Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 3.254
Filtrar
1.
Nature ; 625(7993): 175-180, 2024 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-38093006

RESUMEN

Oxytocin (OXT), a nine-amino-acid peptide produced in the hypothalamus and released by the posterior pituitary, has well-known actions in parturition, lactation and social behaviour1, and has become an intriguing therapeutic target for conditions such as autism and schizophrenia2. Exogenous OXT has also been shown to have effects on body weight, lipid levels and glucose homeostasis1,3, suggesting that it may also have therapeutic potential for metabolic disease1,4. It is unclear, however, whether endogenous OXT participates in metabolic homeostasis. Here we show that OXT is a critical regulator of adipose tissue lipolysis in both mice and humans. In addition, OXT serves to facilitate the ability of ß-adrenergic agonists to fully promote lipolysis. Most surprisingly, the relevant source of OXT in these metabolic actions is a previously unidentified subpopulation of tyrosine hydroxylase-positive sympathetic neurons. Our data reveal that OXT from the peripheral nervous system is an endogenous regulator of adipose and systemic metabolism.


Asunto(s)
Tejido Adiposo , Lipólisis , Neuronas , Oxitocina , Animales , Humanos , Ratones , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Agonistas Adrenérgicos beta/farmacología , Lipólisis/efectos de los fármacos , Neuronas/metabolismo , Oxitocina/metabolismo , Oxitocina/farmacología , Tirosina 3-Monooxigenasa/metabolismo
3.
J Clin Endocrinol Metab ; 108(3): 653-664, 2023 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-36240323

RESUMEN

CONTEXT: Exogenous ketone body administration lowers circulating glucose levels but the underlying mechanisms are uncertain. OBJECTIVE: We tested the hypothesis that administration of the ketone body ß-hydroxybutyrate (ßOHB) acutely increases insulin sensitivity via feedback suppression of circulating free fatty acid (FFA) levels. METHODS: In a randomized, single-blinded crossover design, 8 healthy men were studied twice with a growth hormone (GH) infusion to induce lipolysis in combination with infusion of either ßOHB or saline. Each study day comprised a basal period and a hyperinsulinemic-euglycemic clamp combined with a glucose tracer and adipose tissue and skeletal muscle biopsies. RESULTS: ßOHB administration profoundly suppressed FFA levels concomitantly with a significant increase in glucose disposal and energy expenditure. This was accompanied by a many-fold increase in skeletal muscle content of both ßOHB and its derivative acetoacetate. CONCLUSION: Our data unravel an insulin-sensitizing effect of ßOHB, which we suggest is mediated by concomitant suppression of lipolysis.


Asunto(s)
Hormona de Crecimiento Humana , Resistencia a la Insulina , Cuerpos Cetónicos , Humanos , Masculino , Ácido 3-Hidroxibutírico/farmacología , Ácidos Grasos no Esterificados , Glucosa , Técnica de Clampeo de la Glucosa , Hormona del Crecimiento , Hormona de Crecimiento Humana/farmacología , Insulina/farmacología , Resistencia a la Insulina/fisiología , Cuerpos Cetónicos/farmacología , Cuerpos Cetónicos/uso terapéutico , Lipólisis/efectos de los fármacos , Lipólisis/fisiología
4.
Int J Mol Sci ; 23(13)2022 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-35806278

RESUMEN

Casein kinase 2 (CK2) is a ubiquitously expressed serine/threonine kinase and is upregulated in human obesity. CX-4945 (Silmitasertib) is a CK2 inhibitor with anti-cancerous and anti-adipogenic activities. However, the anti-adipogenic and pro-lipolytic effects and the mode of action of CX-4945 in (pre)adipocytes remain elusive. Here, we explored the effects of CX-4945 on adipogenesis and lipolysis in differentiating and differentiated 3T3-L1 cells, a murine preadipocyte cell line. CX-4945 at 15 µM strongly reduced lipid droplet (LD) accumulation and triglyceride (TG) content in differentiating 3T3-L1 cells, indicating the drug's anti-adipogenic effect. Mechanistically, CX-4945 reduced the expression levels of CCAAT/enhancer-binding protein-α (C/EBP-α), peroxisome proliferator-activated receptor-γ (PPAR-γ), fatty acid synthase (FAS), acetyl-CoA carboxylase (ACC), and perilipin A in differentiating 3T3-L1 cells. Strikingly, CX-4945 further increased the phosphorylation levels of cAMP-activated protein kinase (AMPK) and liver kinase B-1 (LKB-1) while decreasing the intracellular ATP content in differentiating 3T3-L1 cells. In differentiated 3T3-L1 cells, CX-4945 had abilities to stimulate glycerol release and elevate the phosphorylation levels of hormone-sensitive lipase (HSL), pointing to the drug's pro-lipolytic effect. In addition, CX-4945 induced the activation of extracellular signal-regulated kinase-1/2 (ERK-1/2), and PD98059, an inhibitor of ERK-1/2, attenuated the CX4945-induced glycerol release and HSL phosphorylation in differentiated 3T3-L1 cells, indicating the drug's ERK-1/2-dependent lipolysis. In summary, this investigation shows that CX-4945 has strong anti-adipogenic and pro-lipolytic effects on differentiating and differentiated 3T3-L1 cells, mediated by control of the expression and phosphorylation levels of CK2, C/EBP-α, PPAR-γ, FAS, ACC, perilipin A, AMPK, LKB-1, ERK-1/2, and HSL.


Asunto(s)
Adipogénesis , Quinasa de la Caseína II , Naftiridinas , Fenazinas , Células 3T3-L1 , Proteínas Quinasas Activadas por AMP/metabolismo , Animales , Proteína alfa Potenciadora de Unión a CCAAT/metabolismo , Quinasa de la Caseína II/antagonistas & inhibidores , Quinasa de la Caseína II/metabolismo , Diferenciación Celular/efectos de los fármacos , Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , Glicerol/farmacología , Humanos , Lipólisis/efectos de los fármacos , Ratones , Naftiridinas/farmacología , PPAR gamma/metabolismo , Perilipina-1/metabolismo , Fenazinas/farmacología , Esterol Esterasa/metabolismo
5.
Int J Mol Sci ; 23(9)2022 Apr 23.
Artículo en Inglés | MEDLINE | ID: mdl-35563078

RESUMEN

Oncostatin M (OSM) is an immune cell-derived cytokine that is upregulated in adipose tissue in obesity. Upon binding its receptor (OSMR), OSM induces the phosphorylation of the p66 subunit of Src homology 2 domain-containing transforming protein 1 (SHC1), called p66Shc, and activates the extracellular signal-related kinase (ERK) pathway. Mice with adipocyte-specific OSMR deletion (OsmrFKO) are insulin resistant and exhibit adipose tissue inflammation, suggesting that intact adipocyte OSM-OSMR signaling is necessary for maintaining adipose tissue health. How OSM affects specific adipocyte functions is still unclear. Here, we examined the effects of OSM on adipocyte lipolysis. We treated 3T3-L1 adipocytes with OSM, insulin, and/or inhibitors of SHC1 and ERK and measured glycerol release. We also measured phosphorylation of p66Shc, ERK, and insulin receptor substrate-1 (IRS1) and the expression of lipolysis-associated genes in OSM-exposed 3T3-L1 adipocytes and primary adipocytes from control and OsmrFKO mice. We found that OSM induces adipocyte lipolysis via a p66Shc-ERK pathway and inhibits the suppression of lipolysis by insulin. Further, OSM induces phosphorylation of inhibitory IRS1 residues. We conclude that OSM is a stimulator of lipolysis and inhibits adipocyte insulin response. Future studies will determine how these roles of OSM affect adipose tissue function in health and disease.


Asunto(s)
Insulina , Lipólisis , Oncostatina M , Células 3T3-L1/metabolismo , Adipocitos/metabolismo , Animales , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Insulina/metabolismo , Insulina Regular Humana , Lipólisis/efectos de los fármacos , Ratones , Oncostatina M/metabolismo , Proteína Transformadora 1 que Contiene Dominios de Homología 2 de Src/metabolismo
6.
J Nutr Biochem ; 106: 109017, 2022 08.
Artículo en Inglés | MEDLINE | ID: mdl-35461903

RESUMEN

Lipophagy, a form of selective autophagy, degrades lipid droplet (LD) in adipose tissue and the liver. The chemotherapeutic isothiocyanate sulforaphane (SFN) contributes to lipolysis through the activation of hormone-sensitive lipase and the browning of white adipocytes. However, the details concerning the regulation of lipolysis in adipocytes by SFN-mediated autophagy remain unclear. In this study, we investigated the effects of SFN on autophagy in the epididymal fat of mice fed a high-fat diet (HFD) or control-fat diet and on the molecular mechanisms of autophagy in differentiated 3T3-L1 cells. Western blotting revealed that the protein expression of lipidated LC3 (LC3-II), an autophagic substrate, was induced after 3T3-L1 adipocytes treatment with SFN. In addition, SFN increased the LC3-II protein expression in the epididymal fat of mice fed an HFD. Immunofluorescence showed that the SFN-induced LC3 expression was co-localized with LDs in 3T3-L1 adipocytes and with perilipin, the most abundant adipocyte-specific protein, in adipocytes of mice fed an HFD. Next, we confirmed that SFN activates autophagy flux in differentiated 3T3-L1 cells using the mCherry-EGFP-LC3 and GFP-LC3-RFP-LC3ΔG probe. Furthermore, we examined the induction mechanisms of autophagy by SFN in 3T3-L1 adipocytes using western blotting. ATG5 knockdown partially blocked the SFN-induced release of fatty acids from LDs in mature 3T3-L1 adipocytes. SFN time-dependently elicited the phosphorylation of AMPK, the dephosphorylation of mTOR, and the phosphorylation of ULK1 in differentiated 3T3-L1 cells. Taken together, these results suggest that SFN may provoke lipophagy through AMPK-mTOR-ULK1 pathway signaling, resulting in partial lipolysis of adipocytes.


Asunto(s)
Proteínas Quinasas Activadas por AMP , Homólogo de la Proteína 1 Relacionada con la Autofagia , Isotiocianatos , Serina-Treonina Quinasas TOR , Células 3T3-L1 , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Adipocitos Blancos/efectos de los fármacos , Adipocitos Blancos/metabolismo , Animales , Autofagia/efectos de los fármacos , Homólogo de la Proteína 1 Relacionada con la Autofagia/metabolismo , Isotiocianatos/farmacología , Lipólisis/efectos de los fármacos , Ratones , Transducción de Señal/efectos de los fármacos , Sulfóxidos/farmacología , Serina-Treonina Quinasas TOR/metabolismo
7.
Nat Commun ; 13(1): 942, 2022 02 17.
Artículo en Inglés | MEDLINE | ID: mdl-35177603

RESUMEN

Insulin analogs have been developed to treat diabetes with focus primarily on improving the time action profile without affecting ligand-receptor interaction or functional selectivity. As a result, inherent liabilities (e.g. hypoglycemia) of injectable insulin continue to limit the true therapeutic potential of related agents. Insulin dimers were synthesized to investigate whether partial agonism of the insulin receptor (IR) tyrosine kinase is achievable, and to explore the potential for tissue-selective systemic insulin pharmacology. The insulin dimers induced distinct IR conformational changes compared to native monomeric insulin and substrate phosphorylation assays demonstrated partial agonism. Structurally distinct dimers with differences in conjugation sites and linkers were prepared to deliver desirable IR partial agonist (IRPA). Systemic infusions of a B29-B29 dimer in vivo revealed sharp differences compared to native insulin. Suppression of hepatic glucose production and lipolysis were like that attained with regular insulin, albeit with a distinctly shallower dose-response. In contrast, there was highly attenuated stimulation of glucose uptake into muscle. Mechanistic studies indicated that IRPAs exploit tissue differences in receptor density and have additional distinctions pertaining to drug clearance and distribution. The hepato-adipose selective action of IRPAs is a potentially safer approach for treatment of diabetes.


Asunto(s)
Diabetes Mellitus Experimental/tratamiento farmacológico , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Hipoglucemiantes/farmacología , Insulina/farmacología , Receptor de Insulina/agonistas , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Aloxano/administración & dosificación , Aloxano/toxicidad , Animales , Glucemia/efectos de los fármacos , Glucemia/metabolismo , Células CHO , Cricetulus , Diabetes Mellitus Experimental/sangre , Diabetes Mellitus Experimental/inducido químicamente , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/inducido químicamente , Diabetes Mellitus Tipo 1/metabolismo , Células HEK293 , Humanos , Hipoglucemiantes/uso terapéutico , Insulina/uso terapéutico , Lipólisis/efectos de los fármacos , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratas , Proteínas Recombinantes/farmacología , Proteínas Recombinantes/uso terapéutico , Transducción de Señal/efectos de los fármacos , Porcinos , Porcinos Enanos
8.
Int J Mol Sci ; 23(3)2022 Jan 25.
Artículo en Inglés | MEDLINE | ID: mdl-35163277

RESUMEN

Hydrogen sulfide (H2S) and inorganic polysulfides are important signaling molecules; however, little is known about their role in the adipose tissue. We examined the effect of H2S and polysulfides on adipose tissue lipolysis. H2S and polysulfide production by mesenteric adipose tissue explants in rats was measured. The effect of Na2S and Na2S4, the H2S and polysulfide donors, respectively, on lipolysis markers, plasma non-esterified fatty acids (NEFA) and glycerol, was examined. Na2S but not Na2S4 increased plasma NEFA and glycerol in a time- and dose-dependent manner. Na2S increased cyclic AMP but not cyclic GMP concentration in the adipose tissue. The effect of Na2S on NEFA and glycerol was abolished by the specific inhibitor of protein kinase A, KT5720. The effect of Na2S on lipolysis was not abolished by propranolol, suggesting no involvement of ß-adrenergic receptors. In addition, Na2S had no effect on phosphodiesterase activity in the adipose tissue. Obesity induced by feeding rats a highly palatable diet for 1 month was associated with increased plasma NEFA and glycerol concentrations, as well as greater H2S production in the adipose tissue. In conclusion, H2S stimulates lipolysis and may contribute to the enhanced lipolysis associated with obesity.


Asunto(s)
Tejido Adiposo/metabolismo , Sulfuro de Hidrógeno/metabolismo , Lipólisis/fisiología , Tejido Adiposo/efectos de los fármacos , Animales , GMP Cíclico/metabolismo , Ácidos Grasos no Esterificados/metabolismo , Lipólisis/efectos de los fármacos , Masculino , Síndrome Metabólico/genética , Síndrome Metabólico/metabolismo , Síndrome Metabólico/fisiopatología , Obesidad/metabolismo , Ratas , Ratas Wistar , Receptores Adrenérgicos beta/metabolismo , Sulfuros/metabolismo
9.
Biochem Pharmacol ; 197: 114910, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35026188

RESUMEN

Cannabidiol (CBD) is a non-psychoactive phytocannabinoid found in the Cannabis sativa plant. Human exposure to CBD can be through recreational marijuana use, commercially available CBD-containing products, and medical treatments. Previous studies found that cannabidiol may activate the master regulator of adipogenesis, peroxisome proliferator activated receptor gamma (PPARγ). Here we investigated the effects of CBD on adipogenesis in human and mouse multipotent mesenchymal stromal stem cells (MSCs). We tested the effects of CBD on nuclear receptor activation and adipogenic potential to demonstrate the mechanism of CBD effects and employed the in vitro MSC differentiation models to assess adipogenic effects of CBD.Using transient transfection assays, we demonstrated that CBD activated mouse and human PPARγ, but not its heterodimeric partner, the retinoid 'X' receptor, RXR. Our results showed that CBD increased lipid accumulation and the expression of adipogenic genes in mouse and human MSCs in vitro. Adipogenic differentiation induced by CBD was significantly decreased by the PPARγ antagonist T0070907, supporting the hypothesis that CBD promoted differentiation via PPARγ. Taken together, our results indicate that in humans and in mice, CBD induced adipogenic differentiation in MSCs through a PPARγ-dependent mechanism.


Asunto(s)
Adipogénesis/efectos de los fármacos , Cannabidiol/farmacología , Lipogénesis/efectos de los fármacos , Lipólisis/efectos de los fármacos , Células Madre Mesenquimatosas/efectos de los fármacos , PPAR gamma/agonistas , Adipogénesis/fisiología , Animales , Benzamidas/farmacología , Línea Celular Transformada , Humanos , Lipogénesis/fisiología , Lipólisis/fisiología , Células Madre Mesenquimatosas/metabolismo , Ratones , PPAR gamma/antagonistas & inhibidores , PPAR gamma/metabolismo , Piridinas/farmacología
10.
Biochem Pharmacol ; 197: 114933, 2022 03.
Artículo en Inglés | MEDLINE | ID: mdl-35093393

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is becoming an increasingly serious disease worldwide. Unfortunately, no specific drug has been approved to treat NAFLD. Accumulating evidence suggests that lipotoxicity, which is induced by an excess of intracellular triacylglycerols (TAGs), is a potential mechanism underlying the ill-defined progression of NAFLD. Under physiological conditions, a balance is maintained between TAGs and free fatty acids (FFAs) in the liver. TAGs are catabolized to FFAs through neutral lipolysis and/or lipophagy, while FFAs can be anabolized to TAGs through an esterification reaction. However, in the livers of patients with NAFLD, lipophagy appears to fail. Reversing this abnormal state through several lipophagic molecules (mTORC1, AMPK, PLIN, etc.) facilitates NAFLD amelioration; therefore, restoring failed lipophagy may be a highly efficient therapeutic strategy for NAFLD. Here, we outline the lipophagy phases with the relevant important proteins and discuss the roles of lipophagy in the progression of NAFLD. Additionally, the potential candidate drugs with therapeutic value targeting these proteins are discussed to show novel strategies for future treatment of NAFLD.


Asunto(s)
Autofagia/efectos de los fármacos , Sistemas de Liberación de Medicamentos/métodos , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/metabolismo , Enfermedad del Hígado Graso no Alcohólico/tratamiento farmacológico , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Animales , Autofagosomas/efectos de los fármacos , Autofagosomas/metabolismo , Autofagia/fisiología , Berberina/administración & dosificación , Ácidos Grasos no Esterificados/antagonistas & inhibidores , Ácidos Grasos no Esterificados/metabolismo , Factores de Crecimiento de Fibroblastos/administración & dosificación , Humanos , Metabolismo de los Lípidos/fisiología , Lipólisis/efectos de los fármacos , Lipólisis/fisiología , Hígado/efectos de los fármacos , Diana Mecanicista del Complejo 1 de la Rapamicina/administración & dosificación , Canales de Potencial de Receptor Transitorio/administración & dosificación , Triglicéridos/antagonistas & inhibidores , Triglicéridos/metabolismo
11.
Food Funct ; 13(1): 131-142, 2022 Jan 04.
Artículo en Inglés | MEDLINE | ID: mdl-34870663

RESUMEN

Medium-chain triglyceride (MCT) and eicosapentaenoic acid (EPA) have been widely applied in nutritional supplementation. However, when administered individually or mixed, they were unable to maximize their nutritional value. Hence, EPA-rich medium- and long-chain triacylglycerol (MLCT) was synthesized from MCT and EPA-rich fish oil (FO) by enzymatic transesterification. The fatty acids in triglyceride (TAG) were rearranged which resulted in significant changes in TAG profiles compared to the physical mixture of MCT and FO (PM). EPA-containing MML (MML, MLM and LMM) and LLM (LLM, LML and MLL) type TAGs account for 70.21%. The fate of different oils (MCT, FO, PM, and MLCT) across the gastrointestinal tract was subsequently simulated using an in vitro digestion model. The results showed that the physical and structural characteristics of different oils during digestion depended upon the oil type and the microenvironment they were in. After 120 min of small intestine digestion, the degree of hydrolysis for MLCT was higher than that for the other three oils. The final FFA release level was in the following order: MLCT (102.79%) > MCT (95.20%) > PM (85.81%) > FO (74.18%). This can be attributed to the composition and positional distribution of fatty acids in TAGs. What's more, LCFAs (EPA) in MLCT mainly existed in the form of sn-2 MAG, which was conducive to their subsequent absorption and transport. These results may aid in the future rational design of structural lipids, thereby regulating lipid digestion and maximizing the nutritional value of oils.


Asunto(s)
Digestión/efectos de los fármacos , Ácido Eicosapentaenoico , Ácidos Grasos , Triglicéridos , Ácido Eicosapentaenoico/química , Ácido Eicosapentaenoico/metabolismo , Ácido Eicosapentaenoico/farmacología , Esterificación , Ácidos Grasos/química , Ácidos Grasos/metabolismo , Lipólisis/efectos de los fármacos , Modelos Biológicos , Triglicéridos/química , Triglicéridos/metabolismo , Triglicéridos/farmacología
12.
Biomed Pharmacother ; 145: 112440, 2022 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-34839254

RESUMEN

In this study, we investigated whether the activating transcription factor 3 (ATF3) inducer ST32db, a synthetic compound with a chemical structure similar to that of native Danshen compounds, exerts an anti-obesity effect in 3T3-L1 white preadipocytes, D16 beige cells, and mice with obesity induced by a high-fat diet (HFD). The results showed that ST32db inhibited 3T3-L1 preadipocyte differentiation by inhibiting adipogenesis/lipogenesis-related gene (and protein levels) and enhancing lipolysis-related gene (and protein levels) via the activation of ß3-adrenoceptor (ß3-AR)/PKA/p38, AMPK, and ERK pathways. Furthermore, ST32db inhibited triacylglycerol accumulation in D16 adipocytes by suppressing adipogenesis/lipogenesis-related gene (and protein levels) and upregulating browning gene expression by suppressing the ß3-AR/PKA/p38, and AMPK pathways. Intraperitoneally injected ST32db (1 mg kg-1 twice weekly) inhibited body weight gain and reduced the weight of inguinal white adipose tissue (iWAT), epididymal WAT (eWAT), and mesenteric WAT, with no effects on food intake by the obese mice. The adipocyte diameter and area of iWAT and eWAT were decreased in obese mice injected with ST32db compared with those administered only HFD. In addition, ST32db significantly suppressed adipogenesis and activated lipolysis, browning, mitochondrial oxidative phosphorylation, and ß-oxidation-related pathways by suppressing the p38 pathway in the iWAT of the obese mice. These results indicated that the ATF3 inducer ST32db has therapeutic potential for reducing obesity.


Asunto(s)
Factor de Transcripción Activador 3 , Fármacos Antiobesidad , Obesidad , Animales , Masculino , Ratones , Células 3T3-L1 , Factor de Transcripción Activador 3/efectos de los fármacos , Factor de Transcripción Activador 3/metabolismo , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adipogénesis/efectos de los fármacos , Tejido Adiposo Blanco/efectos de los fármacos , Fármacos Antiobesidad/farmacología , Diferenciación Celular/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Lipólisis/efectos de los fármacos , Ratones Endogámicos C57BL , Obesidad/tratamiento farmacológico , Aumento de Peso/efectos de los fármacos
13.
J Clin Invest ; 132(2)2022 01 18.
Artículo en Inglés | MEDLINE | ID: mdl-34847077

RESUMEN

The dysregulation of energy homeostasis in obesity involves multihormone resistance. Although leptin and insulin resistance have been well characterized, catecholamine resistance remains largely unexplored. Murine ß3-adrenergic receptor expression in adipocytes is orders of magnitude higher compared with that of other isoforms. While resistant to classical desensitization pathways, its mRNA (Adrb3) and protein expression are dramatically downregulated after ligand exposure (homologous desensitization). ß3-Adrenergic receptor downregulation also occurs after high-fat diet feeding, concurrent with catecholamine resistance and elevated inflammation. This downregulation is recapitulated in vitro by TNF-α treatment (heterologous desensitization). Both homologous and heterologous desensitization of Adrb3 were triggered by induction of the pseudokinase TRIB1 downstream of the EPAC/RAP2A/PI-PLC pathway. TRIB1 in turn degraded the primary transcriptional activator of Adrb3, CEBPα. EPAC/RAP inhibition enhanced catecholamine-stimulated lipolysis and energy expenditure in obese mice. Moreover, adipose tissue expression of genes in this pathway correlated with body weight extremes in a cohort of genetically diverse mice and with BMI in 2 independent cohorts of humans. These data implicate a signaling axis that may explain reduced hormone-stimulated lipolysis in obesity and resistance to therapeutic interventions with ß3-adrenergic receptor agonists.


Asunto(s)
Adipocitos/metabolismo , Catecolaminas/farmacología , Regulación hacia Abajo/efectos de los fármacos , Resistencia a Medicamentos/efectos de los fármacos , Obesidad/metabolismo , Receptores Adrenérgicos beta 3/metabolismo , Células 3T3-L1 , Animales , Regulación hacia Abajo/genética , Resistencia a Medicamentos/genética , Metabolismo Energético/efectos de los fármacos , Metabolismo Energético/genética , Lipólisis/efectos de los fármacos , Lipólisis/genética , Masculino , Ratones , Obesidad/tratamiento farmacológico , Obesidad/genética , Receptores Adrenérgicos beta 3/genética , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética
14.
Nutrients ; 13(12)2021 Dec 13.
Artículo en Inglés | MEDLINE | ID: mdl-34959999

RESUMEN

In this study, we investigated the pharmacological effect of a water extract of Raphani Semen (RSWE) on alcoholic fatty liver disease (AFLD) using ethanol-induced AFLD mice (the NIAAA model) and palmitic acid (PA)-induced steatosis HepG2 cells. An RSWE supplement improved serum and hepatic triglyceride (TG) levels of AFLD mice, as well as their liver histological structure. To explore the molecular action of RSWE in the improvement of AFLD, we investigated the effect of RSWE on four major pathways for lipid homeostasis in the liver: free fatty acid transport, lipogenesis, lipolysis, and ß-oxidation. Importantly, RSWE decreased the mRNA expression of de novo lipogenesis-related genes, such as Srebf1, Cebpa, Pparg, and Lpin1, as well as the protein levels of these factors, in the liver of AFLD mice. That these actions of RSWE affect lipogenesis was confirmed using PA-induced steatosis HepG2 cells. Overall, our findings suggest that RSWE has the potential for improvement of AFLD by inhibiting de novo lipogenesis.


Asunto(s)
Hígado Graso Alcohólico/tratamiento farmacológico , Lipogénesis/efectos de los fármacos , Extractos Vegetales/farmacología , Raphanus/química , Semillas/química , Animales , Etanol/efectos adversos , Ácidos Grasos no Esterificados/metabolismo , Hígado Graso Alcohólico/metabolismo , Células Hep G2 , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Lipólisis/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Oxidación-Reducción/efectos de los fármacos , Ácido Palmítico/efectos adversos , Fosfatidato Fosfatasa/metabolismo , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/metabolismo , Triglicéridos/sangre
15.
Int J Mol Sci ; 22(23)2021 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-34884912

RESUMEN

Growth hormone and insulin-like growth factors (GH/IGF axis) regulate somatic growth in mammals and fish, although their action on metabolism is not fully understood in the latter. An intraperitoneal injection of extended-release recombinant bovine growth hormone (rbGH, Posilac®) was used in gilthead sea bream fingerlings and juveniles to analyse the metabolic response of liver and red and white muscles by enzymatic, isotopic and proteomic analyses. GH-induced lipolysis and glycogenolysis were reflected in liver composition, and metabolic and redox enzymes reported higher lipid use and lower protein oxidation. In white and red muscle reserves, rBGH increased glycogen while reducing lipid. The isotopic analysis of muscles showed a decrease in the recycling of proteins and a greater recycling of lipids and glycogen in the rBGH groups, which favoured a protein sparing effect. The protein synthesis capacity (RNA/protein) of white muscle increased, while cytochrome-c-oxidase (COX) protein expression decreased in rBGH group. Proteomic analysis of white muscle revealed only downregulation of 8 proteins, related to carbohydrate metabolic processes. The global results corroborated that GH acted by saving dietary proteins for muscle growth mainly by promoting the use of lipids as energy in the muscles of the gilthead sea bream. There was a fuel switch from carbohydrates to lipids with compensatory changes in antioxidant pathways that overall resulted in enhanced somatic growth.


Asunto(s)
Complejo IV de Transporte de Electrones/metabolismo , Hormona del Crecimiento/administración & dosificación , Dorada/crecimiento & desarrollo , Somatomedinas/metabolismo , Animales , Bovinos , Proteínas de Peces/metabolismo , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Glucógeno/metabolismo , Glucogenólisis/efectos de los fármacos , Hormona del Crecimiento/genética , Hormona del Crecimiento/farmacología , Marcaje Isotópico , Lipólisis/efectos de los fármacos , Proteómica , Proteínas Recombinantes/administración & dosificación , Proteínas Recombinantes/farmacología , Dorada/metabolismo
16.
Molecules ; 26(23)2021 Nov 29.
Artículo en Inglés | MEDLINE | ID: mdl-34885819

RESUMEN

The present study reports a cost-effective, environmentally friendly method to increase the bioavailability and bio-efficacy of B. rufescens stem bark extract in the biological system via functional modification as B. rufescens stem bark nanoparticles (BR-TO2-NPs). The biosynthesis of BR- -NPs was confirmed by UV-visible (UV-vis) and Fourier-transform infrared (FT-IR) spectroscopy, transmission electron microscopy (TEM), and X-ray diffraction analyses. The shifts in FT-IR stretching vibrations of carboxylic and nitro groups (1615 cm-1), the O-H of phenolics or carboxylic acids (3405 cm-1), alkanes, and alkyne groups (2925 and 2224 cm-1) of the plant extract and lattice (455) indicated successful biosynthesis of BR- -NPs. Compared with the stem bark extract, 40 ng/dL dose of BR- -NPs led to a reduction in adipogenesis and an increase in mitochondrial biogenesis-related gene expressions, adiponectin-R1, PPARγC1α, UCP-1, and PRDM16, in maturing-adipocytes. This confirmed the intracellular uptake, bioavailability, and bio-efficiency of BR-TiO2-NPs. The lipid-lowering capacity of BR-TiO2-NPs effectively inhibited the metabolic inflammation-related gene markers, IL-6, TNF-α, LTB4-R, and Nf-κb. Further, BR-TiO2-NPs stimulating mitochondrial thermogenesis capacity was proven by the significantly enhanced CREB-1 and AMPK protein levels in adipocytes. In conclusion, BR-TiO2-NPs effectively inhibited lipid accumulation and proinflammatory adipokine levels in maturing adipocytes; it may help to overcome obesity-associated comorbidities.


Asunto(s)
Adipocitos/citología , Adipocitos/metabolismo , Adipoquinas/metabolismo , Bauhinia/química , Metabolismo de los Lípidos , Nanopartículas del Metal/química , Corteza de la Planta/química , Titanio/farmacología , Adipogénesis/efectos de los fármacos , Adipogénesis/genética , Muerte Celular/efectos de los fármacos , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/genética , Forma de la Célula/efectos de los fármacos , Cromatografía de Gases y Espectrometría de Masas , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Lipólisis/efectos de los fármacos , Lipólisis/genética , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/metabolismo , Nanopartículas del Metal/ultraestructura , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Estrés Oxidativo/efectos de los fármacos , Tallos de la Planta/química , Termogénesis/efectos de los fármacos , Termogénesis/genética
17.
Biomolecules ; 11(12)2021 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-34944525

RESUMEN

Obesity is a low-grade systemic inflammatory disease involving adipocytokines. As though Anmyungambi decoction (AMGB) showed significant improvement on obesity in a clinical trial, the molecular mechanism of AMGB in obesity remains unknown. Therefore, we explored the potential mechanisms of action of AMGB on obesity through network pharmacological approaches. We revealed that targets of AMGB are significantly associated with obesity-related and adipocyte-elevated genes. Evodiamine, berberine, genipin, palmitic acid, genistein, and quercetin were shown to regulate adipocytokine signaling pathway proteins which mainly involved tumor necrosis factor receptor 1, leptin receptor. In terms of the regulatory pathway of lipolysis in adipocytes, norephedrine, pseudoephedrine, quercetin, and limonin were shown to affect adrenergic receptor-beta, protein kinase A, etc. We also found that AMGB has the potentials to enhance the insulin signaling pathway thereby preventing type II diabetes mellitus. Additionally, AMGB was discovered to be able to control not only insulin-related proteins but also inflammatory mediators and apoptotic regulators and caspases, hence reducing hepatocyte injury in nonalcoholic fatty liver disease. Our findings help develop a better understanding of how AMGB controls obesity.


Asunto(s)
Adipoquinas/genética , Obesidad/genética , Extractos Vegetales/farmacología , Adipocitos/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Lipólisis/efectos de los fármacos , Farmacología en Red , Obesidad/tratamiento farmacológico , Extractos Vegetales/química , Extractos Vegetales/uso terapéutico , Transducción de Señal/efectos de los fármacos
18.
Biomed Res Int ; 2021: 2521273, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34812408

RESUMEN

Obesity is a public health problem characterized by increased body weight due to abnormal adipose tissue expansion. Bioactive compound consumption from the diet or intake of dietary supplements is one of the possible ways to control obesity. Natural products with adipogenesis-regulating potential act as obesity treatments. We evaluated the synergistic antiangiogenesis, antiadipogenic and antilipogenic efficacy of standardized rebaudioside A, sativoside, and theasaponin E1 formulations (RASE1) in vitro in human umbilical vein endothelial cells (HUVECs), 3T3-L1 preadipocytes respectively, and in vivo using a high-fat and carbohydrate diet-induced obesity mouse model. Orlistat was used as a positive control, while untreated cells and animals were normal controls (NCs). Adipose tissue, liver, and blood were analyzed after dissection. Extracted stevia compounds and green tea seed saponin E1 exhibited pronounced antiobesity effects when combined. RASE1 inhibited HUVEC proliferation and tube formation by suppressing VEGFR2, NF-κB, PIK3, and-catenin beta-1 expression levels. RASE1 inhibited 3T3-L1 adipocyte differentiation and lipid accumulation by downregulating adipogenesis- and lipogenesis-promoting genes. RASE1 oral administration reduced mouse body and body fat pad weight and blood cholesterol, TG, ALT, AST, glucose, insulin, and adipokine levels. RASE1 suppressed adipogenic and lipid metabolism gene expression in mouse adipose and liver tissues and enhanced AMP-activated protein kinase levels in liver and adipose tissues and in serum adiponectin. RASE1 suppressed the NF-κB pathway and proinflammatory cytokines IL-10, IL-6, and TNF-α levels in mice which involve inflammation and progression of obesity. The overall results indicate RASE1 is a potential therapeutic formulation and functional food for treating or preventing obesity and inflammation.


Asunto(s)
Productos Biológicos/uso terapéutico , Inflamación/tratamiento farmacológico , Obesidad/tratamiento farmacológico , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Adipogénesis/efectos de los fármacos , Adipogénesis/genética , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Productos Biológicos/administración & dosificación , Productos Biológicos/toxicidad , Modelos Animales de Enfermedad , Diterpenos de Tipo Kaurano/administración & dosificación , Composición de Medicamentos , Sinergismo Farmacológico , Femenino , Glucósidos/administración & dosificación , Células Endoteliales de la Vena Umbilical Humana , Humanos , Inflamación/metabolismo , Metabolismo de los Lípidos/efectos de los fármacos , Lipogénesis/efectos de los fármacos , Lipogénesis/genética , Lipólisis/efectos de los fármacos , Ratones , Ratones Endogámicos ICR , Obesidad/genética , Obesidad/metabolismo , Ácido Oleanólico/administración & dosificación , Ácido Oleanólico/análogos & derivados , Fitoterapia , ARN Mensajero/genética , ARN Mensajero/metabolismo , Saponinas/administración & dosificación , Transducción de Señal/efectos de los fármacos , Stevia/química , Té/química
19.
Nat Metab ; 3(11): 1445-1465, 2021 11.
Artículo en Inglés | MEDLINE | ID: mdl-34799702

RESUMEN

The perception that intracellular lipolysis is a straightforward process that releases fatty acids from fat stores in adipose tissue to generate energy has experienced major revisions over the last two decades. The discovery of new lipolytic enzymes and coregulators, the demonstration that lipophagy and lysosomal lipolysis contribute to the degradation of cellular lipid stores and the characterization of numerous factors and signalling pathways that regulate lipid hydrolysis on transcriptional and post-transcriptional levels have revolutionized our understanding of lipolysis. In this review, we focus on the mechanisms that facilitate intracellular fatty-acid mobilization, drawing on canonical and noncanonical enzymatic pathways. We summarize how intracellular lipolysis affects lipid-mediated signalling, metabolic regulation and energy homeostasis in multiple organs. Finally, we examine how these processes affect pathogenesis and how lipolysis may be targeted to potentially prevent or treat various diseases.


Asunto(s)
Tejido Adiposo/fisiología , Movilización Lipídica , Lipólisis/fisiología , Tejido Adiposo/efectos de los fármacos , Animales , Biomarcadores , Manejo de la Enfermedad , Susceptibilidad a Enfermedades , Metabolismo Energético/efectos de los fármacos , Ácidos Grasos/metabolismo , Regulación Enzimológica de la Expresión Génica , Humanos , Movilización Lipídica/efectos de los fármacos , Lipólisis/efectos de los fármacos , Redes y Vías Metabólicas/efectos de los fármacos , Terapia Molecular Dirigida , Procesamiento Proteico-Postraduccional , Triglicéridos/metabolismo
20.
Nutrients ; 13(11)2021 Oct 29.
Artículo en Inglés | MEDLINE | ID: mdl-34836142

RESUMEN

The nutritional relevance of food compositional data could be improved by taking the bioaccessibility of these constituents into account. A lack of routine methods to assess the bioaccessibility of fatty acids (FAs) in food is one of the limiting factors of doing so. An analytical protocol is proposed for routine assessment of the extent of lipolysis via in vitro digestion simulation methods in food products. The established method provides specific information on each FA individually. Steps of the protocol including the Bligh and Dyer chloroform/methanol/water extraction of esterified and free FAs from in vitro digesta, methyl ester derivatization, and GC-FID analysis were specifically tailored to help routine work and were harmonized with the Infogest in vitro digestion simulation protocol (both v1.0 and v2.0). The method was applied to assess the degree of FA-specific lipolysis in a baked fish (carp) meal and the results showed that the FA composition of the original food significantly differed from that of the distribution of FFAs in the digesta. The use of gastric lipase (in Infogest v2.0 protocol) increased total FA release by 9.5% and its specific impact on palmitic acid was the most prominent.


Asunto(s)
Cromatografía de Gases/métodos , Digestión , Ácidos Grasos/análisis , Técnicas In Vitro/métodos , Lipólisis/efectos de los fármacos , Ácidos Grasos no Esterificados/análisis , Humanos , Modelos Biológicos , Valor Nutritivo , Alimentos Marinos/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA