Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
Proc Natl Acad Sci U S A ; 109(7): E423-31, 2012 Feb 14.
Artículo en Inglés | MEDLINE | ID: mdl-22308344

RESUMEN

Lamins A and C, alternatively spliced products of the LMNA gene, are key components of the nuclear lamina. The two isoforms are found in similar amounts in most tissues, but we observed an unexpected pattern of expression in the brain. Western blot and immunohistochemistry studies showed that lamin C is abundant in the mouse brain, whereas lamin A and its precursor prelamin A are restricted to endothelial cells and meningeal cells and are absent in neurons and glia. Prelamin A transcript levels were low in the brain, but this finding could not be explained by alternative splicing. In lamin A-only knockin mice, where alternative splicing is absent and all the output of the gene is channeled into prelamin A transcripts, large amounts of lamin A were found in peripheral tissues, but there was very little lamin A in the brain. Also, in knockin mice expressing exclusively progerin (a toxic form of prelamin A found in Hutchinson-Gilford progeria syndrome), the levels of progerin in the brain were extremely low. Further studies showed that prelamin A expression, but not lamin C expression, is down-regulated by a brain-specific microRNA, miR-9. Expression of miR-9 in cultured cells reduced lamin A expression, and this effect was abolished when the miR-9-binding site in the prelamin A 3' UTR was mutated. The down-regulation of prelamin A expression in the brain could explain why mouse models of Hutchinson-Gilford progeria syndrome are free of central nervous system pathology.


Asunto(s)
Encéfalo/metabolismo , Lamina Tipo A/metabolismo , MicroARNs/metabolismo , Animales , Western Blotting , Ratones
2.
Arterioscler Thromb Vasc Biol ; 32(2): 230-5, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22173228

RESUMEN

OBJECTIVE: Gpihbp1-deficient (Gpihbp1-/-) mice lack the ability to transport lipoprotein lipase to the capillary lumen, resulting in mislocalization of lipoprotein lipase within tissues, defective lipolysis of triglyceride-rich lipoproteins, and chylomicronemia. We asked whether GPIHBP1 deficiency and mislocalization of catalytically active lipoprotein lipase would alter the composition of triglycerides in adipose tissue or perturb the expression of lipid biosynthetic genes. We also asked whether perturbations in adipose tissue composition and gene expression, if they occur, would be accompanied by reciprocal metabolic changes in the liver. METHODS AND RESULTS: The chylomicronemia in Gpihbp1-/- mice was associated with reduced levels of essential fatty acids in adipose tissue triglycerides and increased expression of lipid biosynthetic genes. The liver exhibited the opposite changes: increased levels of essential fatty acids in triglycerides and reduced expression of lipid biosynthetic genes. CONCLUSIONS: Defective lipolysis in Gpihbp1-/- mice causes reciprocal metabolic perturbations in adipose tissue and liver. In adipose tissue, the essential fatty acid content of triglycerides is reduced and lipid biosynthetic gene expression is increased, whereas the opposite changes occur in the liver.


Asunto(s)
Tejido Adiposo/metabolismo , Metabolismo de los Lípidos/fisiología , Hígado/metabolismo , Receptores de Lipoproteína/deficiencia , Animales , Ácidos Grasos/metabolismo , Lipólisis/fisiología , Lipoproteína Lipasa/metabolismo , Masculino , Ratones , Ratones Noqueados , Modelos Animales , Receptores de Lipoproteína/genética , Triglicéridos/metabolismo
3.
J Lipid Res ; 53(12): 2690-7, 2012 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-23008484

RESUMEN

Lipoprotein lipase (LPL) is secreted into the interstitial spaces by adipocytes and myocytes but then must be transported to the capillary lumen by GPIHBP1, a glycosylphosphatidylinositol-anchored protein of capillary endothelial cells. The mechanism by which GPIHBP1 and LPL move across endothelial cells remains unclear. We asked whether the transport of GPIHBP1 and LPL across endothelial cells was uni- or bidirectional. We also asked whether GPIHBP1 and LPL are transported across cells in vesicles and whether this transport process requires caveolin-1. The movement of GPIHBP1 and LPL across cultured endothelial cells was bidirectional. Also, GPIHBP1 moved bidirectionally across capillary endothelial cells in live mice. The transport of LPL across endothelial cells was inhibited by dynasore and genistein, consistent with a vesicular transport process. Also, transmission electron microscopy (EM) and dual-axis EM tomography revealed GPIHBP1 and LPL in invaginations of the plasma membrane and in vesicles. The movement of GPIHBP1 across capillary endothelial cells was efficient in the absence of caveolin-1, and there was no defect in the internalization of LPL by caveolin-1-deficient endothelial cells in culture. Our studies show that GPIHBP1 and LPL move bidirectionally across endothelial cells in vesicles and that transport is efficient even when caveolin-1 is absent.


Asunto(s)
Células Endoteliales/metabolismo , Lipoproteína Lipasa/metabolismo , Receptores de Lipoproteína/metabolismo , Animales , Células CHO , Cricetinae , Células Endoteliales/química , Células Endoteliales/enzimología , Genisteína/farmacología , Humanos , Hidrazonas/farmacología , Lipoproteína Lipasa/antagonistas & inhibidores , Ratones , Ratones Noqueados , Ratas , Receptores de Lipoproteína/deficiencia , Relación Estructura-Actividad
4.
Hum Mol Genet ; 19(13): 2682-94, 2010 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-20421363

RESUMEN

Lamin A is formed from prelamin A by four post-translational processing steps-farnesylation, release of the last three amino acids of the protein, methylation of the farnesylcysteine and the endoproteolytic release of the C-terminal 15 amino acids (including the farnesylcysteine methyl ester). When the final processing step does not occur, a farnesylated and methylated prelamin A accumulates in cells, causing a severe progeroid disease, restrictive dermopathy (RD). Whether RD is caused by the retention of farnesyl lipid on prelamin A, or by the retention of the last 15 amino acids of the protein, is unknown. To address this issue, we created knock-in mice harboring a mutant Lmna allele (LmnanPLAO) that yields exclusively non-farnesylated prelamin A (and no lamin C). These mice had no evidence of progeria but succumbed to cardiomyopathy. We suspected that the non-farnesylated prelamin A in the tissues of these mice would be strikingly mislocalized to the nucleoplasm, but this was not the case; most was at the nuclear rim (indistinguishable from the lamin A in wild-type mice). The cardiomyopathy could not be ascribed to an absence of lamin C because mice expressing an otherwise identical knock-in allele yielding only wild-type prelamin A appeared normal. We conclude that lamin C synthesis is dispensable in mice and that the failure to convert prelamin A to mature lamin A causes cardiomyopathy (at least in the absence of lamin C). The latter finding is potentially relevant to the long-term use of protein farnesyltransferase inhibitors, which lead to an accumulation of non-farnesylated prelamin A.


Asunto(s)
Cardiomiopatías/metabolismo , Proteínas Nucleares/genética , Proteínas Nucleares/metabolismo , Progeria/metabolismo , Precursores de Proteínas/genética , Precursores de Proteínas/metabolismo , Animales , Cardiomiopatías/etiología , Fibroblastos/metabolismo , Fibroblastos/patología , Técnicas de Sustitución del Gen , Lamina Tipo A/metabolismo , Ratones , Ratones Endogámicos , Modelos Animales , Progeria/etiología , Prenilación de Proteína
5.
J Biol Chem ; 285(50): 39239-48, 2010 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-20889497

RESUMEN

Glycosylphosphatidylinositol-anchored high density lipoprotein-binding protein 1 (GPIHBP1), a GPI-anchored endothelial cell protein, binds lipoprotein lipase (LPL) and transports it into the lumen of capillaries where it hydrolyzes triglycerides in lipoproteins. GPIHBP1 is assumed to be expressed mainly within the heart, skeletal muscle, and adipose tissue, the sites where most lipolysis occurs, but the tissue pattern of GPIHBP1 expression has never been evaluated systematically. Because GPIHBP1 is found on the luminal face of capillaries, we predicted that it would be possible to define GPIHBP1 expression patterns with radiolabeled GPIHBP1-specific antibodies and positron emission tomography (PET) scanning. In Gpihbp1(-/-) mice, GPIHBP1-specific antibodies were cleared slowly from the blood, and PET imaging showed retention of the antibodies in the blood pools (heart and great vessels). In Gpihbp1(+/+) mice, the antibodies were cleared extremely rapidly from the blood and, to our surprise, were taken up mainly by lung and liver. Immunofluorescence microscopy confirmed the presence of GPIHBP1 in the capillary endothelium of both lung and liver. In most tissues with high levels of Gpihbp1 expression, Lpl expression was also high, but the lung was an exception (very high Gpihbp1 expression and extremely low Lpl expression). Despite low Lpl transcript levels, however, LPL protein was readily detectable in the lung, suggesting that some of that LPL originates elsewhere and then is captured by GPIHBP1 in the lung. In support of this concept, lung LPL levels were significantly lower in Gpihbp1(-/-) mice than in Gpihbp1(+/+) mice. In addition, Lpl(-/-) mice expressing human LPL exclusively in muscle contained high levels of human LPL in the lung.


Asunto(s)
Regulación de la Expresión Génica , Glicosilfosfatidilinositoles/metabolismo , Receptores de Lipoproteína/química , Animales , Sitios de Unión , Capilares/metabolismo , Membrana Celular/metabolismo , Endotelio/metabolismo , Cinética , Pulmón/metabolismo , Ratones , Ratones Transgénicos , Modelos Biológicos , Tomografía de Emisión de Positrones/métodos
6.
J Am Heart Assoc ; 6(7)2017 Jul 22.
Artículo en Inglés | MEDLINE | ID: mdl-28735290

RESUMEN

BACKGROUND: The MMP (matrix metalloproteinase) family plays diverse and critical roles in directing vascular wall remodeling in atherosclerosis. Unlike secreted-type MMPs, a member of the membrane-type MMP family, MT1-MMP (membrane-type 1 MMP; MMP14), mediates pericellular extracellular matrix degradation that is indispensable for maintaining physiological extracellular matrix homeostasis. However, given the premature mortality exhibited by MT1-MMP-null mice, the potential role of the proteinase in atherogenesis remains elusive. We sought to determine the effects of both MT1-MMP heterozygosity and tissue-specific gene targeting on atherogenesis in APOE (apolipoprotein E)-null mice. METHODS AND RESULTS: MT1-MMP heterozygosity in the APOE-null background (Mmp14+/-Apoe-/- ) significantly promoted atherogenesis relative to Mmp14+/+Apoe-/- mice. Furthermore, the tissue-specific deletion of MT1-MMP from vascular smooth muscle cells (VSMCs) in SM22α-Cre(+)Mmp14F/FApoe-/- (VSMC-knockout) mice likewise increased the severity of atherosclerotic lesions. Although VSMC-knockout mice also developed progressive atherosclerotic aneurysms in their iliac arteries, macrophage- and adipose-specific MT1-MMP-knockout mice did not display this sensitized phenotype. In VSMC-knockout mice, atherosclerotic lesions were populated by hyperproliferating VSMCs (smooth muscle actin- and Ki67-double-positive cells) that were characterized by a proinflammatory gene expression profile. Finally, MT1-MMP-null VSMCs cultured in a 3-dimensional spheroid model system designed to mimic in vivo-like cell-cell and cell-extracellular matrix interactions, likewise displayed markedly increased proliferative potential. CONCLUSIONS: MT1-MMP expressed by VSMCs plays a key role in limiting the progression of atherosclerosis in APOE-null mice by regulating proliferative responses and inhibiting the deterioration of VSMC function in atherogenic vascular walls.


Asunto(s)
Enfermedades de la Aorta/enzimología , Aterosclerosis/enzimología , Proliferación Celular , Metaloproteinasa 14 de la Matriz/metabolismo , Músculo Liso Vascular/enzimología , Miocitos del Músculo Liso/enzimología , Animales , Aorta/enzimología , Aorta/patología , Enfermedades de la Aorta/genética , Enfermedades de la Aorta/patología , Aterosclerosis/genética , Aterosclerosis/patología , Comunicación Celular , Uniones Célula-Matriz/enzimología , Uniones Célula-Matriz/patología , Células Cultivadas , Modelos Animales de Enfermedad , Femenino , Predisposición Genética a la Enfermedad , Heterocigoto , Arteria Ilíaca/enzimología , Arteria Ilíaca/patología , Mediadores de Inflamación/metabolismo , Masculino , Metaloproteinasa 14 de la Matriz/deficiencia , Metaloproteinasa 14 de la Matriz/genética , Ratones Endogámicos C57BL , Ratones Noqueados para ApoE , Músculo Liso Vascular/patología , Miocitos del Músculo Liso/patología , Fenotipo , Placa Aterosclerótica , Transducción de Señal , Remodelación Vascular
7.
J Vis Exp ; (114)2016 08 11.
Artículo en Inglés | MEDLINE | ID: mdl-27583550

RESUMEN

The isolation of adipose-derived stem cells (ASCs) is an important method in the field of adipose tissue biology, adipogenesis, and extracellular matrix (ECM) remodeling. In vivo, ECM-rich environment consisting of fibrillar collagens provides a structural support to adipose tissues during the progression and regression of obesity. Physiological ECM remodeling mediated by matrix metalloproteinases (MMPs) plays a major role in regulating adipose tissue size and function(1,2). The loss of physiological collagenolytic ECM remodeling may lead to excessive collagen accumulation (tissue fibrosis), macrophage infiltration, and ultimately, a loss of metabolic homeostasis including insulin resistance(3,4). When a phenotypic change of the adipose tissue is observed in gene-targeted mouse models, isolating primary ASCs from fat depots for in vitro studies is an effective approach to define the role of the specific gene in regulating the function of ASCs. In the following, we define an immunomagnetic separation of Sca1(high) ASCs.


Asunto(s)
Adipocitos , Separación Inmunomagnética/métodos , Adipocitos/citología , Adipogénesis , Tejido Adiposo , Animales , Diferenciación Celular , Células Madre
8.
J Invest Dermatol ; 136(2): 436-443, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26967477

RESUMEN

SLURP1, a member of the lymphocyte antigen 6 protein family, is secreted by suprabasal keratinocytes. Mutations in SLURP1 cause a palmoplantar keratoderma (PPK) known as mal de Meleda. SLURP2, another secreted lymphocyte antigen 6 protein, is encoded by a gene located ?20 kb downstream from SLURP1. SLURP2 is produced by suprabasal keratinocytes. To investigate the importance of SLURP2, we first examined Slurp2 knockout mice in which exon 2-3 sequences had been replaced with lacZ and neo cassettes. Slurp2(-/-) mice exhibited hyperkeratosis on the volar surface of the paws (i.e., palmoplantar keratoderma), increased keratinocyte proliferation, and an accumulation of lipid droplets in the stratum corneum. They also exhibited reduced body weight and hind limb clasping. These phenotypes are similar to those of Slurp1(-/-) mice. To solidify a link between Slurp2 deficiency and palmoplantar keratoderma and to be confident that the disease phenotypes in Slurp2(-/-) mice were not secondary to the effects of the lacZ and neo cassettes on Slurp1 expression, we created a new line of Slurp2 knockout mice (Slurp2X(-/-)) in which Slurp2 was inactivated with a simple nonsense mutation. Slurp2X(-/-) mice exhibited the same disease phenotypes. Thus, Slurp2 deficiency and Slurp1 deficiencies cause the same disease phenotypes.


Asunto(s)
Antígenos Ly/genética , Codón sin Sentido , Proteínas Ligadas a GPI/genética , Regulación de la Expresión Génica , Queratodermia Palmoplantar/genética , Activador de Plasminógeno de Tipo Uroquinasa/genética , Proteínas Adaptadoras Transductoras de Señales , Animales , Células Cultivadas , Modelos Animales de Enfermedad , Proteínas Ligadas a GPI/deficiencia , Inmunohistoquímica , Queratinocitos/citología , Queratinocitos/metabolismo , Queratodermia Palmoplantar/patología , Ratones , Ratones Noqueados , Fenotipo , Distribución Aleatoria , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos
9.
Matrix Biol ; 36: 28-38, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24726953

RESUMEN

Stem cell antigen-1 (Sca1 or Ly6A/E) is a cell surface marker that is widely expressed in mesenchymal stem cells, including adipose-derived stem cells (ASCs). We hypothesized that the fat depot-specific gene signature of Sca1(high) ASCs may play the major role in defining adipose tissue function and extracellular matrix (ECM) remodeling in a depot-specific manner. Herein we aimed to characterize the unique gene signature and ECM remodeling of Sca1(high) ASCs isolated from subcutaneous (inguinal) and visceral (epididymal) adipose tissues. Sca1(high) ASCs are found in the adventitia and perivascular areas of adipose tissues. Sca1(high) ASCs purified with magnetic-activated cell sorting (MACS) demonstrate dendrite or round shape with the higher expression of cytokines and chemokines (e.g., Il6, Cxcl1) and the lower expression of a glucose transporter (Glut1). Subcutaneous and visceral fat-derived Sca1(high) ASCs particularly differ in the gene expressions of adhesion and ECM molecules. While the expression of the major membrane-type collagenase (MMP14) is comparable between the groups, the expressions of secreted collagenases (MMP8 and MMP13) are higher in visceral Sca1(high) ASCs than in subcutaneous ASCs. Consistently, slow but focal MMP-dependent collagenolysis was observed with subcutaneous adipose tissue-derived vascular stromal cells, whereas rapid and bulk collagenolysis was observed with visceral adipose tissue-derived cells in MMP-dependent and -independent manners. These results suggest that the fat depot-specific gene signatures of ASCs may contribute to the distinct patterns of ECM remodeling and adipose function in different fat depots.


Asunto(s)
Adipogénesis/genética , Antígenos Ly/biosíntesis , Grasa Intraabdominal/metabolismo , Proteínas de la Membrana/biosíntesis , Grasa Subcutánea/metabolismo , Tejido Adiposo/crecimiento & desarrollo , Tejido Adiposo/metabolismo , Animales , Diferenciación Celular/genética , Matriz Extracelular/genética , Matriz Extracelular/metabolismo , Regulación del Desarrollo de la Expresión Génica , Humanos , Células Madre Mesenquimatosas/metabolismo , Ratones
10.
J Invest Dermatol ; 134(6): 1589-1598, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24499735

RESUMEN

Mutations in SLURP1 cause mal de Meleda, a rare palmoplantar keratoderma (PPK). SLURP1 is a secreted protein that is expressed highly in keratinocytes but has also been identified elsewhere (e.g., spinal cord neurons). Here, we examined Slurp1-deficient mice (Slurp1(-/-)) created by replacing exon 2 with ß-gal and neo cassettes. Slurp1(-/-) mice developed severe PPK characterized by increased keratinocyte proliferation, an accumulation of lipid droplets in the stratum corneum, and a water barrier defect. In addition, Slurp1(-/-) mice exhibited reduced adiposity, protection from obesity on a high-fat diet, low plasma lipid levels, and a neuromuscular abnormality (hind-limb clasping). Initially, it was unclear whether the metabolic and neuromuscular phenotypes were due to Slurp1 deficiency, because we found that the targeted Slurp1 mutation reduced the expression of several neighboring genes (e.g., Slurp2, Lypd2). We therefore created a new line of knockout mice (Slurp1X(-/-) mice) with a simple nonsense mutation in exon 2. The Slurp1X mutation did not reduce the expression of adjacent genes, but Slurp1X(-/-) mice exhibited all of the phenotypes observed in the original line of knockout mice. Thus, Slurp1 deficiency in mice elicits metabolic and neuromuscular abnormalities in addition to PPK.


Asunto(s)
Antígenos Ly/metabolismo , Queratodermia Palmoplantar/metabolismo , Queratodermia Palmoplantar/fisiopatología , Enfermedades Neuromusculares/fisiopatología , Activador de Plasminógeno de Tipo Uroquinasa/metabolismo , Alelos , Animales , Antígenos Ly/genética , Peso Corporal , Codón sin Sentido , Epidermis/metabolismo , Epidermis/patología , Exones , Femenino , Genotipo , Lípidos/sangre , Masculino , Ratones , Ratones Noqueados , Fenotipo , Activador de Plasminógeno de Tipo Uroquinasa/genética , Agua/metabolismo
11.
Endocrinology ; 154(12): 4548-59, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24140711

RESUMEN

Thrombospondin 1 (THBS1 or TSP-1) is a circulating glycoprotein highly expressed in hypertrophic visceral adipose tissues of humans and mice. High-fat diet (HFD) feeding induces the robust increase of circulating THBS1 in the early stages of HFD challenge. The loss of Thbs1 protects male mice from diet-induced weight gain and adipocyte hypertrophy. Hyperinsulinemic euglycemic clamp study has demonstrated that Thbs1-null mice are protected from HFD-induced insulin resistance. Tissue-specific glucose uptake study has revealed that the insulin-sensitive phenotype of Thbs1-null mice is mostly mediated by skeletal muscles. Further assessments of the muscle phenotype using RNA sequencing, quantitative PCR, and histological studies have demonstrated that Thbs1-null skeletal muscles are protected from the HFD-dependent induction of Col3a1 and Col6a1, coupled with a new collagen deposition. At the same time, the Thbs1-null mice display a better circadian rhythm and higher amplitude of energy expenditure with a browning phenotype in sc adipose tissues. These results suggest that THBS1, which circulates in response to a HFD, may induce insulin resistance and fibrotic tissue damage in skeletal muscles as well as the de-browning of sc adipose tissues in the early stages of a HFD challenge. Our study may shed new light on the pathogenic role played by a circulating extracellular matrix protein in the cross talk between adipose tissues and skeletal muscles during obesity progression.


Asunto(s)
Grasas de la Dieta/efectos adversos , Fibrosis/inducido químicamente , Resistencia a la Insulina/fisiología , Enfermedades Musculares/etiología , Trombospondina 1/metabolismo , Células 3T3-L1 , Tejido Adiposo Blanco/metabolismo , Animales , Grasas de la Dieta/administración & dosificación , Relación Dosis-Respuesta a Droga , Epidídimo , Regulación de la Expresión Génica/efectos de los fármacos , Técnica de Clampeo de la Glucosa , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Músculo Esquelético/metabolismo , Músculo Esquelético/patología , Obesidad , Trombospondina 1/genética , Transcriptoma
12.
Nucleus ; 2(1): 4-9, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21647293

RESUMEN

Lmna yields two major protein products in somatic cells, lamin C and prelamin A. Mature lamin A is produced from prelamin A by four posttranslational processing steps-farnesylation of a carboxyl-terminal cysteine, release of the last three amino acids of the protein, methylation of the farnesylcysteine, and the endoproteolytic release of the carboxyl-terminal 15 amino acids of the protein (including the farnesylcysteine methyl ester). Although the posttranslational processing of prelamin A has been conserved in vertebrate evolution, its physiologic significance remains unclear. Here we review recent studies in which we investigated prelamin A processing with Lmna knock-in mice that produce exclusively prelamin A (Lmna(PLAO)), mature lamin A (Lmna(LAO)) or nonfarnesylated prelamin A (Lmna(nPLAO)). We found that the synthesis of lamin C is dispensable in laboratory mice, that the direct production of mature lamin A (completely bypassing all prelamin A processing) causes no discernable pathology in mice, and that exclusive production of nonfarnesylated prelamin A leads to cardiomyopathy.


Asunto(s)
Laminas/metabolismo , Proteínas Nucleares/metabolismo , Progeria/metabolismo , Precursores de Proteínas/metabolismo , Animales , Lamina Tipo A , Laminas/deficiencia , Laminas/genética , Ratones , Progeria/patología , Prenilación de Proteína , Procesamiento Proteico-Postraduccional
13.
Mol Biol Cell ; 22(23): 4683-93, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-21976703

RESUMEN

Neuronal migration is essential for the development of the mammalian brain. Here, we document severe defects in neuronal migration and reduced numbers of neurons in lamin B1-deficient mice. Lamin B1 deficiency resulted in striking abnormalities in the nuclear shape of cortical neurons; many neurons contained a solitary nuclear bleb and exhibited an asymmetric distribution of lamin B2. In contrast, lamin B2 deficiency led to increased numbers of neurons with elongated nuclei. We used conditional alleles for Lmnb1 and Lmnb2 to create forebrain-specific knockout mice. The forebrain-specific Lmnb1- and Lmnb2-knockout models had a small forebrain with disorganized layering of neurons and nuclear shape abnormalities, similar to abnormalities identified in the conventional knockout mice. A more severe phenotype, complete atrophy of the cortex, was observed in forebrain-specific Lmnb1/Lmnb2 double-knockout mice. This study demonstrates that both lamin B1 and lamin B2 are essential for brain development, with lamin B1 being required for the integrity of the nuclear lamina, and lamin B2 being important for resistance to nuclear elongation in neurons.


Asunto(s)
Encéfalo/embriología , Encéfalo/crecimiento & desarrollo , Lamina Tipo B/metabolismo , Neuronas/patología , Prosencéfalo/anomalías , Animales , Encéfalo/anomalías , Lamina Tipo B/genética , Ratones , Ratones Noqueados , Neuronas/metabolismo , Lámina Nuclear , Prosencéfalo/crecimiento & desarrollo
14.
Cell Metab ; 12(1): 42-52, 2010 Jul 07.
Artículo en Inglés | MEDLINE | ID: mdl-20620994

RESUMEN

The lipolytic processing of triglyceride-rich lipoproteins by lipoprotein lipase (LPL) is the central event in plasma lipid metabolism, providing lipids for storage in adipose tissue and fuel for vital organs such as the heart. LPL is synthesized and secreted by myocytes and adipocytes, but then finds its way into the lumen of capillaries, where it hydrolyzes lipoprotein triglycerides. The mechanism by which LPL reaches the lumen of capillaries has remained an unresolved problem of plasma lipid metabolism. Here, we show that GPIHBP1 is responsible for the transport of LPL into capillaries. In Gpihbp1-deficient mice, LPL is mislocalized to the interstitial spaces surrounding myocytes and adipocytes. Also, we show that GPIHBP1 is located at the basolateral surface of capillary endothelial cells and actively transports LPL across endothelial cells. Our experiments define the function of GPIHBP1 in triglyceride metabolism and provide a mechanism for the transport of LPL into capillaries.


Asunto(s)
Capilares/enzimología , Lipoproteína Lipasa/metabolismo , Receptores de Lipoproteína/metabolismo , Tejido Adiposo/irrigación sanguínea , Animales , Células Endoteliales/enzimología , Células Endoteliales/metabolismo , Metabolismo de los Lípidos , Lipoproteína Lipasa/análisis , Lipoproteínas/metabolismo , Ratones , Ratones Noqueados , Receptores de Lipoproteína/análisis , Receptores de Lipoproteína/genética , Triglicéridos/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA