Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
Free Radic Res ; 54(11-12): 918-930, 2020 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-32623920

RESUMEN

The translocation of transcription factor EB (TFEB) to the nucleus plays a pivotal role in the regulation of basic cellular processes, such as lysosome biogenesis and autophagy. Autophagy is an intracellular degradation system that delivers cytoplasmic constituents to the lysosome, which is important in maintaining cellular homeostasis during environmental stress. Furthermore, oxidative stress is a critical cause for the progression of neurodegenerative diseases. Curcumin has anti-oxidative and anti-inflammatory activities, and is expected to have potential therapeutic effects in various diseases. In this study, we demonstrated that curcumin regulated TFEB export signalling via inhibition of glycogen synthase kinase-3ß (GSK-3ß); GSK-3ß was inactivated by curcumin, leading to reduced phosphorylation of TFEB. We further showed that H2O2-induced oxidative stress was reduced by curcumin via the Nrf2/HO-1 pathway in human neuroblastoma cells. In addition, we showed that curcumin induced the degradation of amyloidogenic proteins, including amyloid-ß precursor protein and α-synuclein, through the TFEB-autophagy/lysosomal pathway. In conclusion, curcumin regulates autophagy by controlling TFEB through the inhibition of GSK-3ß, and increases antioxidant gene expression in human neuroblastoma cells. These results contribute to the development of novel cellular therapies for neurodegenerative diseases.


Asunto(s)
Precursor de Proteína beta-Amiloide/metabolismo , Antineoplásicos/uso terapéutico , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/metabolismo , Curcumina/uso terapéutico , Glucógeno Sintasa Quinasa 3 beta/antagonistas & inhibidores , Neuroblastoma/genética , Antineoplásicos/farmacología , Línea Celular Tumoral , Curcumina/farmacología , Humanos , Especies Reactivas de Oxígeno , Transfección
2.
Mol Cells ; 43(5): 431-437, 2020 May 31.
Artículo en Inglés | MEDLINE | ID: mdl-32392909

RESUMEN

The hypothalamus is a crucial organ for the maintenance of appropriate body fat storage. Neurons in the hypothalamic arcuate nucleus (ARH) detect energy shortage or surplus via the circulating concentrations of metabolic hormones and nutrients, and then coordinate energy intake and expenditure to maintain energy homeostasis. Malfunction or loss of hypothalamic ARH neurons results in obesity. Accumulated evidence suggests that hypothalamic inflammation is a key pathological mechanism that links chronic overconsumption of a high-fat diet (HFD) with the development of obesity and related metabolic complications. Interestingly, overnutrition-induced hypothalamic inflammation occurs specifically in the ARH, where microglia initiate an inflammatory response by releasing proinflammatory cytokines and chemokines in response to excessive fatty acid flux. Upon more prolonged HFD consumption, astrocytes and perivascular macrophages become involved and sustain hypothalamic inflammation. ARH neurons are victims of hypothalamic inflammation, but they may actively participate in hypothalamic inflammation by sending quiescence or stress signals to surrounding glia. In this mini-review, we describe the current state of knowledge regarding the contributions of neurons and glia, and their interactions, to HFD-induced hypothalamic inflammation.


Asunto(s)
Tejido Adiposo/inmunología , Hipotálamo/inmunología , Inflamación/metabolismo , Macrófagos/inmunología , Microglía/inmunología , Neuronas/inmunología , Obesidad/inmunología , Animales , Citocinas/metabolismo , Dieta Alta en Grasa , Metabolismo Energético , Humanos , Inmunidad Celular , Inflamación Neurogénica
3.
J Neuroinflammation ; 16(1): 221, 2019 Nov 14.
Artículo en Inglés | MEDLINE | ID: mdl-31727092

RESUMEN

BACKGROUND: Obese mice on a high-fat diet (HFD) display signs of inflammation in the hypothalamic arcuate nucleus (ARC), a critical area for controlling systemic energy metabolism. This has been suggested as a key mechanism of obesity-associated hypothalamic dysfunction. We reported earlier that bone marrow-derived macrophages accumulate in the ARC to sustain hypothalamic inflammation upon chronic exposure to an HFD. However, the mechanism underlying hypothalamic macrophage accumulation has remained unclear. METHODS: We investigated whether circulating monocytes or myeloid precursors contribute to hypothalamic macrophage expansion during chronic HFD feeding. To trace circulating myeloid cells, we generated mice that express green fluorescent protein (GFP) in their lysozyme M-expressing myeloid cells (LysMGFP mice). We conducted parabiosis and bone marrow transplantation experiments using these animals. Mice received an HFD for 12 or 30 weeks and were then sacrificed to analyze LysMGFP cells in the hypothalamus. Hypothalamic vascular permeability in the HFD-fed obese mice was also tested by examining the extravascular leakage of Evans blue and fluorescence-labeled albumin. The timing of LysMGFP cell entry to the hypothalamus during development was also evaluated. RESULTS: Our parabiosis and bone marrow transplantation experiments revealed a significant infiltration of circulating LysMGFP cells into the liver, skeletal muscle, choroid plexus, and leptomeninges but not in the hypothalamic ARC during chronic HFD feeding, despite increased hypothalamic vascular permeability. These results suggested that the recruitment of circulating monocytes is not a major mechanism for maintaining and expanding the hypothalamic macrophage population in diet-induced obesity. We demonstrated instead that LysMGFP cells infiltrate the hypothalamus during its development. LysMGFP cells appeared in the hypothalamic area from the late embryonic period. This cellular pool suddenly increased immediately after birth, peaked at the postnatal second week, and adopted an adult pattern of distribution after weaning. CONCLUSIONS: Bone marrow-derived macrophages mostly populate the hypothalamus in early postnatal life and may maintain their pool without significant recruitment of circulating monocytes throughout life, even under conditions of chronic HFD feeding.


Asunto(s)
Hipotálamo/metabolismo , Macrófagos/metabolismo , Obesidad/metabolismo , Animales , Trasplante de Médula Ósea , Permeabilidad Capilar , Dieta Alta en Grasa , Metabolismo Energético , Resistencia a la Insulina/fisiología , Hígado/metabolismo , Masculino , Ratones , Parabiosis
4.
Metabolism ; 88: 51-60, 2018 11.
Artículo en Inglés | MEDLINE | ID: mdl-30179604

RESUMEN

BACKGROUND: Nicotinamide adenine dinucleotide (NAD)-dependent deacetylase SIRT1 is an important regulator of hypothalamic neuronal function. Thus, an adequate hypothalamic NAD content is critical for maintaining normal energy homeostasis. METHODS: We investigated whether NAD supplementation increases hypothalamic NAD levels and affects energy metabolism in mice. Furthermore, we investigated the mechanisms underlying the effects of exogenous NAD on central metabolism upon entering the hypothalamus. RESULTS: Central and peripheral NAD administration suppressed fasting-induced hyperphagia and weight gain in mice. Extracellular NAD was imported into N1 hypothalamic neuronal cells in a connexin 43-dependent and CD73-independent manner. Consistent with the in vitro data, inhibition of hypothalamic connexin 43 blocked hypothalamic NAD uptake and NAD-induced anorexia. Exogenous NAD suppressed NPY and AgRP transcriptional activity, which was mediated by SIRT1 and FOXO1. CONCLUSIONS: Exogenous NAD is effectively transported to the hypothalamus via a connexin 43-dependent mechanism and increases hypothalamic NAD content. Therefore, NAD supplementation is a potential therapeutic method for metabolic disorders characterized by hypothalamic NAD depletion.


Asunto(s)
Conexina 43/metabolismo , Metabolismo Energético/efectos de los fármacos , Hipotálamo/efectos de los fármacos , NAD/farmacología , Proteína Relacionada con Agouti/genética , Animales , Transporte Biológico , Hiperfagia/prevención & control , Hipotálamo/citología , Hipotálamo/metabolismo , Inyecciones Intraperitoneales , Inyecciones Intraventriculares , Masculino , Ratones Endogámicos C57BL , NAD/administración & dosificación , Neuronas/metabolismo , Neuropéptido Y/genética , Sirtuina 1/metabolismo , Transcripción Genética/efectos de los fármacos , Aumento de Peso/efectos de los fármacos
5.
J Med Food ; 21(6): 551-559, 2018 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-29569982

RESUMEN

The inflammatory cytokine tumor necrosis factor α (TNFα), upregulated in the obese condition, promotes protein degradation and is implicated in obesity-related skeletal muscle atrophy and age-related sarcopenia. Quercetin, a flavonoid, elicits antioxidative and anti-inflammatory activities. In this study, we investigated the effect of quercetin on TNFα-induced skeletal muscle atrophy as well as its potential mechanism of action. In this study, we observed that quercetin suppressed expression of TNFα-induced atrophic factors such as MAFbx/atrogin-1 and MuRF1 in myotubes, and it enhanced heme oxygenase-1 (HO-1) protein level accompanied by increased nuclear translocation of nuclear factor erythroid 2-related factor 2 (Nrf2) in myotubes. The HO-1 inhibitor ZnPP suppressed the inhibitory actions of quercetin on TNFα-induced atrophic responses and degradation of IκB-α in myotubes. Moreover, quercetin supplementation to high-fat diet-fed obese mice inhibited obesity-induced atrophic responses in skeletal muscle, accompanied by upregulation of HO-1 and inactivation of nuclear factor-kappa B (NF-κB), and the quercetin actions were attenuated in Nrf2-deficient mice. These findings suggest that quercetin protects against TNFα-induced muscle atrophy under obese conditions through Nrf2-mediated HO-1 induction accompanied by inactivation of NF-κB. Quercetin may be used as a dietary supplement to protect against obesity-induced skeletal muscle atrophy.


Asunto(s)
Hemo-Oxigenasa 1/genética , Atrofia Muscular/tratamiento farmacológico , Atrofia Muscular/genética , Obesidad/complicaciones , Quercetina/administración & dosificación , Factor de Necrosis Tumoral alfa/inmunología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Hemo-Oxigenasa 1/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/metabolismo , Atrofia Muscular/etiología , Atrofia Muscular/inmunología , Factor 2 Relacionado con NF-E2/genética , Factor 2 Relacionado con NF-E2/metabolismo , Obesidad/genética , Obesidad/metabolismo , Factor de Necrosis Tumoral alfa/genética , Regulación hacia Arriba
6.
Lipids ; 52(7): 657-664, 2017 07.
Artículo en Inglés | MEDLINE | ID: mdl-28601955

RESUMEN

Although the Apiaceae herb family has been traditionally used for the management of type 2 diabetes, its molecular mechanism has not been clarified. Coumarin derivatives, which are abundant in plants of the Apiaceae family, were evaluated for their effects on adipogenesis. We found that suksdorfin significantly promoted adipocyte differentiation and enhanced production of adiponectin, an anti-diabetic adipokine. We also demonstrated that suksdorfin activates peroxisome proliferator-activated receptor gamma (PPARγ), a master regulator of adipogenesis. Furthermore, we showed metabolic disorders in obese diabetic KK-Ay mice were attenuated by suksdorfin feeding. Suksdorfin intake induced adipocyte miniaturization and increased expression levels of PPARγ target genes related to adipocyte differentiation. These results indicated that suksdorfin induces adipogenesis in white adipose tissue (WAT) via the activation of PPARγ, leading to improvement of obesity-induced metabolic disorders. Therefore, suksdorfin-mediated amelioration of WAT dysfunctions might be responsible for the anti-diabetic effects of traditional herbal medicine therapy with Apiaceae.


Asunto(s)
Adipocitos/efectos de los fármacos , Cumarinas/administración & dosificación , Trastornos del Metabolismo de la Glucosa/tratamiento farmacológico , PPAR gamma/metabolismo , Células 3T3-L1 , Adipocitos/citología , Adipocitos/metabolismo , Adiponectina/metabolismo , Animales , Apiaceae/química , Diferenciación Celular/efectos de los fármacos , Cumarinas/farmacología , Activación Enzimática/efectos de los fármacos , Regulación Enzimológica de la Expresión Génica/efectos de los fármacos , Trastornos del Metabolismo de la Glucosa/enzimología , Ratones , Ratones Obesos , Transducción de Señal/efectos de los fármacos
7.
Nutrients ; 9(7)2017 Jun 23.
Artículo en Inglés | MEDLINE | ID: mdl-28644409

RESUMEN

Obesity-induced hypothalamic inflammation is characterized by activation of microglia, which are resident macrophages of the central nervous system, and is implicated in the derangement of energy homeostasis, metabolic complications, and neurodegenerative diseases. Quercetin, a naturally occurring flavonoid, is known to protect against oxidative stress and inflammation-related metabolic complications. Here, we demonstrate that quercetin reduces obesity-induced hypothalamic inflammation by inhibiting microglia-mediated inflammatory responses, and the beneficial action of quercetin is associated with heme oxygenase (HO-1) induction. Quercetin markedly reduced the production of inflammatory mediators (monocyte chemoattractant protein (MCP)-1, interleukin (IL-6), IL-1ß, nitric oxide) by microglia stimulated with saturated fatty acid palmitate and/or lipid-laden microglia-conditioned medium. Quercetin also upregulated the expression of HO-1 in palmitate-treated lipid-laden microglia, and the actions of quercetin against microglia activation accompanied by IκBα degradation were abolished by a HO-1 inhibitor. Moreover, quercetin supplementation reduced the levels of inflammatory cytokines and microglia activation markers in the hypothalamus of high fat diet (HFD)-fed obese mice, which was accompanied by upregulation of HO-1. These findings indicate that quercetin suppresses microglia-mediated inflammatory responses via the induction of HO-1, and hence protects against obesity-induced hypothalamic inflammation.


Asunto(s)
Hemo-Oxigenasa 1/metabolismo , Hipotálamo/patología , Inflamación/inducido químicamente , Proteínas de la Membrana/metabolismo , Microglía/efectos de los fármacos , Obesidad/complicaciones , Quercetina/farmacología , Animales , Técnicas de Cultivo de Célula , Línea Celular , Medios de Cultivo Condicionados , Dieta Alta en Grasa/efectos adversos , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/efectos adversos , Regulación de la Expresión Génica/efectos de los fármacos , Hemo-Oxigenasa 1/genética , Inflamación/tratamiento farmacológico , Masculino , Proteínas de la Membrana/genética , Ratones , Obesidad/inducido químicamente , Distribución Aleatoria
8.
FEBS Lett ; 591(12): 1742-1751, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28542876

RESUMEN

Obesity-induced hypothalamic inflammation is closely associated with various metabolic complications and neurodegenerative disorders. Astrocytes, the most abundant glial cells in the central nervous system, play a crucial role in pathological hypothalamic inflammatory processes. Here, we demonstrate that hypothalamic astrocytes accumulate lipid droplets under saturated fatty acid-rich conditions, such as obese environment, and that the lipid-laden astrocytes increase astrogliosis markers and inflammatory cytokines (TNFα, IL-1ß, IL-6, MCP-1) at the transcript and/or protein level. Medium conditioned by the lipid-laden astrocytes stimulate microglial chemotactic activity and upregulate transcripts of the microglia activation marker Iba-1 and inflammatory cytokines. These findings indicate that the lipid-laden astrocytes formed in free fatty acid-rich obese condition may participate in obesity-induced hypothalamic inflammation through promoting microglia migration and activation.


Asunto(s)
Astrocitos/metabolismo , Citocinas/metabolismo , Regulación de la Expresión Génica , Hipotálamo/metabolismo , Metabolismo de los Lípidos , Microglía/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Animales , Animales Recién Nacidos , Astrocitos/citología , Astrocitos/inmunología , Astrocitos/patología , Biomarcadores/metabolismo , Línea Celular , Movimiento Celular , Células Cultivadas , Quimiotaxis , Citocinas/genética , Ácidos Grasos no Esterificados/efectos adversos , Hipotálamo/citología , Hipotálamo/inmunología , Hipotálamo/patología , Gotas Lipídicas/inmunología , Gotas Lipídicas/metabolismo , Gotas Lipídicas/patología , Ratones Endogámicos C57BL , Microglía/citología , Microglía/inmunología , Microglía/patología , Proteínas del Tejido Nervioso/genética , Obesidad/inmunología , Obesidad/metabolismo , Obesidad/patología , Ácido Palmítico/efectos adversos , ARN Mensajero
9.
Obesity (Silver Spring) ; 24(11): 2351-2360, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27619735

RESUMEN

OBJECTIVE: Obesity-induced inflammation plays a pivotal role in the pathogenesis of insulin resistance and type 2 diabetes. Xanthoangelol (XA) and 4-hydroxyderrcin (4-HD), phytochemicals extracted from Angelica keiskei, have been reported to possess various biological properties. Whether XA and 4-HD alleviate obesity-induced inflammation and inflammation-induced adipocyte dysfunction was investigated. METHODS: For the in vitro study, a co-culture system composed of macrophages and adipocytes and macrophages stimulated with conditioned medium derived from fully differentiated adipocytes was conducted. For the in vivo study, mice were fed a high-fat diet supplemented with XA for 14 weeks. RESULTS: XA and 4-HD suppressed inflammatory factors in co-culture system. Moreover, treatment of RAW macrophages with XA and 4-HD moderated the suppression of uncoupling protein 1 promoter activity and gene expression in C3H10T1/2 adipocytes, which was induced by conditioned medium derived from LPS-stimulated RAW macrophages. Also, XA and 4-HD inhibited c-Jun N-terminal kinase phosphorylation, nuclear factor-κB, and activator protein 1, the last two being transcription activators in activated macrophages. Furthermore, in mice fed the high-fat diet, XA reduced inflammatory factors within the white adipose tissue. CONCLUSIONS: These results suggest that XA and 4-HD might be promising phytochemicals to suppress obesity-induced inflammation and inflammation-induced adipocyte dysfunction.


Asunto(s)
Angelica/química , Chalcona/análogos & derivados , Obesidad/tratamiento farmacológico , Fitoterapia , Extractos Vegetales/farmacología , Adipocitos/efectos de los fármacos , Animales , Diferenciación Celular/efectos de los fármacos , Chalcona/farmacología , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Dieta Alta en Grasa , Inflamación/tratamiento farmacológico , Inflamación/etiología , Proteínas Quinasas JNK Activadas por Mitógenos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/efectos de los fármacos , Obesidad/complicaciones , Obesidad/fisiopatología , Fosforilación/efectos de los fármacos , Factor de Transcripción AP-1/efectos de los fármacos
10.
J Med Food ; 19(7): 678-85, 2016 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-27322965

RESUMEN

Obesity-induced adipose inflammation plays a crucial role in the development of obesity-induced metabolic disorders such as insulin resistance and type 2 diabetes. In the presence of obesity, hypertrophic adipocytes release inflammatory mediators, including tumor necrosis factor-alpha (TNFα) and monocyte chemoattractant protein-1 (MCP-1), which enhance the recruitment and activation of macrophages, and in turn augment adipose inflammation. We demonstrate that the soy peptide Phe-Leu-Val (FLV) reduces inflammatory responses and insulin resistance in mature adipocytes. Specifically, the soy peptide FLV inhibits the release of inflammatory cytokines (TNFα, MCP-1, and IL-6) from both TNFα-stimulated adipocytes and cocultured adipocytes/macrophages. This inhibition is mediated by the inactivation of the inflammatory signaling molecules c-Jun N-terminal kinase (JNK) and IκB kinase (IKK), and the downregulation of IκBα in the adipocytes. In addition, soy peptide FLV enhances insulin responsiveness and increases glucose uptake in adipocytes. More importantly, we, for the first time, found that adipocytes express peptide transporter 2 (PepT2) protein, and the beneficial action of the soy peptide FLV was disrupted by the peptide transporter inhibitor GlySar. These findings suggest that soy peptide FLV is transported into adipocytes by PepT2 and then downregulates TNFα-induced inflammatory signaling, thereby increasing insulin responsiveness in the cells. The soy peptide FLV, therefore, has the potential to prevent obesity-induced adipose inflammation and insulin resistance.


Asunto(s)
Adipocitos/efectos de los fármacos , Antiinflamatorios , Glycine max/química , Resistencia a la Insulina , Oligopéptidos/farmacología , Factor de Necrosis Tumoral alfa/farmacología , Adipocitos/metabolismo , Quimiocina CCL2/metabolismo , Citocinas/metabolismo , Humanos , Inflamación/prevención & control , Obesidad/metabolismo , Transducción de Señal/efectos de los fármacos , Simportadores/antagonistas & inhibidores , Simportadores/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
11.
Biochem Biophys Res Commun ; 464(3): 840-7, 2015 Aug 28.
Artículo en Inglés | MEDLINE | ID: mdl-26188090

RESUMEN

This study examined whether oral administration of an arginase inhibitor regulates adipose tissue macrophage infiltration and inflammation in mice with high fat diet (HFD)-induced obesity. Male C57BL/6 mice (n = 30) were randomly assigned to control (CTL, n = 10), HFD only (n = 10), and HFD with arginase inhibitor N(ω)-hydroxy-nor-l-arginine (HFD with nor-NOHA, n = 10) groups. Plasma and mRNA levels of cytokines in epididymal adipose tissues (EAT), macrophage infiltration into EAT, and macrophage phenotype polarization were measured in the animals after 12 weeks. Additionally, the effects of nor-NOHA on adipose tissue macrophage infiltration and mRNA expression of cytokines were measured in co-cultured 3T3-L1 adipocytes and RAW 264.7 macrophages. Macrophage infiltration into the adipocytes was significantly suppressed by nor-NOHA treatment in adipocyte/macrophage co-culture system and mice with HFD-induced obesity. Pro-inflammatory cytokines, including monocyte chemoattractant protein-1 (MCP-1), tumor necrosis factor-alpha (TNF-α), and interleukin-6 (IL-6), were significantly downregulated, and the anti-inflammatory cytokine IL-10 was significantly upregulated in nor-NOHA-treated co-cultured cells. In the mice with HFD-induced obesity, plasma and mRNA levels of MCP-1 significantly reduced after supplementation with nor-NOHA. In addition, oral supplement of nor-NOHA modified M1/M2 phenotype ratio in the EAT. Oral supplementation of an arginase inhibitor, nor-NOHA, altered M1/M2 macrophage phenotype and macrophage infiltration into HFD-induced obese adipose tissue, thereby improved adipose tissue inflammatory response. These results may indicate that arginase inhibition ameliorates obesity-induced adipose tissue inflammation.


Asunto(s)
Arginasa/antagonistas & inhibidores , Arginina/análogos & derivados , Inhibidores Enzimáticos/farmacología , Obesidad/complicaciones , Paniculitis/tratamiento farmacológico , Células 3T3-L1/efectos de los fármacos , Células 3T3-L1/metabolismo , Adipocitos/efectos de los fármacos , Tejido Adiposo/metabolismo , Administración Oral , Animales , Arginasa/metabolismo , Arginina/administración & dosificación , Arginina/farmacología , Quimiocina CCL2/genética , Quimiocina CCL2/metabolismo , Técnicas de Cocultivo , Citocinas/sangre , Citocinas/genética , Dieta Alta en Grasa/efectos adversos , Inhibidores Enzimáticos/administración & dosificación , Regulación de la Expresión Génica/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/tratamiento farmacológico , Obesidad/etiología , Paniculitis/etiología
12.
J Med Food ; 17(5): 550-7, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24325454

RESUMEN

Obesity-induced inflammation is characterized by recruitment of adipose tissue macrophages that release inflammatory cytokines and chemokines. MIP-1α (macrophage inflammatory protein 1α)/CCL3, a CC chemokine, induces monocyte/macrophage infiltration and thus is implicated in obesity-induced adipose inflammation. Quercetin has been shown to modulate obesity-induced inflammation, but the mechanism of its action remains unclear. Here we demonstrate that quercetin decreases MIP-1α release from adipocytes and macrophages and from cocultured adipocytes/macrophages; it also opposes MIP-1α-induced macrophage infiltration and activation. The inhibitory action of quercetin on the MIP-1α-induced inflammatory responses of macrophages is mediated by downregulation of CCR1/CCR5, and inhibition of activation of JNK, p38 mitogen-activated-protein kinase (MAPK), and IKK as well as IκBα degradation. These findings suggest that quercetin may be a useful agent against obesity-induced adipose tissue inflammation.


Asunto(s)
Tejido Adiposo , Quimiocina CCL3/antagonistas & inhibidores , Inflamación/prevención & control , Quercetina/farmacología , Receptores CCR/genética , Células 3T3-L1 , Adipocitos/metabolismo , Tejido Adiposo/química , Animales , Línea Celular , Quimiocina CCL3/genética , Quimiocina CCL3/fisiología , Quimiotaxis/efectos de los fármacos , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Regulación hacia Abajo/efectos de los fármacos , Activación Enzimática/efectos de los fármacos , Inflamación/etiología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Obesidad/complicaciones , ARN Mensajero/análisis , Receptores CCR1/genética , Receptores CCR5/genética , Transducción de Señal/efectos de los fármacos , Proteínas Quinasas p38 Activadas por Mitógenos
13.
Obesity (Silver Spring) ; 21(1): 115-22, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23505175

RESUMEN

OBJECTIVE: Visceral obesity contributes to the development of obesity-related disorders such as diabetes, hyperlipidemia, and fatty liver disease, as well as cardiovascular diseases. In this study, we determined whether topical application of capsaicin can reduce fat accumulation in visceral adipose tissues. METHODS AND RESULTS: We first observed that topical application of 0.075% capsaicin to male mice fed a high-fat diet significantly reduced weight gain and visceral fat. Fat cells were markedly smaller in the mesenteric and epididymal adipose tissues of mice treated with capsaicin cream. The capsaicin treatment also lowered serum levels of fasting glucose, total cholesterol, and triglycerides. Immunoblot analysis and RT-PCR revealed increased expression of adiponectin and other adipokines including peroxisome proliferator-activated receptor (PPAR) α, PPARγ, visfatin, and adipsin, but reduced expression of tumor necrosis factor-α and IL-6. CONCLUSIONS: These results indicate that topical application of capsaicin to obese mice limits fat accumulation in adipose tissues and may reduce inflammation and increase insulin sensitivity.


Asunto(s)
Adipoquinas/metabolismo , Capsaicina/uso terapéutico , Dieta Alta en Grasa/efectos adversos , Grasa Intraabdominal/efectos de los fármacos , Obesidad/tratamiento farmacológico , Fitoterapia , Extractos Vegetales/uso terapéutico , Adipocitos/efectos de los fármacos , Adiponectina/metabolismo , Administración Tópica , Animales , Glucemia/metabolismo , Capsaicina/administración & dosificación , Capsaicina/farmacología , Capsicum/química , Colesterol/sangre , Factor D del Complemento/metabolismo , Epidídimo , Interleucina-6/metabolismo , Grasa Intraabdominal/citología , Grasa Intraabdominal/metabolismo , Masculino , Mesenterio , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Nicotinamida Fosforribosiltransferasa/metabolismo , Obesidad/etiología , Obesidad/metabolismo , PPAR alfa/metabolismo , PPAR gamma/metabolismo , Extractos Vegetales/farmacología , Triglicéridos/sangre , Factor de Necrosis Tumoral alfa/metabolismo , Aumento de Peso/efectos de los fármacos
14.
Mol Nutr Food Res ; 57(7): 1135-44, 2013 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-23495198

RESUMEN

SCOPE: Inflammation plays a key role in obesity-related pathologies such as insulin resistance and type 2 diabetes. Hypertrophied adipocytes trigger the enhancement of macrophage infiltration and the release of various proinflammatory factors in obese adipose tissue. In this study, we examined whether auraptene, a citrus-fruit-derived compound, could suppress the production of inflammatory factors that mediate the interaction between adipocytes and macrophages. METHODS AND RESULTS: Experiments using a co-culture system of 3T3-L1 adipocytes and RAW264 macrophages showed that auraptene reduced the production of nitric oxide and tumor necrosis factor-α. In RAW264 macrophages, auraptene also suppressed the inflammation induced by either LPS or the conditioned medium derived from 3T3-L1 adipocytes. In addition, auraptene inhibited the phosphorylation of the p38 mitogen-activated protein kinase and suppressed the production of proinflammatory mediators in activated macrophages. CONCLUSION: Our findings indicate that auraptene exhibits anti-inflammatory properties by suppressing the production of inflammatory factors that mediate the interaction between adipocytes and macrophages, suggesting that auraptene is a valuable food-derived compound with a potential to attenuate chronic inflammation in adipose tissue and to improve obesity-related insulin resistance.


Asunto(s)
Antiinflamatorios/farmacología , Cumarinas/farmacología , Macrófagos/efectos de los fármacos , Extractos Vegetales/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/genética , Células 3T3-L1 , Adipocitos/efectos de los fármacos , Adipocitos/metabolismo , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Animales , Línea Celular Tumoral , Quimiocina CCL2/metabolismo , Citrus/química , Técnicas de Cocultivo , Medios de Cultivo Condicionados , Frutas/química , Inflamación/tratamiento farmacológico , Inflamación/patología , Resistencia a la Insulina , Lipopolisacáridos/efectos adversos , Macrófagos/metabolismo , Ratones , Óxido Nítrico/antagonistas & inhibidores , Óxido Nítrico/metabolismo , Obesidad/metabolismo , Fosforilación , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/biosíntesis , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
15.
J Agric Food Chem ; 60(48): 11935-41, 2012 Dec 05.
Artículo en Inglés | MEDLINE | ID: mdl-23140571

RESUMEN

In this study, we investigated effects of pine nut oil (PNO) on high-fat-diet (HFD)-induced obesity and metabolic dysfunction in skeletal muscle and brown adipose tissue (BAT). Male C57BL/6 mice were fed a HFD with 15% energy from lard and 30% energy from either soybean oil (SBO-HFD) or PNO (PNO-HFD) for 12 weeks. The PNO-HFD resulted in less weight gain and intramuscular lipid accumulation than the SBO-HFD and was accompanied by upregulation of transcripts and proteins related to oxidative metabolism and phosphorylation of AMP-activated protein kinase (AMPK), as well as molecules selectively expressed in type I and type IIa muscle fibers. In addition, uncoupling protein-1 was upregulated in BAT. These beneficial metabolic effects were partly associated with the dual ligand activity of pinolenic acid, which is abundant in PNO, for peroxisome proliferator-activated receptors α and δ. Our findings suggest that PNO may have potential as a dietary supplement for counteracting obesity and metabolic dysregulation.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Mitocondrias/metabolismo , Músculo Esquelético/metabolismo , Obesidad/metabolismo , Pinus/química , Aceites de Plantas/farmacología , Proteínas Quinasas Activadas por AMP/metabolismo , Tejido Adiposo Pardo/efectos de los fármacos , Animales , Dieta , Dieta Alta en Grasa/efectos adversos , Grasas de la Dieta/farmacología , Canales Iónicos/metabolismo , Ácidos Linolénicos/farmacología , Masculino , Ratones , Ratones Endogámicos C57BL , Mitocondrias/efectos de los fármacos , Proteínas Mitocondriales/metabolismo , Músculo Esquelético/efectos de los fármacos , Nueces/química , Obesidad/inducido químicamente , Obesidad/dietoterapia , PPAR alfa/metabolismo , PPAR delta/metabolismo , Fosforilación/efectos de los fármacos , Aceite de Soja/farmacología , Termogénesis/efectos de los fármacos , Proteína Desacopladora 1 , Aumento de Peso/efectos de los fármacos
16.
J Nutr Biochem ; 23(7): 768-76, 2012 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21889885

RESUMEN

Tiliroside contained in several dietary plants, such as rose hips, strawberry and raspberry, is a glycosidic flavonoid and possesses anti-inflammatory, antioxidant, anticarcinogenic and hepatoprotective activities. Recently, it has been reported that the administration of tiliroside significantly inhibited body weight gain and visceral fat accumulation in normal mice. In this study, we evaluated the effects of tiliroside on obesity-induced metabolic disorders in obese-diabetic KK-A(y) mice. In KK-A(y) mice, the administration of tiliroside (100 mg/kg body weight/day) for 21 days failed to suppress body weight gain and visceral fat accumulation. Although tiliroside did not affect oxygen consumption, respiratory exchange ratio was significantly decreased in mice treated with tiliroside. In the analysis of metabolic characteristics, it was shown that plasma insulin, free fatty acid and triglyceride levels were decreased, and plasma adiponectin levels were increased in mice administered tiliroside. The messenger RNA expression levels of hepatic adiponectin receptor (AdipoR)-1 and AdipoR2 and skeletal muscular AdipoR1 were up-regulated by tiliroside treatment. Furthermore, it was indicated that tiliroside treatment activated AMP-activated protein kinase in both the liver and skeletal muscle and peroxisome proliferator-activated receptor α in the liver. Finally, tiliroside inhibited obesity-induced hepatic and muscular triglyceride accumulation. These findings suggest that tiliroside enhances fatty acid oxidation via the enhancement adiponectin signaling associated with the activation of both AMP-activated protein kinase and peroxisome proliferator-activated receptor α and ameliorates obesity-induced metabolic disorders, such as hyperinsulinemia and hyperlipidemia, although it does not suppress body weight gain and visceral fat accumulation in obese-diabetic model mice.


Asunto(s)
Adiponectina/sangre , Flavonoides/farmacología , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/efectos de los fármacos , Músculo Esquelético/efectos de los fármacos , Extractos Vegetales/farmacología , Proteínas Quinasas Activadas por AMP/genética , Proteínas Quinasas Activadas por AMP/metabolismo , Adiponectina/genética , Animales , Ácidos Grasos/sangre , Femenino , Insulina/sangre , Grasa Intraabdominal/metabolismo , Hígado/metabolismo , Enfermedades Metabólicas/tratamiento farmacológico , Enfermedades Metabólicas/etiología , Enfermedades Metabólicas/fisiopatología , Ratones , Ratones Obesos , Músculo Esquelético/metabolismo , Obesidad/complicaciones , Obesidad/tratamiento farmacológico , Obesidad/fisiopatología , PPAR alfa/genética , PPAR alfa/metabolismo , Receptores de Adiponectina/genética , Receptores de Adiponectina/metabolismo , Transducción de Señal , Triglicéridos/sangre , Regulación hacia Arriba , Aumento de Peso/efectos de los fármacos
17.
J Med Food ; 14(3): 310-5, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21332406

RESUMEN

Metabolic dysregulation (e.g., hyperglycemia, hyperinsulinemia, hyperlipidemia, etc.) is a hallmark of obesity-related diseases such as insulin resistance, type 2 diabetes, and fatty liver disease. In this study, we assessed whether dietary capsaicin attenuated the metabolic dysregulation in genetically obese diabetic KKAy mice, which have severe diabetic phenotypes. Male KKAy mice fed a high-fat diet for 2 weeks received a 0.015% capsaicin supplement for a further 3 weeks and were compared with nonsupplemented controls. Dietary capsaicin markedly decreased fasting glucose/insulin and triglyceride levels in the plasma and/or liver, as well as expression of inflammatory adipocytokine genes (e.g., monocyte chemoattractant protein-1 and interleukin-6) and macrophage infiltration. At the same time expression of the adiponectin gene/protein and its receptor, AdipoR2, increased in adipose tissue and/or plasma, accompanied by increased activation of hepatic AMP-activated protein kinase, a marker of fatty acid oxidation. These findings suggest that dietary capsaicin reduces metabolic dysregulation in obese/diabetic KKAy mice by enhancing expression of adiponectin and its receptor. Capsaicin may be useful as a dietary factor for reducing obesity-related metabolic dysregulation.


Asunto(s)
Adiponectina/metabolismo , Capsaicina/uso terapéutico , Capsicum/química , Diabetes Mellitus Experimental/tratamiento farmacológico , Obesidad/tratamiento farmacológico , Fitoterapia , Extractos Vegetales/uso terapéutico , Proteínas Quinasas Activadas por AMP/metabolismo , Adipoquinas/metabolismo , Adiponectina/genética , Tejido Adiposo/metabolismo , Animales , Glucemia/metabolismo , Capsaicina/farmacología , Diabetes Mellitus Experimental/metabolismo , Suplementos Dietéticos , Insulina/sangre , Peroxidación de Lípido/efectos de los fármacos , Hígado/metabolismo , Macrófagos/efectos de los fármacos , Masculino , Ratones , Ratones Obesos , Obesidad/metabolismo , Extractos Vegetales/farmacología , Receptores de Adiponectina/metabolismo , Triglicéridos/metabolismo
18.
Mediators Inflamm ; 2010: 367838, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20508825

RESUMEN

Obesity is associated with a low-grade systemic chronic inflammatory state, characterized by the abnormal production of pro- and anti-inflammatory adipocytokines. It has been found that immune cells such as macrophages can infiltrate adipose tissue and are responsible for the majority of inflammatory cytokine production. Obesity-induced inflammation is considered a potential mechanism linking obesity to its related pathologies, such as insulin resistance, cardiovascular diseases, type-2 diabetes, and some immune disorders. Therefore, targeting obesity-related inflammatory components may be a useful strategy to prevent or ameliorate the development of such obesity-related diseases. It has been shown that several food components can modulate inflammatory responses in adipose tissue via various mechanisms, some of which are dependent on peroxisome proliferator-activated receptor gamma (PPARgamma), whereas others are independent on PPARgamma, by attenuating signals of nuclear factor-kappaB (NF-kappaB) and/or c-Jun amino-terminal kinase (JNK). In this review, we introduce the beneficial effects of anti-inflammatory phytochemicals that can help prevent obesity-induced inflammatory responses and pathologies.


Asunto(s)
Alimentos Funcionales , Inflamación , Obesidad , Adipoquinas/inmunología , Tejido Adiposo/inmunología , Animales , Humanos , Inflamación/dietoterapia , Inflamación/etiología , Inflamación/inmunología , Resistencia a la Insulina , Obesidad/complicaciones , Obesidad/inmunología , Obesidad/patología , PPAR gamma/metabolismo , Extractos Vegetales/metabolismo , Transducción de Señal
19.
Obesity (Silver Spring) ; 18(4): 780-7, 2010 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19798065

RESUMEN

Obesity-induced inflammation contributes to the development of obesity-related metabolic disorders such as insulin resistance, type 2 diabetes, fatty liver disease, and cardiovascular disease. In this study, we investigated whether dietary capsaicin can reduce obesity-induced inflammation and metabolic disorders such as insulin resistance and hepatic steatosis. Male C57BL/6 obese mice fed a high-fat diet for 10 weeks received a supplement of 0.015% capsaicin for a further 10 weeks and were compared with unsupplemented controls. Glucose intolerance was estimated by glucose tolerance tests. Transcripts of adipocytokine genes and the corresponding proteins were measured by reverse transcription-PCR and enzyme-linked immunosorbent assay, and macrophage numbers were determined by flow cytometric analysis. Transient receptor potential vanilloid type-1 (TRPV-1), peroxisome proliferator-activated receptor (PPAR)-alpha, and PPARgamma coactivator-1alpha (PGC-1alpha) mRNAs were also measured by RT-PCR, and PPARalpha luciferase assays were performed. Dietary capsaicin lowered fasting glucose, insulin, leptin levels, and markedly reduced the impairment of glucose tolerance in obese mice. Levels of tumor necrosis factor-alpha (TNFalpha), monocyte chemoattractant protein-1 (MCP-1), and interleukin (IL)-6 mRNAs and proteins in adipose tissue and liver decreased markedly, as did macrophage infiltration, hepatic triglycerides, and TRPV-1 expression in adipose tissue. At the same time, the mRNA/protein of adiponectin in the adipose tissue and PPARalpha/PGC-1alpha mRNA in the liver increased. Moreover, luciferase assays revealed that capsaicin is capable of binding PPARalpha. Our data suggest that dietary capsaicin may reduce obesity-induced glucose intolerance by not only suppressing inflammatory responses but also enhancing fatty acid oxidation in adipose tissue and/or liver, both of which are important peripheral tissues affecting insulin resistance. The effects of capsaicin in adipose tissue and liver are related to its dual action on PPARalpha and TRPV-1 expression/activation.


Asunto(s)
Antiinflamatorios/uso terapéutico , Capsaicina/uso terapéutico , Hígado Graso/tratamiento farmacológico , Hipoglucemiantes/uso terapéutico , Resistencia a la Insulina , Obesidad/complicaciones , Adiponectina/genética , Adiponectina/metabolismo , Tejido Adiposo/metabolismo , Animales , Antiinflamatorios/farmacología , Glucemia/metabolismo , Capsaicina/farmacología , Grasas de la Dieta/administración & dosificación , Grasas de la Dieta/metabolismo , Suplementos Dietéticos , Modelos Animales de Enfermedad , Hígado Graso/etiología , Hígado Graso/metabolismo , Citometría de Flujo , Expresión Génica , Intolerancia a la Glucosa/etiología , Intolerancia a la Glucosa/metabolismo , Hipoglucemiantes/farmacología , Inflamación/genética , Insulina/sangre , Leptina/sangre , Metabolismo de los Lípidos/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/tratamiento farmacológico , Obesidad/metabolismo , PPAR alfa/química , PPAR alfa/genética , PPAR alfa/metabolismo , ARN Mensajero/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Canales Catiónicos TRPV/genética , Canales Catiónicos TRPV/metabolismo , Triglicéridos/metabolismo
20.
J Med Food ; 12(4): 770-5, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19735175

RESUMEN

Rheumatoid arthritis (RA) is characterized by chronic inflammation of the synovial joints. This study investigated whether or not a diet deficient in vitamin E is a possible risk factor in the development of RA and evaluated the efficacy of antioxidant supplementation. Male DBA/1J mice were maintained on either a control diet (C) or a vitamin E-depleted (-VE) diet for 4 weeks before arthritis induction. The mice in the control group were subdivided into the control group (C/C), the 0.05% alpha-tocopherol-supplemented group (C/+VE), and the 0.5% quercetin-supplemented group (C/+Q). The vitamin E-depleted group was subdivided into the -VE group (-VE/-VE), the 0.05% alpha-tocopherol-supplemented group (-VE/+VE), and the 0.5% quercetin-supplemented group (-VE/+Q) (in total, six groups, 27 mice per group). The mice were maintained on the experimental diets for 9 weeks. Study results indicate that the -VE/-VE group showed higher joint tissue tumor necrosis factor-alpha and interleukin-1beta mRNA expressions, whereas alpha-tocopherol or quercetin supplementation reduced tissue cytokine mRNA levels to values comparable to those of the C/C group. The mice fed the -VE/-VE diet exhibited higher levels of circulating macrophage chemoattractant protein 1, nitric oxide, and prostaglandin E(2) compared to those in other groups. Supplementation with alpha-tocopherol or quercetin in mice fed -VE diet decreased these markers similar to those of the mice in the C/C group. No supplementation effect was observed, however, in the mice fed with the control diet prior to RA induction. These results suggest that dietary deficiency of vitamin E increases inflammatory responses and that antioxidants successfully suppress the inflammatory responses. However, significant clinical improvement may require longer observation.


Asunto(s)
Antiinflamatorios/uso terapéutico , Antioxidantes/uso terapéutico , Artritis Experimental/tratamiento farmacológico , Mediadores de Inflamación/antagonistas & inhibidores , Quercetina/uso terapéutico , Deficiencia de Vitamina E/complicaciones , alfa-Tocoferol/uso terapéutico , Animales , Antiinflamatorios/farmacología , Antioxidantes/farmacología , Artritis Experimental/complicaciones , Artritis Experimental/metabolismo , Biomarcadores/metabolismo , Colágeno , Citocinas/genética , Citocinas/metabolismo , Dieta , Modelos Animales de Enfermedad , Masculino , Ratones , Ratones Endogámicos DBA , Quercetina/administración & dosificación , Quercetina/farmacología , ARN Mensajero/metabolismo , Factores de Riesgo , alfa-Tocoferol/administración & dosificación , alfa-Tocoferol/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA