Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 16 de 16
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
2.
Science ; 360(6393): 1087-1092, 2018 06 08.
Artículo en Inglés | MEDLINE | ID: mdl-29880681

RESUMEN

A high concentration of low-density lipoprotein cholesterol (LDL-C) is a major risk factor for cardiovascular disease. Although LDL-C levels vary among humans and are heritable, the genetic factors affecting LDL-C are not fully characterized. We identified a rare frameshift variant in the LIMA1 (also known as EPLIN or SREBP3) gene from a Chinese family of Kazakh ethnicity with inherited low LDL-C and reduced cholesterol absorption. In a mouse model, LIMA1 was mainly expressed in the small intestine and localized on the brush border membrane. LIMA1 bridged NPC1L1, an essential protein for cholesterol absorption, to a transportation complex containing myosin Vb and facilitated cholesterol uptake. Similar to the human phenotype, Lima1-deficient mice displayed reduced cholesterol absorption and were resistant to diet-induced hypercholesterolemia. Through our study of both mice and humans, we identify LIMA1 as a key protein regulating intestinal cholesterol absorption.


Asunto(s)
Pueblo Asiatico/genética , LDL-Colesterol/metabolismo , Proteínas del Citoesqueleto/metabolismo , Mutación del Sistema de Lectura , Absorción Intestinal/genética , Mucosa Intestinal/metabolismo , Animales , China , LDL-Colesterol/sangre , Proteínas del Citoesqueleto/genética , Variación Genética , Células Hep G2 , Humanos , Kazajstán/etnología , Proteínas de la Membrana/metabolismo , Proteínas de Transporte de Membrana , Ratones , Ratones Noqueados , Cadenas Pesadas de Miosina/metabolismo , Miosina Tipo V/metabolismo , Linaje , Unión Proteica , Transporte de Proteínas
3.
Cell Rep ; 19(13): 2823-2835, 2017 06 27.
Artículo en Inglés | MEDLINE | ID: mdl-28658628

RESUMEN

Proper intracellular cholesterol trafficking is critical for cellular function. Two lysosome-resident proteins, NPC1 and NPC2, mediate the egress of low-density lipoprotein-derived cholesterol from lysosomes. However, other proteins involved in this process remain largely unknown. Through amphotericin B-based selection, we isolated two cholesterol transport-defective cell lines. Subsequent whole-transcriptome-sequencing analysis revealed two cell lines bearing the same mutation in the vacuolar protein sorting 53 (Vps53) gene. Depletion of VPS53 or other subunits of the Golgi-associated retrograde protein (GARP) complex impaired NPC2 sorting to lysosomes and caused cholesterol accumulation. GARP deficiency blocked the retrieval of the cation-independent mannose 6-phosphate receptor (CI-MPR) to the trans-Golgi network. Further, Vps54 mutant mice displayed reduced cellular NPC2 protein levels and increased cholesterol accumulation, underscoring the physiological role of the GARP complex in cholesterol transport. We conclude that the GARP complex contributes to intracellular cholesterol transport by targeting NPC2 to lysosomes in a CI-MPR-dependent manner.


Asunto(s)
Colesterol/metabolismo , Lisosomas/metabolismo , Proteínas de la Membrana/genética , Proteínas de Transporte Vesicular/metabolismo , Animales , Transporte Biológico , Humanos , Proteínas de la Membrana/metabolismo , Ratones
4.
Hepatology ; 65(6): 1936-1947, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28027595

RESUMEN

Obesity is a critical risk factor for hepatocellular carcinoma (HCC). However, it remains unknown whether inhibition of de novo lipid biosynthesis can suppress HCC. In this study, we blocked the sterol regulatory element-binding protein (SREBP) pathway, one of the key determinants of lipid homeostasis, by ablating 78-kDa cell-surface glycoprotein or SREBP cleavage-activating protein in hepatocytes, as well as by administering a chemical compound called betulin. We found that either genetically or pharmacologically inhibiting the SREBP pathway dramatically reduced diethylnitrosamine-induced HCC progression by down-regulating tumor-promoting cytokines, including interleukin (IL)-6, tumor necrosis factor alpha, and IL-1ß. CONCLUSION: Inhibition of de novo lipid biosynthesis by suppressing the SREBP pathway prevents HCC. This study identifies a previously underappreciated role of the SREBP pathway in HCC and suggests a novel metabolic strategy to control liver cancer. (Hepatology 2017;65:1936-1947).


Asunto(s)
Carcinogénesis/patología , Carcinoma Hepatocelular/patología , Interleucina-1beta/metabolismo , Neoplasias Hepáticas/patología , Proteína 1 de Unión a los Elementos Reguladores de Esteroles/genética , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Carcinoma Hepatocelular/fisiopatología , Modelos Animales de Enfermedad , Regulación Neoplásica de la Expresión Génica , Hepatocitos/metabolismo , Hepatocitos/patología , Inflamación/patología , Inflamación/prevención & control , Neoplasias Hepáticas/fisiopatología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neoplasias Experimentales , Obesidad/complicaciones , Obesidad/patología , Unión Proteica/genética , Distribución Aleatoria , Reacción en Cadena en Tiempo Real de la Polimerasa/métodos , Valores de Referencia , Factores de Riesgo , Células Tumorales Cultivadas
5.
Biochem Biophys Res Commun ; 479(4): 628-635, 2016 Oct 28.
Artículo en Inglés | MEDLINE | ID: mdl-27697530

RESUMEN

BACKGROUND: Plasma levels of low-density lipoprotein cholesterol (LDL-C) are a major risk factor for cardiovascular disease and are influenced by both heredity and dietary habits. The Niemann-Pick C1 like 1 (NPC1L1) protein mediates efficient dietary cholesterol absorption and contributes to variations in human LDL-C levels. METHODS: In the present study, using high throughput sequencing we identified three non-synonymous (NS) variations and 64 synonymous variations in the NPC1L1 gene from subsets of Chinese Han, Uygur and Kazakh populations with high or low LDL-C. Subsequently, three NS variations encoding R174H, V177I and V1284L substitutions were observed only in Uygur and Kazakh individuals with limited maximal plasma LDL-C levels. RESULTS: In further experiments, we investigated cholesterol-regulated recycling and glycosylation and stability of these NS NPC1L1 variants. However, no significant differences between WT and variant NPC1L1 proteins were observed using in vivo assays in mouse livers with adenovirus-mediated expression, demonstrating that none of the three NPC1L1 NS variants caused decreased uptake of biliary cholesterol. CONCLUSIONS: Simultaneously, these data indicate that R174H, V177I and V1284L NPC1L1 variations in high or low LDL-C individuals may not directly influence cholesterol absorption by NPC1L1.


Asunto(s)
VLDL-Colesterol/sangre , Etnicidad/genética , Variación Genética , Hipercolesterolemia/genética , Proteínas de la Membrana/genética , Adulto , Animales , Línea Celular Tumoral , China/etnología , VLDL-Colesterol/genética , VLDL-Colesterol/metabolismo , Femenino , Humanos , Hipercolesterolemia/sangre , Reabsorción Intestinal/genética , Kazajstán/etnología , Hígado/metabolismo , Masculino , Proteínas de la Membrana/metabolismo , Proteínas de Transporte de Membrana , Ratones Endogámicos ICR , Persona de Mediana Edad , Sistemas de Lectura Abierta/genética , Ratas
6.
J Cell Sci ; 129(16): 3104-14, 2016 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-27358480

RESUMEN

Excitatory amino acid transporter type 3 (EAAT3, also known as SLC1A1) is a high-affinity, Na(+)-dependent glutamate carrier that localizes primarily within the cell and at the apical plasma membrane. Although previous studies have reported proteins and sequence regions involved in EAAT3 trafficking, the detailed molecular mechanism by which EAAT3 is distributed to the correct location still remains elusive. Here, we identify that the YVNGGF sequence in the C-terminus of EAAT3 is responsible for its intracellular localization and apical sorting in rat hepatoma cells CRL1601 and Madin-Darby canine kidney (MDCK) cells, respectively. We further demonstrate that Numb, a clathrin adaptor protein, directly binds the YVNGGF motif and regulates the localization of EAAT3. Mutation of Y503, N505 and F508 within the YVNGGF motif to alanine residues or silencing Numb by use of small interfering RNA (siRNA) results in the aberrant localization of EAAT3. Moreover, both Numb and the YVNGGF motif mediate EAAT3 endocytosis in CRL1601 cells. In summary, our study suggests that Numb is a pivotal adaptor protein that mediates the subcellular localization of EAAT3 through binding the YxNxxF (where x stands for any amino acid) motif.


Asunto(s)
Transportador 3 de Aminoácidos Excitadores/química , Transportador 3 de Aminoácidos Excitadores/metabolismo , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Secuencias de Aminoácidos , Animales , Perros , Endocitosis , Técnicas de Silenciamiento del Gen , Células HEK293 , Humanos , Masculino , Ratones Endogámicos BALB C , Mutación/genética , Unión Proteica , Transporte de Proteínas , Ratas , Relación Estructura-Actividad , Fracciones Subcelulares/metabolismo
7.
Cell ; 161(2): 291-306, 2015 Apr 09.
Artículo en Inglés | MEDLINE | ID: mdl-25860611

RESUMEN

Cholesterol is dynamically transported among organelles, which is essential for multiple cellular functions. However, the mechanism underlying intracellular cholesterol transport has remained largely unknown. We established an amphotericin B-based assay enabling a genome-wide shRNA screen for delayed LDL-cholesterol transport and identified 341 hits with particular enrichment of peroxisome genes, suggesting a previously unappreciated pathway for cholesterol transport. We show dynamic membrane contacts between peroxisome and lysosome, which are mediated by lysosomal Synaptotagmin VII binding to the lipid PI(4,5)P2 on peroxisomal membrane. LDL-cholesterol enhances such contacts, and cholesterol is transported from lysosome to peroxisome. Disruption of critical peroxisome genes leads to cholesterol accumulation in lysosome. Together, these findings reveal an unexpected role of peroxisome in intracellular cholesterol transport. We further demonstrate massive cholesterol accumulation in human patient cells and mouse model of peroxisomal disorders, suggesting a contribution of abnormal cholesterol accumulation to these diseases.


Asunto(s)
Colesterol/metabolismo , Lisosomas/metabolismo , Peroxisomas/metabolismo , ARN Interferente Pequeño/metabolismo , Transportadoras de Casetes de Unión a ATP/metabolismo , Adrenoleucodistrofia/metabolismo , Anfotericina B/farmacología , Animales , Transporte Biológico , Estudio de Asociación del Genoma Completo , Humanos , Ratones , Trastorno Peroxisomal/metabolismo , Trastorno Peroxisomal/patología , Fosfatidilinositol 4,5-Difosfato/metabolismo , Sinaptotagminas/metabolismo , Pez Cebra
8.
PLoS One ; 9(11): e112632, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25426949

RESUMEN

Somatic cell genetics is a powerful approach for unraveling the regulatory mechanism of cholesterol metabolism. However, it is difficult to identify the mutant gene(s) due to cells are usually mutagenized chemically or physically. To identify important genes controlling cholesterol biosynthesis, an unbiased forward genetics approach named validation-based insertional mutagenesis (VBIM) system was used to isolate and characterize the 25-hydroxycholesterol (25-HC)-resistant and SR-12813-resistant mutants. Here we report that five mutant cell lines were isolated. Among which, four sterol-resistant mutants either contain a truncated NH2-terminal domain of sterol regulatory element-binding protein (SREBP)-2 terminating at amino acids (aa) 400, or harbor an overexpressed SREBP cleavage-activating protein (SCAP). Besides, one SR-12813 resistant mutant was identified to contain a truncated COOH-terminal catalytic domain of 3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMG-CoA reductase). This study demonstrates that the VBIM system can be a powerful tool to screen novel regulatory genes in cholesterol biosynthesis.


Asunto(s)
Colesterol/biosíntesis , Hidroximetilglutaril-CoA Reductasas/genética , Péptidos y Proteínas de Señalización Intracelular/genética , Proteínas de la Membrana/genética , Mutagénesis Insercional/métodos , Proteína 2 de Unión a Elementos Reguladores de Esteroles/genética , Animales , Células CHO , Cricetulus , Difosfonatos/farmacología , Regulación de la Expresión Génica/efectos de los fármacos , Pruebas Genéticas/métodos , Vectores Genéticos , Células HEK293 , Células HeLa , Humanos , Hidroxicolesteroles/farmacología , Hidroximetilglutaril-CoA Reductasas/química , Hidroximetilglutaril-CoA Reductasas/metabolismo , Péptidos y Proteínas de Señalización Intracelular/química , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lentivirus/genética , Proteínas de la Membrana/química , Proteínas de la Membrana/metabolismo , Estructura Terciaria de Proteína , Transducción de Señal/efectos de los fármacos , Proteína 2 de Unión a Elementos Reguladores de Esteroles/química , Proteína 2 de Unión a Elementos Reguladores de Esteroles/metabolismo
9.
J Biol Chem ; 289(48): 33689-700, 2014 Nov 28.
Artículo en Inglés | MEDLINE | ID: mdl-25331956

RESUMEN

The uptake of circulating low density lipoproteins (LDL) is mediated by LDL receptor (LDLR) through clathrin-dependent endocytosis. At the early stage of this process, adaptor proteins ARH and Dab2 specifically bind the endocytic signal motif in LDLR and recruit clathrin/AP2 to initiate internalization. On the other hand, intestinal cholesterol is absorbed by Niemann-Pick C1-Like 1 (NPC1L1) through clathrin-dependent endocytosis. Another adaptor protein, Numb recognizes the endocytic motif in NPC1L1 C terminus and couples NPC1L1 to endocytic machinery. The ARH, Dab2, and Numb proteins contain a homogeneous phosphotyrosine binding (PTB) domain that directly binds endocytic motifs. Because ARH, Dab2, and Numb are all PTB domain family members, the emerging mystery is whether these adaptors act complementally in LDLR and NPC1L1 endocytosis. Here, we found that ARH and Dab2 did not bind NPC1L1 and were not required for NPC1L1 internalization. Similarly, Numb lacked the ability to interact with the LDLR C terminus and was dispensable for LDL uptake. Only the Numb isoforms with shorter PTB domain could facilitate NPC1L1 endocytosis. Besides the reported function in intestinal cholesterol absorption, Numb also mediated cholesterol reabsorption from bile in liver. We further identified a Numb variant with G595D substitution in humans of low blood LDL-cholesterol. The G595D substitution impaired NPC1L1 internalization and cholesterol reabsorption, due to attenuating affinity of Numb to clathrin/AP2. These results demonstrate that Numb specifically regulates NPC1L1-mediated cholesterol absorption both in human intestine and liver, distinct from ARH and Dab2, which selectively participate in LDLR-mediated LDL uptake.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , LDL-Colesterol/metabolismo , Endocitosis/fisiología , Proteínas de la Membrana/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Receptores de LDL/metabolismo , Proteínas Supresoras de Tumor/metabolismo , Proteínas Adaptadoras Transductoras de Señales/genética , Sustitución de Aminoácidos , Animales , Proteínas Reguladoras de la Apoptosis , Transporte Biológico Activo/fisiología , Línea Celular Tumoral , LDL-Colesterol/genética , Humanos , Proteínas de la Membrana/genética , Proteínas de Transporte de Membrana , Mutación Missense , Proteínas del Tejido Nervioso/genética , Ratas , Receptores de LDL/genética , Proteínas Supresoras de Tumor/genética
10.
Cell Metab ; 16(2): 213-25, 2012 Aug 08.
Artículo en Inglés | MEDLINE | ID: mdl-22863805

RESUMEN

gp78 is a membrane-anchored ubiquitin ligase mediating the degradation of HMG-CoA reductase (HMGCR) and Insig-1. As a rate-limiting enzyme in cholesterol biosynthesis, HMGCR undergoes rapid sterol-promoted degradation. In contrast, destruction of Insig-1 releases its inhibition on SREBP and stimulates the expression of lipogenic genes. Thus, gp78 has opposite effects on lipid biosynthesis. We here generated liver-specific gp78 knockout (L-gp78(-/-)) mice and showed that although the degradation of HMGCR was blunted, SREBP was suppressed due to the elevation of Insig-1/-2, and therefore the lipid biosynthesis was decreased. The L-gp78(-/-) mice were protected from diet-/age-induced obesity and glucose intolerance. The livers of L-gp78(-/-) mice produced more FGF21, which activated thermogenesis in brown adipocytes and enhanced energy expenditure. Together, the major function of gp78 in liver is regulating lipid biosynthesis through SREBP pathway. Ablation of gp78 decreases the lipid levels and increases FGF21, and is beneficial to patients with metabolic diseases.


Asunto(s)
Hiperlipidemias/genética , Resistencia a la Insulina/genética , Lípidos/biosíntesis , Hígado/metabolismo , Receptores del Factor Autocrino de Motilidad/deficiencia , Proteínas de Unión a los Elementos Reguladores de Esteroles/antagonistas & inhibidores , Animales , Glucemia , Cromatografía Liquida , Factores de Crecimiento de Fibroblastos/metabolismo , Hidroximetilglutaril-CoA Reductasas/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Noqueados , Receptores del Factor Autocrino de Motilidad/metabolismo
11.
J Biol Chem ; 286(28): 25088-97, 2011 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-21602275

RESUMEN

Niemann-Pick C1-like 1 (NPC1L1) is a multitransmembrane protein playing a crucial role in dietary and biliary cholesterol absorption. Cholesterol promotes the formation and endocytosis of NPC1L1-flotillin-cholesterol membrane microdomains, which is an early step in cholesterol uptake. How cholesterol is sensed in this step is unknown. Here, we find that the N-terminal domain (NTD) of NPC1L1 binds cholesterol. Mutation of residue Leu-216 in NPC1L1-NTD eliminates cholesterol binding, decreases the formation of NPC1L1-flotillin-cholesterol membrane microdomains, and prevents NPC1L1-mediated cholesterol uptake in culture cells and mice livers. NPC1L1-NTD specifically binds cholesterol but not plant sterols, which may account for the selective cholesterol absorption in intestine. Furthermore, 25- or 27-hydroxycholesterol competes with cholesterol to bind NPC1L1-NTD and inhibits the cholesterol induced endocytosis of NPC1L1. Together, these results demonstrate that plasma membrane-localized NPC1L1 binds exogenous cholesterol via its NTD, and facilitates the formation of NPC1L1-flotillin-cholesterol membrane microdomains that are then internalized into cells through the clathrin-AP2 pathway. Our study uncovers the mechanism of cholesterol sensing by NPC1L1 and proposes a mechanism for selective cholesterol absorption.


Asunto(s)
Colesterol/metabolismo , Endocitosis/fisiología , Hígado/metabolismo , Microdominios de Membrana/metabolismo , Proteínas de la Membrana/metabolismo , Proteínas de Transporte de Membrana/metabolismo , Absorción/fisiología , Complejo 2 de Proteína Adaptadora/genética , Complejo 2 de Proteína Adaptadora/metabolismo , Animales , Transporte Biológico Activo/fisiología , Colesterol/genética , Clatrina/genética , Clatrina/metabolismo , Células HEK293 , Humanos , Microdominios de Membrana/genética , Proteínas de la Membrana/genética , Proteínas de Transporte de Membrana/genética , Ratones , Unión Proteica , Estructura Terciaria de Proteína
12.
Proc Natl Acad Sci U S A ; 108(2): 551-6, 2011 Jan 11.
Artículo en Inglés | MEDLINE | ID: mdl-21187433

RESUMEN

Dietary absorption is a major way for mammals to obtain cholesterol, which is mediated by Niemann-Pick C1-like 1 (NPC1L1) via vesicular endocytosis. One fundamental question in this process is how free cholesterol is efficiently taken up through the internalization of NPC1L1. Using exogenously expressed NPC1L1-EGFP, we show that the lipid raft proteins flotillins associate with NPC1L1 and their localization is regulated by NPC1L1 during intracellular trafficking. Furthermore, flotillins are essential for NPC1L1-mediated cellular cholesterol uptake, biliary cholesterol reabsorption, and the regulation of lipid levels in mice. Together with NPC1L1, they form cholesterol-enriched membrane microdomains, which function as carriers for bulk of cholesterol. The hypocholesterolemic drug ezetimibe disrupts the association between NPC1L1 and flotillins, which blocks the formation of the cholesterol-enriched microdomains. Our findings reveal a functional role of flotillins in NPC1L1-mediated cholesterol uptake and elucidate the formation of NPC1L1-flotillins-postive cholesterol-enriched membrane microdomains as a mechanism for efficient cholesterol absorption.


Asunto(s)
Colesterol/metabolismo , Proteínas de la Membrana/metabolismo , Enfermedades de Niemann-Pick/metabolismo , Animales , Anticolesterolemiantes/farmacología , Azetidinas/farmacología , Colesterol/química , Ezetimiba , Proteínas Fluorescentes Verdes/metabolismo , Homeostasis , Humanos , Proteínas de Transporte de Membrana/metabolismo , Ratones , Microscopía Confocal/métodos , Microscopía Fluorescente/métodos , Ratas
13.
J Biol Chem ; 284(33): 22481-22490, 2009 Aug 14.
Artículo en Inglés | MEDLINE | ID: mdl-19542231

RESUMEN

Niemann-Pick C1-like 1 (NPC1L1) plays a critical role in the enterohepatic absorption of free cholesterol. Cellular cholesterol depletion induces the transport of NPC1L1 from the endocytic recycling compartment to the plasma membrane (PM), and cholesterol replenishment causes the internalization of NPC1L1 together with cholesterol via clathrin-mediated endocytosis. Although NPC1L1 has been characterized, the other proteins involved in cholesterol absorption and the endocytic recycling of NPC1L1 are largely unknown. Most of the vesicular trafficking events are dependent on the cytoskeleton and motor proteins. Here, we investigated the roles of the microfilament and microfilament-associated triple complex composed of myosin Vb, Rab11a, and Rab11-FIP2 in the transport of NPC1L1 from the endocytic recycling compartment to the PM. Interfering with the dynamics of the microfilament by pharmacological treatment delayed the transport of NPC1L1 to the cell surface. Meanwhile, inactivation of any component of the myosin Vb.Rab11a.Rab11-FIP2 triple complex inhibited the export of NPC1L1. Expression of the dominant-negative mutants of myosin Vb, Rab11a, or Rab11-FIP2 decreased the cellular cholesterol uptake by blocking the transport of NPC1L1 to the PM. These results suggest that the efficient transport of NPC1L1 to the PM is dependent on the microfilament-associated myosin Vb.Rab11a.Rab11-FIP2 triple complex.


Asunto(s)
Proteínas Portadoras/química , Colesterol/metabolismo , Proteínas de la Membrana/química , Proteínas de Transporte de Membrana/química , Miosinas/química , Proteínas de Unión al GTP rab/química , Citoesqueleto de Actina/química , Animales , Línea Celular Tumoral , Membrana Celular/metabolismo , Colesterol/química , Endocitosis , Genes Dominantes , Modelos Biológicos , Transporte de Proteínas , ARN Interferente Pequeño/metabolismo , Ratas
14.
Cell Metab ; 7(6): 508-19, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18522832

RESUMEN

Niemann-Pick C1-like 1 (NPC1L1) is a polytopic transmembrane protein that plays a critical role in cholesterol absorption. Ezetimibe, a hypocholesterolemic drug, has been reported to bind NPC1L1 and block cholesterol absorption. However, the molecular mechanism of NPC1L1-mediated cholesterol uptake and how ezetimibe inhibits this process are poorly defined. Here we find that cholesterol specifically promotes the internalization of NPC1L1 and that this process requires microfilaments and the clathrin/AP2 complex. Blocking NPC1L1 endocytosis dramatically decreases cholesterol internalization, indicating that NPC1L1 mediates cholesterol uptake via its vesicular endocytosis. Ezetimibe prevents NPC1L1 from incorporating into clathrin-coated vesicles and thus inhibits cholesterol uptake. Together, our data suggest a model wherein cholesterol is internalized into cells with NPC1L1 through clathrin/AP2-mediated endocytosis and ezetimibe inhibits cholesterol absorption by blocking the internalization of NPC1L1.


Asunto(s)
Azetidinas/farmacología , Colesterol/metabolismo , Endocitosis/efectos de los fármacos , Absorción Intestinal/efectos de los fármacos , Proteínas de la Membrana/metabolismo , Animales , Anticolesterolemiantes/farmacología , Línea Celular , Colesterol/fisiología , Vesículas Cubiertas por Clatrina/metabolismo , Clonación Molecular , Ezetimiba , Humanos , Hígado/citología , Proteínas de la Membrana/antagonistas & inhibidores , Proteínas de la Membrana/genética , Proteínas de Transporte de Membrana , Ratas
15.
Cell Metab ; 6(2): 115-28, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17681147

RESUMEN

The membrane-anchored ubiquitin ligase gp78 promotes degradation of misfolded endoplasmic reticulum (ER) proteins and sterol-regulated degradation of HMG-CoA reductase. It was known previously that Ufd1 plays a critical role in ER-associated degradation (ERAD) together with Npl4 and VCP. The VCP-Ufd1-Npl4 complex recognizes polyubiquitin chains and transfers the ubiquitinated proteins to the proteasome. Here we show that Ufd1 directly interacts with gp78 and functions as a cofactor. Ufd1 enhances the E3 activity of gp78, accelerates the ubiquitination and degradation of reductase, and eventually promotes receptor-mediated uptake of low-density lipoprotein. Furthermore, we demonstrate that the monoubiquitin-binding site in Ufd1 is required for the enhancement of gp78 activity and that the polyubiquitin-binding site in Ufd1 is critical for a postubiquitination step in ERAD. In summary, our study identifies Ufd1 as a cofactor of gp78, reveals an unappreciated function of Ufd1 in the ubiquitination reaction during ERAD, and illustrates that Ufd1 plays a critical role in cholesterol metabolism.


Asunto(s)
Colesterol/metabolismo , Hidroximetilglutaril-CoA Reductasas/metabolismo , Proteínas/metabolismo , Receptores de Citocinas/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo , Proteínas Adaptadoras del Transporte Vesicular , Aminoácidos , Animales , Sitios de Unión , Células CHO , Línea Celular , Cricetinae , Cricetulus , Estabilidad de Enzimas , Humanos , Péptidos y Proteínas de Señalización Intracelular , Lipoproteínas LDL/metabolismo , Modelos Biológicos , Unión Proteica , Mapeo de Interacción de Proteínas , Procesamiento Proteico-Postraduccional , Estructura Terciaria de Proteína , Receptores del Factor Autocrino de Motilidad , Receptores de Citocinas/química , Ubiquitina/metabolismo , Ubiquitina-Proteína Ligasas/química
16.
Biochemistry ; 42(17): 5143-50, 2003 May 06.
Artículo en Inglés | MEDLINE | ID: mdl-12718558

RESUMEN

Neurogranin/RC3 is a neuron-specific, Ca(2+)-sensitive calmodulin binding protein and a specific protein kinase C substrate. Neurogranin may function to regulate calmodulin levels at specific sites in neurons through phosphorylation at serine residue within its IQ motif, oxidation outside the IQ motif, or changes in local cellular Ca(2+) concentration. To gain insight into the functional role of neurogranin in the regulation of calmodulin-dependent activities, we investigated the structure and dynamics of a full-length rat neurogranin protein with 78 amino acids using triple resonance NMR techniques. In the absence of calmodulin or PKC, neurogranin exists in an unfolded form as evidenced by high backbone mobility and the absence of long-range nuclear Overhauser effect (NOE). Analyses of the chemical shifts (13)C(alpha), (13)C(beta), and (1)H(alpha) reveal the presence of a local alpha-helical structure for the region between residues G25-A42. Three-bond (1)H(N)-(1)H(alpha) coupling constants support the finding that the sequence between residues G25 and A42 populates a non-native helical structure in the unfolded neurogranin. Homonuclear NOE results are consistent with the conclusions drawn from chemical shifts and coupling constants. (15)N relaxation data indicate motional restrictions on a nanosecond time scale in the region from D15 to S48. Spectral densities and order parameters data further confirm that the unfolded neurogranin exists in conformation with residual secondary structures. The medium mobility of the nascent helical region may help to reduce the entropy loss when neurogranin binds to its targets, but the complex between neurogranin and calmodulin is not stable enough for structural determination by NMR. Calmodulin titration of neurogranin indicates that residues D15-G52 of neurogranin undergo significant structural changes upon binding to calmodulin.


Asunto(s)
Proteínas de Unión a Calmodulina/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Proteína Quinasa C/metabolismo , Serina , Animales , Sitios de Unión , Proteínas de Unión a Calmodulina/química , Proteínas de Unión a Calmodulina/genética , Clonación Molecular , ADN Complementario/genética , ADN Complementario/metabolismo , Espectroscopía de Resonancia por Spin del Electrón , Espectroscopía de Resonancia Magnética , Proteínas del Tejido Nervioso/química , Proteínas del Tejido Nervioso/genética , Neurogranina , Neuronas/metabolismo , Fosforilación , Conformación Proteica , Ratas , Proteínas Recombinantes/química , Proteínas Recombinantes/metabolismo , Especificidad por Sustrato , Termodinámica , Transfección
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...