-treated mice. Adiponectin, PPARγ, TNF-α, Emr1 or M1/M2 levels were significantly restored in WAT from HF1 % compared to HF mice. CONCLUSIONS: RBEE-supplemented diet attenuated insulin resistance, dyslipidemia and morphological and functional alterations of adipose tissue in DIO mice. These benefits were accompanied by a modulating effect in adipocytes secretion and some biomarkers associated with macrophage polarization. Therefore, RBEE may be considered an alternative nutritional complement over metabolic syndrome and its complications.


Assuntos
Tecido Adiposo Branco/metabolismo , Dieta Hiperlipídica/efeitos adversos , Fibras na Dieta/administração & dosagem , Inflamação/dietoterapia , Macrófagos/metabolismo , Oryza/química , Adipócitos , Adiponectina/metabolismo , Animais , Biomarcadores/sangue , Proteínas de Ligação ao Cálcio , Colesterol/sangue , Dislipidemias/dietoterapia , Dislipidemias/etiologia , Inflamação/etiologia , Insulina/sangue , Resistência à Insulina , Interleucina-1beta/sangue , Interleucina-6/sangue , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Obesidade/dietoterapia , Obesidade/etiologia , PPAR gama/metabolismo , Receptores de Superfície Celular/metabolismo , Receptores Acoplados a Proteínas G , Triglicerídeos/sangue , Fator de Necrose Tumoral alfa/metabolismo
11.
Biochim Biophys Acta ; 1832(8): 1241-8, 2013 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-23507144

RESUMO

The role of peroxisome proliferator activator receptor (PPAR)ß/δ in the pathogenesis of Alzheimer's disease has only recently been explored through the use of PPARß/δ agonists. Here we evaluated the effects of PPARß/δ deficiency on the amyloidogenic pathway and tau hyperphosphorylation. PPARß/δ-null mice showed cognitive impairment in the object recognition task, accompanied by enhanced DNA-binding activity of NF-κB in the cortex and increased expression of IL-6. In addition, two NF-κB-target genes involved in ß-amyloid (Aß) synthesis and deposition, the ß site APP cleaving enzyme 1 (Bace1) and the receptor for advanced glycation endproducts (Rage), respectively, increased in PPARß/δ-null mice compared to wild type animals. The protein levels of glial fibrillary acidic protein (GFAP) increased in the cortex of PPARß/δ-null mice, which would suggest the presence of astrogliosis. Finally, tau hyperphosphorylation at Ser199 and enhanced levels of PHF-tau were associated with increased levels of the tau kinases CDK5 and phospho-ERK1/2 in the cortex of PPARß/δ(-/-) mice. Collectively, our findings indicate that PPARß/δ deficiency results in cognitive impairment associated with enhanced inflammation, astrogliosis and tau hyperphosphorylation in the cortex.


Assuntos
Secretases da Proteína Precursora do Amiloide/metabolismo , Ácido Aspártico Endopeptidases/metabolismo , Córtex Cerebral/metabolismo , PPAR beta/deficiência , Receptores Imunológicos/metabolismo , Proteínas tau/metabolismo , Secretases da Proteína Precursora do Amiloide/genética , Peptídeos beta-Amiloides/genética , Peptídeos beta-Amiloides/metabolismo , Animais , Ácido Aspártico Endopeptidases/genética , Cognição/fisiologia , Disfunção Cognitiva/genética , Disfunção Cognitiva/metabolismo , Quinase 5 Dependente de Ciclina/genética , Quinase 5 Dependente de Ciclina/metabolismo , Proteínas de Ligação a DNA/genética , Proteínas de Ligação a DNA/metabolismo , Proteína Glial Fibrilar Ácida , Inflamação , Interleucina-6/genética , Interleucina-6/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Masculino , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/genética , NF-kappa B/metabolismo , Proteínas do Tecido Nervoso/genética , Proteínas do Tecido Nervoso/metabolismo , PPAR beta/genética , PPAR beta/metabolismo , Fosforilação , Receptor para Produtos Finais de Glicação Avançada , Receptores Imunológicos/genética , Proteínas tau/genética
12.
Eur J Nutr ; 52(2): 789-97, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22661284

RESUMO

BACKGROUND AND PURPOSE: Rice bran enzymatic extract (RBEE) has advantages compared to the original rice bran or its oils including water solubility, lack of rancidity and increased content in high nutritional proteins and nutraceutical compounds, particularly phytosterols, γ-oryzanol and tocols. Our aim was to determine the beneficial effects of RBEE in the pathogenesis of metabolic syndrome in obese Zucker rats. METHODS: Obese Zucker rats and their lean littermates were fed a 1 and 5 % RBEE-supplemented diet (O1, O5, L1 and L5). Simultaneously, obese and lean Zucker rats, fed a standard diet, were used as controls (OC and LC, respectively). Body weight, food and water intake, and systolic blood pressure were weekly evaluated. After treatment, biochemical assays of serum glucose, insulin, triglycerides (TG), total cholesterol (TC), non-esterified fatty acids (NEFA), adiponectin and nitrates (NO((x))) were determined. RESULTS: RBEE treatment reduced circulating levels of TG and TC, whereas increased HDL-cholesterol without altering NEFA values in obese rats. The extract also induced a significant dose-dependent reduction of hypertension linked to obesity. RBEE of 5 % improved insulin resistance and subsequently reduced HOMA-IR index without altering serum glucose levels. Obese animals treated with RBEE showed partial restoration of adiponectin levels and a significant attenuation of pro-inflammatory values of NO((x)). CONCLUSION: These findings evidence the nutraceutical properties of RBEE against the pathogenesis of metabolic syndrome by attenuating dyslipidemia, hypertension and insulin resistance as well as by restoring hypoadiponectinemia associated to obesity.


Assuntos
Dislipidemias/tratamento farmacológico , Hipertensão/tratamento farmacológico , Resistência à Insulina , Obesidade/tratamento farmacológico , Oryza/química , Extratos Vegetais/farmacologia , Adiponectina/sangue , Animais , Glicemia/análise , Pressão Sanguínea/efeitos dos fármacos , Peso Corporal/efeitos dos fármacos , Colesterol/sangue , Dieta , Ácidos Graxos não Esterificados/sangue , Insulina/sangue , Síndrome Metabólica/tratamento farmacológico , Nitratos/sangue , Obesidade/sangue , Fenilpropionatos/análise , Fitosteróis/análise , Ratos , Ratos Zucker , Triglicerídeos/sangue , Água/química
13.
J Neuroendocrinol ; 35(9): e13234, 2023 09.
Artigo em Inglês | MEDLINE | ID: mdl-36735894

RESUMO

Tackling the growing incidence and prevalence of obesity urgently requires uncovering new molecular pathways with therapeutic potential. The brain, and in particular the hypothalamus, is a major integrator of metabolic signals from peripheral tissues that regulate functions such as feeding behavior and energy expenditure. In obesity, hypothalamic capacity to sense nutritional status and regulate these functions is altered. An emerging line of research is that hypothalamic lipid metabolism plays a critical role in regulating energy balance. Here, we focus on the carnitine palmitoyltransferase 1 (CPT1) enzyme family responsible for long-chain fatty acid metabolism. The evidence suggests that two of its isoforms expressed in the brain, CPT1A and CPT1C, play a crucial role in hypothalamic lipid metabolism, and their promise as targets in food intake and bodyweight management is currently being intensively investigated. In this review we describe and discuss the metabolic actions and potential up- and downstream effectors of hypothalamic CPT1 isoforms, and posit the need to develop innovative nanomedicine platforms for selective targeting of CPT1 and related nutrient sensors in specific brain areas as potential next-generation therapy to treat obesity.


Assuntos
Carnitina O-Palmitoiltransferase , Metabolismo Energético , Humanos , Carnitina O-Palmitoiltransferase/metabolismo , Metabolismo Energético/fisiologia , Obesidade/metabolismo , Isoformas de Proteínas/metabolismo , Hipotálamo/metabolismo
14.
Cell Death Dis ; 14(1): 57, 2023 01 24.
Artigo em Inglês | MEDLINE | ID: mdl-36693836

RESUMO

There is an urgent need to identify reliable genetic biomarkers for accurate diagnosis, prognosis, and treatment of different tumor types. Described as a prognostic marker for many tumors is the neuronal protein carnitine palmitoyltransferase 1 C (CPT1C). Several studies report that CPT1C is involved in cancer cell adaptation to nutrient depletion and hypoxia. However, the molecular role played by CPT1C in cancer cells is controversial. Most published studies assume that, like canonical CPT1 isoforms, CPT1C is a mediator of fatty acid transport to mitochondria for beta-oxidation, despite the fact that CPT1C has inefficient catalytic activity and is located in the endoplasmic reticulum. In this review, we collate existing evidence on CPT1C in neurons, showing that CPT1C is a sensor of nutrients that interacts with and regulates other proteins involved in lipid metabolism and transport, lysosome motility, and the secretory pathway. We argue, therefore, that CPT1C expression in cancer cells is not a direct regulator of fat burn, but rather is a regulator of lipid metabolic reprograming and cell adaptation to environmental stressors. We also review the clinical relevance of CPT1C as a prognostic indicator and its contribution to tumor growth, cancer invasiveness, and cell senescence. This new and integrated vision of CPT1C function can help better understand the metabolic plasticity of cancer cells and improve the design of therapeutic strategies.


Assuntos
Carnitina O-Palmitoiltransferase , Neoplasias , Humanos , Carnitina O-Palmitoiltransferase/genética , Carnitina O-Palmitoiltransferase/metabolismo , Hipóxia/metabolismo , Neoplasias/genética , Neoplasias/metabolismo , Neoplasias/fisiopatologia , Neurônios/metabolismo , Oxirredução
15.
Front Mol Biosci ; 10: 1274221, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-38053578

RESUMO

With rapid industrialization, urbanization, and climate change, the impact of environmental factors on human health is becoming increasingly evident and understanding the complex mechanisms involved is vital from a healthcare perspective. Nevertheless, the relationship between physiological stress resulting from environmental stressors and environmental disease is complex and not well understood. Chronic exposure to environmental stressors, such as air and water contaminants, pesticides, and toxic metals, has been recognized as a potent elicitor of physiological responses ranging from systemic inflammation to immune system dysregulation causing or progressing environmental diseases. Conversely, physiological stress can exacerbate susceptibility to environmental diseases. Stress-induced alterations in immune function and hormonal balance may impair the ability to detoxify harmful substances and combat pathogens. Additionally, prolonged stress can impact lifestyle choices, leading to harmful behaviors. Understanding the link between physiological stress and environmental disease requires a systematic, multidisciplinary approach. Addressing this complex relationship necessitates the establishment of a global research network. This perspective discusses the intricate interplay between physiological stress and environmental disease, focusing on common environmental diseases, cancer, diabetes, and cognitive degeneration. Furthermore, we highlight the intricate and reciprocal nature of the connection between physiological stress and these environmental diseases giving a perspective on the current state of knowledge as well as identifying where further information is necessary. Recognizing the role of physiological stress in environmental health outcomes will aid in the development of comprehensive strategies to safeguard public health and promote ecological balance.

16.
Biomater Sci ; 11(7): 2336-2347, 2023 Mar 28.
Artigo em Inglês | MEDLINE | ID: mdl-36804651

RESUMO

Targeting brain lipid metabolism is a promising strategy to regulate the energy balance and fight metabolic diseases such as obesity. The development of stable platforms for selective delivery of drugs, particularly to the hypothalamus, is a challenge but a possible solution for these metabolic diseases. Attenuating fatty acid oxidation in the hypothalamus via CPT1A inhibition leads to satiety, but this target is difficult to reach in vivo with the current drugs. We propose using an advanced crosslinked polymeric micelle-type nanomedicine that can stably load the CPT1A inhibitor C75-CoA for in vivo control of the energy balance. Central administration of the nanomedicine induced a rapid attenuation of food intake and body weight in mice via regulation of appetite-related neuropeptides and neuronal activation of specific hypothalamic regions driving changes in the liver and adipose tissue. This nanomedicine targeting brain lipid metabolism was successful in the modulation of food intake and peripheral metabolism in mice.


Assuntos
Metabolismo dos Lipídeos , Nanomedicina , Camundongos , Animais , Metabolismo Energético , Obesidade/metabolismo , Hipotálamo/metabolismo
17.
Nutrients ; 14(8)2022 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-35458168

RESUMO

Dietary components exert protective effects against obesity and related metabolic and cardiovascular disturbances by interfering with the molecular pathways leading to these pathologies. Dietary biomolecules are currently promising strategies to help in the management of obesity and metabolic syndrome, which are still unmet medical issues. Olive oil, a key component of the Mediterranean diet, provides an exceptional lipid matrix highly rich in bioactive molecules. Among them, the pentacyclic triterpenic acids (i.e., oleanolic acid) have gained clinical relevance in the last decade due to their wide range of biological actions, particularly in terms of vascular function, obesity and insulin resistance. Considering the promising effects of these triterpenic compounds as nutraceuticals and components of functional foods against obesity and associated complications, the aim of our review is to decipher and discuss the main molecular mechanisms underlying these effects driven by olive oil triterpenes, in particular by oleanolic acid. Special attention is paid to their signaling and targets related to glucose and insulin homeostasis, lipid metabolism, adiposity and cardiovascular dysfunction in obesity. Our study is aimed at providing a better understanding of the impact of dietary components of olive oil in the long-term management of obesity and metabolic syndrome in humans.


Assuntos
Dieta Mediterrânea , Síndrome Metabólica , Ácido Oleanólico , Humanos , Síndrome Metabólica/tratamento farmacológico , Obesidade/tratamento farmacológico , Ácido Oleanólico/farmacologia , Ácido Oleanólico/uso terapêutico , Azeite de Oliva/farmacologia
18.
Antioxidants (Basel) ; 11(11)2022 Oct 31.
Artigo em Inglês | MEDLINE | ID: mdl-36358537

RESUMO

The increasing prevalence of obesity worldwide has promoted research on human metabolism and foods such as sofrito, a tomato and olive oil-based sauce from the Mediterranean diet, has shown beneficial effects on obesity and related complications. Sofrito has been associated with better cardiovascular health, metabolic syndrome, and anti-inflammatory effects. The aim of this study was to understand how sofrito intake could contribute to the control of energy metabolism in obese rats. For this purpose, integrative untargeted lipidomics, metabolomics, and targeted gene expression approaches were used in the liver and adipose tissue to identify metabolic changes and the mechanism of action promoted by sofrito intake. A new biomarker was identified in the liver, butanediol glucuronide, an indicator of ketogenic activation and lipid oxidation after the sofrito intervention. Gene expression analysis revealed an increase in the uptake and liver oxidation of lipids for energy production and ketogenesis activation as fuel for other tissues in sofrito-fed animals. Sofrito altered the lipidomic profile in the fat depots of obese rats. This multiomics study identifies a new biomarker linked to the beneficial actions of sofrito against obesity and provides further insight into the beneficial effect of the Mediterranean diet components.

19.
Prog Lipid Res ; 81: 101071, 2021 01.
Artigo em Inglês | MEDLINE | ID: mdl-33186641

RESUMO

Nutrients, hormones and the energy sensor AMP-activated protein kinase (AMPK) tightly regulate the intracellular levels of the metabolic intermediary malonyl-CoA, which is a precursor of fatty acid synthesis and a negative regulator of fatty acid oxidation. In the brain, the involvement of malonyl-CoA in the control of food intake and energy homeostasis has been known for decades. However, recent data uncover a new role in cognition and brain development. The sensing of malonyl-CoA by carnitine palmitoyltransferase 1 (CPT1) proteins regulates a variety of functions, such as the fate of neuronal stem cell precursors, the motility of lysosomes in developing axons, the trafficking of glutamate receptors to the neuron surface (necessary for proper synaptic function) and the metabolic coupling between astrocytes and neurons. We discuss the relevance of those recent findings evidencing how nutrients and metabolic disorders impact cognition. We also enumerate all nutritional and hormonal conditions that are known to regulate malonyl-CoA levels in the brain, reflect on protein malonylation as a new post-translational modification, and give a reasoned vision of the opportunities and challenges that future research in the field could address.


Assuntos
Carnitina O-Palmitoiltransferase , Malonil Coenzima A , Encéfalo/metabolismo , Carnitina O-Palmitoiltransferase/metabolismo , Cognição , Homeostase , Malonil Coenzima A/metabolismo
20.
Br J Pharmacol ; 178(7): 1507-1523, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33444462

RESUMO

BACKGROUND AND PURPOSE: The enzyme α/ß-hydrolase domain containing 6 (ABHD6), a new member of the endocannabinoid system, is a promising therapeutic target against neuronal-related diseases. However, how ABHD6 activity is regulated is not known. ABHD6 coexists in protein complexes with the brain-specific carnitine palmitoyltransferase 1C (CPT1C). CPT1C is involved in neuro-metabolic functions, depending on brain malonyl-CoA levels. Our aim was to study CPT1C-ABHD6 interaction and determine whether CPT1C is a key regulator of ABHD6 activity depending on nutritional status. EXPERIMENTAL APPROACH: Co-immunoprecipitation and FRET assays were used to explore ABHD6 interaction with CPT1C or modified malonyl-CoA-insensitive or C-terminal truncated CPT1C forms. Cannabinoid CB1 receptor-mediated signalling was investigated by determining cAMP levels. A novel highly sensitive fluorescent method was optimized to measure ABHD6 activity in non-neuronal and neuronal cells and in brain tissues from wild-type (WT) and CPT1C-KO mice. KEY RESULTS: CPT1C interacted with ABHD6 and negatively regulated its hydrolase activity, thereby regulating 2-AG downstream signalling. Accordingly, brain tissues of CPT1C-KO mice showed increased ABHD6 activity. CPT1C malonyl-CoA sensing was key to the regulatory role on ABHD6 activity and CB1 receptor signalling. Fasting, which attenuates brain malonyl-CoA, significantly increased ABHD6 activity in hypothalamus from WT, but not CPT1C-KO, mice. CONCLUSIONS AND IMPLICATIONS: Our finding that negative regulation of ABHD6 activity, particularly in the hypothalamus, is sensitive to nutritional status throws new light on the characterization and the importance of the proteins involved as potential targets against diseases affecting the CNS.


Assuntos
Carnitina O-Palmitoiltransferase , Monoacilglicerol Lipases/metabolismo , Estado Nutricional , Animais , Carnitina O-Palmitoiltransferase/genética , Hidrolases , Malonil Coenzima A , Camundongos
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA