Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 138
Filtrar
1.
J Nutr Biochem ; 100: 108908, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34801687

RESUMEN

Succinic acid widely exists in foods and is used as a food additive. Succinate not only serves as an energy substrate, but also induces protein succinylation. Histone succinylation activates gene transcription. The brown adipose tissue (BAT) is critical for prevention of obesity and metabolic dysfunction, and the fetal stage is pivotal for BAT development. Up to now, the role of maternal succinate supplementation on fetal BAT development and offspring BAT function remains unexamined. To test, female C57BL/6J mice (2-month-old) were separated into 2 groups, received with or without 0.5% succinic acid in drinking water during gestation and lactation. After weaning, female offspring were challenged with high fat diet (HFD) for 12 weeks. Newborn, female weanling, and HFD female offspring mice were analyzed. For neonatal and weaning mice, the BAT weight relative to the whole body weight was significantly increased in the succinate group. The expression of PGC-1α, a key transcription co-activator promoting mitochondrial biogenesis, was elevated in BAT of female neonatal and offspring born to succinate-fed dams. Consistently, maternal succinate supplementation enhanced thermogenesis and the expression of thermogenic genes in offspring BAT. Additionally, maternal succinate supplementation protected female offspring against HFD-induced obesity. Furthermore, in C3H10T1/2 cells, succinate supplementation promoted PGC-1α expression and brown adipogenesis. Mechanistically, succinate supplementation increased permissive histone succinylation and H3K4me3 modification in the Ppargc1a promoter, which correlated with the higher expression of Ppargc1a. In conclusion, maternal succinate supplementation during pregnancy and lactation enhanced fetal BAT development and offspring BAT thermogenesis, which prevented HFD-induced obesity and metabolism dysfunction in offspring.


Asunto(s)
Adipogénesis , Tejido Adiposo Pardo/embriología , Suplementos Dietéticos , Ácido Succínico/administración & dosificación , Termogénesis , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Pardo/fisiología , Animales , Animales Recién Nacidos , Línea Celular , Dieta Alta en Grasa , Femenino , Código de Histonas , Histonas/metabolismo , Lactancia , Ratones , Ratones Endogámicos C57BL , Obesidad/prevención & control , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/genética , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma/metabolismo , Embarazo , Regiones Promotoras Genéticas
2.
Genes Dev ; 35(9-10): 713-728, 2021 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-33888555

RESUMEN

MED1 often serves as a surrogate of the general transcription coactivator complex Mediator for identifying active enhancers. MED1 is required for phenotypic conversion of fibroblasts to adipocytes in vitro, but its role in adipose development and expansion in vivo has not been reported. Here, we show that MED1 is not generally required for transcription during adipogenesis in culture and that MED1 is dispensable for adipose development in mice. Instead, MED1 is required for postnatal adipose expansion and the induction of fatty acid and triglyceride synthesis genes after pups switch diet from high-fat maternal milk to carbohydrate-based chow. During adipogenesis, MED1 is dispensable for induction of lineage-determining transcription factors (TFs) PPARγ and C/EBPα but is required for lipid accumulation in the late phase of differentiation. Mechanistically, MED1 controls the induction of lipogenesis genes by facilitating lipogenic TF ChREBP- and SREBP1a-dependent recruitment of Mediator to active enhancers. Together, our findings identify a cell- and gene-specific regulatory role of MED1 as a lipogenesis coactivator required for postnatal adipose expansion.


Asunto(s)
Tejido Adiposo/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica/genética , Lipogénesis/genética , Subunidad 1 del Complejo Mediador/genética , Subunidad 1 del Complejo Mediador/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo Pardo/embriología , Animales , Células Cultivadas , Dieta , Ratones , Unión Proteica/genética
3.
Biochim Biophys Acta Gene Regul Mech ; 1863(1): 194437, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31730826

RESUMEN

Browning of white adipocytes (WAs) (also referred as beige cells) was demonstrated to execute thermogenesis by consuming stored lipids as do brown adipocytes (BAs), and this is highly related to metabolic homeostasis. Alternative splicing (AS) constitutes a pivotal mechanism for defining cellular fates and functional specifications. Nevertheless, the impacts of AS regulation on the browning of WAs have not been comprehensively investigated. In this study, we first identified the discriminative expression and splicing profiles of the muscleblind-like 1 (MBNL1) gene in postnatal brown adipose tissues (BATs) compared to those of embryonic BATs. A shift in the MBNL1+ex 5 isoform 7 (MBNL17) to MBNL1-ex 5 isoform 1 (MBNL11) was characterized throughout BAT development or during the in vitro browning of pre-WAs, 3T3-L1 cells. The interplay between MBNL1 and the exonic CCUG motif constitutes an autoregulatory mechanism for excluding MBNL1 exon 5. The simultaneous association of RNA-binding motif protein 4a (RBM4a) with exonic and intronic CU elements collaboratively mediates the skipping of MBNL1 exon 5. Overexpressing the MBNL11 isoform exhibited a more-prominent effect than that of the MBNL17 isoform on programming its own transcripts and beige cell-related splicing events in a CCUG motif-mediated manner. In addition to splicing regulation, overexpression of the MBNL11 and MBNL17 isoforms differentially enhanced beige adipogenic signatures of 3T3-L1 cells. Our findings demonstrated that MBNL1 constitutes an emerging and autoregulatory mechanism involved in development of beige cells.


Asunto(s)
Adipogénesis/genética , Tejido Adiposo Pardo/embriología , Tejido Adiposo Pardo/crecimiento & desarrollo , Empalme Alternativo , Proteínas de Unión al ADN/genética , Proteínas de Unión al ARN/genética , Células 3T3-L1 , Tejido Adiposo Pardo/metabolismo , Animales , Proteínas de Unión al ADN/metabolismo , Exones , Ratones , Ratones Endogámicos C57BL , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Proteínas de Unión al ARN/metabolismo
4.
Cell Rep ; 29(6): 1410-1418.e6, 2019 11 05.
Artículo en Inglés | MEDLINE | ID: mdl-31693883

RESUMEN

Browning induction or transplantation of brown adipose tissue (BAT) or brown/beige adipocytes derived from progenitor or induced pluripotent stem cells (iPSCs) can represent a powerful strategy to treat metabolic diseases. However, our poor understanding of the mechanisms that govern the differentiation and activation of brown adipocytes limits the development of such therapy. Various genetic factors controlling the differentiation of brown adipocytes have been identified, although most studies have been performed using in vitro cultured pre-adipocytes. We investigate here the differentiation of brown adipocytes from adipose progenitors in the mouse embryo. We demonstrate that the formation of multiple lipid droplets (LDs) is initiated within clusters of glycogen, which is degraded through glycophagy to provide the metabolic substrates essential for de novo lipogenesis and LD formation. Therefore, this study uncovers the role of glycogen in the generation of LDs.


Asunto(s)
Adipocitos Marrones/metabolismo , Adipogénesis/genética , Tejido Adiposo Pardo/metabolismo , Embrión de Mamíferos/metabolismo , Glucógeno/metabolismo , Gotas Lipídicas/metabolismo , Adipocitos Marrones/ultraestructura , Tejido Adiposo Pardo/embriología , Tejido Adiposo Pardo/ultraestructura , Animales , Autofagia/efectos de los fármacos , Autofagia/genética , Proteínas Potenciadoras de Unión a CCAAT/genética , Proteínas Potenciadoras de Unión a CCAAT/metabolismo , Células Cultivadas , Proteínas de Unión a Ácidos Grasos/genética , Proteínas de Unión a Ácidos Grasos/metabolismo , Glucógeno/ultraestructura , Humanos , Gotas Lipídicas/ultraestructura , Ratones , Ratones Endogámicos C57BL , Microscopía Electrónica de Transmisión , PPAR gamma/genética , PPAR gamma/metabolismo , ARN Interferente Pequeño , Transcriptoma
5.
Biochim Biophys Acta Mol Cell Biol Lipids ; 1863(12): 1488-1497, 2018 12.
Artículo en Inglés | MEDLINE | ID: mdl-30266429

RESUMEN

Brown adipose tissue (BAT) is a crucial regulator of energy expenditure. Emerging evidence suggests that n-3 PUFA potentiate brown adipogenesis in vitro. Since the pregnancy and lactation is a critical time for brown fat formation, we hypothesized that maternal supplementation of n-3 PUFA promotes BAT development in offspring. Female C57BL/6 mice were fed a diet containing n-3 PUFA (3%) derived from fish oil (FO), or an isocaloric diet devoid of n-3 PUFA (Cont) during pregnancy and lactation. Maternal n-3 PUFA intake was delivered to the BAT of neonates significantly reducing the n-6/n-3 ratio. The maternal n-3 PUFA exposure was linked with upregulated brown-specific gene and protein profiles and the functional cluster of brown-specific miRNAs. In addition, maternal n-3 PUFA induced histone modifications in the BAT evidenced by 1) increased epigenetic signature of brown adipogenesis, i.e., H3K27Ac and H3K9me2, 2) modified chromatin-remodeling enzymes, and 3) enriched the H3K27Ac in the promoter region of Ucp1. The offspring received maternal n-3 PUFA nutrition exhibited a significant increase in whole-body energy expenditure and better maintenance of core body temperature against acute cold treatment. Collectively, our results suggest that maternal n-3 PUFA supplementation potentiates fetal BAT development via the synergistic action of miRNA production and histone modifications, which may confer long-lasting metabolic benefits to offspring.


Asunto(s)
Tejido Adiposo Pardo/embriología , Epigénesis Genética/efectos de los fármacos , Ácidos Grasos Omega-3/administración & dosificación , Aceites de Pescado/química , Tejido Adiposo Pardo/efectos de los fármacos , Tejido Adiposo Pardo/metabolismo , Animales , Ensamble y Desensamble de Cromatina/efectos de los fármacos , Suplementos Dietéticos , Metabolismo Energético , Femenino , Aceites de Pescado/administración & dosificación , Regulación del Desarrollo de la Expresión Génica/efectos de los fármacos , Código de Histonas/efectos de los fármacos , Histonas/metabolismo , Ratones , Ratones Endogámicos C57BL , MicroARNs/genética , Embarazo , Proteína Desacopladora 1/genética
6.
J Endocrinol ; 238(1): R53-R62, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29789429

RESUMEN

Although brown adipose tissue (BAT) is one of the smallest organs in the body, it has the potential to have a substantial impact on both heat production as well as fat and carbohydrate metabolism. This is most apparent at birth, which is characterised with the rapid appearance and activation of the BAT specific mitochondrial uncoupling protein (UCP)1 in many large mammals. The amount of brown fat then gradually declines with age, an adaptation that can be modulated by the thermal environment. Given the increased incidence of maternal obesity and its potential transmission to the mother's offspring, increasing BAT activity in the mother could be one mechanism to prevent this cycle. To date, however, all rodent studies investigating maternal obesity have been conducted at standard laboratory temperature (21°C), which represents an appreciable cold challenge. This could also explain why offspring weight is rarely increased, suggesting that future studies would benefit from being conducted at thermoneutrality (~28°C). It is also becoming apparent that each fat depot has a unique transcriptome and show different developmental pattern, which is not readily apparent macroscopically. These differences could contribute to the retention of UCP1 within the supraclavicular fat depot, the most active depot in adult humans, increasing heat production following a meal. Despite the rapid increase in publications on BAT over the past decade, the extent to which modifications in diet and/or environment can be utilised to promote its activity in the mother and/or her offspring remains to be established.


Asunto(s)
Tejido Adiposo Pardo/embriología , Tejido Adiposo Pardo/fisiología , Reproducción/fisiología , Tejido Adiposo Pardo/crecimiento & desarrollo , Animales , Peso Corporal/fisiología , Femenino , Humanos , Masculino , Obesidad/metabolismo , Obesidad/fisiopatología , Embarazo , Complicaciones del Embarazo/metabolismo , Complicaciones del Embarazo/fisiopatología , Efectos Tardíos de la Exposición Prenatal/metabolismo , Efectos Tardíos de la Exposición Prenatal/fisiopatología , Termogénesis/fisiología
7.
Endocrinology ; 159(5): 2050-2061, 2018 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-29579167

RESUMEN

Regulation of adipogenesis is of major interest given that adipose tissue expansion and dysfunction are central to metabolic syndrome. Glucocorticoids (GCs) are important for adipogenesis in vitro. However, establishing a role for GCs in adipogenesis in vivo has been difficult. GC receptor (GR)‒null mice die at birth, a time at which wild-type (WT) mice do not have fully developed white adipose depots. We conducted several studies aimed at defining the role of GC signaling in adipogenesis in vitro and in vivo. First, we showed that GR-null mouse embryonic fibroblasts (MEFs) have compromised ability to form adipocytes in vitro, a phenotype that can be partially rescued with a peroxisome proliferator-activated receptor γ agonist. Next, we demonstrated that MEFs are capable of forming de novo fat pads in mice despite the absence of GR or circulating GCs [by bilateral adrenalectomy (ADX)]. However, ADX and GR-null fat pads and their associated adipocyte areas were smaller than those in controls. Second, using adipocyte-specific luciferase reporter mice, we identified adipocytes in both WT and GR-null embryonic day (E)18 mouse embryos. Lastly, positive perilipin staining in WT and GR-null E18 embryos confirmed the presence of early white inguinal and brown adipocytes. Taken together, these results provide compelling evidence that GCs and GR augment but are not required for the development of functional adipose tissue in vivo.


Asunto(s)
Adipogénesis/genética , Tejido Adiposo Pardo/embriología , Tejido Adiposo Blanco/embriología , Fibroblastos/metabolismo , Glucocorticoides/metabolismo , Receptores de Glucocorticoides/genética , Adipocitos Marrones , Adipocitos Blancos , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Adrenalectomía , Animales , Técnicas In Vitro , Ratones , PPAR gamma/agonistas , Perilipina-1/metabolismo , Transducción de Señal
8.
Breast J ; 24(2): 199-202, 2018 03.
Artículo en Inglés | MEDLINE | ID: mdl-28786166

RESUMEN

This paper aims to review the concept of hibernomas, with focus on their occurrence, in the breast. It will make reference to a specific case from the Helen Joseph Hospital's Breast Clinic situated in Johannesburg, South Africa. We describe the clinical, radiological and pathological findings in a patient as well as the final diagnosis and treatment (in the form of surgery). This report emphasizes the distinguishable features of hibernomas, and gives guidance as to the surgical approach in large hibernomas stressing the ease of achieving cosmesis without major oncoplastic techniques. More specifically, discussion as whether these rapidly growing, nonrecurring, usually encapsulated growths consisting of brown fat tissue similar to that found in hibernating animals arise from the breast or the underlying muscle is considered.


Asunto(s)
Neoplasias de la Mama/diagnóstico , Lipoma/diagnóstico , Neoplasias de los Tejidos Blandos/diagnóstico , Tejido Adiposo Pardo/embriología , Tejido Adiposo Pardo/patología , Biopsia , Neoplasias de la Mama/embriología , Neoplasias de la Mama/patología , Neoplasias de la Mama/cirugía , Femenino , Humanos , Lipoma/embriología , Lipoma/patología , Lipoma/cirugía , Mamografía , Persona de Mediana Edad , Neoplasias de los Tejidos Blandos/embriología , Neoplasias de los Tejidos Blandos/patología , Neoplasias de los Tejidos Blandos/cirugía , Ultrasonografía
9.
Dev Cell ; 41(4): 382-391.e5, 2017 05 22.
Artículo en Inglés | MEDLINE | ID: mdl-28535373

RESUMEN

During mouse embryo development, both muscle progenitor cells (MPCs) and brown adipocytes (BAs) are known to derive from the same Pax7+/Myf5+ progenitor cells. However, the underlying mechanisms for the cell fate control remain unclear. In Pax7-null MPCs from young mice, several BA-specific genes, including Prdm16 and Ucp1 and many other adipocyte-related genes, were upregulated with a concomitant reduction of Myod and Myf5, two muscle lineage-determining genes. This suggests a cell fate switch from MPC to BA. Consistently, freshly isolated Pax7-null but not wild-type MPCs formed lipid-droplet-containing UCP1+ BA in culture. Mechanistically, MyoD and Myf5, both known transcription targets of Pax7 in MPC, potently repress Prdm16, a BA-specific lineage-determining gene, via the E2F4/p107/p130 transcription repressor complex. Importantly, inducible Pax7 ablation in developing mouse embryos promoted brown fat development. Thus, the MyoD/Myf5-E2F4/p107/p130 axis functions in both the Pax7+/Myf5+ embryonic progenitor cells and postnatal myoblasts to repress the alternative BA fate.


Asunto(s)
Adipocitos Marrones/citología , Adipocitos Marrones/metabolismo , Linaje de la Célula , Músculos/citología , Células Madre/citología , Células Madre/metabolismo , Tejido Adiposo Pardo/embriología , Tejido Adiposo Pardo/metabolismo , Animales , Línea Celular , Linaje de la Célula/genética , Células Cultivadas , Proteínas de Unión al ADN/metabolismo , Factor de Transcripción E2F4/metabolismo , Embrión de Mamíferos/metabolismo , Eliminación de Gen , Técnicas de Silenciamiento del Gen , Ratones , Proteína MioD/metabolismo , Factor 5 Regulador Miogénico/metabolismo , Factor de Transcripción PAX7/metabolismo , Proteínas Represoras/metabolismo , Factores de Transcripción/metabolismo , Transcripción Genética , Regulación hacia Arriba/genética
10.
Dev Cell ; 35(5): 568-583, 2015 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-26625958

RESUMEN

Progression from brown preadipocytes to adipocytes engages two transcriptional programs: the expression of adipogenic genes common to both brown fat (BAT) and white fat (WAT), and the expression of BAT-selective genes. However, the dynamics of chromatin states and epigenetic enzymes involved remain poorly understood. Here we show that BAT development is selectively marked and guided by repressive H3K27me3 and is executed by its demethylase Jmjd3. We find that a significant subset of BAT-selective genes, but not common fat genes or WAT-selective genes, are demarcated by H3K27me3 in both brown and white preadipocytes. Jmjd3-catalyzed removal of H3K27me3, in part through Rreb1-mediated recruitment, is required for expression of BAT-selective genes and for development of beige adipocytes both in vitro and in vivo. Moreover, gain- and loss-of-function Jmjd3 transgenic mice show age-dependent body weight reduction and cold intolerance, respectively. Together, we identify an epigenetic mechanism governing BAT fate determination and WAT plasticity.


Asunto(s)
Tejido Adiposo Pardo/embriología , Tejido Adiposo Blanco/embriología , Regulación del Desarrollo de la Expresión Génica , Histona Demetilasas con Dominio de Jumonji/metabolismo , Adipocitos/citología , Adipocitos/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Peso Corporal , Proteínas de Unión al ADN/metabolismo , Canales Iónicos/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Proteínas Mitocondriales/metabolismo , Mutación Puntual , Regiones Promotoras Genéticas , Análisis de Secuencia de ARN , Termogénesis/genética , Factores de Transcripción/metabolismo , Transgenes , Proteína Desacopladora 1
11.
Sci Rep ; 5: 14487, 2015 Sep 28.
Artículo en Inglés | MEDLINE | ID: mdl-26411793

RESUMEN

High-Mobility-Group-A1 (HMGA1) proteins are non-histone proteins that regulate chromatin structure and gene expression during embryogenesis, tumourigenesis and immune responses. In vitro studies suggest that HMGA1 proteins may be required to regulate adipogenesis. To examine the role of HMGA1 in vivo, we generated transgenic mice overexpressing HMGA1 in adipose tissues. HMGA1 transgenic mice showed a marked reduction in white and brown adipose tissue mass that was associated with downregulation of genes involved in adipogenesis and concomitant upregulation of preadipocyte markers. Reduced adipogenesis and decreased fat mass were not associated with altered glucose homeostasis since HMGA1 transgenic mice fed a regular-chow diet exhibited normal glucose tolerance and insulin sensitivity. However, when fed a high-fat diet, overexpression of HMGA1 resulted in decreased body-weight gain, reduced fat mass, but improved insulin sensitivity and glucose tolerance. Although HMGA1 transgenic mice exhibited impaired glucose uptake in adipose tissue due to impaired adipogenesis, the increased glucose uptake observed in skeletal muscle may account for the improved glucose homeostasis. Our results indicate that HMGA1 plays an important function in the regulation of white and brown adipogenesis in vivo and suggests that impaired adipocyte differentiation and decreased fat mass is not always associated with impaired whole-body glucose homeostasis.


Asunto(s)
Adipogénesis/genética , Tejido Adiposo/metabolismo , Expresión Génica , Proteínas HMGA/genética , Resistencia a la Insulina/genética , Obesidad/etiología , Tejido Adiposo/embriología , Tejido Adiposo Pardo/embriología , Tejido Adiposo Pardo/metabolismo , Adiposidad/genética , Animales , Dieta Alta en Grasa , Modelos Animales de Enfermedad , Glucosa/metabolismo , Prueba de Tolerancia a la Glucosa , Masculino , Ratones , Ratones Transgénicos , Obesidad/metabolismo , Especificidad de Órganos/genética
12.
Annu Rev Nutr ; 35: 295-320, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26076904

RESUMEN

There are three different types of adipose tissue (AT)-brown, white, and beige-that differ with stage of development, species, and anatomical location. Of these, brown AT (BAT) is the least abundant but has the greatest potential impact on energy balance. BAT is capable of rapidly producing large amounts of heat through activation of the unique uncoupling protein 1 (UCP1) located within the inner mitochondrial membrane. White AT is an endocrine organ and site of lipid storage, whereas beige AT is primarily white but contains some cells that possess UCP1. BAT first appears in the fetus around mid-gestation and is then gradually lost through childhood, adolescence, and adulthood. We focus on the interrelationships between adipocyte classification, anatomical location, and impact of diet in early life together with the extent to which fat development differs between the major species examined. Ultimately, novel dietary interventions designed to reactivate BAT could be possible.


Asunto(s)
Tejido Adiposo Pardo/crecimiento & desarrollo , Tejido Adiposo Pardo/fisiología , Adipocitos/clasificación , Adipocitos/fisiología , Tejido Adiposo/embriología , Tejido Adiposo/crecimiento & desarrollo , Tejido Adiposo Pardo/embriología , Tejido Adiposo Blanco/fisiología , Animales , Dieta , Metabolismo Energético/fisiología , Epigénesis Genética , Femenino , Desarrollo Fetal , Edad Gestacional , Humanos , Canales Iónicos/fisiología , Fenómenos Fisiologicos Nutricionales Maternos , Proteínas Mitocondriales/fisiología , Embarazo , Termogénesis/fisiología , Proteína Desacopladora 1
13.
Am J Physiol Endocrinol Metab ; 308(12): E1043-55, 2015 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-25898954

RESUMEN

Traditional therapies for type 1 diabetes (T1D) involve insulin replacement or islet/pancreas transplantation and have numerous limitations. Our previous work demonstrated the ability of embryonic brown adipose tissue (BAT) transplants to establish normoglycemia without insulin in chemically induced models of insulin-deficient diabetes. The current study sought to extend the technique to an autoimmune-mediated T1D model and document the underlying mechanisms. In nonobese diabetic (NOD) mice, BAT transplants result in complete reversal of T1D associated with rapid and long-lasting euglycemia. In addition, BAT transplants placed prior to the onset of diabetes on NOD mice can prevent or significantly delay the onset of diabetes. As with streptozotocin (STZ)-diabetic models, euglycemia is independent of insulin and strongly correlates with decrease of inflammation and increase of adipokines. Plasma insulin-like growth factor-I (IGF-I) is the first hormone to increase following BAT transplants. Adipose tissue of transplant recipients consistently express IGF-I compared with little or no expression in controls, and plasma IGF-I levels show a direct negative correlation with glucose, glucagon, and inflammatory cytokines. Adipogenic and anti-inflammatory properties of IGF-I may stimulate regeneration of new healthy white adipose tissue, which in turn secretes hypoglycemic adipokines that substitute for insulin. IGF-I can also directly decrease blood glucose through activating insulin receptor. These data demonstrate the potential for insulin-independent reversal of autoimmune-induced T1D with BAT transplants and implicate IGF-I as a likely mediator in the resulting equilibrium.


Asunto(s)
Tejido Adiposo Pardo/trasplante , Diabetes Mellitus Experimental/terapia , Diabetes Mellitus Tipo 1/terapia , Insulina/metabolismo , Tejido Adiposo Pardo/embriología , Animales , Glucemia/metabolismo , Embrión de Mamíferos , Femenino , Trasplante de Tejido Fetal , Resistencia a la Insulina , Factor I del Crecimiento Similar a la Insulina/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Embarazo
14.
Proc Natl Acad Sci U S A ; 112(16): 5069-74, 2015 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-25848030

RESUMEN

Although recent studies have shown that brown adipose tissue (BAT) arises from progenitor cells that also give rise to skeletal muscle, the developmental signals that control the formation of BAT remain largely unknown. Here, we show that brown preadipocytes possess primary cilia and can respond to Hedgehog (Hh) signaling. Furthermore, cell-autonomous activation of Hh signaling blocks early brown-preadipocyte differentiation, inhibits BAT formation in vivo, and results in replacement of neck BAT with poorly differentiated skeletal muscle. Finally, we show that Hh signaling inhibits BAT formation partially through up-regulation of chicken ovalbumin upstream promoter transcription factor II (COUP-TFII). Taken together, our studies uncover a previously unidentified role for Hh as an inhibitor of BAT development.


Asunto(s)
Tejido Adiposo Pardo/embriología , Tejido Adiposo Pardo/metabolismo , Proteínas Hedgehog/metabolismo , Transducción de Señal , Adipocitos Marrones/citología , Adipocitos Marrones/metabolismo , Tejido Adiposo Pardo/citología , Animales , Factor de Transcripción COUP II/metabolismo , Diferenciación Celular/genética , Cilios/metabolismo , Ratones , Transducción de Señal/genética , Regulación hacia Arriba/genética
15.
Biochim Biophys Acta ; 1851(5): 686-96, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25668679

RESUMEN

In this review we discuss the role of developmental transcription factors in adipose tissue biology with a focus on how these developmental genes may contribute to regional variation in adipose tissue distribution and function. Regional, depot-specific, differences in lipid handling and signalling (lipolysis, lipid storage and adipokine/lipokine signalling) are important determinants of metabolic health. At a cellular level, preadipocytes removed from their original depot and cultured in vitro retain depot-specific functional properties, implying that these are intrinsic to the cells and not a function of their environment in situ. High throughput screening has identified a number of developmental transcription factors involved in embryological development, including members of the Homeobox and T-Box gene families, that are strongly differentially expressed between regional white adipose tissue depots and also between brown and white adipose tissue. However, the significance of depot-specific developmental signatures remains unclear. Developmental transcription factors determine body patterning during embryogenesis. The divergent developmental origins of regional adipose tissue depots may explain their differing functional characteristics. There is evidence from human genetics that developmental genes determine adipose tissue distribution: in GWAS studies a number of developmental genes have been identified as being correlated with anthropometric measures of adiposity and fat distribution. Additionally, compelling functional studies have recently implicated developmental genes in both white adipogenesis and the so-called 'browning' of white adipose tissue. Understanding the genetic and developmental pathways in adipose tissue may help uncover novel ways to intervene with the function of adipose tissue in order to promote health.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Factores de Transcripción/metabolismo , Adipoquinas/metabolismo , Tejido Adiposo Pardo/embriología , Tejido Adiposo Pardo/crecimiento & desarrollo , Tejido Adiposo Blanco/embriología , Tejido Adiposo Blanco/crecimiento & desarrollo , Adiposidad , Animales , Metabolismo Energético , Regulación del Desarrollo de la Expresión Génica , Humanos , Lipogénesis , Lipólisis , Morfogénesis , Transducción de Señal , Factores de Transcripción/genética
16.
Proc Natl Acad Sci U S A ; 111(40): 14466-71, 2014 Oct 07.
Artículo en Inglés | MEDLINE | ID: mdl-25197048

RESUMEN

Brown adipocytes and muscle and dorsal dermis descend from precursor cells in the dermomyotome, but the factors that regulate commitment to the brown adipose lineage are unknown. Here, we prospectively isolated and determined the molecular profile of embryonic brown preadipose cells. Brown adipogenic precursor activity in embryos was confined to platelet-derived growth factor α(+), myogenic factor 5(Cre)-lineage-marked cells. RNA-sequence analysis identified early B-cell factor 2 (Ebf2) as one of the most selectively expressed genes in this cell fraction. Importantly, Ebf2-expressing cells purified from Ebf2(GFP) embryos or brown fat tissue did not express myoblast or dermal cell markers and uniformly differentiated into brown adipocytes. Interestingly, Ebf2-expressing cells from white fat tissue in adult animals differentiated into brown-like (or beige) adipocytes. Loss of Ebf2 in brown preadipose cells reduced the expression levels of brown preadipose-signature genes, whereas ectopic Ebf2 expression in myoblasts activated brown preadipose-specific genes. Altogether, these results indicate that Ebf2 specifically marks and regulates the molecular profile of brown preadipose cells.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo/metabolismo , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Adipocitos/citología , Adipogénesis/genética , Tejido Adiposo/citología , Tejido Adiposo/embriología , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/embriología , Tejido Adiposo Blanco/citología , Tejido Adiposo Blanco/embriología , Tejido Adiposo Blanco/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Biomarcadores/metabolismo , Linaje de la Célula/genética , Células Cultivadas , Embrión de Mamíferos/citología , Embrión de Mamíferos/embriología , Embrión de Mamíferos/metabolismo , Citometría de Flujo , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Inmunohistoquímica , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Factor 5 Regulador Miogénico/genética , Factor 5 Regulador Miogénico/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
17.
BMC Dev Biol ; 14: 24, 2014 May 28.
Artículo en Inglés | MEDLINE | ID: mdl-24886590

RESUMEN

BACKGROUND: Chloride Intracellular Channel 4 (CLIC4) is one of seven members in the closely related CLIC protein family. CLIC4 is involved in multiple cellular processes including apoptosis, cellular differentiation, inflammation and endothelial tubulogenesis. Despite over a decade of research, no comprehensive in situ expression analysis of CLIC4 in a living organism has been reported. In order to fulfill this goal, we generated a knock-in mouse to express Green Fluorescent Protein (GFP) from the CLIC4 locus, thus substituting the GFP coding region for CLIC4. We used GFP protein expression to eliminate cross reaction with other CLIC family members. RESULTS: We analyzed CLIC4 expression during embryonic development and adult organs. During mid and late gestation, CLIC4 expression is modulated particularly in fetal brain, heart, thymus, liver and kidney as well as in developing brown adipose tissue and stratifying epidermis. In the adult mouse, CLIC4 is highly expressed globally in vascular endothelial cells as well as in liver, lung alveolar septae, pancreatic acini, spermatogonia, renal proximal tubules, cardiomyocytes and thymic epithelial cells. Neural expression included axonal tracks, olfactory bulb, Purkinje cell layer and dentate gyrus. Renal CLIC4 expression was most pronounced in proximal tubules, although altered renal function was not detected in the absence of CLIC4. Myeloid cells and B cells of the spleen are rich in CLIC4 expression as are CD4 and CD8 positive T cells. CONCLUSIONS: In a comprehensive study detailing CLIC4 expression in situ in a mouse model that excludes cross reaction with other family members, we were able to document previously unreported expression for CLIC4 in developing fetus, particularly the brain. In addition, compartmentalized expression of CLIC4 in specific adult tissues and cells provides a focus to explore potential functions of this protein not addressed previously.


Asunto(s)
Canales de Cloruro/genética , Perfilación de la Expresión Génica , Regulación del Desarrollo de la Expresión Génica , Proteínas Fluorescentes Verdes/genética , Proteínas Mitocondriales/genética , Tejido Adiposo Pardo/embriología , Tejido Adiposo Pardo/crecimiento & desarrollo , Tejido Adiposo Pardo/metabolismo , Animales , Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Encéfalo/metabolismo , Canales de Cloruro/metabolismo , Epidermis/embriología , Epidermis/crecimiento & desarrollo , Epidermis/metabolismo , Corazón Fetal/embriología , Corazón Fetal/metabolismo , Proteínas Fluorescentes Verdes/metabolismo , Corazón/crecimiento & desarrollo , Immunoblotting , Inmunohistoquímica , Hibridación in Situ , Riñón/embriología , Riñón/crecimiento & desarrollo , Riñón/metabolismo , Hígado/embriología , Hígado/crecimiento & desarrollo , Hígado/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Microscopía Confocal , Proteínas Mitocondriales/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Timo/embriología , Timo/crecimiento & desarrollo , Timo/metabolismo
18.
Nature ; 510(7503): 76-83, 2014 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-24899307

RESUMEN

Our understanding of adipose tissue biology has progressed rapidly since the turn of the century. White adipose tissue has emerged as a key determinant of healthy metabolism and metabolic dysfunction. This realization is paralleled only by the confirmation that adult humans have heat-dissipating brown adipose tissue, an important contributor to energy balance and a possible therapeutic target for the treatment of metabolic disease. We propose that the development of successful strategies to target brown and white adipose tissues will depend on investigations that elucidate their developmental origins and cell-type-specific functional regulators.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Adipocitos/citología , Adipocitos/metabolismo , Adipocitos/patología , Adipogénesis , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/embriología , Tejido Adiposo Pardo/inervación , Tejido Adiposo Pardo/patología , Tejido Adiposo Blanco/citología , Tejido Adiposo Blanco/embriología , Tejido Adiposo Blanco/inervación , Tejido Adiposo Blanco/patología , Animales , Humanos , Lipólisis , Obesidad/metabolismo , Obesidad/patología , Sistema Nervioso Simpático/metabolismo , Termogénesis
19.
Nat Cell Biol ; 16(4): 367-75, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24609269

RESUMEN

Fuelled by the obesity epidemic, there is considerable interest in the developmental origins of white adipose tissue (WAT) and the stem and progenitor cells from which it arises. Whereas increased visceral fat mass is associated with metabolic dysfunction, increased subcutaneous WAT is protective. There are six visceral fat depots: perirenal, gonadal, epicardial, retroperitoneal, omental and mesenteric, and it is a subject of much debate whether these have a common developmental origin and whether this differs from that for subcutaneous WAT. Here we show that all six visceral WAT depots receive a significant contribution from cells expressing Wt1 late in gestation. Conversely, no subcutaneous WAT or brown adipose tissue arises from Wt1-expressing cells. Postnatally, a subset of visceral WAT continues to arise from Wt1-expressing cells, consistent with the finding that Wt1 marks a proportion of cell populations enriched in WAT progenitors. We show that all visceral fat depots have a mesothelial layer like the visceral organs with which they are associated, and provide several lines of evidence that Wt1-expressing mesothelium can produce adipocytes. These results reveal a major ontogenetic difference between visceral and subcutaneous WAT, and pinpoint the lateral plate mesoderm as a major source of visceral WAT. They also support the notion that visceral WAT progenitors are heterogeneous, and suggest that mesothelium is a source of adipocytes.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Proteínas WT1/metabolismo , Adipocitos/citología , Tejido Adiposo Pardo/citología , Tejido Adiposo Pardo/embriología , Tejido Adiposo Blanco/citología , Tejido Adiposo Blanco/embriología , Animales , Antineoplásicos Hormonales/farmacología , Linaje de la Célula/genética , Técnicas de Sustitución del Gen , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Mesodermo/citología , Mesodermo/metabolismo , Ratones , Tamoxifeno/farmacología , Proteínas WT1/genética
20.
J Clin Endocrinol Metab ; 99(1): 151-9, 2014 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-24217905

RESUMEN

CONTEXT: Brown adipose tissue (BAT) has the unique ability of generating heat due to the expression of mitochondrial uncoupling protein 1 (UCP1). A recent discovery regarding functional BAT in adult humans has increased interest in the molecular pathways of BAT development and functionality. An important role for estrogen in white adipose tissue was shown, but the possible role of estrogen in human fetal BAT (fBAT) is unclear. OBJECTIVE: The objective of this study was to determine whether human fBAT expresses estrogen receptor α (ERα) and ERß. In addition, we examined their localization as well as their correlation with crucial proteins involved in BAT differentiation, proliferation, mitochondriogenesis and thermogenesis including peroxisome proliferator-activated receptor γ (PPARγ), proliferating cell nuclear antigen (PCNA), PPARγ-coactivator-1α (PGC-1α), and UCP1. DESIGN: The fBAT was obtained from 4 human male fetuses aged 15, 17, 20, and 23 weeks gestation. ERα and ERß expression was assessed using Western blotting, immunohistochemistry, and immunocytochemistry. Possible correlations with PPARγ, PCNA, PGC-1α, and UCP1 were examined by double immunofluorescence. RESULTS: Both ERα and ERß were expressed in human fBAT, with ERα being dominant. Unlike ERß, which was present only in mature brown adipocytes, we detected ERα in mature adipocytes, preadipocytes, mesenchymal and endothelial cells. In addition, double immunofluorescence supported the notion that differentiation in fBAT probably involves ERα. Immunocytochemical analysis revealed mitochondrial localization of both receptors. CONCLUSION: The expression of both ERα and ERß in human fBAT suggests a role for estrogen in its development, primarily via ERα. In addition, our results indicate that fBAT mitochondria could be targeted by estrogens and pointed out the possible role of both ERs in mitochondriogenesis.


Asunto(s)
Tejido Adiposo Pardo/metabolismo , Receptor alfa de Estrógeno/metabolismo , Receptor beta de Estrógeno/metabolismo , Feto/metabolismo , Tejido Adiposo Pardo/embriología , Tejido Adiposo Pardo/crecimiento & desarrollo , Edad Gestacional , Humanos , Inmunohistoquímica , Canales Iónicos/metabolismo , Masculino , Proteínas Mitocondriales/metabolismo , PPAR gamma/metabolismo , Coactivador 1-alfa del Receptor Activado por Proliferadores de Peroxisomas gamma , Antígeno Nuclear de Célula en Proliferación/metabolismo , Distribución Tisular , Factores de Transcripción/metabolismo , Proteína Desacopladora 1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...