Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 37
Filtrar
1.
Biochem Pharmacol ; 192: 114723, 2021 10.
Artículo en Inglés | MEDLINE | ID: mdl-34364887

RESUMEN

Both obesity and aging are associated with the development of metabolic diseases such as type 2 diabetes and cardiovascular disease. Chronic low-grade inflammation of adipose tissue is one of the mechanisms implicated in the progression of these diseases. Obesity and aging trigger adipose tissue alterations that ultimately lead to a pro-inflammatory phenotype of the adipose tissue-resident immune cells. Obesity and aging also share other features such as a higher visceral vs. subcutaneous adipose tissue ratio and a decreased lifespan. Here, we review the common characteristics of obesity and aging and the alterations in white adipose tissue and resident immune cells. We focus on the adipose tissue metabolic derangements in obesity and aging such as inflammation and adipose tissue remodeling.


Asunto(s)
Adipocitos Blancos/inmunología , Tejido Adiposo Blanco/inmunología , Envejecimiento/inmunología , Distribución de la Grasa Corporal/métodos , Obesidad/inmunología , Adipocitos Blancos/metabolismo , Adipocitos Blancos/patología , Tejido Adiposo Blanco/metabolismo , Tejido Adiposo Blanco/patología , Envejecimiento/metabolismo , Envejecimiento/patología , Animales , Humanos , Grasa Intraabdominal/inmunología , Grasa Intraabdominal/metabolismo , Grasa Intraabdominal/patología , Obesidad/metabolismo , Obesidad/patología
2.
Int J Mol Sci ; 22(4)2021 Feb 16.
Artículo en Inglés | MEDLINE | ID: mdl-33669239

RESUMEN

Irregular inflammatory responses are a major contributor to tissue dysfunction and inefficient repair. Skin has proven to be a powerful model to study mechanisms that regulate inflammation. In particular, skin wound healing is dependent on a rapid, robust immune response and subsequent dampening of inflammatory signaling. While injury-induced inflammation has historically been attributed to keratinocytes and immune cells, a vast body of evidence supports the ability of non-immune cells to coordinate inflammation in numerous tissues and diseases. In this review, we concentrate on the active participation of tissue-resident adipocytes and fibroblasts in pro-inflammatory signaling after injury, and how altered cellular communication from these cells can contribute to irregular inflammation associated with aberrant wound healing. Furthering our understanding of how tissue-resident mesenchymal cells contribute to inflammation will likely reveal new targets that can be manipulated to regulate inflammation and repair.


Asunto(s)
Adipocitos Blancos/inmunología , Dermis/citología , Dermis/lesiones , Fibroblastos/inmunología , Cicatrización de Heridas/inmunología , Envejecimiento/inmunología , Envejecimiento/metabolismo , Animales , Comunicación Celular/inmunología , Polaridad Celular/inmunología , Citocinas/metabolismo , Diabetes Mellitus/inmunología , Diabetes Mellitus/metabolismo , Humanos , Inflamación/inmunología , Inflamación/metabolismo , Células Madre Mesenquimatosas/metabolismo , Transducción de Señal/inmunología
3.
Physiol Rep ; 9(3): e14721, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33527668

RESUMEN

BACKGROUND: LDL-cholesterol lowering variants that upregulate receptor uptake of LDL, such as in PCSK9 and HMGCR, are associated with diabetes via unclear mechanisms. Activation of the NLRP3 inflammasome/interleukin-1 beta (IL-1ß) pathway promotes white adipose tissue (WAT) dysfunction and type 2 diabetes (T2D) and is regulated by LDL receptors (LDLR and CD36). We hypothesized that: (a) normocholesterolemic subjects with lower plasma PCSK9, identifying those with higher WAT surface-expression of LDLR and CD36, have higher activation of WAT NLRP3 inflammasome and T2D risk factors, and; (b) LDL upregulate adipocyte NLRP3 inflammasome and inhibit adipocyte function. METHODOLOGY: Post hoc analysis was conducted in 27 overweight/ obese subjects with normal plasma LDL-C and measures of disposition index (DI during Botnia clamps) and postprandial fat metabolism. WAT was assessed for surface-expression of LDLR and CD36 (immunohistochemistry), protein expression (immunoblot), IL-1ß secretion (AlphaLISA), and function (3 H-triolein storage). RESULTS: Compared to subjects with higher than median plasma PCSK9, subjects with lower PCSK9 had higher WAT surface-expression of LDLR (+81%) and CD36 (+36%), WAT IL-1ß secretion (+284%), plasma IL-1 receptor-antagonist (+85%), and postprandial hypertriglyceridemia, and lower WAT pro-IL-1ß protein (-66%), WAT function (-62%), and DI (-28%), without group-differences in body composition, energy intake or expenditure. Adjusting for WAT LDLR or CD36 eliminated group-differences in WAT function, DI, and postprandial hypertriglyceridemia. Native LDL inhibited Simpson-Golabi Behmel-syndrome (SGBS) adipocyte differentiation and function and increased inflammation. CONCLUSION: Normocholesterolemic subjects with lower plasma PCSK9 and higher WAT surface-expression of LDLR and CD36 have higher WAT NLRP3 inflammasome activation and T2D risk factors. This may be due to LDL-induced inhibition of adipocyte function.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Antígenos CD36/metabolismo , Colesterol/sangre , Inflamasomas/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Obesidad/sangre , Proproteína Convertasa 9/sangre , Receptores de LDL/metabolismo , Adipocitos Blancos/inmunología , Adipocitos Blancos/metabolismo , Adipogénesis , Tejido Adiposo Blanco/inmunología , Anciano , Biomarcadores/sangre , Células Cultivadas , Diabetes Mellitus Tipo 2/etiología , Regulación hacia Abajo , Femenino , Humanos , Interleucina-1beta/metabolismo , Masculino , Persona de Mediana Edad , Obesidad/complicaciones , Obesidad/enzimología , Obesidad/inmunología , Medición de Riesgo , Factores de Riesgo
4.
Cells ; 8(7)2019 06 30.
Artículo en Inglés | MEDLINE | ID: mdl-31262098

RESUMEN

Obesity is characterized by chronic and low-grade systemic inflammation, an increase of adipose tissue, hypertrophy, and hyperplasia of adipocytes. Adipose tissues can be classified into white, brown, beige and pink adipose tissues, which display different regulatory, morphological and functional characteristics of their adipocyte and immune cells. Brown and white adipocytes can play a key role not only in the control of energy homeostasis, or through the balance between energy storage and expenditure, but also by the modulation of immune and inflammatory responses. Therefore, brown and white adipocytes can orchestrate important immunological crosstalk that may deeply impact the tumor microenvironment and be crucial for cancer establishment and progression. Recent works have indicated that white adipose tissues can undergo a process called browning, in which an inducible brown adipocyte develops. In this review, we depict the mechanisms involved in the differential role of brown, white and pink adipocytes, highlighting their structural, morphological, regulatory and functional characteristics and correlation with cancer predisposition, establishment, and progression. We also discuss the impact of the increased adiposity in the inflammatory and immunological modulation. Moreover, we focused on the plasticity of adipocytes, describing the molecules produced and secreted by those cells, the modulation of the signaling pathways involved in the browning phenomena of white adipose tissue and its impact on inflammation and cancer.


Asunto(s)
Adiposidad/inmunología , Carcinogénesis/inmunología , Inflamación/inmunología , Neoplasias/inmunología , Obesidad/inmunología , Adipocitos Marrones/inmunología , Adipocitos Marrones/metabolismo , Adipocitos Blancos/inmunología , Adipocitos Blancos/metabolismo , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/inmunología , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/citología , Tejido Adiposo Blanco/inmunología , Tejido Adiposo Blanco/metabolismo , Animales , Carcinogénesis/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Metabolismo Energético/inmunología , Humanos , Inflamación/metabolismo , Inflamación/patología , Neoplasias/metabolismo , Neoplasias/patología , Obesidad/complicaciones , Obesidad/metabolismo , Microambiente Tumoral/inmunología
5.
Endocrinology ; 160(7): 1645-1658, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31107528

RESUMEN

The immune system plays a critical role in white adipose tissue (WAT) energy homeostasis and, by extension, whole-body metabolism. Substantial evidence from mouse and human studies firmly establishes that insulin sensitivity deteriorates as a result of subclinical inflammation in the adipose tissue of individuals with diabetes. However, the relationship between adipose tissue expandability and immune cell infiltration remains a complex problem important for understanding the pathogenesis of obesity. Notably, a large body of work challenges the idea that all immune responses are deleterious to WAT function. This review highlights recent advances that describe how immune cells and adipocytes coordinately enable WAT expansion and regulation of energy homeostasis.


Asunto(s)
Adipocitos Blancos/inmunología , Tejido Adiposo Blanco/inmunología , Metabolismo Energético/inmunología , Sistema Inmunológico/inmunología , Obesidad/inmunología , Animales , Inflamación/inmunología , Resistencia a la Insulina/fisiología
6.
J. physiol. biochem ; 74(4): 559-568, nov. 2018. tab, graf
Artículo en Inglés | IBECS | ID: ibc-179034

RESUMEN

Obesity is usually associated with low-grade inflammation, which determines the appearance of comorbidities like atherosclerosis and insulin resistance. Infiltrated macrophages in adipose tissue are partly responsible of this inflammatory condition. Numerous studies point to the existence of close intercommunication between macrophages and adipocytes and pay particular attention to the proinflammatory cytokines released by both cell types. However, it has been recently described that in both, circulation and tissue level, there are extracellular vesicles (including microvesicles and exosomes) containing miRNAs, mRNAs, and proteins that can influence the inflammatory response. The objective of the present research is to investigate the effect of exosomes released by lipopolysaccharide (LPS)-activated macrophages on gene expression and cell metabolism of adipocytes, focusing on the differential exosomal miRNA pattern between LPS- and non-activated macrophages. The results show that the exosomes secreted by the macrophages do not influence the preadipocyte-to-adipocyte differentiation process, fat storage, and insulin-mediated glucose uptake in adipocytes. However, exosomes induce changes in adipocyte gene expression depending on their origin (LPS- or non-activated macrophages), including genes such as CXCL5, SOD, TNFAIP3, C3, and CD34. Some of the pathways or metabolic processes upregulated by exosomes from LPS-activated macrophages are related to inflammation (complement activation, regulation of reactive oxygen species, migration and activation of leukocyte, and monocyte chemotaxis), carbohydrate catabolism, and cell activation. miR-530, chr9_22532, and chr16_34840 are more abundant in exosomes from LPS-activated macrophages, whereas miR-127, miR-143, and miR-486 are more abundant in those secreted by non-activated macrophages


Asunto(s)
Humanos , Adipocitos Blancos/fisiología , Adipogénesis , Regulación de la Expresión Génica , Resistencia a la Insulina , Activación de Macrófagos , Macrófagos/fisiología , Absorción Fisiológica , Adipocitos Blancos/citología , Adipocitos Blancos/inmunología , Antígenos CD34/metabolismo , Comunicación Celular , Quimiocina CXCL5/genética , Quimiocina CXCL5/metabolismo , Técnicas de Cocultivo , Complemento C3/genética , Complemento C3/metabolismo
7.
J Physiol Biochem ; 74(4): 559-568, 2018 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-29560554

RESUMEN

Obesity is usually associated with low-grade inflammation, which determines the appearance of comorbidities like atherosclerosis and insulin resistance. Infiltrated macrophages in adipose tissue are partly responsible of this inflammatory condition. Numerous studies point to the existence of close intercommunication between macrophages and adipocytes and pay particular attention to the proinflammatory cytokines released by both cell types. However, it has been recently described that in both, circulation and tissue level, there are extracellular vesicles (including microvesicles and exosomes) containing miRNAs, mRNAs, and proteins that can influence the inflammatory response. The objective of the present research is to investigate the effect of exosomes released by lipopolysaccharide (LPS)-activated macrophages on gene expression and cell metabolism of adipocytes, focusing on the differential exosomal miRNA pattern between LPS- and non-activated macrophages. The results show that the exosomes secreted by the macrophages do not influence the preadipocyte-to-adipocyte differentiation process, fat storage, and insulin-mediated glucose uptake in adipocytes. However, exosomes induce changes in adipocyte gene expression depending on their origin (LPS- or non-activated macrophages), including genes such as CXCL5, SOD, TNFAIP3, C3, and CD34. Some of the pathways or metabolic processes upregulated by exosomes from LPS-activated macrophages are related to inflammation (complement activation, regulation of reactive oxygen species, migration and activation of leukocyte, and monocyte chemotaxis), carbohydrate catabolism, and cell activation. miR-530, chr9_22532, and chr16_34840 are more abundant in exosomes from LPS-activated macrophages, whereas miR-127, miR-143, and miR-486 are more abundant in those secreted by non-activated macrophages.


Asunto(s)
Adipocitos Blancos/fisiología , Adipogénesis , Exosomas/fisiología , Regulación de la Expresión Génica , Resistencia a la Insulina , Activación de Macrófagos , Macrófagos/fisiología , Absorción Fisiológica , Adipocitos Blancos/citología , Adipocitos Blancos/inmunología , Antígenos CD34/genética , Antígenos CD34/metabolismo , Comunicación Celular/efectos de los fármacos , Células Cultivadas , Quimiocina CXCL5/genética , Quimiocina CXCL5/metabolismo , Técnicas de Cocultivo , Complemento C3/genética , Complemento C3/metabolismo , Desoxiglucosa/metabolismo , Exosomas/efectos de los fármacos , Exosomas/inmunología , Humanos , Lipopolisacáridos/toxicidad , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , MicroARNs/metabolismo , Células THP-1 , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/genética , Proteína 3 Inducida por el Factor de Necrosis Tumoral alfa/metabolismo
8.
Georgian Med News ; (274): 116-124, 2018 Jan.
Artículo en Ruso | MEDLINE | ID: mdl-29461239

RESUMEN

In addition to accumulation and metabolism of triglycerides, white adipose tissue is recognized as the active endocrine organ, whose dysfunction is associated with the development of a wide range of diseases. The secretome of adipocytes is represented by a wide range of adipokines, which vary in depot and sex-specific manner. In addition, adipokines have diverse biological effects, correlations with different metabolic features and functions. In this review, the data on biological effects, origin and the clinical significance of adipokines are discussed. The influence of adipokines on metabolism, sensitivity to insulin, vascular homeostasis, angiogenesis, repair, inflammation and immune cells are shown. Visceral adipose tissue accumulation is accompanied with adipocytes hypertrophy and overproduction of such proinflammatory and proaterogenic molecules like resistin, visfatin, vaspin, tumor necrosis factor, interleukin 6, lipocalin, glypican 4, RBP4 etc. There is a tight correlation between these adipokines level and development of insulin resistance, type 2 diabetes, cardiometabolic complications and cancer. Thus, adipokines represent a group of informative biomarkers for the diagnostics of metabolic disorders and the prediction of the outcome of the wide range of diseases. The study of the effects and mechanisms of the action of adipokines is the basis for determining new targets for therapy.


Asunto(s)
Adipocitos Blancos/metabolismo , Adipoquinas/genética , Tejido Adiposo Blanco/metabolismo , Enfermedades Cardiovasculares/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Neoplasias/metabolismo , Adipocitos Blancos/citología , Adipocitos Blancos/inmunología , Adipoquinas/inmunología , Tejido Adiposo Blanco/citología , Tejido Adiposo Blanco/inmunología , Biomarcadores/metabolismo , Enfermedades Cardiovasculares/genética , Enfermedades Cardiovasculares/inmunología , Enfermedades Cardiovasculares/patología , Diabetes Mellitus Tipo 2/genética , Diabetes Mellitus Tipo 2/inmunología , Diabetes Mellitus Tipo 2/patología , Regulación de la Expresión Génica , Humanos , Resistencia a la Insulina , Metaboloma/inmunología , Neoplasias/genética , Neoplasias/inmunología , Neoplasias/patología , Neovascularización Fisiológica/inmunología , Factores Sexuales , Transducción de Señal , Triglicéridos/inmunología , Triglicéridos/metabolismo
9.
Diabetes ; 67(3): 400-411, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29298809

RESUMEN

Sucrose nonfermenting-related kinase (SNRK) is a member of the AMPK-related kinase family, and its physiological role in adipose energy homeostasis and inflammation remains unknown. We previously reported that SNRK is ubiquitously and abundantly expressed in both white adipose tissue (WAT) and brown adipose tissue (BAT), but SNRK expression diminishes in adipose tissue in obesity. In this study we report novel experimental findings from both animal models and human genetics. SNRK is essential for survival; SNRK globally deficient pups die within 24 h after birth. Heterozygous mice are characterized by inflamed WAT and less BAT. Adipocyte-specific ablation of SNRK causes inflammation in WAT, ectopic lipid deposition in liver and muscle, and impaired adaptive thermogenesis in BAT. These metabolic disorders subsequently lead to decreased energy expenditure, higher body weight, and insulin resistance. We further confirm the significant association of common variants of the SNRK gene with obesity risk in humans. Through applying a phosphoproteomic approach, we identified eukaryotic elongation factor 1δ and histone deacetylase 1/2 as potential SNRK substrates. Taking these data together, we conclude that SNRK represses WAT inflammation and is essential to maintain BAT thermogenesis, making it a novel therapeutic target for treating obesity and associated metabolic disorders.


Asunto(s)
Adipocitos Marrones/metabolismo , Adipocitos Blancos/metabolismo , Metabolismo Energético , Mitocondrias/metabolismo , Paniculitis/metabolismo , Proteínas Serina-Treonina Quinasas/metabolismo , Adipocitos Marrones/inmunología , Adipocitos Marrones/patología , Adipocitos Marrones/ultraestructura , Adipocitos Blancos/inmunología , Adipocitos Blancos/patología , Adipocitos Blancos/ultraestructura , Animales , Índice de Masa Corporal , Células Cultivadas , Cruzamientos Genéticos , Femenino , Regulación de la Expresión Génica , Predisposición Genética a la Enfermedad , Estudio de Asociación del Genoma Completo , Humanos , Masculino , Ratones Noqueados , Ratones Transgénicos , Microscopía Electrónica de Transmisión , Mitocondrias/inmunología , Mitocondrias/patología , Mitocondrias/ultraestructura , Obesidad/genética , Obesidad/fisiopatología , Paniculitis/etiología , Paniculitis/inmunología , Paniculitis/patología , Polimorfismo de Nucleótido Simple , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Proteínas Serina-Treonina Quinasas/genética , Interferencia de ARN , Termogénesis
10.
Diabetes ; 67(3): 360-371, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-29233935

RESUMEN

Peroxisome proliferator-activated receptor (PPAR) δ plays a pivotal role in metabolic homeostasis through its effect on insulin signaling. Although diverse genomic actions of PPARδ are postulated, the specific molecular mechanisms whereby PPARδ controls insulin signaling have not been fully elucidated. We demonstrate here that short-term activation of PPARδ results in the formation of a stable complex with nuclear T-cell protein tyrosine phosphatase 45 (TCPTP45) isoform. This interaction of PPARδ with TCPTP45 blocked translocation of TCPTP45 into the cytoplasm, thereby preventing its interaction with the insulin receptor, which inhibits insulin signaling. Interaction of PPARδ with TCPTP45 blunted interleukin 6-induced insulin resistance, leading to retention of TCPTP45 in the nucleus, thereby facilitating deactivation of the signal transducer and activator of transcription 3 (STAT3)-suppressor of cytokine signaling 3 (SOCS3) signal. Finally, GW501516-activated PPARδ improved insulin signaling and glucose intolerance in mice fed a high-fat diet through its interaction with TCPTP45. This novel interaction of PPARδ constitutes the most upstream component identified of the mechanism downregulating insulin signaling.


Asunto(s)
Intolerancia a la Glucosa/prevención & control , Hepatocitos/efectos de los fármacos , Resistencia a la Insulina , Obesidad/tratamiento farmacológico , PPAR delta/agonistas , Proteína Tirosina Fosfatasa no Receptora Tipo 2/metabolismo , Tiazoles/farmacología , Transporte Activo de Núcleo Celular/efectos de los fármacos , Adipocitos Blancos/efectos de los fármacos , Adipocitos Blancos/inmunología , Adipocitos Blancos/metabolismo , Adipocitos Blancos/patología , Empalme Alternativo , Animales , Antiinflamatorios no Esteroideos/farmacología , Antiinflamatorios no Esteroideos/uso terapéutico , Línea Celular , Células Cultivadas , Intolerancia a la Glucosa/etiología , Intolerancia a la Glucosa/inmunología , Hepatocitos/inmunología , Hepatocitos/metabolismo , Hepatocitos/patología , Humanos , Masculino , Ratones Endogámicos ICR , Fibras Musculares Esqueléticas/efectos de los fármacos , Fibras Musculares Esqueléticas/inmunología , Fibras Musculares Esqueléticas/metabolismo , Fibras Musculares Esqueléticas/patología , Obesidad/metabolismo , Obesidad/patología , Obesidad/fisiopatología , PPAR delta/antagonistas & inhibidores , PPAR delta/genética , PPAR delta/metabolismo , Multimerización de Proteína/efectos de los fármacos , Proteína Tirosina Fosfatasa no Receptora Tipo 2/química , Proteína Tirosina Fosfatasa no Receptora Tipo 2/genética , Interferencia de ARN , Proteínas Recombinantes de Fusión/química , Proteínas Recombinantes de Fusión/metabolismo , Organismos Libres de Patógenos Específicos , Tiazoles/uso terapéutico
11.
Diabetes ; 67(2): 235-247, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29133512

RESUMEN

Adipose-derived stem cells (ADSCs) play critical roles in controlling obesity-associated inflammation and metabolic disorders. Exosomes from ADSCs exert protective effects in several diseases, but their roles in obesity and related pathological conditions remain unclear. In this study, we showed that treatment of obese mice with ADSC-derived exosomes facilitated their metabolic homeostasis, including improved insulin sensitivity (27.8% improvement), reduced obesity, and alleviated hepatic steatosis. ADSC-derived exosomes drove alternatively activated M2 macrophage polarization, inflammation reduction, and beiging in white adipose tissue (WAT) of diet-induced obese mice. Mechanistically, exosomes from ADSCs transferred into macrophages to induce anti-inflammatory M2 phenotypes through the transactivation of arginase-1 by exosome-carried active STAT3. Moreover, M2 macrophages induced by ADSC-derived exosomes not only expressed high levels of tyrosine hydroxylase responsible for catecholamine release, but also promoted ADSC proliferation and lactate production, thereby favoring WAT beiging and homeostasis in response to high-fat challenge. These findings delineate a novel exosome-mediated mechanism for ADSC-macrophage cross talk that facilitates immune and metabolic homeostasis in WAT, thus providing potential therapy for obesity and diabetes.


Asunto(s)
Adipocitos Beige/patología , Adipocitos Blancos/patología , Adipogénesis , Células Madre Adultas/patología , Exosomas/trasplante , Macrófagos Peritoneales/trasplante , Obesidad/terapia , Adipocitos Beige/inmunología , Adipocitos Beige/metabolismo , Adipocitos Blancos/inmunología , Adipocitos Blancos/metabolismo , Células Madre Adultas/inmunología , Células Madre Adultas/metabolismo , Animales , Biomarcadores/metabolismo , Comunicación Celular , Polaridad Celular , Proliferación Celular , Células Cultivadas , Dieta Alta en Grasa/efectos adversos , Exosomas/inmunología , Exosomas/metabolismo , Exosomas/patología , Resistencia a la Insulina , Activación de Macrófagos , Macrófagos Peritoneales/inmunología , Macrófagos Peritoneales/metabolismo , Macrófagos Peritoneales/patología , Masculino , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , Microscopía Fluorescente , Enfermedad del Hígado Graso no Alcohólico/etiología , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Obesidad/inmunología , Obesidad/patología , Obesidad/fisiopatología , Fagocitosis
12.
J Biol Chem ; 292(34): 13959-13969, 2017 08 25.
Artículo en Inglés | MEDLINE | ID: mdl-28698385

RESUMEN

Adipose tissue inflammation has been linked to metabolic diseases such as obesity and type 2 diabetes. However, the molecules that mediate inflammation in adipose tissue have not been addressed. Although retinoic acid receptor-related orphan receptor α (RORα) is known to be involved in the regulation of inflammatory response in some tissues, its role is largely unknown in adipose tissue. Conversely, it is known that endoplasmic reticulum (ER) stress and unfolding protein response (UPR) signaling affect the inflammatory response in obese adipose tissue, but whether RORα regulates these processes remains unknown. In this study, we investigate the link between RORα and adipose tissue inflammation. We showed that the inflammatory response in macrophages or 3T3-L1 adipocytes stimulated by lipopolysaccharide, as well as adipose tissue in obese mice, markedly increased the expression of RORα. Adenovirus-mediated overexpression of RORα or treatment with the RORα-specific agonist SR1078 enhanced the expression of inflammatory cytokines and increased the number of infiltrated macrophages into adipose tissue. Furthermore, SR1078 up-regulated the mRNA expression of ER stress response genes and enhanced phosphorylations of two of the three mediators of major UPR signaling pathways, PERK and IRE1α. Finally, we found that alleviation of ER stress using a chemical chaperone followed by the suppression of RORα induced inflammation in adipose tissue. Our data suggest that RORα-induced ER stress response potentially contributes to the adipose tissue inflammation that can be mitigated by treatment with chemical chaperones. The relationships established here between RORα expression, inflammation, and UPR signaling may have implications for therapeutic targeting of obesity-related metabolic diseases.


Asunto(s)
Adipocitos Blancos/metabolismo , Estrés del Retículo Endoplásmico , Macrófagos/metabolismo , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Paniculitis/metabolismo , Transducción de Señal/efectos de los fármacos , Respuesta de Proteína Desplegada , Células 3T3-L1 , Adipocitos Blancos/efectos de los fármacos , Adipocitos Blancos/inmunología , Adipocitos Blancos/patología , Animales , Fármacos Antiobesidad/farmacología , Fármacos Antiobesidad/uso terapéutico , Benzamidas/farmacología , Benzamidas/uso terapéutico , Estrés del Retículo Endoplásmico/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Resistencia a la Insulina , Lipopolisacáridos/toxicidad , Activación de Macrófagos/efectos de los fármacos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/agonistas , Miembro 1 del Grupo F de la Subfamilia 1 de Receptores Nucleares/genética , Obesidad/tratamiento farmacológico , Obesidad/fisiopatología , Paniculitis/inmunología , Paniculitis/patología , Paniculitis/prevención & control , Fenilbutiratos/farmacología , Fenilbutiratos/uso terapéutico , Células RAW 264.7 , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Respuesta de Proteína Desplegada/efectos de los fármacos
13.
Biochem Pharmacol ; 140: 73-88, 2017 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-28642037

RESUMEN

Low testosterone levels are strongly related to obesity in males. The balance between the classically M1 and alternatively M2 polarized macrophages also plays a critical role in obesity. It is not clear whether testosterone regulates macrophage polarization and then affects adipocyte differentiation. In this report, we demonstrate that testosterone strengthens interleukin (IL) -4-induced M2 polarization and inhibits lipopolysaccharide (LPS)-induced M1 polarization, but has no direct effect on adipocyte differentiation. Cellular signaling studies indicate that testosterone regulates macrophage polarization through the inhibitory regulative G-protein (Gαi) mainly, rather than via androgen receptors, and phosphorylation of Akt. Moreover, testosterone inhibits pre-adipocyte differentiation induced by M1 macrophage medium. Lowering of serum testosterone in mice by injecting a luteinizing hormone receptor (LHR) peptide increases epididymal white adipose tissue. Testosterone supplementation reverses this effect. Therefore, our findings indicate that testosterone inhibits pre-adipocyte differentiation by switching macrophages to M2 polarization through the Gαi and Akt signaling pathways.


Asunto(s)
Adipocitos Blancos/efectos de los fármacos , Adipogénesis/efectos de los fármacos , Adiposidad/efectos de los fármacos , Terapia de Reemplazo de Hormonas , Macrófagos/efectos de los fármacos , Obesidad/prevención & control , Testosterona/uso terapéutico , Células 3T3-L1 , Adipocitos Blancos/inmunología , Adipocitos Blancos/metabolismo , Adipocitos Blancos/patología , Animales , Polaridad Celular/efectos de los fármacos , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/agonistas , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/antagonistas & inhibidores , Subunidades alfa de la Proteína de Unión al GTP Gi-Go/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Obesidad/inmunología , Obesidad/metabolismo , Obesidad/patología , Proteínas Proto-Oncogénicas c-akt/antagonistas & inhibidores , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , Células RAW 264.7 , Interferencia de ARN , Transducción de Señal/efectos de los fármacos , Organismos Libres de Patógenos Específicos , Testosterona/sangre , Testosterona/farmacología
14.
Mediators Inflamm ; 2017: 9067049, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28458470

RESUMEN

Obesity is marked by chronic, low-grade inflammation. Here, we examined whether intrinsic differences between white and brown adipocytes influence the inflammatory status of macrophages. White and brown adipocytes were characterized by transcriptional regulation of UCP-1, PGC1α, PGC1ß, and CIDEA and their level of IL-6 secretion. The inflammatory profile of PMA-differentiated U937 and THP-1 macrophages, in resting state and after stimulation with LPS/IFN-gamma and IL-4, was assessed by measuring IL-6 secretion and transcriptional regulation of a panel of inflammatory genes after mono- or indirect coculture with white and brown adipocytes. White adipocyte monocultures show increased IL-6 secretion compared to brown adipocytes. White adipocytes cocultured with U937 and THP-1 macrophages induced a greater increase in IL-6 secretion compared to brown adipocytes cocultured with both macrophages. White adipocytes cocultured with macrophages increased inflammatory gene expression in both types. In contrast, macrophages cocultured with brown adipocytes induced downregulation or no alterations in inflammatory gene expression. The effects of adipocytes on macrophages appear to be independent of stimulation state. Brown adipocytes exhibit an intrinsic ability to dampen inflammatory profile of macrophages, while white adipocytes enhance it. These data suggest that brown adipocytes may be less prone to adipose tissue inflammation that is associated with obesity.


Asunto(s)
Adipocitos Marrones/metabolismo , Adipocitos Blancos/metabolismo , Inflamación/metabolismo , Macrófagos/metabolismo , Adipocitos Marrones/efectos de los fármacos , Adipocitos Marrones/inmunología , Adipocitos Blancos/efectos de los fármacos , Adipocitos Blancos/inmunología , Adulto , Diferenciación Celular/efectos de los fármacos , Línea Celular , Células Cultivadas , Ensayo de Inmunoadsorción Enzimática , Femenino , Perfilación de la Expresión Génica , Humanos , Inflamación/inmunología , Interleucina-4/farmacología , Interleucina-6/metabolismo , Lipopolisacáridos/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Persona de Mediana Edad
15.
Arch Physiol Biochem ; 123(1): 61-67, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27855518

RESUMEN

The role of IL-1ß in regulating the expression of extracellular matrix (ECM) and cell adhesion genes in human adipocytes has been examined. Adipocytes differentiated in culture were incubated with IL-1ß for 4 or 24 h and RNA probed with PCR arrays for 84 ECM and cell adhesion genes. Treatment with IL-1ß resulted in changes in the expression at one or both time points of ∼50% of the genes probed by the arrays, the majority being down-regulated. Genes whose expression was down-regulated by IL-1ß included those encoding several collagen chains and integrin subunits. In contrast, IL-1ß induced substantial increases (>10-fold) in the expression of ICAM1, VCAM1, MMP1 and MMP3; the secretion of the encoded proteins was also markedly stimulated. IL-1ß has a pervasive effect on the expression of ECM and cell adhesion genes in human adipocytes, consistent with the derangement of tissue structure during inflammation in white fat.


Asunto(s)
Adipocitos Blancos/metabolismo , Moléculas de Adhesión Celular/metabolismo , Proteínas de la Matriz Extracelular/metabolismo , Regulación de la Expresión Génica , Interleucina-1beta/metabolismo , Adipocitos Blancos/inmunología , Adipocitos Blancos/patología , Moléculas de Adhesión Celular/genética , Diferenciación Celular , Células Cultivadas , Proteínas de la Matriz Extracelular/genética , Femenino , Humanos , Molécula 1 de Adhesión Intercelular/química , Molécula 1 de Adhesión Intercelular/genética , Molécula 1 de Adhesión Intercelular/metabolismo , Interleucina-1beta/genética , Metaloproteinasa 1 de la Matriz/química , Metaloproteinasa 1 de la Matriz/genética , Metaloproteinasa 1 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/química , Metaloproteinasa 3 de la Matriz/genética , Metaloproteinasa 3 de la Matriz/metabolismo , Persona de Mediana Edad , Obesidad/inmunología , Obesidad/metabolismo , Obesidad/patología , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Recombinantes/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Grasa Subcutánea/inmunología , Grasa Subcutánea/metabolismo , Grasa Subcutánea/patología , Regulación hacia Arriba , Molécula 1 de Adhesión Celular Vascular/química , Molécula 1 de Adhesión Celular Vascular/genética , Molécula 1 de Adhesión Celular Vascular/metabolismo
16.
J Biol Chem ; 291(45): 23390-23402, 2016 Nov 04.
Artículo en Inglés | MEDLINE | ID: mdl-27621315

RESUMEN

Adrenomedullin 2 (ADM2) is an endogenous bioactive peptide belonging to the calcitonin gene-related peptide family. Our previous studies showed that overexpression of ADM2 in mice reduced obesity and insulin resistance by increasing thermogenesis in brown adipose tissue. However, the effects of ADM2 in another type of thermogenic adipocyte, beige adipocytes, remain to be understood. The plasma ADM2 levels were inversely correlated with obesity in humans, and adipo-ADM2-transgenic (tg) mice displayed resistance to high-fat diet-induced obesity with increased energy expenditure. Beiging of subcutaneous white adipose tissues (WAT) was more noticeably induced in high-fat diet-fed transgenic mice with adipocyte-ADM2 overexpression (adipo-ADM2-tg mice) than in WT animals. ADM2 treatment in primary rat subcutaneous adipocytes induced beiging with up-regulation of UCP1 and beiging-related marker genes and increased mitochondrial uncoupling respiration, which was mainly mediated by activation of the calcitonin receptor-like receptor (CRLR)·receptor activity-modifying protein 1 (RAMP1) complex and PKA and p38 MAPK signaling pathways. Importantly, this adipocyte-autonomous beiging effect by ADM2 was translatable to human primary adipocytes. In addition, M2 macrophage activation also contributed to the beiging effects of ADM2 through catecholamine secretion. Therefore, our study reveals that ADM2 enhances subcutaneous WAT beiging via a direct effect by activating the CRLR·RAMP1-cAMP/PKA and p38 MAPK pathways in white adipocytes and via an indirect effect by stimulating alternative M2 polarization in macrophages. Through both mechanisms, beiging of WAT by ADM2 results in increased energy expenditure and reduced obesity, suggesting ADM2 as a novel anti-obesity target.


Asunto(s)
Tejido Adiposo Pardo/inmunología , Tejido Adiposo Blanco/inmunología , Activación de Macrófagos , Macrófagos/inmunología , Neuropéptidos/inmunología , Obesidad/inmunología , Hormonas Peptídicas/inmunología , Adipocitos Blancos/inmunología , Adipocitos Blancos/patología , Tejido Adiposo Pardo/patología , Tejido Adiposo Blanco/patología , Animales , Células Cultivadas , Dieta Alta en Grasa/efectos adversos , Metabolismo Energético , Femenino , Regulación de la Expresión Génica , Humanos , Macrófagos/patología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , Neuropéptidos/genética , Obesidad/etiología , Obesidad/genética , Obesidad/patología , Hormonas Peptídicas/genética , Ratas Sprague-Dawley , Transducción de Señal , Termogénesis , Regulación hacia Arriba
17.
J Nutr Biochem ; 34: 61-72, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27208584

RESUMEN

Adipocyte-macrophage cross-talk propagates immune responses in obese adipose tissue (AT). Long-chain n-3 polyunsaturated fatty acids (LC n-3 PUFA) mitigate inflammation, partly through up-regulation of adiponectin; however, specific mechanisms are unclear. We determined if adipocyte-macrophage cross-talk could be mitigated by dietary LC n-3 PUFA and if this was dependent on adiponectin-mediated signaling. We utilized an in vitro co-culture model mimicking the ratio of adipocytes:macrophages in obese AT, whereby 3T3-L1 adipocytes were co-cultured with splenic CD11b(+) macrophages from C57BL/6 mice fed high-fat control (HF-CON; 34% w/w fat) or fish oil diets (HF-FO; 34% w/w fat containing 7.6% w/w FO), as well as mice fed low-fat control (LF-CON; 10% w/w fat) or FO diets (LF-FO; 10% w/w fat containing 3% w/w FO). Co-culture conditions tested effects of soluble mediator-driven mechanisms (trans-well system), cell contact and low-dose lipopolysaccharide (LPS) mimicking acute or chronic inflammatory conditions. HF-FO macrophages from acute LPS-stimulated trans-well co-cultures had decreased mRNA expression of Casp1, Il1ß and Il18, as well as cellular caspase-1 activity compared to HF-CON macrophages (P≤.05). Moreover, adipocytes from acute LPS-stimulated HF-FO co-cultures had decreased caspase-1 activity and decreased IL-1ß/IL-18 levels following chronic LPS pretreatment compared to HF-CON co-cultures (P≤.05). Additionally, in contact co-cultures with adiponectin-neutralizing antibody, the FO-mediated modulation of NFκB activity and decrease in phosphorylated p65 NFκB, expression of NLRP3 inflammasome genes, M1 macrophage marker genes and inflammatory cytokine/chemokine secretion were controlled partly through adiponectin, while cellular caspase-1 activity and IL-1ß/1L-18 levels were decreased independently of adiponectin (P≤.05). LC n-3 PUFA may decrease the intensity of adipocyte-macrophage cross-talk to mitigate obesity-associated pathologies.


Asunto(s)
Adipocitos Blancos/metabolismo , Suplementos Dietéticos , Ácidos Grasos Omega-3/uso terapéutico , Inflamasomas/metabolismo , Macrófagos/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/antagonistas & inhibidores , Obesidad/dietoterapia , Células 3T3-L1 , Adipocitos Blancos/inmunología , Adipocitos Blancos/patología , Animales , Antiinflamatorios no Esteroideos/análisis , Antiinflamatorios no Esteroideos/química , Antiinflamatorios no Esteroideos/metabolismo , Antiinflamatorios no Esteroideos/uso terapéutico , Antígeno CD11b/metabolismo , Comunicación Celular , Células Cultivadas , Técnicas de Cocultivo , Dieta Alta en Grasa/efectos adversos , Suplementos Dietéticos/análisis , Ácidos Grasos Omega-3/análisis , Ácidos Grasos Omega-3/metabolismo , Femenino , Aceites de Pescado/química , Aceites de Pescado/uso terapéutico , Regulación de la Expresión Génica , Inflamasomas/inmunología , Macrófagos/inmunología , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Obesidad/inmunología , Obesidad/metabolismo , Obesidad/patología , Bazo/inmunología , Bazo/metabolismo , Bazo/patología
18.
J Clin Endocrinol Metab ; 100(11): E1467-76, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26252355

RESUMEN

CONTEXT: Molecular mechanisms associated with physiological variations in adipose tissue (AT) are not fully recognized. The most recent reports highlight the critical relevance of microRNAs (miRNAs) found in AT. OBJECTIVE: To identify changes in messenger RNA (mRNA) and miRNA expressions and their interaction in human AT before and after surgery-induced weight loss. Research Design and Setting: Genome-wide mRNA and miRNA expressions were assessed by microarrays in abdominal subcutaneous AT of 16 morbidly obese women before and 2 years after laparoscopic Roux-en-Y gastric bypass. The association of changes in miRNAs with their respective mRNA targets was studied. The results were replicated in publicly available microarray datasets. Validation was made by real-time polymerase chain reaction in additional fat samples from 26 age-matched lean women and in isolated human adipocytes. RESULTS: A total of 5018 different mRNA probe sets and 15 miRNAs were differentially expressed after surgery-induced weight loss. The clustering of similar expression patterns for gene products with related functions revealed molecular footprints that elucidate significant changes in cell cycle, development, lipid metabolism, and the inflammatory response. The participation of inflammation was demonstrated by results assessed in isolated adipocytes. Interestingly, when transcriptomes were analyzed taking into account the presence of miRNA target sites, miRNA target mRNAs were upregulated in obese AT (P value = 2 × 10(-181)) and inflamed adipocytes (P value = 4 × 10(-61)), according to the number of target sites harbored by each transcript. CONCLUSIONS: Current findings suggest impaired miRNA target gene expression in obese AT in close association with inflammation, both improving after weight loss.


Asunto(s)
Regulación hacia Abajo , Derivación Gástrica , MicroARNs/metabolismo , Obesidad Mórbida/cirugía , Grasa Subcutánea Abdominal/metabolismo , Adipocitos Blancos/citología , Adipocitos Blancos/inmunología , Adipocitos Blancos/metabolismo , Adulto , Índice de Masa Corporal , Línea Celular , Células Cultivadas , Estudios de Cohortes , Estudios Transversales , Femenino , Perfilación de la Expresión Génica , Estudio de Asociación del Genoma Completo , Humanos , Estudios Longitudinales , Persona de Mediana Edad , Monocitos/inmunología , Monocitos/metabolismo , Obesidad Mórbida/genética , Obesidad Mórbida/inmunología , Obesidad Mórbida/metabolismo , ARN Mensajero/metabolismo , Grasa Subcutánea Abdominal/inmunología , Pérdida de Peso
19.
Am J Physiol Endocrinol Metab ; 309(3): E214-23, 2015 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-26015438

RESUMEN

Dipeptidyl peptidase IV (DPP-IV) expression in visceral adipose tissue is reportedly increased in obese patients, suggesting an association of DPP-IV with inflammation. In this study, first, lipopolysaccharide (LPS)- or palmitate-induced elevations of inflammatory cytokine mRNA expressions in RAW264.7 macrophages were shown to be significantly suppressed by coincubation with a DPP-IV inhibitor, anagliptin (10 µM), despite low DPP-IV expression in the RAW264.7 cells. Regarding the molecular mechanism, LPS-induced degradation of IκBα and phosphorylations of p65, JNK, and p38, as well as NF-κB and AP-1 promoter activities, were revealed to be suppressed by incubation with anagliptin, indicating suppressive effects of anagliptin on both NF-κB and AP-1 signaling pathways. Anagliptin also acted on 3T3-L1 adipocytes, weakly suppressing the inflammatory cytokine expressions induced by LPS and TNFα. When 3T3-L1 and RAW cells were cocultured and stimulated with LPS, the effects of anagliptin on the suppression of cytokine expressions in 3T3-L1 adipocytes were more marked and became evident at the 10 µM concentration. Anti-inflammatory effects of anagliptin were also observed in vivo on the elevated hepatic and adipose expressions and serum concentrations of inflammatory cytokines in association with the suppression of hepatic NF-κB transcriptional activity in LPS-infused mice. Taking these observations together, the anti-inflammatory properties of anagliptin may be beneficial in terms of preventing exacerbation of diabetes and cardiovascular events.


Asunto(s)
Adipocitos Blancos/efectos de los fármacos , Antiinflamatorios no Esteroideos/farmacología , Inhibidores de la Dipeptidil-Peptidasa IV/farmacología , Hígado/efectos de los fármacos , Macrófagos/efectos de los fármacos , FN-kappa B/antagonistas & inhibidores , Pirimidinas/farmacología , Células 3T3-L1 , Adipocitos Blancos/inmunología , Adipocitos Blancos/metabolismo , Animales , Antiinflamatorios no Esteroideos/uso terapéutico , Línea Celular Transformada , Técnicas de Cocultivo , Citocinas/agonistas , Citocinas/antagonistas & inhibidores , Citocinas/genética , Citocinas/metabolismo , Dipeptidil Peptidasa 4/química , Dipeptidil Peptidasa 4/genética , Dipeptidil Peptidasa 4/metabolismo , Inhibidores de la Dipeptidil-Peptidasa IV/uso terapéutico , Regulación de la Expresión Génica/efectos de los fármacos , Genes Reporteros/efectos de los fármacos , Hígado/inmunología , Hígado/metabolismo , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/agonistas , FN-kappa B/genética , FN-kappa B/metabolismo , Fosforilación/efectos de los fármacos , Procesamiento Proteico-Postraduccional/efectos de los fármacos , Pirimidinas/uso terapéutico , Elementos de Respuesta/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Síndrome de Respuesta Inflamatoria Sistémica/sangre , Síndrome de Respuesta Inflamatoria Sistémica/inmunología , Síndrome de Respuesta Inflamatoria Sistémica/metabolismo , Síndrome de Respuesta Inflamatoria Sistémica/prevención & control
20.
Biochem Biophys Res Commun ; 461(2): 287-92, 2015 May 29.
Artículo en Inglés | MEDLINE | ID: mdl-25881506

RESUMEN

Obesity involves hypoxic adipose tissue and low-grade chronic inflammation. We investigated the impact of hypoxia on inflammatory response to TNF-α in white and brown adipocytes. In response to TNF-α, the expression of the inducible enzymes iNOS and COX-2 was prominently and selectively potentiated during hypoxia while only moderately under normoxia. Levels of their products, nitrite and prostaglandinE2 were elevated accordingly. NS398, a selective COX-2 inhibitor, reduced nitrite levels. The expression of PGC-1α, a transcriptional co-activator involved in mitochondrial biogenesis, and PPARγ, a transcription factor involved in adipocyte homeostasis, was reduced by TNF-α during hypoxia. These results suggest that hypoxia potentiates the inflammatory response by TNF-α in both white and brown adipocytes and downregulates the transcription factors involved in adipocyte function.


Asunto(s)
Adipocitos/inmunología , Ciclooxigenasa 2/genética , Regulación de la Expresión Génica , Hipoxia/inmunología , Óxido Nítrico Sintasa de Tipo II/genética , Factor de Necrosis Tumoral alfa/inmunología , Adipocitos/metabolismo , Adipocitos/patología , Adipocitos Marrones/inmunología , Adipocitos Marrones/metabolismo , Adipocitos Marrones/patología , Adipocitos Blancos/inmunología , Adipocitos Blancos/metabolismo , Adipocitos Blancos/patología , Animales , Hipoxia de la Célula , Línea Celular , Ciclooxigenasa 2/análisis , Hipoxia/genética , Hipoxia/patología , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Ratones , Óxido Nítrico Sintasa de Tipo II/análisis
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...