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











Base de dados
Intervalo de ano de publicação
1.
Lipids ; 54(11-12): 707-714, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31574565

RESUMO

Siphonaxanthin is a carotenoid found in certain green algae, and its promising beneficial properties, such as its anti-obesity effect, have recently been demonstrated. However, there is little information about the molecular mechanisms underlying intestinal absorption of siphonaxanthin. In this study, we aimed to elucidate how siphonaxanthin is transported across the intestinal epithelium using differentiated Caco-2 cells (dCaco-2 cells), recombinant proteins, and an animal model. Siphonaxanthin was taken up by dCaco-2 cells, a model of intestinal epithelial cells, and its uptake linearly increased up to at least 6 h. Pharmacological inhibition of Nieman-Pick C1-like 1 (NPC1L1), but not that of scavenger receptor class B type 1 (SR-B1), significantly suppressed siphonaxanthin uptake by dCaco-2 cells. Results from an in vitro binding assay suggested that the N-terminal domain of NPC1L1, which is an extracellular domain of NPC1L1, binds with siphonaxanthin. Moreover, pretreatment with ezetimibe, an inhibitor of NPC1L1, significantly decreased the plasma level of siphonaxanthin following oral administration in mice. Considered together, we concluded that NPC1L1 promotes siphonaxanthin transport across the intestinal epithelium.


Assuntos
Proteínas de Membrana Transportadoras/metabolismo , Xantofilas/metabolismo , Administração Oral , Animais , Células CACO-2 , Ezetimiba/administração & dosagem , Ezetimiba/farmacologia , Humanos , Absorção Intestinal/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Estrutura Molecular , Células Tumorais Cultivadas , Xantofilas/sangue , Xantofilas/química
2.
Am J Physiol Cell Physiol ; 316(4): C559-C566, 2019 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-30789754

RESUMO

Intestinal Niemann-Pick C1 Like 1 (NPC1L1) protein plays a key role in cholesterol absorption. A decrease in NPC1L1 expression has been implicated in lowering plasma cholesterol and mitigating the risk for coronary heart disease. Little is known about the mechanisms responsible for NPC1L1 protein degradation that upon activation may lead to a reduction in NPC1L1 protein levels in intestinal epithelial cells (IECs). In current studies, the human intestinal Caco-2 and HuTu-80 cell lines expressing NPC1L1-hemagglutinin fusion protein were used to investigate the mechanisms of NPC1L1 protein degradation. Incubation with the proteasome inhibitors MG-132 and lactacystin (10 µM, 24 h) significantly increased NPC1L1 protein levels in IECs. Also, the inhibition of the lysosomal pathway with bafilomycin A1 (80 nM, 24 h) resulted in a significant increase in NPC1L1 protein levels. Immunoprecipitation studies showed that NPC1L1 protein is both a poly- and monoubiquinated polypeptide and that the inhibition of the proteasomal pathway remarkably increased the level of the polyubiquinated NPC1L1. The surface expression of NPC1L1 was increased by the inhibition of both proteasomal and lysosomal pathways. Furthermore, the pharmacological inhibition of mitogen-activated protein kinase pathway (PD-98059, 15 µM, 24 h) and siRNA silencing of ERK1/2 resulted in a significant decrease in NPC1L1 protein levels in IECs. In conclusion, our results showed that basal level of intestinal cholesterol transporter NPC1L1 protein is modulated by both ubiquitin proteasome- and lysosome-dependent degradation as well as by ERK1/2-dependent pathway. The modulation of these pathways may provide novel clues for therapeutic intervention to inhibit cholesterol absorption and lower plasma cholesterol.


Assuntos
Células Epiteliais/metabolismo , Mucosa Intestinal/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Proteólise , Células CACO-2 , Células Epiteliais/patologia , Humanos , Mucosa Intestinal/patologia
3.
Sci Rep ; 7(1): 633, 2017 04 04.
Artigo em Inglês | MEDLINE | ID: mdl-28377633

RESUMO

Only free drugs have been believed to be carried into tissues through active or passive transport. However, considering that lipoproteins function as carriers of serum lipids such as cholesterol and triglycerides, we hypothesized that lipoproteins can associate with certain drugs and mediate their transport into tissues in lipid-associated form. Here, in vitro and in vivo studies with low density lipoprotein receptor (LDLR)-overexpressing or -knockdown cells and wild-type or LDLR-mutant mice were used to show the association of various drugs with lipoproteins and the uptake of lipoprotein-associated drugs through a lipoprotein receptor-mediated process. In clinical studies, investigation of the effect of lipoprotein apheresis on serum drug concentrations in patients with familial hypercholesterolemia demonstrated that lipoprotein-mediated drug transport occurs in humans as well as in mice. These findings represent a new concept regarding the transport and metabolism of drugs in the body and suggest that the role of lipoprotein-mediated drug transport should be considered when developing effective and safe pharmacotherapies.


Assuntos
Portadores de Fármacos , Lipoproteínas LDL/metabolismo , Lipoproteínas VLDL/metabolismo , Preparações Farmacêuticas/metabolismo , Anlodipino/farmacocinética , Animais , Transporte Biológico , Colesterol/sangue , Hiperlipoproteinemia Tipo II/sangue , Hiperlipoproteinemia Tipo II/tratamento farmacológico , Hiperlipoproteinemia Tipo II/metabolismo , Masculino , Camundongos , Ligação Proteica , Receptores de LDL/genética , Receptores de LDL/metabolismo , Ticlopidina/farmacocinética
4.
Artigo em Inglês | MEDLINE | ID: mdl-24940679

RESUMO

ATP-binding cassette transporter G2 (ABCG2), also known as breast cancer resistance protein (BCRP), is identified as a high-capacity urate exporter and its dysfunction has an association with serum uric acid (SUA) levels and gout/hyperuricemia risk. However, pathophysiologically important pathway(s) responsible for the ABCG2-mediated urate excretion were unknown. In this study, we investigated how ABCG2 dysfunction affected the urate excretion pathways. First, we revealed that mouse Abcg2 mediates urate transport using the membrane vesicle system. The export process by mouse Abcg2 was ATP-dependent and not saturable under the physiological concentration of urate. Then, we characterized the excretion of urate into urine, bile, and intestinal lumen using in vivo mouse model. SUA of Abcg2-knockout mice was significantly higher than that of control mice. Under this condition, the renal urate excretion was increased in Abcg2-knockout mice, whereas the urate excretion from the intestine was decreased to less than a half. Biliary urate excretion showed no significant difference regardless of Abcg2 genotype. From these results, we estimated the relative contribution of each pathway to total urate excretion; in wild-type mice, the renal excretion pathway contributes approximately two-thirds, the intestinal excretion pathway contributes one-third of the total urate excretion, and the urate excretion into bile is minor. Decreased intestinal excretion could account for the increased SUA of Abcg2-knockout mice. Thus, ABCG2 is suggested to have an important role in extra-renal urate excretion, especially in intestinal excretion. Accordingly, increased SUA in patients with ABCG2 dysfunction could be explained by the decreased excretion of urate from the intestine.


Assuntos
Transportadores de Cassetes de Ligação de ATP/genética , Mucosa Intestinal/metabolismo , Ácido Úrico/sangue , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/deficiência , Animais , Técnicas de Inativação de Genes , Células HEK293 , Humanos , Masculino , Camundongos , Ácido Úrico/metabolismo
5.
Mol Ther ; 22(1): 112-22, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24025748

RESUMO

Tumors are theoretically capable of eliciting an antitumor immune response, but are often poorly immunogenic. Oncolytic viruses (OVs) have recently emerged as a promising strategy for the immunogenic delivery of tumor-associated antigens (TAAs) to cancer patients. However, safe and effective OV/TAA therapies have not yet been established. We have previously demonstrated that vectors based on Sindbis virus (SV) can inhibit tumor growth and activate the innate immune system in mice. Here, we demonstrate that SV vectors carrying a TAA generate a dramatically enhanced therapeutic effect in mice bearing subcutaneous, intraperitoneal, and lung cancers. Notably, SV/TAA efficacy was not dependent on tumor cell targeting, but was characterized by the transient expression of TAAs in lymph nodes draining the injection site. Early T-cell activation at this site was followed by a robust influx of NKG2D expressing antigen-specific cytotoxic CD8+ T cells into the tumor site, subsequently leading to the generation of long-lasting memory T cells which conferred protection against rechallenge with TAA-positive as well as TAA-negative tumor cells. By combining in vivo imaging, flow cytometry, cytotoxicity/cytokine assays, and tetramer analysis, we investigated the relationship between these events and propose a model for CD8+ T-cell activation during SV/TAA therapy.


Assuntos
Antígenos de Neoplasias/genética , Antígenos de Neoplasias/imunologia , Linfócitos T CD8-Positivos/imunologia , Vetores Genéticos/genética , Linfonodos/imunologia , Neoplasias/genética , Neoplasias/imunologia , Sindbis virus/genética , Animais , Cricetinae , Citotoxicidade Imunológica , Modelos Animais de Doenças , Epitopos de Linfócito T/imunologia , Feminino , Expressão Gênica , Técnicas de Transferência de Genes , Genes Reporter , Vetores Genéticos/administração & dosagem , Vetores Genéticos/imunologia , Memória Imunológica , Linfonodos/metabolismo , Ativação Linfocitária , Camundongos , Neoplasias/patologia , Neoplasias/terapia , Vírus Oncolíticos/genética , Vírus Oncolíticos/imunologia , Sindbis virus/imunologia , Carga Tumoral/genética , Carga Tumoral/imunologia
6.
Biochim Biophys Acta ; 1831(2): 459-68, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23200860

RESUMO

STARD10, a member of the steroidogenic acute regulatory protein (StAR)-related lipid transfer (START) protein family, is highly expressed in the liver and has been shown to transfer phosphatidylcholine. Therefore it has been assumed that STARD10 may function in the secretion of phospholipids into the bile. To help elucidate the physiological role of STARD10, we produced Stard10 knockout mice (Stard10(-/-)) and studied their phenotype. Neither liver content nor biliary secretion of phosphatidylcholine was altered in Stard10(-/-) mice. Unexpectedly, the biliary secretion of bile acids from the liver and the level of taurine-conjugated bile acids in the bile were significantly higher in Stard10(-/-) mice than wild type (WT) mice. In contrast, the levels of the secondary bile acids were lower in the liver of Stard10(-/-) mice, suggesting that the enterohepatic cycling is impaired. STARD10 was also expressed in the gallbladder and small intestine where the expression level of apical sodium dependent bile acid transporter (ASBT) turned out to be markedly lower in Stard10(-/-) mice than in WT mice when measured under fed condition. Consistent with the above results, the fecal excretion of bile acids was significantly increased in Stard10(-/-) mice. Interestingly, PPARα-dependent genes responsible for the regulation of bile acid metabolism were down-regulated in the liver of Stard10(-/-) mice. The loss of STARD10 impaired the PPARα activity and the expression of a PPARα-target gene such as Cyp8b1 in mouse hepatoma cells. These results indicate that STARD10 is involved in regulating bile acid metabolism through the modulation of PPARα-mediated mechanism.


Assuntos
Ácidos e Sais Biliares/fisiologia , Homeostase , PPAR alfa/fisiologia , Fosfoproteínas/fisiologia , Animais , Primers do DNA , Camundongos , Camundongos Knockout , Fosfoproteínas/genética , RNA Interferente Pequeno
7.
Biochem Pharmacol ; 84(7): 925-35, 2012 Oct 01.
Artigo em Inglês | MEDLINE | ID: mdl-22820248

RESUMO

Ursodeoxycholic acid (UDCA) is a hepatoprotective bile acid used in the treatment of chronic liver diseases. Although several pharmacological effects, including choleresis and inhibition of apoptosis, have been proposed, the impact of UDCA on hepatic structure is not well understood. Here, the influence of UDCA on bile canalicular (BC) morphology was evaluated in vitro in immortalized rat hepatocytes (McA-RH 7777 cells) and primary rat hepatocytes. Cells cultured for 3 days in the presence of UDCA, the BC lumen was enlarged and the bile canaliculi were surrounded by multiple cells (≥5) with a continuous canal-like structure, reminiscent of the in vivo BC network. The effects were dependent on p38MAPK and conventional PKC in McA-RH cells, and partially dependent on p38MAPK, MAPK/ERK kinase, and conventional PKC in primary rat hepatocytes. These findings were then studied in vivo in a rat model of dimethylnitrosamine-induced hepatic injury, in which the BC network is significantly disrupted. In accordance with the in vitro observations, administration of UDCA (40 mg/kg/day) to the injured rats for 18 days improved the BC network compared with the vehicle control. Serum hepatic markers were not altered by UDCA treatment, suggesting that the morphological effects were due to the direct actions of UDCA on network formation. Our data provide new evidence of the pharmacological potential of UDCA in accelerating or regenerating BC network formation in vitro, in hepatic cell culture models, and in vivo in a rat model of hepatic injury, and provide a basis for understanding its hepatoprotective effects.


Assuntos
Sistema Biliar/efeitos dos fármacos , Doença Hepática Induzida por Substâncias e Drogas/tratamento farmacológico , Hepatócitos/efeitos dos fármacos , Ácido Ursodesoxicólico/farmacologia , Animais , Sistema Biliar/crescimento & desenvolvimento , Linhagem Celular Tumoral , Relação Dose-Resposta a Droga , MAP Quinase Quinase Quinases/genética , MAP Quinase Quinase Quinases/metabolismo , Masculino , Proteína Quinase C/genética , Proteína Quinase C/metabolismo , Ratos , Ratos Wistar , Ácido Taurocólico/toxicidade , Proteínas Quinases p38 Ativadas por Mitógeno/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
8.
Nat Commun ; 3: 764, 2012 Apr 03.
Artigo em Inglês | MEDLINE | ID: mdl-22473008

RESUMO

ABCG2, also known as BCRP, is a high-capacity urate exporter, the dysfunction of which raises gout/hyperuricemia risk. Generally, hyperuricemia has been classified into urate 'overproduction type' and/or 'underexcretion type' based solely on renal urate excretion, without considering an extra-renal pathway. Here we show that decreased extra-renal urate excretion caused by ABCG2 dysfunction is a common mechanism of hyperuricemia. Clinical parameters, including urinary urate excretion, are examined in 644 male outpatients with hyperuricemia. Paradoxically, ABCG2 export dysfunction significantly increases urinary urate excretion and risk ratio of urate overproduction. Abcg2-knockout mice show increased serum uric acid levels and renal urate excretion, and decreased intestinal urate excretion. Together with high ABCG2 expression in extra-renal tissues, our data suggest that the 'overproduction type' in the current concept of hyperuricemia be renamed 'renal overload type', which consists of two subtypes-'extra-renal urate underexcretion' and genuine 'urate overproduction'-providing a new concept valuable for the treatment of hyperuricemia and gout.


Assuntos
Hiperuricemia/metabolismo , Rim/metabolismo , Ácido Úrico/metabolismo , Membro 2 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Transportadores de Cassetes de Ligação de ATP/metabolismo , Adulto , Idoso , Animais , Transporte Biológico , Regulação para Baixo , Humanos , Hiperuricemia/genética , Hiperuricemia/fisiopatologia , Rim/fisiopatologia , Masculino , Camundongos , Camundongos Knockout , Pessoa de Meia-Idade , Proteínas de Neoplasias/genética , Proteínas de Neoplasias/metabolismo
9.
Hepatology ; 55(3): 953-64, 2012 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-22095670

RESUMO

UNLABELLED: The hepatic expression of Niemann-Pick C1-like 1 (NPC1L1), which is a key molecule in intestinal cholesterol absorption, is high in humans. In addition to NPC1L1, Niemann-Pick C2 (NPC2), a secretory cholesterol-binding protein involved in intracellular cholesterol trafficking and the stimulation of biliary cholesterol secretion, is also expressed in the liver. In this study, we examined the molecular interaction and functional association between NPC1L1 and NPC2. In vitro studies with adenovirus-based or plasmid-mediated gene transfer systems revealed that NPC1L1 negatively regulated the protein expression and secretion of NPC2 without affecting the level of NPC2 messenger RNA. Experiments with small interfering RNA against NPC1L1 confirmed the endogenous association of these proteins. In addition, endocytosed NPC2 could compensate for the reduction of NPC2 in NPC1L1-overexpressing cells, and this demonstrated that the posttranscriptional regulation of NPC2 was dependent on a novel ability of NPC1L1 to inhibit the maturation of NPC2 and accelerate the degradation of NPC2 during its maturation. Furthermore, to confirm the physiological relevance of NPC1L1-mediated regulation, we analyzed human liver specimens and found a negative correlation between the protein levels of hepatic NPC1L1 and hepatic NPC2. CONCLUSION: NPC1L1 down-regulates the expression and secretion of NPC2 by inhibiting its maturation and accelerating its degradation. NPC2 functions as a regulator of intracellular cholesterol trafficking and biliary cholesterol secretion; therefore, in addition to its role in cholesterol re-uptake from the bile by hepatocytes, hepatic NPC1L1 may control cholesterol homeostasis via the down-regulation of NPC2.


Assuntos
Proteínas de Transporte/metabolismo , Glicoproteínas/metabolismo , Fígado/metabolismo , Proteínas de Membrana/metabolismo , Animais , Células CHO , Proteínas de Transporte/genética , Colesterol/metabolismo , Cricetinae , Cricetulus , Regulação para Baixo , Técnicas de Transferência de Genes , Glicoproteínas/genética , Homeostase , Humanos , Técnicas In Vitro , Proteínas de Membrana/efeitos dos fármacos , Proteínas de Membrana/genética , Proteínas de Membrana Transportadoras , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/farmacologia , Proteínas de Transporte Vesicular
10.
Gastroenterology ; 140(5): 1664-74, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21315718

RESUMO

BACKGROUND & AIMS: Biliary cholesterol secretion helps maintain cholesterol homeostasis; it is regulated by the cholesterol exporter adenosine triphosphate-binding cassettes G5 and G8 (ABCG5/G8) and the cholesterol importer Niemann-Pick C1-like 1 (NPC1L1). We studied another putative regulator of cholesterol secretion into bile, Niemann-Pick C2 (NPC2)--a cholesterol-binding protein secreted by the biliary system--and determined its effects on transporter-mediated biliary secretion of cholesterol. METHODS: Mice with hepatic knockdown of Npc2 or that overexpressed NPC2 were created using adenovirus-mediated gene transfer; biliary lipids were characterized. The effects of secreted NPC2 on cholesterol transporter activity were examined in vitro using cells that overexpressed ABCG5/G8 or NPC1L1. RESULTS: Studies of mice with altered hepatic expression of NPC2 revealed that this expression positively regulates the biliary secretion of cholesterol, supported by the correlation between levels of NPC2 protein and cholesterol in human bile. In vitro analysis showed that secreted NPC2 stimulated ABCG5/G8-mediated cholesterol efflux but not NPC1L1-mediated cholesterol uptake. Consistent with these observations, no significant changes in biliary cholesterol secretion were observed on hepatic overexpression of NPC2 in ABCG5/G8-null mice, indicating that NPC2 requires ABCG5/G8 to stimulate cholesterol secretion. Analyses of NPC2 mutants showed that the stimulatory effect of biliary NPC2 was independent of the function of lysosomal NPC2 as a regulator of intracellular cholesterol trafficking. CONCLUSIONS: NPC2 is a positive regulator of biliary cholesterol secretion via stimulation of ABCG5/G8-mediated cholesterol transport.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Bile/química , Colesterol/metabolismo , Regulação da Expressão Gênica , Lipoproteínas/metabolismo , RNA Mensageiro , Proteínas de Transporte Vesicular/genética , Membro 5 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Membro 8 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Animais , Transporte Biológico , Modelos Animais de Doenças , Humanos , Camundongos , Camundongos Knockout , Reação em Cadeia da Polimerase , Proteínas de Transporte Vesicular/biossíntese
11.
Pharm Res ; 28(2): 405-12, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-20953676

RESUMO

PURPOSE: Niemann-Pick C1-like 1 (NPC1L1), a pharmacological target of ezetimibe, is responsible for cholesterol absorption in enterocytes and hepatocytes. In the present study, the involvement of peroxisome proliferator-activated receptor α (PPARα) and its cofactor, PPARγ coactivator 1α (PGC1α) in the transcriptional regulation of human NPC1L1 was analyzed. METHODS: Reporter gene assays and electrophoretic mobility shift assays (EMSAs) were performed with the 5'-flanking region of the human NPC1L1 gene and the effect of siPPARα was examined. RESULTS: PPARα-mediated transactivation was observed with human NPC1L1 promoter constructs. Detailed analyses using deletion- and mutated-promoter constructs revealed the presence of a functional PPARα-response element (PPRE) upstream of the human NPC1L1 gene (-846/-834), a direct binding of PPARα and RXRα to which was confirmed by EMSAs. Moreover, PPARα-specific knockdown resulted in a significant decrease in the endogenous expression of NPC1L1 mRNA and protein in human-derived HepG2 cells. Furthermore, cotransfection of PGC1α stimulated the SREBP2/HNF4α- and PPARα/RXRα-mediated activation of the human NPC1L1 promoter. CONCLUSIONS: We found that PPARα positively regulates human NPC1L1 transcription via direct binding to a PPRE. Additionally, PGC1α stimulates the SREBP2/HNF4α- and PPARα/RXRα-mediated transactivation of human NPC1L1. These findings may provide new insights into the close relationship of glucose, fatty acids and cholesterol homeostasis.


Assuntos
Mucosa Intestinal/metabolismo , Proteínas de Membrana , PPAR alfa/metabolismo , PPAR gama/metabolismo , Azetidinas/farmacologia , Colesterol/genética , Colesterol/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Enterócitos/metabolismo , Ezetimiba , Fenofibrato/farmacologia , Regulação da Expressão Gênica , Células Hep G2 , Humanos , Absorção Intestinal/efeitos dos fármacos , Absorção Intestinal/genética , Intestinos/efeitos dos fármacos , Proteínas de Membrana/genética , Proteínas de Membrana/metabolismo , Proteínas de Membrana Transportadoras , Doenças de Niemann-Pick/genética , Doenças de Niemann-Pick/metabolismo , PPAR alfa/genética , PPAR gama/genética , Ligação Proteica , Elementos de Resposta/genética
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA