Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 6 de 6
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Subcell Biochem ; 81: 21-76, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-27830500

RESUMEN

Multiple binding and transport proteins facilitate many aspects of retinoid biology through effects on retinoid transport, cellular uptake, metabolism, and nuclear delivery. These include the serum retinol binding protein sRBP (aka Rbp4), the plasma membrane sRBP receptor Stra6, and the intracellular retinoid binding-proteins such as cellular retinol-binding proteins (CRBP) and cellular retinoic acid binding-proteins (CRABP). sRBP transports the highly lipophilic retinol through an aqueous medium. The major intracellular retinol-binding protein, CRBP1, likely enhances efficient retinoid use by providing a sink to facilitate retinol uptake from sRBP through the plasma membrane or via Stra6, delivering retinol or retinal to select enzymes that generate retinyl esters or retinoic acid, and protecting retinol/retinal from excess catabolism or opportunistic metabolism. Intracellular retinoic acid binding-proteins (CRABP1 and 2, and FABP5) seem to have more diverse functions distinctive to each, such as directing retinoic acid to catabolism, delivering retinoic acid to specific nuclear receptors, and generating non-canonical actions. Gene ablation of intracellular retinoid binding-proteins does not cause embryonic lethality or gross morphological defects. Metabolic and functional defects manifested in knockouts of CRBP1, CRBP2 and CRBP3, however, illustrate their essentiality to health, and in the case of CRBP2, to survival during limited dietary vitamin A. Future studies should continue to address the specific molecular interactions that occur between retinoid binding-proteins and their targets and their precise physiologic contributions to retinoid homeostasis and function.


Asunto(s)
Retinoides/fisiología , Proteínas Celulares de Unión al Retinol/fisiología , Oxidorreductasas de Alcohol/metabolismo , Aldehído Deshidrogenasa/metabolismo , Animales , Transporte Biológico , Núcleo Celular/metabolismo , Ojo/metabolismo , Técnicas de Inactivación de Genes , Homeostasis , Humanos , Mucosa Intestinal/metabolismo , Ratones , Ratones Noqueados , Modelos Moleculares , Proteínas de Neoplasias/metabolismo , Conformación Proteica , Receptores Citoplasmáticos y Nucleares/metabolismo , Retinaldehído/metabolismo , Proteínas Celulares de Unión al Retinol/química , Proteínas Celulares de Unión al Retinol/deficiencia , Proteínas Celulares de Unión al Retinol/genética , Transducción de Señal/fisiología , Tretinoina/metabolismo , Vitamina A/metabolismo , Vitamina A/toxicidad
2.
FASEB J ; 27(5): 1904-16, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23362116

RESUMEN

Cellular retinol-binding protein, type I (CrbpI), encoded by retinol-binding protein, type 1 (Rbp1), is a chaperone of vitamin A (retinol) that is epigenetically silenced in ~25% of human breast cancers. CrbpI delivers vitamin A to enzymes for metabolism into an active metabolite, all-trans retinoic acid (atRA), where atRA is essential to cell proliferation, apoptosis, differentiation, and migration. Here, we show the effect of CrbpI loss on mammary atRA homeostasis using the Rbp1(-/-) mouse model. Rbp1(-/-) mouse mammary tissue has disrupted retinoid homeostasis that results in 40% depleted endogenous atRA. CrbpI loss and atRA depletion precede defects in atRA biosynthesis enzyme expression. Compensation by CrbpIII as a retinoid chaperone does not functionally replace CrbpI. Mammary subcellular fractions isolated from Rbp1(-/-) mice have altered retinol dehydrogenase/reductase (Rdh) enzyme activity that results in 24-42% less atRA production. Rbp1(-/-) mammary tissue has epithelial hyperplasia, stromal hypercellularity, increased collagen, and increased oxidative stress characteristic of atRA deficiency and early tissue dysfunction that precedes tumor formation. Consistent with the findings from the Rbp1(-/-) mouse, tumorigenic epithelial cells lacking CrbpI expression produce 51% less atRA. Together, these data show that CrbpI loss disrupts atRA homeostasis in mammary tissue, resulting in microenvironmental defects similar to those observed at the early stages of tumorigenesis.


Asunto(s)
Glándulas Mamarias Animales/metabolismo , Proteínas Celulares de Unión al Retinol/fisiología , Oxidorreductasas de Alcohol/metabolismo , Animales , Femenino , Homeostasis , Ratones , Proteínas Celulares de Unión al Retinol/deficiencia , Proteínas Celulares de Unión al Retinol/metabolismo , Tretinoina/metabolismo
3.
Int J Biochem Cell Biol ; 44(4): 612-9, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22230368

RESUMEN

Mesenchymal stem cells (MSCs) are multipotent adult stem cells that can differentiate into osteoblasts, chondrocytes and adipocytes, providing a potential source for musculoskeletal tissue engineering. Retinoid signaling plays very important roles in skeletal development. CRBP1 (cellular retinol binding protein 1), a key component of retinoid signaling pathway, is known to take part in vitamin A metabolism and intracellular transporting of retinoids. However, the role of CRBP1 in MSCs remains still obscure. In this study, we investigated the cellular effects of CRBP1 on osteogenic and adipogenic differentiation of bone marrow derived MSCs in vitro and in vivo. Our results showed that CRBP1 overexpression promoted osteogenic differentiation of bone marrow derived MSCs, while inhibited their adipogenic differentiation. We also demonstrated that the possible underlying mechanism for CRBP1 promoting osteogenic differentiation of MSCs was by inhibiting RXRα-induced ß-catenin degradation, maintaining ß-catenin and pERK1/2 at higher levels. These findings reveal a potential role of CRBP1 in the regulation of ß-catenin turnover which can greatly affect the process of osteogenesis and adipogenesis of MSCs.


Asunto(s)
Adipogénesis , Células Madre Mesenquimatosas/citología , Células Madre Mesenquimatosas/metabolismo , Osteogénesis , Receptor alfa X Retinoide/metabolismo , Proteínas Celulares de Unión al Retinol/metabolismo , beta Catenina/metabolismo , Animales , Células de la Médula Ósea/citología , Silenciador del Gen , Células HEK293 , Humanos , Lentivirus/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Fosfoproteínas/metabolismo , Estabilidad Proteica , Proteolisis , ARN Interferente Pequeño/genética , Ratas , Ratas Sprague-Dawley , Proteínas Celulares de Unión al Retinol/deficiencia , Proteínas Celulares de Unión al Retinol/genética , Transducción de Señal
4.
Proc Natl Acad Sci U S A ; 108(33): 13379-86, 2011 Aug 16.
Artículo en Inglés | MEDLINE | ID: mdl-21788502

RESUMEN

Aberrations in epigenetic processes, such as histone methylation, can cause cancer. Retinoblastoma binding protein 2 (RBP2; also called JARID1A or KDM5A) can demethylate tri- and dimethylated lysine 4 in histone H3, which are epigenetic marks for transcriptionally active chromatin, whereas the multiple endocrine neoplasia type 1 (MEN1) tumor suppressor promotes H3K4 methylation. Previous studies suggested that inhibition of RBP2 contributed to tumor suppression by the retinoblastoma protein (pRB). Here, we show that genetic ablation of Rbp2 decreases tumor formation and prolongs survival in Rb1(+/-) mice and Men1-defective mice. These studies link RBP2 histone demethylase activity to tumorigenesis and nominate RBP2 as a potential target for cancer therapy.


Asunto(s)
Neoplasias/prevención & control , Proteínas Proto-Oncogénicas/deficiencia , Proteína de Retinoblastoma/deficiencia , Proteínas Celulares de Unión al Retinol/deficiencia , Animales , Inhibidores Enzimáticos/uso terapéutico , Epigenómica , Histona Demetilasas , Histonas/metabolismo , Metilación , Ratones , Ratones Noqueados , Neoplasias/enzimología , Neoplasias/etiología , Proteínas Celulares de Unión al Retinol/antagonistas & inhibidores , Tasa de Supervivencia
5.
Mol Cell Biol ; 31(16): 3277-85, 2011 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-21670153

RESUMEN

Cellular retinol-binding protein type I (CrbpI), encoded by Rpb1, serves as a chaperone of retinol homeostasis, but its physiological effects remain incompletely understood. We show here that the Rbp1(-/-) mouse has disrupted retinoid homeostasis in multiple tissues, with abnormally high 9-cis-retinoic acid (9cRA), a pancreas autacoid that attenuates glucose-stimulated insulin secretion. The Rbp1(-/-) pancreas has increased retinol and intense ectopic expression of Rpb2 mRNA, which encodes CrbpII: both would contribute to increased ß-cell 9cRA biosynthesis. 9cRA in Rbp1(-/-) pancreas resists postprandial and glucose-induced decreases. Rbp1(-/-) mice have defective islet expression of genes involved in glucose sensing and insulin secretion, as well as islet α-cell infiltration, which contribute to reduced glucose-stimulated insulin secretion, high glucagon secretion, an abnormally high rate of gluconeogenesis, and hyperglycemia. A diet rich in vitamin A (as in a standard chow diet) increases pancreas 9cRA and impairs glucose tolerance. Crbp1 attenuates the negative impact of vitamin A (retinol) on glucose tolerance, regardless of the dietary retinol content. Rbp1(-/-) mice have an increased rate of fatty acid oxidation and resist obesity when fed a high-fat diet. Thus, glucose homeostasis and energy metabolism rely on Rbp1 expression and its moderation of pancreas retinol and of the autacoid 9cRA.


Asunto(s)
Glucosa/metabolismo , Homeostasis , Páncreas/metabolismo , Proteínas Celulares de Unión al Retinol/fisiología , Tretinoina/metabolismo , Alitretinoína , Animales , Antineoplásicos , Metabolismo Energético , Islotes Pancreáticos/metabolismo , Ratones , Ratones Noqueados , Proteínas Celulares de Unión al Retinol/deficiencia , Vitamina A/metabolismo
6.
Mol Cell Biol ; 30(14): 3412-20, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20498279

RESUMEN

Adipogenesis is governed by a well-documented cascade of transcription factors. However, less is known about non-transcription factors that govern early stages of adipogenesis. Here we show that cellular retinol-binding protein type I (CRBP-I), a small cytosolic binding protein for retinol and retinaldehyde, is specifically restricted to preadipocytes in white adipose tissue. The absence of CRBP-I in mice (CRBP-I-KO mice) leads to increased adiposity. Despite increased adiposity, CRBP-I-KO mice remain more glucose tolerant and insulin sensitive during high-fat-diet feeding. 3T3-L1 cells deficient in CRBP-I or mouse embryonic fibroblasts derived from CRBP-I-KO mice had increased adipocyte differentiation and triglyceride (TG) accumulation. This was due to increased expression and activity of PPAR gamma, while other transcription factor pathways in early and late differentiation remained unchanged. Conversely, the overexpression of CRBP-I in 3T3-L1 cells results in decreased TG accumulation. In conclusion, CRBP-I is a cytosolic protein specifically expressed in preadipocytes that regulates adipocyte differentiation in part by affecting PPAR gamma activity.


Asunto(s)
Adipogénesis/fisiología , Proteínas Celulares de Unión al Retinol/fisiología , Células 3T3-L1 , Adipocitos/citología , Adipocitos/metabolismo , Adiposidad , Animales , Secuencia de Bases , Diferenciación Celular/fisiología , Células Madre Embrionarias/citología , Células Madre Embrionarias/metabolismo , Prueba de Tolerancia a la Glucosa , Resistencia a la Insulina , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Obesidad/metabolismo , Obesidad/patología , PPAR gamma/metabolismo , Fenotipo , ARN Interferente Pequeño/genética , Proteínas Celulares de Unión al Retinol/antagonistas & inhibidores , Proteínas Celulares de Unión al Retinol/deficiencia , Proteínas Celulares de Unión al Retinol/genética , Triglicéridos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA