Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Cancer Res ; 84(4): 545-559, 2024 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-38117489

RESUMEN

Adipose tissue within the tumor microenvironment (TME) plays a critical role in supporting breast cancer progression. In this study, we identified FAM3 metabolism-regulating signaling molecule C (FAM3C) produced by cancer-associated adipocytes (CAA) as a key regulator of tumor progression. FAM3C overexpression in cultured adipocytes significantly reduced cell death in both adipocytes and cocultured breast cancer cells while suppressing markers of fibrosis. Conversely, FAM3C depletion in CAAs resulted in adipocyte-mesenchymal transition (AMT) and increased fibrosis within the TME. Adipocyte FAM3C expression was driven by TGFß signaling from breast cancer cells and was reduced upon treatment with a TGFß-neutralizing antibody. FAM3C knockdown in CAAs early in tumorigenesis in a genetically engineered mouse model of breast cancer significantly inhibited primary and metastatic tumor growth. Circulating FAM3C levels were elevated in patients with metastatic breast cancer compared with those with nonmetastatic breast cancer. These results suggest that therapeutic inhibition of FAM3C expression levels in CAAs during early tumor development could be a promising approach in the treatment of patients with breast cancer. SIGNIFICANCE: High FAM3C levels in cancer-associated adipocytes contribute to tumor-supportive niches and are tightly associated with metastatic growth, indicating that FAM3C inhibition could be beneficial for treating patients with breast cancer.


Asunto(s)
Neoplasias de la Mama , Citocinas , Proteínas de Neoplasias , Animales , Femenino , Humanos , Ratones , Adipocitos/metabolismo , Neoplasias de la Mama/patología , Supervivencia Celular , Citocinas/metabolismo , Fibrosis , Proteínas de Neoplasias/metabolismo , Factor de Crecimiento Transformador beta/metabolismo , Microambiente Tumoral
2.
Nat Commun ; 14(1): 439, 2023 01 27.
Artículo en Inglés | MEDLINE | ID: mdl-36707514

RESUMEN

Hyperglycemia is a risk factor for breast cancer-related morbidity and mortality. Hyperglycemia induces Neuregulin 1 (Nrg1) overexpression in breast cancer, which subsequently promotes tumor progression. However, molecular mechanisms underlying hyperglycemia-induced Nrg1 overexpression remain poorly understood. Here, we show that hyperglycemia causes active histone modifications at the Nrg1 enhancer, forming enhanceosome complexes where recombination signal binding protein for immunoglobulin kappa J region (RBPJ), E1A binding protein p300 (P300), and SET domain containing 1 A (SETD1A) are recruited to upregulate Nrg1 expression. Deletions in RBPJ-binding sites causes hyperglycemia-controlled Nrg1 levels to be downregulated, resulting in decreased tumor growth in vitro and in vivo. Mice with modest-temporary hyperglycemia, induced by low-dose short-exposure streptozotocin, display accelerated tumor growth and lapatinib resistance, whereas combining lapatinib with N-[N-(3,5-difluorophenacetyl)-l-alanyl]-S42 phenylglycine t-butyl ester (DAPT) ameliorates tumor growth under these modest hyperglycemic conditions by inhibiting NOTCH and EGFR superfamilies. NOTCH activity is correlated with NRG1 levels, and high NRG1 levels predicts poor outcomes, particularly in HER2-positive breast cancer patients. Our findings highlight the hyperglycemia-linked epigenetic modulation of NRG1 as a potential therapeutic strategy for treating breast cancer patients with diabetes.


Asunto(s)
Hiperglucemia , Neoplasias , Animales , Ratones , Lapatinib , Epigénesis Genética , Neurregulina-1/genética , Neurregulina-1/metabolismo , Línea Celular Tumoral , Hiperglucemia/genética
3.
Diabetes ; 71(8): 1746-1762, 2022 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-35167651

RESUMEN

Dysregulation of extracellular matrix proteins in obese adipose tissue (AT) induces systemic insulin resistance. The metabolic roles of type VI collagen and its cleavage peptide endotrophin in obese AT are well established. However, the mechanisms regulating endotrophin generation remain elusive. Herein, we identified that several endotrophin-containing peptides (pre-endotrophins) were generated from the COL6A3 chain in a stepwise manner for the efficient production of mature endotrophin, partly through the action of hypoxia-induced matrix metalloproteinases (MMPs), including MMP2, MMP9, and MMP16. Hypoxia is an upstream regulator of COL6A3 expression and the proteolytic processing that regulates endotrophin generation. Hypoxia-inducible factor 1α (HIF1α) and the hypoxia-associated suppression of microRNA-29 (miR-29) cooperatively control the levels of COL6A3 and MMPs, which are responsible for endotrophin generation in hypoxic ATs. Adipocyte-specific Hif1α knock-out (APN-HIF1αKO) mice fed a chronic high-fat diet exhibited the significant amelioration of both local fibro-inflammation in AT and systemic insulin resistance compared with their control littermates, partly through the inhibition of endotrophin generation. Strikingly, adenovirus-mediated miR-29 overexpression in the ATs of APN-HIF1αKO mice in obesity significantly decreased endotrophin levels, suggesting that miR-29, combined with HIF1α inhibition in AT, could be a promising therapeutic strategy for treating obesity and related metabolic diseases.


Asunto(s)
Subunidad alfa del Factor 1 Inducible por Hipoxia , Resistencia a la Insulina , MicroARNs , Tejido Adiposo/metabolismo , Animales , Colágeno Tipo VI/metabolismo , Hipoxia/genética , Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/deficiencia , Inflamación/genética , Inflamación/metabolismo , Resistencia a la Insulina/genética , Ratones , MicroARNs/genética , MicroARNs/metabolismo , Obesidad/genética , Obesidad/metabolismo
4.
Metabolism ; 114: 154430, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-33188786

RESUMEN

OBJECTIVE: Obesity-induced adipose tissue remodeling is closely associated with systemic insulin resistance. However, the mechanistic involvement of adipocyte-derived extracellular matrix proteins under pathophysiological conditions remains unclear. Our aim was to investigate the distinctive contributions of each chain of type VI collagens (Col6) and its cleavage protein endotrophin to adipocyte functions and insulin sensitivity. METHODS: Col6 comprises three alpha chains: Col6a1, Col6a2, and Col6a3. We generated Col6a1-, Col6a2-, and Col6a3-deficient 3T3-L1 adipocytes using the CRISPR-Cas9 system as well as a novel Col6a3-deficient (Col6a3KO) mouse model for loss-of-function studies. Adenoviral-endotrophin and adipocyte-specific doxycycline-inducible endotrophin transgenic mice were utilized for the gain-of-function analysis. RESULTS: The holo-Col6 fibrils were found to be required for mature adipocyte differentiation. Only Col6a3-deficient 3T3-L1 adipocytes showed decreased inflammation and basal adipocyte lipolysis and prevented ER-stress-induced insulin resistance. Consistently, Col6a3KO mice showed decreased adipocyte size and fat mass of epididymal adipose tissues due to a defect in adipogenic and lipolytic capacity of adipocytes. Beyond the structural role of Col6a3, overexpression of endotrophin in obese mice further augmented insulin resistance, which was tightly associated with a significant increase in lipolysis, inflammation, and cellular apoptosis in adipose tissues, whereas this showed a limited effect on adipogenesis. CONCLUSIONS: These novel findings corroborate our previous observations suggesting that adipose tissue extracellular matrix regulates adipocyte function and insulin sensitivity in pathophysiological conditions. Mechanistically, holo-Col6 fibrils and their signaling derivative endotrophin govern adipocyte function independently of their role as structural supports via MAPK signaling pathways, and the latter could be an important metabolic effector in obesity-related metabolic diseases.


Asunto(s)
Adipocitos/metabolismo , Adipogénesis/fisiología , Colágeno Tipo VI/metabolismo , Lipólisis/fisiología , Fragmentos de Péptidos/metabolismo , Células 3T3-L1 , Tejido Adiposo/metabolismo , Animales , Colágeno Tipo VI/genética , Interleucina-6/metabolismo , Ratones , Ratones Transgénicos , Fragmentos de Péptidos/genética
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.
J Inflamm (Lond) ; 14: 9, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28503098

RESUMEN

Obesity-induced inflammation causes skeletal muscle atrophy accompanied by disruption of oxidative metabolism and is implicated in metabolic complications such as insulin resistance and type 2 diabetes. We previously reported that 4-1BB, a member of the tumor necrosis factor receptor superfamily, participated in obesity-induced skeletal muscle inflammation. Here, we show that the absence of 4-1BB in obese mice fed a high-fat diet led to a decrease in expression of atrophic factors (MuRF1 and Atrogin-1) with suppression of NF-κB activity, and that this was accompanied by increases in mitochondrial oxidative metabolic genes/proteins (e.g., PGC-1α, CPT1ß, etc.) expression and oxidative muscle fibers marker genes/proteins in the skeletal muscle. These findings suggest that 4-1BB-mediated inflammatory signaling could be a potential target for combating obesity-related muscle atrophy and metabolic derangement in skeletal muscle.

7.
Mol Nutr Food Res ; 61(10)2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-28544341

RESUMEN

SCOPE: Recent reports indicate that gut microbiota and their metabolites may regulate host inflammatory conditions, including the chronic inflammation of obese adipose tissues. In this study, we investigated whether specific synthesized fatty acids, identical to the metabolites generated by gut microbiota, act as anti-inflammatory factors in obesity-induced inflammation. METHODS AND RESULTS: We first used lipopolysaccharide (LPS)-stimulated RAW 264.7 macrophages to examine the anti-inflammatory effect of fatty acids synthesized to resemble representative polyunsaturated fatty acid metabolites from gut microbiota. Fatty acids containing an enone structure showed the most potent anti-inflammatory activity. Enone fatty acids also displayed anti-inflammatory effects on macrophages cocultured with hypertrophied 3T3-L1 or immortalized primary adipocytes; and macrophages stimulated with 3T3-L1 adipocyte conditioned medium. Consistently, the beneficial outcome was revealed in the case of LPS- and obesity-induced inflammatory cytokine stimulation in ex vivo adipose tissues. Furthermore, these fatty acids recovered the suppression of ß-adrenergic receptor-stimulated uncoupling protein 1 expression and secretion of adiponectin in C3H10T1/2 and 3T3-L1 adipocytes, respectively, under inflammatory conditions, suggesting that enone fatty acids can ameliorate dysfunctions of adipocytes induced by inflammation. CONCLUSION: These findings indicate that synthesized enone fatty acids show potent anti-inflammatory effects, leading to the improvement of inflammation-induced dysfunctions in adipocytes.


Asunto(s)
Adipocitos/citología , Antiinflamatorios/farmacología , Ácidos Grasos Insaturados/farmacología , Microbioma Gastrointestinal , Inflamación/terapia , Macrófagos/citología , Células 3T3-L1 , Adiponectina/metabolismo , Animales , Quimiocina CCL2/metabolismo , Técnicas de Cocultivo , Relación Dosis-Respuesta a Droga , Regulación de la Expresión Génica , Lactobacillus/metabolismo , Ratones , Óxido Nítrico/metabolismo , Obesidad/terapia , Células RAW 264.7 , Factor de Necrosis Tumoral alfa/metabolismo
8.
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
9.
Nutr Metab (Lond) ; 12: 33, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26445592

RESUMEN

BACKGROUND: Obesity-induced hepatic lipid accumulation causes lipotoxicity, mitochondrial dysfunction, oxidative stress, and insulin resistance, and is implicated in non-alcoholic hepatic pathologies such as steatohepatitis and fibrosis. Heme oxygenase-1 (HO-1), an important antioxidant enzyme catalyzing the rate-limiting step in heme degradation, protects against oxidative stress, inflammation, and metabolic dysregulation. Here, we demonstrate that the phytochemical, quercetin, a natural polyphenol flavonoid, protects against hepatic steatosis in obese mice fed a high-fat diet, and that it does so by inducing HO-1 and stimulating increased hepatic mitochondrial oxidative metabolism. METHODS: Male C57BL/6 mice were fed a regular diet (RD), a high-fat diet (HFD), and an HFD supplemented with quercetin for 9 weeks. Levels of mitochondrial biogenesis and oxidative metabolic transcripts/proteins were measured by real-time PCR and/or Western blotting. HO-1 transcripts/proteins were measured real-time PCR and/or Western blotting. RESULTS: Quercetin upregulated genes involved in mitochondrial biogenesis and oxidative metabolism in lipid-laden hepatocytes and the livers of HFD-fed obese mice, and this was accompanied by increased levels of the transcription factor, nuclear erythroid 2-related factor 2 (Nrf-2), and HO-1 protein. The HO-1 inducer hemin and the HO-1 byproduct carbon monoxide (CO) also enhanced hepatic oxidative metabolism in HFD-fed obese mice. Moreover, the metabolic changes and the lipid-lowering effects of quercetin were completely blocked by the HO-1 inhibitor ZnPP and by deficiency of Nrf-2. CONCLUSION: These findings suggest that quercetin stimulates hepatic mitochondrial oxidative metabolism by inducing HO-1 via the Nrf-2 pathway. Quercetin may be useful in protecting against obesity-induced hepatosteatosis.

10.
FEBS J ; 282(8): 1468-80, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25691217

RESUMEN

Obesity-induced monocyte/macrophage proliferation and activation play a crucial role in various chronic inflammatory metabolic disorders, such as insulin resistance, diabetes mellitus, and atherosclerosis. 4-1BBL, a member of the tumor necrosis factor superfamily expressed on monocytes/macrophages, provides inflammatory signals to modulate their proliferation, survival, and cytokine release. Previously, we demonstrated that 4-1BBL signaling promotes adipose inflammation through enhancement of macrophage activation. Here, we show that 4-1BBL stimulation on monocytes/macrophages enhanced reprogramming of glucose metabolism in the cells, and that this was accompanied by cell proliferation. 4-1BBL stimulation on macrophages increased glucose uptake, transcript/protein levels of glucose transporter 1 and glycolytic enzymes, and lactate production. It also enhanced transcript levels of genes involved in the pentose phosphate pathway and lipogenesis. The 4-1BBL-induced metabolic reprogramming was mediated by AKT-mammalian target of rapamycin signaling. The effect of 4-1BBL-induced macrophage proliferation was completely abolished by 2-deoxyglucose, a glycolytic inhibitor. These findings suggest that 4-1BBL signaling promotes cell proliferation through reprogramming of glucose metabolism in monocytes/macrophages to support their energy demands and biomass production. The 4-1BBL signaling pathway may be a valid target for controlling macrophage-mediated chronic inflammation in obesity and metabolic diseases.


Asunto(s)
Ligando 4-1BB/metabolismo , Proliferación Celular , Glucosa/metabolismo , Inflamación/metabolismo , Macrófagos/metabolismo , Monocitos/metabolismo , Obesidad/metabolismo , Ligando 4-1BB/genética , Tejido Adiposo/citología , Tejido Adiposo/metabolismo , Animales , Apoptosis , Western Blotting , Células Cultivadas , Citocinas/metabolismo , Femenino , Citometría de Flujo , Humanos , Inflamación/genética , Inflamación/patología , Resistencia a la Insulina , Macrófagos/citología , Masculino , Ratones , Monocitos/citología , Obesidad/genética , Obesidad/patología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal
11.
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
12.
Am J Physiol Endocrinol Metab ; 306(3): E247-55, 2014 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-24326418

RESUMEN

Recently, fibrosis is observed in obese adipose tissue; however, the pathogenesis remains to be clarified. Obese adipose tissue is characterized by chronic inflammation with massive accumulation of immune cells including mast cells. The objective of the present study was to clarify the relationship between fibrosis and mast cells in obese adipose tissue, as well as to determine the origin of infiltrating mast cells. We observed the enhancement of mast cell accumulation and fibrosis in adipose tissue of severely obese diabetic db/db mice. Furthermore, adipose tissue-conditioned medium (ATCM) from severely obese diabetic db/db mice significantly enhanced collagen 5 mRNA expression in NIH-3T3 fibroblasts, and this enhancement was suppressed by the addition of an anti-mast cell protease 6 (MCP-6) antibody. An in vitro study showed that only collagen V among various types of collagen inhibited preadipocyte differentiation. Moreover, we found that ATCM from the nonobese but not obese stages of db/db mice significantly enhanced the migration of bone marrow-derived mast cells (BMMCs). These findings suggest that immature mast cells that infiltrate into adipose tissue at the nonobese stage gradually mature with the progression of obesity and diabetes and that MCP-6 secreted from mature mast cells induces collagen V expression in obese adipose tissue, which may contribute to the process of adipose tissue fibrosis. Induction of collagen V by MCP-6 might accelerate insulin resistance via the suppression of preadipocyte differentiation.


Asunto(s)
Tejido Adiposo/patología , Mastocitos/fisiología , Células 3T3-L1 , Tejido Adiposo/inmunología , Animales , Células de la Médula Ósea/fisiología , Movimiento Celular , Células Cultivadas , Diabetes Mellitus Experimental/inmunología , Diabetes Mellitus Experimental/patología , Fibrosis , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Células 3T3 NIH
13.
Mediators Inflamm ; 2014: 834294, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25614714

RESUMEN

Skeletal muscle inflammation and atrophy are closely associated with metabolic impairment such as insulin resistance. Quercetin, a natural polyphenol flavonoid, is known to elicit anti-inflammatory and antioxidant activities. In this study, we investigated its effect on obesity-induced skeletal muscle inflammation and atrophy in mice. Male C57BL/6 mice were fed a regular diet, a high-fat diet (HFD), and an HFD supplemented with quercetin for nine weeks. Quercetin reduced levels of inflammatory cytokines and macrophage accumulation in the skeletal muscle of the HFD-fed obese mice. It also reduced transcript and protein levels of the specific atrophic factors, Atrogin-1 and MuRF1, in the skeletal muscle of the HFD-fed obese mice, and protected against the reduction of muscle mass and muscle fiber size. In vitro, quercetin markedly diminished transcript levels of inflammatory receptors and activation of their signaling molecules (ERK, p38 MAPK, and NF-κB) in cocultured myotubes/macrophages, and this was accompanied by reduced expression of the atrophic factors. Together, these findings suggest that quercetin reduces obesity-induced skeletal muscle atrophy by inhibiting inflammatory receptors and their signaling pathway. Quercetin may be useful for preventing obesity-induced muscle inflammation and sarcopenia.


Asunto(s)
Antioxidantes/química , Atrofia/patología , Inflamación/patología , Músculo Esquelético/patología , Obesidad/complicaciones , Quercetina/química , Animales , Secuencia de Bases , Línea Celular , Citocinas/metabolismo , Inflamación/metabolismo , Resistencia a la Insulina , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Musculares/metabolismo , Músculo Esquelético/metabolismo , Proteínas Ligasas SKP Cullina F-box/metabolismo , Sarcopenia/metabolismo , Transducción de Señal , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas/metabolismo
14.
Mediators Inflamm ; 2013: 865159, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-24453430

RESUMEN

Obesity-induced skeletal muscle inflammation is characterized by increased macrophage infiltration and inflammatory cytokine production. In this study, we investigated whether 4-1BB, a member of the TNF receptor superfamily (TNFRSF9) that provides inflammatory signals, participates in obesity-induced skeletal muscle inflammation. Expression of the 4-1BB gene, accompanied by increased levels of inflammatory cytokines, was markedly upregulated in the skeletal muscle of obese mice fed a high-fat diet, in muscle cells treated with obesity factors, and in cocultured muscle cells/macrophages. In vitro stimulation of 4-1BB with agonistic antibody increased inflammatory cytokine levels in TNFα-pretreated muscle cells, and this effect was absent in cells derived from 4-1BB-deficient mice. Conversely, disruption of the interaction between 4-1BB and its ligand (4-1BBL) with blocking antibody decreased the release of inflammatory cytokines from cocultured muscle cells/macrophages. Moreover, deficiency of 4-1BB markedly reduced macrophage infiltration and inflammatory cytokine production in the skeletal muscle of mice fed a high-fat diet. These findings indicate that 4-1BB mediates the inflammatory responses in obese skeletal muscle by interacting with its ligand 4-1BBL on macrophages. Therefore, 4-1BB and 4-1BBL may be useful targets for prevention of obesity-induced inflammation in skeletal muscle.


Asunto(s)
Ligando 4-1BB/fisiología , Inflamación/etiología , Músculo Esquelético/patología , Obesidad/complicaciones , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/fisiología , Ligando 4-1BB/antagonistas & inhibidores , Ligando 4-1BB/genética , Animales , Células Cultivadas , Inflamación/prevención & control , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/análisis , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/antagonistas & inhibidores , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/genética
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.
Mediators Inflamm ; 2012: 972629, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23316108

RESUMEN

Obesity-induced adipose inflammation is characterized by recruitment of macrophages to adipose tissue and release of inflammatory cytokines. 4-1BB, a costimulatory receptor, modulates inflammatory processes through interaction with its ligand 4-1BBL on immune cell surfaces. In this study, we examined whether a 4-1BB/4-1BBL interaction between adipocytes and macrophages participates in obesity-induced adipose inflammation. We found that 4-1BB was expressed on adipocytes and was upregulated by obesity-related factors, which also enhanced 4-1BBL expression on macrophages. 4-1BB and/or 4-1BBL agonists, respectively, activated inflammatory signaling molecules (MAPK/IκBα and MAPK/Akt) in adipocytes and macrophages and enhanced the release of inflammatory cytokines (MCP-1, TNF-α, and IL-6). Moreover, disruption of the 4-1BB/4-1BBL interaction decreased the release of inflammatory cytokines from contact cocultured adipocytes/macrophages. These findings indicate that 4-1BB/4-1BBL-mediated bidirectional signaling in adipocytes/macrophages promotes adipose inflammation. 4-1BB and 4-1BBL may be useful targets for protection against obesity-induced adipose inflammation.


Asunto(s)
Ligando 4-1BB/fisiología , Adipocitos/fisiología , Comunicación Celular , Inflamación/etiología , Macrófagos/fisiología , Obesidad/complicaciones , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/fisiología , Animales , Células Cultivadas , Citocinas/biosíntesis , Masculino , Ratones , Ratones Endogámicos C57BL
17.
Diabetes ; 60(12): 3159-68, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21998397

RESUMEN

OBJECTIVE: Inflammation is an important factor in the development of insulin resistance, type 2 diabetes, and fatty liver disease. As a member of the tumor necrosis factor receptor superfamily (TNFRSF9) expressed on immune cells, 4-1BB/CD137 provides a bidirectional inflammatory signal through binding to its ligand 4-1BBL. Both 4-1BB and 4-1BBL have been shown to play an important role in the pathogenesis of various inflammatory diseases. RESEARCH DESIGN AND METHODS: Eight-week-old male 4-1BB-deficient and wild-type (WT) mice were fed a high-fat diet (HFD) or a regular diet for 9 weeks. RESULTS: We demonstrate that 4-1BB deficiency protects against HFD-induced obesity, glucose intolerance, and fatty liver disease. The 4-1BB-deficient mice fed an HFD showed less body weight gain, adiposity, adipose infiltration of macrophages/T cells, and tissue levels of inflammatory cytokines (e.g., TNF-α, interleukin-6, and monocyte chemoattractant protein-1 [MCP-1]) compared with HFD-fed control mice. HFD-induced glucose intolerance/insulin resistance and fatty liver were also markedly attenuated in the 4-1BB-deficient mice. CONCLUSIONS: These findings suggest that 4-1BB and 4-1BBL may be useful therapeutic targets for combating obesity-induced inflammation and metabolic disorders.


Asunto(s)
Dieta Alta en Grasa/efectos adversos , Inflamación/metabolismo , Obesidad/fisiopatología , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/deficiencia , Miembro 9 de la Superfamilia de Receptores de Factores de Necrosis Tumoral/metabolismo , Ligando 4-1BB/metabolismo , Tejido Adiposo/efectos de los fármacos , Tejido Adiposo/metabolismo , Adiposidad/genética , Adiposidad/fisiología , Animales , Western Blotting , Peso Corporal/genética , Peso Corporal/fisiología , Calorimetría Indirecta , Quimiocina CCL2/metabolismo , Citometría de Flujo , Intolerancia a la Glucosa , Inflamación/genética , Interleucina-6/metabolismo , Hígado/efectos de los fármacos , Hígado/metabolismo , Masculino , Ratones , FN-kappa B/metabolismo , Reacción en Cadena en Tiempo Real de la Polimerasa , Factor de Necrosis Tumoral alfa/metabolismo
18.
FEBS Lett ; 585(14): 2285-90, 2011 Jul 21.
Artículo en Inglés | MEDLINE | ID: mdl-21679708

RESUMEN

HVEM is a member of the TNF receptor superfamily that plays a role in the development of various inflammatory diseases. In this study, we show that HVEM deficiency attenuates adipose tissue inflammatory responses and glucose intolerance in diet-induced obesity. Feeding a high-fat diet (HFD) to HVEM-deficient mice elicited a reduction in the number of macrophages and T cells infiltrated into adipose tissue. Proinflammatory cytokine levels in the adipose tissue decreased in HFD-fed HVEM-deficient mice, while levels of the anti-inflammatory cytokine IL-10 increased. Moreover, glucose intolerance and insulin sensitivity were markedly improved in the HFD-fed HVEM-deficient mice. These findings indicate that HVEM may be a useful target for combating obesity-induced inflammatory responses and insulin resistance.


Asunto(s)
Tejido Adiposo/fisiopatología , Grasas de la Dieta/farmacología , Intolerancia a la Glucosa , Inflamación/fisiopatología , Miembro 14 de Receptores del Factor de Necrosis Tumoral/deficiencia , Adipocitos/citología , Adipocitos/metabolismo , Animales , Glucemia/metabolismo , Citocinas/metabolismo , Insulina/metabolismo , Resistencia a la Insulina/fisiología , Ratones , Obesidad/fisiopatología
19.
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
20.
Endocrinology ; 151(10): 4725-35, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20719857

RESUMEN

Immune cells (e.g. macrophages and T cells) in adipose tissue play a crucial role in the development of obesity-induced inflammation and metabolic disorders. Here we report findings suggesting that the immune signaling molecule 4-1BB/CD137 is a novel target for treatment of obesity and metabolic disorders. 4-1BB stimulation with agonistic antibody reduced body weight and adiposity and markedly improved glucose intolerance and hepatosteatosis in diet-induced obese mice and genetically obese/diabetic mice. Increases in lymphoid T cell expansion/activation and adipose/hepatic CD8+ T cell recruitment were evident in the anti-4-1BB antibody-treated obese mice. Glycolysis, ß-oxidation, and oxygen consumption rates also increased in the treated mice. These findings suggest that 4-1BB-stimulation accompanied by CD8+ T cell expansion/activation enhances glucose/lipid metabolism, leading to increased energy expenditure. Manipulation of 4-1BB may provide a unique immunological strategy against obesity and metabolic disorders.


Asunto(s)
Ligando 4-1BB/agonistas , Ligando 4-1BB/inmunología , Adiposidad/efectos de los fármacos , Anticuerpos/farmacología , Hígado Graso/prevención & control , Resistencia a la Insulina , Obesidad/terapia , Ligando 4-1BB/fisiología , Adiposidad/inmunología , Animales , Anticuerpos/uso terapéutico , Linfocitos T CD8-positivos/efectos de los fármacos , Linfocitos T CD8-positivos/inmunología , Hígado Graso/complicaciones , Intolerancia a la Glucosa/inmunología , Intolerancia a la Glucosa/prevención & control , Inmunoterapia , Resistencia a la Insulina/inmunología , Activación de Linfocitos/fisiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Obesos , Obesidad/complicaciones , Obesidad/inmunología , Obesidad/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA