Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
1.
J Lipid Res ; 54(6): 1550-1559, 2013 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-23564733

RESUMO

The LDL receptor (LDLR) relies upon endocytic adaptor proteins for internalization of lipoproteins. The results of this study show that the LDLR adaptor autosomal recessive hypercholesterolemia protein (ARH) requires nitric oxide to support LDL uptake. Nitric oxide nitrosylates ARH at C199 and C286, and these posttranslational modifications are necessary for association of ARH with the adaptor protein 2 (AP-2) component of clathrin-coated pits. In the absence of nitrosylation, ARH is unable to target LDL-LDLR complexes to coated pits, resulting in poor LDL uptake. The role of nitric oxide on LDLR function is specific for ARH because inhibition of nitric oxide synthase activity impairs ARH-supported LDL uptake but has no effect on other LDLR-dependent lipoprotein uptake processes, including VLDL remnant uptake and dab2-supported LDL uptake. These findings suggest that cells that depend upon ARH for LDL uptake can control which lipoproteins are internalized by their LDLRs through changes in nitric oxide.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Lipoproteínas LDL/metabolismo , Óxido Nítrico/metabolismo , Receptores de LDL/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Células HEK293 , Humanos , Lipoproteínas LDL/genética , Lipoproteínas VLDL/genética , Lipoproteínas VLDL/metabolismo , Microdomínios da Membrana/genética , Microdomínios da Membrana/metabolismo , Camundongos , Camundongos Knockout , Óxido Nítrico/genética , Receptores de LDL/genética
2.
J Biol Chem ; 285(9): 6706-15, 2010 Feb 26.
Artigo em Inglês | MEDLINE | ID: mdl-20034933

RESUMO

Obesity and insulin resistance are associated with deposition of triglycerides in tissues other than adipose tissue. Previously, we showed that a missense mutation (I148M) in PNPLA3 (patatin-like phospholipase domain-containing 3 protein) is associated with increased hepatic triglyceride content in humans. Here we examined the effect of the I148M substitution on the enzymatic activity and cellular location of PNPLA3. Structural modeling predicted that the substitution of methionine for isoleucine at residue 148 would restrict access of substrate to the catalytic serine at residue 47. In vitro assays using recombinant PNPLA3 partially purified from Sf9 cells confirmed that the wild type enzyme hydrolyzes emulsified triglyceride and that the I148M substitution abolishes this activity. Expression of PNPLA3-I148M, but not wild type PNPLA3, in cultured hepatocytes or in the livers of mice increased cellular triglyceride content. Cell fractionation studies revealed that approximately 90% of wild type PNPLA3 partitioned between membranes and lipid droplets; substitution of isoleucine for methionine at position 148 did not alter the subcellular distribution of the protein. These data are consistent with PNPLA3-I148M promoting triglyceride accumulation by limiting triglyceride hydrolysis.


Assuntos
Fígado Gorduroso/genética , Lipase/genética , Proteínas de Membrana/genética , Mutação de Sentido Incorreto , Triglicerídeos/metabolismo , Animais , Linhagem Celular , Hepatócitos/metabolismo , Humanos , Hidrólise , Fígado/metabolismo , Camundongos
3.
Proc Natl Acad Sci U S A ; 105(35): 13045-50, 2008 Sep 02.
Artigo em Inglês | MEDLINE | ID: mdl-18753623

RESUMO

Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a secreted protein that controls plasma LDL cholesterol levels by posttranslational regulation of the LDL receptor (LDLR). Previously, we showed that PCSK9 binds specifically to an EGF-like repeat (EGF-A) in LDLR and reroutes the receptor from endosomes to lysosomes rather than to the cell surface. Here, we defined the regions in LDLR and PCSK9 that are required for receptor degradation and examined the relationship between PCSK9 binding and LDLR conformation. Addition of PCSK9 to cultured hepatocytes promoted degradation of WT LDLR and of receptors lacking up to four ligand binding domains, EGF-B or the clustered O-linked sugar region. In contrast, LDLRs lacking the entire ligand binding domain or the beta-propeller domain failed to be degraded, although they bound and internalized PCSK9. Using gel filtration chromatography, we assessed the effects of PCSK9 binding on an acid-dependent conformational change that happens in the extracellular domain of the LDLR. Although PCSK9 prevented the reduction in hydrodynamic radius of the receptor that occurs at a reduced pH, the effect was not sufficient for LDLR degradation. A truncated version of PCSK9 containing the prodomain and the catalytic domain, but not the C-terminal domain, bound the receptor but did not stimulate LDLR degradation. Thus, domains in both the LDLR and PCSK9 that are not required for binding (or internalization) are essential for PCSK9-mediated degradation of the LDLR.


Assuntos
Processamento de Proteína Pós-Traducional , Receptores de LDL/metabolismo , Serina Endopeptidases/química , Serina Endopeptidases/metabolismo , Ácidos , Sequência de Aminoácidos , Animais , Domínio Catalítico , Linhagem Celular , Endocitose , Fator de Crescimento Epidérmico/química , Hepatócitos/metabolismo , Humanos , Radioisótopos do Iodo , Camundongos , Dados de Sequência Molecular , Proteínas Mutantes/metabolismo , Pró-Proteína Convertase 9 , Pró-Proteína Convertases , Ligação Proteica , Homologia de Sequência de Aminoácidos
4.
J Clin Invest ; 117(1): 165-74, 2007 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-17200716

RESUMO

Genetic defects in LDL clearance result in severe hypercholesterolemia and premature atherosclerosis. Mutations in the LDL receptor (LDLR) cause familial hypercholesterolemia (FH), the most severe form of genetic hypercholesterolemia. A phenocopy of FH, autosomal recessive hypercholesterolemia (ARH), is due to mutations in an adaptor protein involved in LDLR internalization. Despite comparable reductions in LDL clearance rates, plasma LDL levels are substantially lower in ARH than in FH. To determine the metabolic basis for this difference, we examined the synthesis and catabolism of VLDL in murine models of FH (Ldlr(-/-)) and ARH (Arh(-/-)). The hyperlipidemic response to a high-sucrose diet was greatly attenuated in Arh(-/-) mice compared with Ldlr(-/-) mice despite similar rates of VLDL secretion. The rate of VLDL clearance was significantly higher in Arh(-/-) mice than in Ldlr(-/-) mice, suggesting that LDLR-dependent uptake of VLDL is maintained in the absence of ARH. Consistent with these findings, hepatocytes from Arh(-/-) mice (but not Ldlr(-/-) mice) internalized beta-migrating VLDL (beta-VLDL). These results demonstrate that ARH is not required for LDLR-dependent uptake of VLDL by the liver. The preservation of VLDL remnant clearance attenuates the phenotype of ARH and likely contributes to greater responsiveness to statins in ARH compared with FH.


Assuntos
Hiperlipoproteinemia Tipo II/genética , Lipoproteínas LDL/sangue , Lipoproteínas VLDL/sangue , Receptores de LDL/sangue , Animais , Colesterol/sangue , Colesterol/metabolismo , Genes Recessivos , Hiperlipoproteinemia Tipo II/sangue , Hiperlipoproteinemia Tipo II/metabolismo , Lipoproteínas/sangue , Lipoproteínas/metabolismo , Lipoproteínas VLDL/metabolismo , Fígado/metabolismo , Camundongos , Camundongos Knockout , Receptores de LDL/deficiência , Receptores de LDL/genética
5.
J Clin Invest ; 116(11): 2995-3005, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17080197

RESUMO

Proprotein convertase subtilisin/kexin type 9 (PCSK9) is a member of the proteinase K subfamily of subtilases that reduces the number of LDL receptors (LDLRs) in liver through an undefined posttranscriptional mechanism. We show that purified PCSK9 added to the medium of HepG2 cells reduces the number of cell-surface LDLRs in a dose- and time-dependent manner. This activity was approximately 10-fold greater for a gain-of-function mutant, PCSK9(D374Y), that causes hypercholesterolemia. Binding and uptake of PCSK9 were largely dependent on the presence of LDLRs. Coimmunoprecipitation and ligand blotting studies indicated that PCSK9 and LDLR directly associate; both proteins colocalized to late endocytic compartments. Purified PCSK9 had no effect on cell-surface LDLRs in hepatocytes lacking autosomal recessive hypercholesterolemia (ARH), an adaptor protein required for endocytosis of the receptor. Transgenic mice overexpressing human PCSK9 in liver secreted large amounts of the protein into plasma, which increased plasma LDL cholesterol concentrations to levels similar to those of LDLR-knockout mice. To determine whether PCSK9 was active in plasma, transgenic PCSK9 mice were parabiosed with wild-type littermates. After parabiosis, secreted PCSK9 was transferred to the circulation of wild-type mice and reduced the number of hepatic LDLRs to nearly undetectable levels. We conclude that secreted PCSK9 associates with the LDLR and reduces hepatic LDLR protein levels.


Assuntos
Hepatócitos/metabolismo , Fígado/metabolismo , Receptores de LDL/metabolismo , Serina Endopeptidases/metabolismo , Animais , Ácido Aspártico/genética , Ácido Aspártico/metabolismo , Linhagem Celular , LDL-Colesterol/sangue , Endocitose , Regulação da Expressão Gênica , Genótipo , Humanos , Fígado/citologia , Camundongos , Mutação/genética , Oxirredução , Pró-Proteína Convertase 9 , Pró-Proteína Convertases , Ligação Proteica , Serina Endopeptidases/genética
6.
Mol Cell Biol ; 24(18): 8154-66, 2004 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-15340076

RESUMO

Cholesterol uptake and efflux are key metabolic processes associated with macrophage physiology and atherosclerosis. Peroxisome proliferator-activated receptor gamma (PPARgamma) and liver X receptor alpha (LXRalpha) have been linked to the regulation of these processes. It remains to be identified how activation of these receptors is connected and regulated by endogenous lipid molecules. We identified CYP27, a p450 enzyme, as a link between retinoid, PPARgamma, and LXR signaling. We show that the human CYP27 gene is under coupled regulation by retinoids and ligands of PPARs via a PPAR-retinoic acid receptor response element in its promoter. Induction of the enzyme's expression results in an increased level of 27-hydroxycholesterol and upregulation of LXR-mediated processes. Upregulated CYP27 activity also leads to LXR-independent elimination of CYP27 metabolites as an alternative means of cholesterol efflux. Moreover, human macrophage-rich atherosclerotic lesions have an increased level of retinoid-, PPARgamma-, and LXR-regulated gene expression and also enhanced CYP27 levels. Our findings suggest that nuclear receptor-regulated CYP27 expression is likely to be a key integrator of retinoic acid receptor-PPARgamma-LXR signaling, relying on natural ligands and contributing to lipid metabolism in macrophages.


Assuntos
Macrófagos/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Esteroide Hidroxilases/genética , Animais , Arteriosclerose/enzimologia , Arteriosclerose/metabolismo , Células Cultivadas , Colestanotriol 26-Mono-Oxigenase , Proteínas de Ligação a DNA , Regulação Enzimológica da Expressão Gênica/efeitos dos fármacos , Humanos , Hidroxicolesteróis/metabolismo , Ligantes , Receptores X do Fígado , Macrófagos/efeitos dos fármacos , Camundongos , Receptores Nucleares Órfãos , Regiões Promotoras Genéticas , Receptores Citoplasmáticos e Nucleares/agonistas , Receptores do Ácido Retinoico/metabolismo , Receptor alfa de Ácido Retinoico , Receptores X de Retinoides , Retinoides/farmacologia , Transdução de Sinais , Fatores de Transcrição/agonistas , Fatores de Transcrição/metabolismo , Transcrição Gênica
7.
Gene ; 283(1-2): 133-43, 2002 Jan 23.
Artigo em Inglês | MEDLINE | ID: mdl-11867220

RESUMO

Human sterol 27-hydroxylase catalyses the first step in the alternative pathway of bile acids biosynthesis in hepatocytes. However the gene encoding this enzyme (CYP27 gene) is expressed in every tissue and some evidence suggests that this enzyme plays a role in cholesterol homeostasis. Although modulation of CYP27 expression has been reported, the mechanisms underlying the regulation of this gene in human tissues is still poorly understood. To elucidate the mechanism governing CYP27 expression we cloned a 4.3 kb fragment of the 5' flanking region of the human CYP27 gene and constructed deletion mutants which were transfected into HepG2 cells. Functional assays showed that the -217/-10 nucleotide region from the translation start site (minimal promoter), devoid of TATA and CAAT boxes, contains all the elements for basal transcription. Foot-printing analysis of minimal promoter showed four protected regions (A-D). Regions A, B and D each contain one Sp1 binding site, and region C contains a HNF4 site. Electrophoretic mobility shift assays demonstrated that Sp1, Sp3 and HNF4 transcription factors bind these sites. Mutagenesis of any of these sites resulted in the loss of promoter activity. Co-transfection of the minimal promoter with Sp1 and Sp3 expression vectors transactivated CYP27 gene promoter in Drosophila SL2 cells, which lack endogenous Sp proteins. Transactivation of the minimal promoter was also observed in HeLa cells co-transfected with HNF4 expression vector. Therefore, Sp1, Sp3 and HNF4 co-operate in the expression of the human CYP27 gene in HepG2 cells.


Assuntos
Sistema Enzimático do Citocromo P-450/genética , Proteínas de Ligação a DNA , Regiões Promotoras Genéticas/genética , Esteroide Hidroxilases/genética , Região 5'-Flanqueadora/genética , Animais , Sequência de Bases , Fatores de Transcrição de Zíper de Leucina e Hélice-Alça-Hélix Básicos , Sítios de Ligação/genética , Carcinoma Hepatocelular/genética , Carcinoma Hepatocelular/patologia , Linhagem Celular , Cloranfenicol O-Acetiltransferase/genética , Cloranfenicol O-Acetiltransferase/metabolismo , Colestanotriol 26-Mono-Oxigenase , DNA/genética , DNA/metabolismo , Pegada de DNA , Desoxirribonuclease I/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Células HeLa , Fator 4 Nuclear de Hepatócito , Humanos , Dados de Sequência Molecular , Mutagênese Sítio-Dirigida , Mutação , Fosfoproteínas/genética , Fosfoproteínas/metabolismo , Ligação Proteica , Proteínas Recombinantes de Fusão/genética , Proteínas Recombinantes de Fusão/metabolismo , Deleção de Sequência , Fator de Transcrição Sp1/genética , Fator de Transcrição Sp1/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Células Tumorais Cultivadas
8.
Orphanet J Rare Dis ; 5: 27, 2010 Oct 06.
Artigo em Inglês | MEDLINE | ID: mdl-20925952

RESUMO

Mutations of the gene encoding the mitochondrial enzyme sterol 27-hydroxylase (CYP27A1 gene) cause defects in the cholesterol pathway to bile acids that lead to the storage of cholestanol and cholesterol in tendons, lenses and the central nervous system. This disorder is the cause of a clinical syndrome known as cerebrotendinous xanthomatosis (CTX). Since 1991 several mutations of the CYP27A1 gene have been reported. We diagnosed the clinical features of CTX in a caucasian woman. Serum levels of cholestanol and 7α-hydroxycholesterol were elevated and the concentration of 27-hydroxycholesterol was reduced. Bile alcohols in the urine and faeces were increased. The analysis of the CYP27A1 gene showed that the patient was a compound heterozygote carrying two mutations both located in exon 8. One mutation is a novel four nucleotide deletion (c.1330-1333delTTCC) that results in a frameshift and the occurrence of a premature stop codon leading to the formation of a truncated protein of 448 amino acids. The other mutation, previously reported, is a C - > T transition (c. c.1381C > T) that converts the glutamine codon at position 461 into a termination codon (p.Q461X). These truncated proteins are expected to have no biological function being devoid of the cysteine residue at position 476 of the normal enzyme that is crucial for heme binding and enzyme activity.


Assuntos
Colestanotriol 26-Mono-Oxigenase/genética , Mutação , Xantomatose Cerebrotendinosa/genética , Adulto , Sequência de Bases , Éxons , Feminino , Genes Recessivos , Heterozigoto , Humanos , Reação em Cadeia da Polimerase , Análise de Sequência de DNA , Xantomatose Cerebrotendinosa/tratamento farmacológico , Xantomatose Cerebrotendinosa/enzimologia , Xantomatose Cerebrotendinosa/fisiopatologia
9.
J Clin Invest ; 119(11): 3278-89, 2009 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-19841541

RESUMO

Renal outer medullary potassium (ROMK) channels are exquisitely regulated to adjust renal potassium excretion and maintain potassium balance. Clathrin-dependent endocytosis plays a critical role, limiting urinary potassium loss in potassium deficiency. In renal disease, aberrant ROMK endocytosis may contribute to potassium retention and hyperkalemia. Previous work has indicated that ROMK endocytosis is stimulated by with-no-lysine (WNK) kinases, but the endocytotic signal and the internalization machinery have not been defined. Here, we found that ROMK bound directly to the clathrin adaptor molecule autosomal recessive hypercholesterolemia (ARH), and this interaction was mediated by what we believe to be a novel variant of the canonical "NPXY" endocytotic signal, YxNPxFV. ARH recruits ROMK to clathrin-coated pits for constitutive and WNK1-stimuated endocytosis, and ARH knockdown decreased basal rates of ROMK endocytosis, in a heterologous expression system, COS-7 cells. We found that ARH was predominantly expressed in the distal nephron where it coimmunoprecipitated and colocalized with ROMK. In mice, the abundance of kidney ARH protein was modulated by dietary potassium and inversely correlated with changes in ROMK. Furthermore, ARH-knockout mice exhibited an altered ROMK response to potassium intake. These data suggest that ARH marks ROMK for clathrin-dependent endocytosis, in concert with the demands of potassium homeostasis.


Assuntos
Proteínas Adaptadoras de Transporte Vesicular/metabolismo , Endocitose/fisiologia , Rim/metabolismo , Canais de Potássio/metabolismo , Proteínas Adaptadoras de Transporte Vesicular/genética , Motivos de Aminoácidos , Animais , Células COS , Chlorocebus aethiops , Regulação da Expressão Gênica , Técnicas de Silenciamento de Genes , Camundongos , Potássio/metabolismo , Isoformas de Proteínas
10.
EMBO J ; 26(14): 3273-82, 2007 Jul 25.
Artigo em Inglês | MEDLINE | ID: mdl-17581630

RESUMO

The low-density lipoprotein (LDL) receptor (LDLR) binds to and internalizes lipoproteins that contain apolipoproteinB100 (apoB100) or apolipoproteinE (apoE). Internalization of the apoB100 lipoprotein ligand, LDL, requires the FDNPVY(807) sequence on the LDLR cytoplasmic domain, which binds to the endocytic machinery of coated pits. We show here that inactivation of the FDNPVY(807) sequence by mutation of Y807 to cysteine prevented the uptake of LDL; however, this mutation did not prevent LDLR-dependent uptake of the apoE lipoprotein ligand, beta-VLDL. Comparison of the surface localization of the LDLR-Y807C using LDLR-immunogold, LDL-gold and beta-VLDL-gold probes revealed enrichment of LDLR-Y807C-bound beta-VLDL in coated pits, suggesting that beta-VLDL binding promoted the internalization of the LDLR-Y807C. Consistent with this possibility, treatment with monensin, which traps internalized LDLR in endosomes, resulted in the loss of surface LDLR-Y807C only when beta-VLDL was present. Reconstitution experiments in which LDLR variants were introduced into LDLR-deficient cells showed that the HIC(818) sequence is involved in beta-VLDL uptake by the LDLR-Y807C. Together, these experiments demonstrate that the LDLR has a very low-density lipoprotein (VLDL)-induced, FDNPVY-independent internalization mechanism.


Assuntos
Endocitose/efeitos dos fármacos , Lipoproteínas VLDL/farmacologia , Receptores de LDL/metabolismo , Sequência de Aminoácidos , Animais , Células Cultivadas , Vesículas Revestidas por Clatrina/efeitos dos fármacos , Vesículas Revestidas por Clatrina/ultraestrutura , Endossomos/efeitos dos fármacos , Endossomos/ultraestrutura , Fibroblastos/citologia , Fibroblastos/efeitos dos fármacos , Fibroblastos/ultraestrutura , Lipoproteínas LDL/metabolismo , Camundongos , Dados de Sequência Molecular , Mutação/genética , Receptores de LDL/química , Receptores de LDL/deficiência
11.
J Biol Chem ; 282(25): 18602-18612, 2007 Jun 22.
Artigo em Inglês | MEDLINE | ID: mdl-17452316

RESUMO

Proprotein convertase subtilisin/kexin type 9 (PCSK9) promotes degradation of hepatic low density lipoprotein receptors (LDLR), the major route of clearance of circulating cholesterol. Gain-of-function mutations in PCSK9 cause hypercholesterolemia and premature atherosclerosis, whereas loss-of-function mutations result in hypocholesterolemia and protection from heart disease. Recombinant human PCSK9 binds the LDLR on the surface of cultured hepatocytes and promotes degradation of the receptor after internalization. Here we localized the site of binding of PCSK9 within the extracellular domain of the LDLR and determined the fate of the receptor after PCSK9 binding. Recombinant human PCSK9 interacted in a sequence-specific manner with the first epidermal growth factor-like repeat (EGF-A) in the EGF homology domain of the human LDLR. Similar binding specificity was observed between PCSK9 and purified EGF-A. Binding to EGF-A was calcium-dependent and increased dramatically with reduction in pH from 7 to 5.2. The addition of PCSK9, but not heat-inactivated PCSK9, to the medium of cultured hepatocytes resulted in redistribution of the receptor from the plasma membrane to lysosomes. These data are consistent with a model in which PCSK9 binding to EGF-A interferes with an acid-dependent conformational change required for receptor recycling. As a consequence, the LDLR is rerouted from the endosome to the lysosome where it is degraded.


Assuntos
Fator de Crescimento Epidérmico/metabolismo , Receptores de LDL/metabolismo , Serina Endopeptidases/fisiologia , Subtilisina/fisiologia , Sequência de Aminoácidos , Animais , Células COS , Células Cultivadas/metabolismo , Chlorocebus aethiops , Hepatócitos/metabolismo , Humanos , Lisossomos/metabolismo , Dados de Sequência Molecular , Pró-Proteína Convertase 9 , Pró-Proteína Convertases , Ligação Proteica , Conformação Proteica , Proteínas Recombinantes/química , Serina Endopeptidases/química
12.
Proc Natl Acad Sci U S A ; 102(15): 5374-9, 2005 Apr 12.
Artigo em Inglês | MEDLINE | ID: mdl-15805190

RESUMO

PCSK9 encodes proprotein convertase subtilisin/kexin type 9a (PCSK9), a member of the proteinase K subfamily of subtilases. Missense mutations in PCSK9 cause an autosomal dominant form of hypercholesterolemia in humans, likely due to a gain-of-function mechanism because overexpression of either WT or mutant PCSK9 reduces hepatic LDL receptor protein (LDLR) in mice. Here, we show that livers of knockout mice lacking PCSK9 manifest increased LDLR protein but not mRNA. Increased LDLR protein led to increased clearance of circulating lipoproteins and decreased plasma cholesterol levels (46 mg/dl in Pcsk9(-/-) mice versus 96 mg/dl in WT mice). Statins, a class of drugs that inhibit cholesterol synthesis, increase expression of sterol regulatory element-binding protein-2 (SREBP-2), a transcription factor that activates both the Ldlr and Pcsk9 genes. Statin administration to Pcsk9(-/-) mice produced an exaggerated increase in LDLRs in liver and enhanced LDL clearance from plasma. These data demonstrate that PCSK9 regulates the amount of LDLR protein in liver and suggest that inhibitors of PCSK9 may act synergistically with statins to enhance LDLRs and reduce plasma cholesterol.


Assuntos
Colesterol/sangue , Inibidores de Hidroximetilglutaril-CoA Redutases/farmacologia , Serina Endopeptidases/deficiência , Ração Animal , Animais , Apolipoproteínas B/sangue , Apolipoproteínas B/metabolismo , Células Cultivadas , Colesterol/metabolismo , Sinergismo Farmacológico , Eletroforese em Gel de Poliacrilamida , Feminino , Deleção de Genes , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Fígado/citologia , Fígado/efeitos dos fármacos , Fígado/metabolismo , Lovastatina/farmacologia , Masculino , Camundongos , Camundongos Knockout , Fenótipo , Pró-Proteína Convertase 9 , Pró-Proteína Convertases , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Receptores de LDL/deficiência , Receptores de LDL/genética , Receptores de LDL/metabolismo , Serina Endopeptidases/genética , Serina Endopeptidases/metabolismo
13.
Cell ; 122(6): 927-39, 2005 Sep 23.
Artigo em Inglês | MEDLINE | ID: mdl-16179260

RESUMO

Apoptosis and autophagy are both tightly regulated biological processes that play a central role in tissue homeostasis, development, and disease. The anti-apoptotic protein, Bcl-2, interacts with the evolutionarily conserved autophagy protein, Beclin 1. However, little is known about the functional significance of this interaction. Here, we show that wild-type Bcl-2 antiapoptotic proteins, but not Beclin 1 binding defective mutants of Bcl-2, inhibit Beclin 1-dependent autophagy in yeast and mammalian cells and that cardiac Bcl-2 transgenic expression inhibits autophagy in mouse heart muscle. Furthermore, Beclin 1 mutants that cannot bind to Bcl-2 induce more autophagy than wild-type Beclin 1 and, unlike wild-type Beclin 1, promote cell death. Thus, Bcl-2 not only functions as an antiapoptotic protein, but also as an antiautophagy protein via its inhibitory interaction with Beclin 1. This antiautophagy function of Bcl-2 may help maintain autophagy at levels that are compatible with cell survival, rather than cell death.


Assuntos
Autofagia/efeitos dos fármacos , Proteínas/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/farmacologia , Proteínas de Saccharomyces cerevisiae/efeitos dos fármacos , Animais , Apoptose/efeitos dos fármacos , Proteínas Reguladoras de Apoptose , Proteína Beclina-1 , Linhagem Celular , Retículo Endoplasmático/química , Regulação da Expressão Gênica , Células HeLa , Humanos , Proteínas de Membrana , Camundongos , Camundongos Transgênicos , Mitocôndrias/química , Mutação , Miócitos Cardíacos/citologia , Miócitos Cardíacos/efeitos dos fármacos , Miócitos Cardíacos/metabolismo , Fosfatidilinositol 3-Quinases/classificação , Fosfatidilinositol 3-Quinases/metabolismo , Inibidores de Fosfoinositídeo-3 Quinase , Proteínas/genética , Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/fisiologia , Saccharomyces cerevisiae/metabolismo , Proteínas de Saccharomyces cerevisiae/metabolismo , Inanição/prevenção & controle
14.
J Biol Chem ; 280(49): 40996-1004, 2005 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-16179341

RESUMO

Autosomal recessive hypercholesterolemia is characterized by a cell type-specific defect in low density lipoprotein receptor (LDLR) endocytosis. LDLR-mediated uptake of LDL is impaired in the liver, but not in fibroblasts of subjects with this disorder. The disease is caused by mutations in ARH, which encodes a putative adaptor protein that interacts with the cytoplasmic tail of the LDLR, phospholipids, and two components of the clathrin endocytic machinery, clathrin and adaptor protein-2 (AP-2) in vitro. To determine the physiological relevance of these interactions, we examined the effect of mutations in the ARH on LDLR location and function in polarized hepatocytes (WIF-B). The integrity of the FDNPVY sequence in the LDLR cytoplasmic tail was required for ARH-associated LDLR clustering into clathrin-coated pits. The phosphotyrosine binding domain of ARH plus either the clathrin box or the AP-2 binding region were required for both clustering and internalization of the LDLR. Parallel studies performed in vivo with the same recombinant forms of ARH in livers of Arh(-/-) mice confirmed the relevance of the cell culture findings. These results demonstrate that ARH must bind the LDLR tail and either clathrin or AP-2 to promote receptor clustering and internalization of LDL.


Assuntos
Proteínas Adaptadoras de Transdução de Sinal/fisiologia , Vesículas Revestidas por Clatrina/metabolismo , Receptores de LDL/metabolismo , Complexo 2 de Proteínas Adaptadoras/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/química , Proteínas Adaptadoras de Transdução de Sinal/deficiência , Proteínas Adaptadoras de Transdução de Sinal/genética , Animais , Sítios de Ligação , Fusão Celular , Linhagem Celular Transformada , Membrana Celular/química , Clatrina/metabolismo , Citoplasma/química , Fibroblastos , Imunofluorescência , Expressão Gênica , Hepatócitos , Humanos , Camundongos , Camundongos Knockout , Microscopia de Fluorescência , Microscopia Imunoeletrônica , Mutação , Ratos , Receptores de LDL/química , Receptores de LDL/genética , Proteínas Recombinantes/análise , Relação Estrutura-Atividade , Transfecção
15.
J Lipid Res ; 44(2): 254-64, 2003 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-12576507

RESUMO

Two point mutations of ABCA1 gene were found in a patient with Tangier disease (TD): i) G>C in intron 2 (IVS2 +5G>C) and ii) c.844 C>T in exon 9 (R282X). The IVS2 +5G>C mutation was also found in the brother of another deceased TD patient, but not in 78 controls and 33 subjects with low HDL. The IVS2 +5G>C mutation disrupts ABCA1 pre-mRNA splicing in fibroblasts, leading to three abnormal mRNAs: devoid of exon 2 (Ex2-/mRNA), exon 4 (Ex4-/mRNA), or both these exons (Ex2-/Ex4-/mRNA), each containing a translation initiation site. These mRNAs are expected either not to be translated or generate short peptides. To investigate the in vitro effect of IVS2 +5G>C mutation, we constructed two ABCA1 minigenes encompassing Ex1-Ex3 region, one with wild-type (WTgene) and the other with mutant (MTgene) intron 2. These minigenes were transfected into COS1 and NIH3T3, two cell lines with a different ABCA1 gene expression. In COS1 cells, WTgene pre-mRNA was spliced correctly, while the splicing of MTgene pre-mRNA resulted in Ex2-/mRNA. In NIH3T3, no splicing of MTgene pre-mRNA was observed, whereas WTgene pre-mRNA was spliced correctly. These results stress the complexity of ABCA1 pre-mRNA splicing in the presence of splice site mutations.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Processamento Alternativo , Mutação Puntual , RNA Mensageiro/metabolismo , Doença de Tangier/genética , Transportador 1 de Cassete de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Adulto , Idoso , Células Cultivadas , Criança , Colesterol/metabolismo , Feminino , Fibroblastos/citologia , Fibroblastos/metabolismo , Haplótipos , Humanos , Íntrons , Masculino , Pessoa de Meia-Idade , Linhagem , RNA Mensageiro/genética , Análise de Sequência de DNA , Doença de Tangier/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA