Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Cell Metab ; 35(10): 1679-1680, 2023 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-37793344

RESUMEN

Male and female mice display highly divergent responses to cold-induced thermogenic beiging of subcutaneous adipose tissues. Recently in Nature, Patel et al. showed that mammary duct epithelial cells respond to cold-induced sympathetic activity, triggering the secretion of lipocalin 2 (LCN2) to inhibit thermogenic differentiation of adjacent mammary adipocytes.


Asunto(s)
Adipocitos , Termogénesis , Masculino , Femenino , Ratones , Animales , Diferenciación Celular , Termogénesis/fisiología , Tejido Adiposo Pardo , Tejido Adiposo Blanco
2.
Cell Rep ; 39(11): 110942, 2022 06 14.
Artículo en Inglés | MEDLINE | ID: mdl-35705048

RESUMEN

Age-related macular degeneration (AMD), the leading cause of irreversible blindness among Americans over 50, is characterized by dysfunction and death of retinal pigment epithelial (RPE) cells. The RPE accumulates iron in AMD, and iron overload triggers RPE cell death in vitro and in vivo. However, the mechanism of RPE iron accumulation in AMD is unknown. We show that high-fat-diet-induced obesity, a risk factor for AMD, drives systemic and local inflammatory circuits upregulating interleukin-1ß (IL-1ß). IL-1ß upregulates RPE iron importers and downregulates iron exporters, causing iron accumulation, oxidative stress, and dysfunction. We term this maladaptive, chronic activation of a nutritional immunity pathway the cellular iron sequestration response (CISR). RNA sequencing (RNA-seq) analysis of choroid and retina from human donors revealed that hallmarks of this pathway are present in AMD microglia and macrophages. Together, these data suggest that inflamed adipose tissue, through the CISR, can lead to RPE pathology in AMD.


Asunto(s)
Degeneración Macular , Epitelio Pigmentado de la Retina , Tejido Adiposo/metabolismo , Humanos , Hierro/metabolismo , Degeneración Macular/metabolismo , Estrés Oxidativo , Retina/metabolismo , Epitelio Pigmentado de la Retina/metabolismo
3.
Mol Metab ; 54: 101357, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34662714

RESUMEN

OBJECTIVE: The capacity to generate new adipocytes from precursor cells is critical for maintaining metabolic health. Adipocyte precursor cells (APCs) constitute a heterogenous collection of cell types; however, the contribution of these various cell types to adipose tissue expansion in vivo remains unknown. The aim of the current study is to investigate the contribution of Dpp4+ progenitors to de novo adipogenesis. METHODS: Single cell analysis has identified several transcriptionally distinct subpopulations of APCs, including Dpp4+ progenitor cells concentrated in the connective tissue surrounding many organs, including white adipose tissue (WAT). Here, we generated a Dpp4CreER mouse model for in vivo lineage tracing of these cells and their downstream progeny in the setting of basal or high fat diet (HFD)-stimulated adipogenesis. RESULTS: Dpp4CreER mice enabled specific temporal labeling of Dpp4+ progenitor cells within their native connective tissue niche. Following a dietary chase period consisting of chow or HFD feeding for 18 weeks, Dpp4+ progenitors differentiated into mature adipocytes within the gonadal and subcutaneous WAT. HFD stimulated adipogenic contribution from Dpp4+ cells in the gonadal but not the subcutaneous depot. Flow cytometry analysis revealed that Dpp4+ progenitors give rise to DPP4(-)/ICAM1+ preadipocytes in vivo. HFD feeding did not perturb the flux of Dpp4+ cell conversion into ICAM1+ preadipocytes in gonadal WAT. Conversely, in subcutaneous WAT, HFD feeding/obesity led to an accumulation of ICAM1+ preadipocytes without a corresponding increase in mature adipocyte differentiation. Examination of non-classical murine visceral depots with relevance to humans, including omentum and retroperitoneal WAT, revealed robust contribution of Dpp4+ progenitors to de novo adipogenesis, which was further stimulated by HFD. CONCLUSION: Our data demonstrate that Dpp4+ interstitial progenitor cells contribute to basal adipogenesis in all fat depots and are recruited to support de novo adipogenic expansion of visceral WAT in the setting of HFD-induced obesity.


Asunto(s)
Adipogénesis/efectos de los fármacos , Dieta Alta en Grasa/efectos adversos , Dipeptidil Peptidasa 4/metabolismo , Intestinos/metabolismo , Células Madre/metabolismo , Animales , Ratones , Ratones Endogámicos
4.
Cell Stem Cell ; 26(6): 801-803, 2020 06 04.
Artículo en Inglés | MEDLINE | ID: mdl-32502400

RESUMEN

In this issue of Cell Stem Cell, Shook et al. (2020) reveal that dermal adipocytes regulate skin wound repair via release of fatty acids that promote macrophage recruitment and accelerated revascularization. Furthermore, mature dermal adipocytes dedifferentiate into migratory extracellularmatrix-producing myofibroblasts.


Asunto(s)
Lipólisis , Miofibroblastos , Adipocitos , Piel , Cicatrización de Heridas
5.
Cell Signal ; 72: 109634, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32283256

RESUMEN

The polycystin-1 and 2 proteins, encoded by the genes mutated in Autosomal Dominant Polycystic Kidney Disease, are connected to a large number of biological pathways. While the nature of these connections and their relevance to the primary functions of the polycystin proteins have yet to be fully elucidated, it is clear that many of them are mediated by or depend upon cleavage of the polycystin-1 protein. Cleavage of polycystin-1 at its G protein coupled receptor proteolytic site is an obligate step in the protein's maturation and in aspects of its trafficking. This cleavage may also serve to prime polycystin-1 to play a role as a non-canonical G protein coupled receptor. Cleavage of the cytoplasmic polycystin-1C terminal tail releases fragments that are able to enter the nucleus and the mitochondria and to influence their activities. Understanding the nature of these cleavages, their regulation and their consequences is likely to provide valuable insights into both the physiological functions served by the polycystin proteins and the pathological consequences of their absence.


Asunto(s)
Transducción de Señal , Canales Catiónicos TRPP/metabolismo , Animales , Adhesión Celular , Humanos , Osteogénesis , Transporte de Proteínas , Proteolisis , Canales Catiónicos TRPP/química
6.
Science ; 364(6438)2019 04 26.
Artículo en Inglés | MEDLINE | ID: mdl-31023895

RESUMEN

Metabolic health depends on the capacity of adipose tissue progenitor cells to undergo de novo adipogenesis. The cellular hierarchy and mechanisms governing adipocyte progenitor differentiation are incompletely understood. Through single-cell RNA sequence analyses, we show that the lineage hierarchy of adipocyte progenitors consists of distinct mesenchymal cell types that are present in both mouse and human adipose tissues. Cells marked by dipeptidyl peptidase-4 (DPP4)/CD26 expression are highly proliferative, multipotent progenitors. During the development of subcutaneous adipose tissue in mice, these progenitor cells give rise to intercellular adhesion molecule-1 (ICAM1)/CD54-expressing (CD54+) committed preadipocytes and a related adipogenic cell population marked by Clec11a and F3/CD142 expression. Transforming growth factor-ß maintains DPP4+ cell identity and inhibits adipogenic commitment of DPP4+ and CD142+ cells. Notably, DPP4+ progenitors reside in the reticular interstitium, a recently appreciated fluid-filled space within and between tissues, including adipose depots.


Asunto(s)
Adipocitos/citología , Adipogénesis , Tejido Adiposo/citología , Células Madre Mesenquimatosas/citología , Adipocitos/enzimología , Animales , Dipeptidil Peptidasa 4/metabolismo , Factores de Crecimiento de Célula Hematopoyética/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Lectinas Tipo C/metabolismo , Células Madre Mesenquimatosas/enzimología , Ratones , Análisis de Secuencia de ARN , Análisis de la Célula Individual , Tromboplastina/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
7.
Hum Mol Genet ; 28(1): 16-30, 2019 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-30215740

RESUMEN

Polycystin-1 (PC1), encoded by the PKD1 gene that is mutated in the autosomal dominant polycystic kidney disease, regulates a number of processes including bone development. Activity of the transcription factor RunX2, which controls osteoblast differentiation, is reduced in Pkd1 mutant mice but the mechanism governing PC1 activation of RunX2 is unclear. PC1 undergoes regulated cleavage that releases its C-terminal tail (CTT), which translocates to the nucleus to modulate transcriptional pathways involved in proliferation and apoptosis. We find that the cleaved CTT of PC1 (PC1-CTT) stimulates the transcriptional coactivator TAZ (Wwtr1), an essential coactivator of RunX2. PC1-CTT physically interacts with TAZ, stimulating RunX2 transcriptional activity in pre-osteoblast cells in a TAZ-dependent manner. The PC1-CTT increases the interaction between TAZ and RunX2 and enhances the recruitment of the p300 transcriptional co-regulatory protein to the TAZ/RunX2/PC1-CTT complex. Zebrafish injected with morpholinos directed against pkd1 manifest severe bone calcification defects and a curly tail phenotype. Injection of messenger RNA (mRNA) encoding the PC1-CTT into pkd1-morphant fish restores bone mineralization and reduces the severity of the curly tail phenotype. These effects are abolished by co-injection of morpholinos directed against TAZ. Injection of mRNA encoding a dominant-active TAZ construct is sufficient to rescue both the curly tail phenotype and the skeletal defects observed in pkd1-morpholino treated fish. Thus, TAZ constitutes a key mechanistic link through which PC1 mediates its physiological functions.


Asunto(s)
Desarrollo Óseo/genética , Péptidos y Proteínas de Señalización Intracelular/fisiología , Canales Catiónicos TRPP/fisiología , Animales , Apoptosis , Desarrollo Óseo/fisiología , Diferenciación Celular , Proteína p300 Asociada a E1A/fisiología , Regulación de la Expresión Génica , Genes Reguladores , Células HEK293 , Humanos , Riñón/metabolismo , Modelos Animales , Morfolinos , Osteoblastos/metabolismo , Osteogénesis/fisiología , Riñón Poliquístico Autosómico Dominante/genética , Canales Catiónicos TRPP/genética , Transactivadores , Factores de Transcripción , Proteínas Coactivadoras Transcripcionales con Motivo de Unión a PDZ , Pez Cebra/genética , Proteínas de Pez Cebra/genética
8.
Pediatr Nephrol ; 29(4): 505-11, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23824180

RESUMEN

Autosomal dominant polycystic kidney disease (ADPKD) is the most common genetic cause of end-stage renal disease, affecting approximately 1 in 1,000 people. The disease is characterized by the development of numerous large fluid-filled renal cysts over the course of decades. These cysts compress the surrounding renal parenchyma and impair its function. Mutations in two genes are responsible for ADPKD. The protein products of both of these genes, polycystin-1 and polycystin-2, localize to the primary cilium and participate in a wide variety of signaling pathways. Polycystin-1 undergoes several proteolytic cleavages that produce fragments which manifest biological activities. Recent results suggest that the production of polycystin-1 cleavage fragments is necessary and sufficient to account for at least some, although certainly not all, of the physiological functions of the parent protein.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Riñón Poliquístico Autosómico Dominante/metabolismo , Canales Catiónicos TRPP/metabolismo , Animales , Humanos , Mutación , Riñón Poliquístico Autosómico Dominante/genética , Canales Catiónicos TRPP/genética , Transcripción Genética
9.
Dev Cell ; 22(1): 197-210, 2012 Jan 17.
Artículo en Inglés | MEDLINE | ID: mdl-22178500

RESUMEN

Mutations in Pkd1, encoding polycystin-1 (PC1), cause autosomal-dominant polycystic kidney disease (ADPKD). We show that the carboxy-terminal tail (CTT) of PC1 is released by γ-secretase-mediated cleavage and regulates the Wnt and CHOP pathways by binding the transcription factors TCF and CHOP, disrupting their interaction with the common transcriptional coactivator p300. Loss of PC1 causes increased proliferation and apoptosis, while reintroducing PC1-CTT into cultured Pkd1 null cells reestablishes normal growth rate, suppresses apoptosis, and prevents cyst formation. Inhibition of γ-secretase activity impairs the ability of PC1 to suppress growth and apoptosis and leads to cyst formation in cultured renal epithelial cells. Expression of the PC1-CTT is sufficient to rescue the dorsal body curvature phenotype in zebrafish embryos resulting from either γ-secretase inhibition or suppression of Pkd1 expression. Thus, γ-secretase-dependent release of the PC1-CTT creates a protein fragment whose expression is sufficient to suppress ADPKD-related phenotypes in vitro and in vivo.


Asunto(s)
Secretasas de la Proteína Precursora del Amiloide/metabolismo , Apoptosis , Factores de Transcripción TCF/metabolismo , Canales Catiónicos TRPP/fisiología , Factor de Transcripción CHOP/metabolismo , Pez Cebra/metabolismo , Factores de Transcripción p300-CBP/genética , Animales , Proliferación Celular , Células Cultivadas , Quistes/etiología , Quistes/metabolismo , Quistes/patología , Embrión no Mamífero/citología , Embrión no Mamífero/metabolismo , Humanos , Immunoblotting , Inmunoprecipitación , Riñón/metabolismo , Riñón/patología , Fenotipo , Riñón Poliquístico Autosómico Dominante/fisiopatología , Factores de Transcripción TCF/genética , Canales Catiónicos TRPP/antagonistas & inhibidores , Factor de Transcripción CHOP/genética , Activación Transcripcional , Vía de Señalización Wnt , Pez Cebra/genética , Pez Cebra/crecimiento & desarrollo , Factores de Transcripción p300-CBP/metabolismo
10.
J Am Soc Nephrol ; 22(10): 1809-14, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21921140

RESUMEN

Polycystic kidney disease (PKD) exhibits an inflammatory component, but the contribution of inflammation to cyst progression is unknown. Macrophages promote the proliferation of tubular cells following ischemic injury, suggesting that they may have a role in cystogenesis. Furthermore, cultured Pkd1-deficient cells express the macrophage chemoattractants Mcp1 and Cxcl16 and stimulate macrophage migration. Here, in orthologous models of both PKD1 and PKD2, abnormally large numbers of alternatively activated macrophages surrounded the cysts. To determine whether pericystic macrophages contribute to the proliferation of cyst-lining cells, we depleted phagocytic cells from Pkd1(fl/fl);Pkhd1-Cre mice by treating with liposomal clodronate from postnatal day 10 until day 24. Compared with vehicle-treated controls, macrophage-depleted mice had a significantly lower cystic index, reduced proliferation of cyst-lining cells, better-preserved renal parenchyma, and improved renal function. In conclusion, these data suggest that macrophages home to cystic areas and contribute to cyst growth. Interruption of these homing and proliferative signals could have therapeutic potential for PKD.


Asunto(s)
Movimiento Celular , Macrófagos/fisiología , Riñón Poliquístico Autosómico Dominante/inmunología , Animales , Antígenos Ly/metabolismo , Línea Celular , Proliferación Celular , Quimiocina CCL2/metabolismo , Quimiocina CXCL16 , Quimiocina CXCL6/metabolismo , Ratones , Ratones Endogámicos C57BL
11.
Hum Mol Genet ; 17(20): 3105-17, 2008 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-18632682

RESUMEN

Polycystin-1 (PC1), the product of the PKD1 gene mutated in the majority of autosomal dominant polycystic kidney disease (ADPKD) cases, undergoes a cleavage resulting in the intracellular release of its C-terminal tail (CTT). Here, we demonstrate that the PC1 CTT co-localizes with and binds to beta-catenin in the nucleus. This interaction requires a nuclear localization motif present in the PC1 CTT as well as the N-terminal portion of beta-catenin. The PC1 CTT inhibits the ability of both beta-catenin and Wnt ligands to activate T-cell factor (TCF)-dependent gene transcription, a major effector of the canonical Wnt signaling pathway. The PC1 CTT may produce this effect by reducing the apparent affinity of the interaction between beta-catenin and the TCF protein. DNA microarray analysis reveals that the canonical Wnt signaling pathway is activated in ADPKD patient cysts. Our results suggest a novel mechanism through which PC1 cleavage may impact upon Wnt-dependent signaling and thereby modulate both developmental processes and cystogenesis.


Asunto(s)
Riñón Poliquístico Autosómico Dominante/genética , Riñón Poliquístico Autosómico Dominante/metabolismo , Canales Catiónicos TRPP/química , Canales Catiónicos TRPP/metabolismo , Proteínas Wnt/metabolismo , beta Catenina/metabolismo , Animales , Sitios de Unión , Células CHO , Línea Celular , Núcleo Celular/metabolismo , Cricetinae , Cricetulus , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Ligandos , Análisis de Secuencia por Matrices de Oligonucleótidos , Fragmentos de Péptidos/química , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/metabolismo , Riñón Poliquístico Autosómico Dominante/etiología , Proteínas Recombinantes/química , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Transducción de Señal , Biología de Sistemas , Factores de Transcripción TCF/genética , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...