Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 10 de 10
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Nat Commun ; 9(1): 3592, 2018 09 04.
Artículo en Inglés | MEDLINE | ID: mdl-30181538

RESUMEN

Adipocytes undergo pronounced changes in size and behavior to support diverse tissue functions, but the mechanisms that control these changes are not well understood. Mammary gland-associated white adipose tissue (mgWAT) regresses in support of milk fat production during lactation and expands during the subsequent involution of milk-producing epithelial cells, providing one of the most marked physiological examples of adipose growth. We examined cellular mechanisms and functional implications of adipocyte and lipid dynamics in the mouse mammary gland (MG). Using in vivo analysis of adipocyte precursors and genetic tracing of mature adipocytes, we find mature adipocyte hypertrophy to be a primary mechanism of mgWAT expansion during involution. Lipid tracking and lipidomics demonstrate that adipocytes fill with epithelial-derived milk lipid. Furthermore, ablation of mgWAT during involution reveals an essential role for adipocytes in milk trafficking from, and proper restructuring of, the mammary epithelium. This work advances our understanding of MG remodeling and tissue-specific roles for adipocytes.


Asunto(s)
Adipocitos/citología , Metabolismo de los Lípidos , Glándulas Mamarias Animales/citología , Glándulas Mamarias Humanas/citología , Adipocitos/metabolismo , Adipocitos Blancos/citología , Adipocitos Blancos/fisiología , Animales , Lactancia Materna , Tamaño de la Célula , Células Epiteliales/citología , Células Epiteliales/fisiología , Ácidos Grasos/metabolismo , Femenino , Humanos , Lactancia/fisiología , Glándulas Mamarias Animales/fisiología , Glándulas Mamarias Humanas/fisiología , Ratones Endogámicos C57BL , Ratones Transgénicos , Embarazo
2.
Development ; 145(17)2018 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-30045918

RESUMEN

The embryonic origin of distinct fat depots and the role for ontogeny in specifying the functional differences among adipocyte lineages between and within depots is unclear. Using a Cre/Lox-based strategy to track the fate of major mesodermal subcompartments in mice we present evidence that <50% of interscapular brown adipocytes are derived from progenitors of the central dermomyotome. Furthermore, we demonstrate that depot-specific adipocyte lineages spatially diverge as early as gastrulation, and that perigonadal adipocytes arise from separate mesodermal subcompartments in males and females. Last, we show adipocyte precursors (APs) of distinct lineages within the same depot exhibit indistinguishable responses to a high fat diet, indicating that ontogenetic differences between APs do not necessarily correspond to functional differences in this context. Altogether, these findings shed light on adipose tissue patterning and suggest that the behavior of adipocyte lineage cells is not strictly determined by developmental history.


Asunto(s)
Adipocitos/metabolismo , Tejido Adiposo/embriología , Linaje de la Célula/fisiología , Embrión de Mamíferos/embriología , Mesodermo/embriología , Células Madre/metabolismo , Adipocitos/citología , Tejido Adiposo/citología , Animales , Embrión de Mamíferos/citología , Mesodermo/citología , Ratones , Ratones Transgénicos , Células Madre/citología
3.
Adipocyte ; 6(3): 193-204, 2017 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-28872979

RESUMEN

Adipocytes were identified in human bone marrow more than a century ago, yet until recently little has been known about their origin, development, function or interactions with other cells in the bone marrow. Little functional significance has been attributed to these cells, a paradigm that still persists today. However, we now know that marrow adipose tissue increases with age and in response to a variety of physiologic induction signals. Bone marrow adipocytes have recently been shown to influence other cell populations within the marrow and can affect whole body metabolism by the secretion of a defined set of adipokines. Recent research shows that marrow adipocytes are distinct from white, brown and beige adipocytes, indicating that the bone marrow is a distinct adipose depot. This review will highlight recent data regarding these areas and the interactions of marrow adipose tissue (MAT) with cells within and outside of the bone marrow.


Asunto(s)
Adipocitos/fisiología , Tejido Adiposo/fisiología , Células de la Médula Ósea/citología , Adipocitos/metabolismo , Adipocitos Marrones/metabolismo , Adipocitos Blancos/metabolismo , Adipogénesis/fisiología , Adipoquinas/fisiología , Tejido Adiposo Pardo/metabolismo , Tejido Adiposo Blanco/metabolismo , Animales , Médula Ósea/metabolismo , Médula Ósea/fisiología , Células de la Médula Ósea/fisiología , Diferenciación Celular/fisiología , Humanos , Ratones , Termogénesis
4.
Adipocyte ; 6(3): 224-233, 2017 07 03.
Artículo en Inglés | MEDLINE | ID: mdl-28792785

RESUMEN

Over the past 2 decades, the incidence of childhood obesity has risen dramatically. This recent rise in childhood obesity is particularly concerning as adults who were obese during childhood develop type II diabetes that is intractable to current forms of treatment compared with individuals who develop obesity in adulthood. While the mechanisms responsible for the exacerbated diabetic phenotype associated with childhood obesity is not clear, it is well known that childhood is an important time period for the establishment of normal white adipose tissue in humans. This association suggests that exposure to obesogenic stimuli during adipose development may have detrimental effects on adipose function and metabolic homeostasis. In this study, we identify the period of development associated with puberty, postnatal days 18-34, as critical for the establishment of normal adipose mass in mice. Exposure of mice to high fat diet only during this time period results in metabolic dysfunction, increased leptin expression, and increased adipocyte size in adulthood in the absence of sustained increased fat mass or body weight. These findings indicate that exposure to obesogenic stimuli during critical developmental periods have prolonged effects on adipose tissue function that may contribute to the exacerbated metabolic dysfunctions associated with childhood obesity.


Asunto(s)
Tejido Adiposo Blanco/metabolismo , Pubertad/fisiología , Adipocitos/metabolismo , Tejido Adiposo/metabolismo , Tejido Adiposo Blanco/fisiología , Adiposidad/fisiología , Animales , Diabetes Mellitus Tipo 2/etiología , Diabetes Mellitus Tipo 2/metabolismo , Dieta Alta en Grasa/efectos adversos , Femenino , Homeostasis/fisiología , Humanos , Leptina/metabolismo , Masculino , Ratones , Obesidad/metabolismo , Pubertad/metabolismo
5.
Cell Stem Cell ; 19(6): 738-751, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27746098

RESUMEN

Tissue growth and maintenance requires stem cell populations that self-renew, proliferate, and differentiate. Maintenance of white adipose tissue (WAT) requires the proliferation and differentiation of adipocyte stem cells (ASCs) to form postmitotic, lipid-filled mature adipocytes. Here we use the dynamic adipogenic program that occurs during hair growth to uncover an unrecognized regulator of ASC self-renewal and proliferation, PDGFA, which activates AKT signaling to drive and maintain the adipogenic program in the skin. Pdgfa expression is reduced in aged ASCs and is required for ASC proliferation and maintenance in the dermis, but not in other WATs. Our molecular and genetic studies uncover PI3K/AKT2 as a direct PDGFA target that is activated in ASCs during WAT hyperplasia and is functionally required for dermal ASC proliferation. Our data therefore reveal active mechanisms that regulate ASC self-renewal in the skin and show that distinct regulatory mechanisms operate in different WAT depots.


Asunto(s)
Adipocitos/citología , Adipocitos/enzimología , Autorrenovación de las Células , Factor de Crecimiento Derivado de Plaquetas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Piel/citología , Células Madre/citología , Adipogénesis , Animales , Antígeno CD24/metabolismo , Proliferación Celular , Dermis/metabolismo , Perfilación de la Expresión Génica , Hiperplasia , Ratones Endogámicos C57BL , Modelos Biológicos , Fosfatidilinositol 3-Quinasas/metabolismo , Receptor alfa de Factor de Crecimiento Derivado de Plaquetas/metabolismo
6.
Cell Metab ; 24(1): 142-50, 2016 07 12.
Artículo en Inglés | MEDLINE | ID: mdl-27320063

RESUMEN

The sexually dimorphic distribution of adipose tissue influences the development of obesity-associated pathologies. The accumulation of visceral white adipose tissue (VWAT) that occurs in males is detrimental to metabolic health, while accumulation of subcutaneous adipose tissue (SWAT) seen in females may be protective. Here, we show that adipocyte hyperplasia contributes directly to the differential fat distribution between the sexes. In male mice, high-fat diet (HFD) induces adipogenesis specifically in VWAT, while in females HFD induces adipogenesis in both VWAT and SWAT in a sex hormone-dependent manner. We also show that the activation of adipocyte precursors (APs), which drives adipocyte hyperplasia in obesity, is regulated by the adipose depot microenvironment and not by cell-intrinsic mechanisms. These findings indicate that APs are plastic cells, which respond to both local and systemic signals that influence their differentiation potential independent of depot origin. Therefore, depot-specific AP niches coordinate adipose tissue growth and distribution.


Asunto(s)
Adipogénesis , Tejido Adiposo/fisiología , Obesidad/fisiopatología , Adipocitos/metabolismo , Adipocitos/patología , Tejido Adiposo Blanco/crecimiento & desarrollo , Tejido Adiposo Blanco/metabolismo , Animales , Dieta Alta en Grasa , Femenino , Hiperplasia , Masculino , Ratones , Obesidad/patología , Tamaño de los Órganos , Caracteres Sexuales
7.
Mol Cell Biol ; 36(7): 1180-93, 2016 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-26830228

RESUMEN

White adipose tissue (WAT) is essential for maintaining metabolic function, especially during obesity. The intronic microRNAs miR-33a and miR-33b, located within the genes encoding sterol regulatory element-binding protein 2 (SREBP-2) and SREBP-1, respectively, are transcribed in concert with their host genes and function alongside them to regulate cholesterol, fatty acid, and glucose metabolism. SREBP-1 is highly expressed in mature WAT and plays a critical role in promoting in vitro adipocyte differentiation. It is unknown whether miR-33b is induced during or involved in adipogenesis. This is in part due to loss of miR-33b in rodents, precluding in vivo assessment of the impact of miR-33b using standard mouse models. This work demonstrates that miR-33b is highly induced upon differentiation of human preadipocytes, along with SREBP-1. We further report that miR-33b is an important regulator of adipogenesis, as inhibition of miR-33b enhanced lipid droplet accumulation. Conversely, overexpression of miR-33b impaired preadipocyte proliferation and reduced lipid droplet formation and the induction of peroxisome proliferator-activated receptor γ (PPARγ) target genes during differentiation. These effects may be mediated by targeting of HMGA2, cyclin-dependent kinase 6 (CDK6), and other predicted miR-33b targets. Together, these findings demonstrate a novel role of miR-33b in the regulation of adipocyte differentiation, with important implications for the development of obesity and metabolic disease.


Asunto(s)
Adipogénesis/genética , Tejido Adiposo Blanco/citología , MicroARNs/genética , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Sitios Genéticos , Humanos , MicroARNs/biosíntesis , MicroARNs/fisiología , Células Madre/citología
8.
Nat Cell Biol ; 17(4): 376-85, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25730471

RESUMEN

Excessive accumulation of white adipose tissue (WAT) is the defining characteristic of obesity. WAT mass is composed primarily of mature adipocytes, which are generated through the proliferation and differentiation of adipocyte precursors (APs). Although the production of new adipocytes contributes to WAT growth in obesity, little is known about the cellular and molecular mechanisms underlying adipogenesis in vivo. Here, we show that high-fat diet feeding in mice rapidly and transiently induces proliferation of APs within WAT to produce new adipocytes. Importantly, the activation of adipogenesis is specific to the perigonadal visceral depot in male mice, consistent with the patterns of obesogenic WAT growth observed in humans. Furthermore, we find that in multiple models of obesity, the activation of APs is dependent on the phosphoinositide 3-kinase (PI3K)-AKT2 pathway; however, the development of WAT does not require AKT2. These data indicate that developmental and obesogenic adipogenesis are regulated through distinct molecular mechanisms.


Asunto(s)
Adipocitos Blancos/citología , Adipogénesis/fisiología , Obesidad/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Proto-Oncogénicas c-akt/metabolismo , Adipocitos Blancos/metabolismo , Adipogénesis/efectos de los fármacos , Adipogénesis/genética , Tejido Adiposo Blanco , Androstadienos/farmacología , Animales , Proliferación Celular/efectos de los fármacos , Proliferación Celular/genética , Dieta Alta en Grasa , Ingestión de Alimentos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Inhibidores de las Quinasa Fosfoinosítidos-3 , Proteínas Proto-Oncogénicas c-akt/genética , Distribución Aleatoria , Tamoxifeno/farmacología , Wortmanina
9.
Curr Biol ; 24(19): 2238-46, 2014 Oct 06.
Artículo en Inglés | MEDLINE | ID: mdl-25242029

RESUMEN

BACKGROUND: Dietary restriction (DR) has been shown to prolong longevity across diverse taxa, yet the mechanistic relationship between DR and longevity remains unclear. MicroRNAs (miRNAs) control aging-related functions such as metabolism and lifespan through regulation of genes in insulin signaling, mitochondrial respiration, and protein homeostasis. RESULTS: We have conducted a network analysis of aging-associated miRNAs connected to transcription factors PHA-4/FOXA and SKN-1/Nrf, which are both necessary for DR-induced lifespan extension in Caenorhabditis elegans. Our network analysis has revealed extensive regulatory interactions between PHA-4, SKN-1, and miRNAs and points to two aging-associated miRNAs, miR-71 and miR-228, as key nodes of this network. We show that miR-71 and miR-228 are critical for the response to DR in C. elegans. DR induces the expression of miR-71 and miR-228, and the regulation of these miRNAs depends on PHA-4 and SKN-1. In turn, we show that PHA-4 and SKN-1 are negatively regulated by miR-228, whereas miR-71 represses PHA-4. CONCLUSIONS: Based on our findings, we have discovered new links in an important pathway connecting DR to aging. By interacting with PHA-4 and SKN-1, miRNAs transduce the effect of dietary-restriction-mediated lifespan extension in C. elegans. Given the conservation of miRNAs, PHA-4, and SKN-1 across phylogeny, these interactions are likely to be conserved in more-complex species.


Asunto(s)
Envejecimiento , Proteínas de Caenorhabditis elegans/genética , Caenorhabditis elegans/fisiología , Proteínas de Unión al ADN/genética , MicroARNs/genética , Transactivadores/genética , Factores de Transcripción/genética , Animales , Caenorhabditis elegans/genética , Proteínas de Caenorhabditis elegans/metabolismo , Proteínas de Unión al ADN/metabolismo , MicroARNs/metabolismo , Reacción en Cadena de la Polimerasa , Transactivadores/metabolismo , Factores de Transcripción/metabolismo
10.
BMC Dev Biol ; 10: 104, 2010 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-20950450

RESUMEN

BACKGROUND: Vertebrate limb development involves a reciprocal feedback loop between limb mesenchyme and the overlying apical ectodermal ridge (AER). Several gene pathways participate in this feedback loop, including Fgf signaling. In the forelimb lateral plate mesenchyme, Tbx5 activates Fgf10 expression, which in turn initiates and maintains the mesenchyme/AER Fgf signaling loop. Recent findings have revealed that Tbx5 transcriptional activity is regulated by dynamic nucleocytoplasmic shuttling and interaction with Pdlim7, a PDZ-LIM protein family member, along actin filaments. This Tbx5 regulation is critical in heart formation, but the coexpression of both proteins in other developing tissues suggests a broader functional role. RESULTS: Knock-down of Pdlim7 function leads to decreased pectoral fin cell proliferation resulting in a severely stunted fin phenotype. While early gene induction and patterning in the presumptive fin field appear normal, the pectoral fin precursor cells display compaction and migration defects between 18 and 24 hours post-fertilization (hpf). During fin growth fgf24 is sequentially expressed in the mesenchyme and then in the apical ectodermal ridge (AER). However, in pdlim7 antisense morpholino-treated embryos this switch of expression is prevented and fgf24 remains ectopically active in the mesenchymal cells. Along with the lack of fgf24 in the AER, other critical factors including fgf8 are reduced, suggesting signaling problems to the underlying mesenchyme. As a consequence of perturbed AER function in the absence of Pdlim7, pathway components in the fin mesenchyme are misregulated or absent, indicating a breakdown of the Fgf signaling feedback loop, which is ultimately responsible for the loss of fin outgrowth. CONCLUSION: This work provides the first evidence for the involvement of Pdlim7 in pectoral fin development. Proper fin outgrowth requires fgf24 downregulation in the fin mesenchyme with subsequent activation in the AER, and Pdlim7 appears to regulate this transition, potentially through Tbx5 regulation. By controlling Tbx5 subcellular localization and transcriptional activity and possibly additional yet unknown means, Pdlim7 is required for proper development of the heart and the fins. These new regulatory mechanisms may have important implications how we interpret Tbx5 function in congenital hand/heart syndromes in humans.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Aletas de Animales/embriología , Epidermis/metabolismo , Factores de Crecimiento de Fibroblastos/metabolismo , Mesodermo/metabolismo , Proteínas de Pez Cebra/metabolismo , Pez Cebra/embriología , Pez Cebra/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Animales , Movimiento Celular , Proliferación Celular , Epidermis/anatomía & histología , Retroalimentación Fisiológica , Regulación del Desarrollo de la Expresión Génica , Humanos , Mesodermo/anatomía & histología , Morfogénesis , Oligonucleótidos Antisentido/genética , Oligonucleótidos Antisentido/metabolismo , Transducción de Señal , Proteínas de Dominio T Box/genética , Proteínas de Dominio T Box/metabolismo , Pez Cebra/anatomía & histología , Proteínas de Pez Cebra/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...