Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 47
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Arterioscler Thromb Vasc Biol ; 43(4): 562-580, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36756879

RESUMEN

BACKGROUND: Postprandial dyslipidemia is a causative risk factor for cardiovascular disease. The majority of absorbed dietary lipids are packaged into chylomicron and then delivered to circulation. Previous studies showed that Surf4 (surfeit locus protein 4) mediates very low-density lipoprotein secretion from hepatocytes. Silencing hepatic Surf4 markedly reduces the development of atherosclerosis in different mouse models of atherosclerosis without causing hepatic steatosis. However, the role of Surf4 in chylomicron secretion is unknown. METHODS: We developed inducible intestinal-specific Surf4 knockdown mice (Surf4IKO) using Vil1Cre-ERT2 and Surf4flox mice. Metabolic cages were used to monitor mouse metabolism. Enzymatic kits were employed to measure serum and tissue lipid levels. The expression of target genes was detected by qRT-PCR and Western Blot. Transmission electron microscopy and radiolabeled oleic acid were used to assess the structure of enterocytes and intestinal lipid absorption and secretion, respectively. Proteomics was performed to determine changes in protein expression in serum and jejunum. RESULTS: Surf4IKO mice, especially male Surf4IKO mice, displayed significant body weight loss, increased mortality, and reduced metabolism. Surf4IKO mice exhibited lipid accumulation in enterocytes and impaired fat absorption and secretion. Lipid droplets and small lipid vacuoles were accumulated in the cytosol and the endoplasmic reticulum lumen of the enterocytes of Surf4IKO mice, respectively. Surf4 colocalized with apoB and co-immunoprecipitated with apoB48 in differentiated Caco-2 cells. Intestinal Surf4 deficiency also significantly reduced serum triglyceride, cholesterol, and free fatty acid levels in mice. Proteomics data revealed that diverse pathways were altered in Surf4IKO mice. In addition, Surf4IKO mice had mild liver damage, decreased liver size and weight, and reduced hepatic triglyceride levels. CONCLUSIONS: Our findings demonstrate that intestinal Surf4 plays an essential role in lipid absorption and chylomicron secretion and suggest that the therapeutic use of Surf4 inhibition requires highly cell/tissue-specific targeting.


Asunto(s)
Aterosclerosis , Mucosa Intestinal , Humanos , Masculino , Animales , Ratones , Mucosa Intestinal/metabolismo , Células CACO-2 , Absorción Intestinal/fisiología , Grasas de la Dieta , Quilomicrones/metabolismo , Metabolismo de los Lípidos/genética , Triglicéridos/metabolismo , Aterosclerosis/metabolismo , Proteínas de la Membrana/genética , Proteínas de la Membrana/metabolismo
2.
Phytother Res ; 37(11): 4932-4962, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37461256

RESUMEN

Extracellular matrix (ECM) resolution by matrix metalloproteinases (MMPs) is a well-documented mechanism. MMPs play a dual and complex role in modulating ECM degradation at different stages of liver fibrosis, depending on the timing and levels of their expression. Increased MMP-1 combats disease progression by cleaving the fibrillar ECM. Activated hepatic stellate cells (HSCs) increase expression of MMP-2, -9, and -13 in different chemicals-induced animal models, which may alleviate or worsen disease progression based on animal models and the stage of liver fibrosis. In the early stage, elevated expression of certain MMPs may damage surrounding tissue and activate HSCs, promoting fibrosis progression. At the later stage, downregulation of MMPs can facilitate ECM accumulation and disease progression. A number of phytochemicals modulate MMP activity and ECM turnover, alleviating disease progression. However, the effects of phytochemicals on the expression of different MMPs are variable and may depend on the disease models and stage, and the dosage, timing and duration of phytochemicals used in each study. Here, we review the most recent advances in the role of MMPs in the effects of phytochemicals on liver fibrogenesis, which indicates that further studies are warranted to confirm and define the potential clinical efficacy of these phytochemicals.


Asunto(s)
Matriz Extracelular , Cirrosis Hepática , Animales , Cirrosis Hepática/tratamiento farmacológico , Matriz Extracelular/metabolismo , Fitoquímicos/farmacología , Fitoquímicos/uso terapéutico , Progresión de la Enfermedad
3.
J Lipid Res ; 62: 100091, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34118252

RESUMEN

Plasma LDL is produced from catabolism of VLDL and cleared from circulation mainly via the hepatic LDL receptor (LDLR). Proprotein convertase subtilisin/kexin type 9 (PCSK9) promotes LDLR degradation, increasing plasma LDL-C levels. Circulating PCSK9 is mainly secreted by the liver, whereas VLDL is exclusively secreted by hepatocytes. However, the mechanism regulating their secretion is not completely understood. Surfeit 4 (Surf4) is a cargo receptor localized in the ER membrane. It recruits cargos into coat protein complex II vesicles to facilitate their secretion. Here, we investigated the role of Surf4 in VLDL and PCSK9 secretion. We generated Surf4 liver-specific knockout mice and found that knockout of Surf4 did not affect PCSK9 secretion, whereas it significantly reduced plasma levels of cholesterol, triglyceride, and lipid-binding protein apolipoprotein B (apoB). In cultured human hepatocytes, Surf4 coimmunoprecipitated and colocalized with apolipoprotein B100, and Surf4 silencing reduced secretion of apolipoprotein B100. Furthermore, knockdown of Surf4 in LDLR knockout (Ldlr-/-) mice significantly reduced triglyceride secretion, plasma levels of apoB and non-HDL-C, and the development of atherosclerosis. However, Surf4 liver-specific knockout mice and Surf4 knockdown in Ldlr-/- mice displayed similar levels of liver lipids and plasma alanine aminotransferase activity as control mice, indicating that inhibition of Surf4 does not cause notable liver damage. Expression of stearoyl-CoA desaturase-1 was also reduced in the liver of these mice, suggesting a reduction in de novo lipogenesis. In summary, hepatic deficiency of Surf4 reduced VLDL secretion and the development of atherosclerosis but did not cause significant hepatic lipid accumulation or liver damage.


Asunto(s)
Aterosclerosis/metabolismo , Lipoproteínas VLDL/metabolismo , Proteínas de la Membrana/metabolismo , Animales , Células Cultivadas , Proteínas de la Membrana/deficiencia , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proproteína Convertasa 9/deficiencia , Proproteína Convertasa 9/metabolismo , Receptores de LDL/deficiencia , Receptores de LDL/metabolismo
4.
Adv Exp Med Biol ; 1276: 137-156, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32705598

RESUMEN

Plasma levels of cholesterol, especially low-density lipoprotein cholesterol (LDL-C), are positively correlated with the risk of cardiovascular disease. Buildup of LDL in the intima promotes the formation of foam cells and consequently initiates atherosclerosis, one of the main underlying causes of cardiovascular disease. Hepatic LDL receptor (LDLR) is mainly responsible for the clearance of plasma LDL. Mutations in LDLR cause familiar hypercholesterolemia and increase the risk of premature coronary heart disease. Proprotein convertase subtilisin/kexin-type 9 (PCSK9) promotes LDLR degradation and thereby plays a critical role in the regulation of plasma cholesterol metabolism. PCSK9 can bind to LDLR and reroute the receptor to lysosomes for degradation, increasing both circulating LDL-C levels and the risk of cardiovascular disease. PCSK9 is mainly regulated by sterol response element binding protein 2 (SREBP2) at the transcriptional level. Furthermore, many proteins have been identified as interacting with PCSK9, regulating plasma cholesterol levels. Pharmacotherapeutic inhibition of PCSK9 dramatically reduces plasma levels of LDL cholesterol and significantly reduces cardiovascular events. In this article, we summarize the latest advances in PCSK9, mainly focusing on the structure, function, and regulation of the protein, the underlying molecular mechanisms, and its pharmacotherapeutic applications.


Asunto(s)
Metabolismo de los Lípidos , Proproteína Convertasa 9/metabolismo , Subtilisina/metabolismo , Humanos , Proproteína Convertasa 9/química , Receptores de LDL/metabolismo , Subtilisina/química
5.
J Lipid Res ; 60(3): 516-527, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30617148

RESUMEN

Proprotein convertase subtilisin/kexin type 9 (PCSK9) promotes LDL receptor (LDLR) degradation, increasing plasma levels of LDL cholesterol and the risk of cardiovascular disease. We have previously shown that, in addition to the epidermal growth factor precursor homology repeat-A of LDLR, at least three ligand-binding repeats (LRs) of LDLR are required for PCSK9-promoted LDLR degradation. However, how exactly the LRs contribute to PCSK9's action on the receptor is not completely understood. Here, we found that substitution of Asp at position 172 in the linker between the LR4 and LR5 of full-length LDLR with Asn (D172N) reduced PCSK9 binding at pH 7.4 (mimic cell surface), but not at pH 6.0 (mimic endosomal environment). On the other hand, mutation of Asp at position 203 in the LR5 of full-length LDLR to Asn (D203N) significantly reduced PCSK9 binding at both pH 7.4 and pH 6.0. D203N also significantly reduced the ability of LDLR to mediate cellular LDL uptake, whereas D172N had no detectable effect. These findings indicate that amino acid residues in the LRs of LDLR play an important role in PCSK9 binding to the receptor.


Asunto(s)
Proproteína Convertasa 9/metabolismo , Receptores de LDL/química , Receptores de LDL/metabolismo , Secuencias Repetitivas de Aminoácido , Células HEK293 , Humanos , Ligandos , Lipoproteínas LDL/metabolismo , Mutación , Unión Proteica , Receptores de LDL/genética
6.
Liver Int ; 39(11): 2102-2111, 2019 11.
Artículo en Inglés | MEDLINE | ID: mdl-31379118

RESUMEN

BACKGROUND: Drug-induced liver injury (DILI) is the most common reason for a drug to be withdrawn from the market. Apart from stopping the offending drug, no regimens are available for treating idiosyncratic DILI in clinical practice. METHODS: We carried out a randomized, double-blind, multidoses, active drug controlled, multicentre phase II trial to assess the safety and efficacy of the study drug, magnesium isoglycyrrhizinate (MgIG), as compared to tiopronin, a standard therapy for DILI in China. The primary outcome was the proportion of alanine aminotransferase (ALT) normalization at week 4 after study drug administration. Logistic regression was used to examine the odds of ALT normalization between low dose (Group A) and high dose (Group B) vs active control (Group C). RESULTS: One hundred and seventy-four eligible subjects were randomized and enrolled into three groups: 59 in group A, 56 in group B and 59 in group C. It was shown that group A and group B lowered ALT level even at early stage of study drug administration; when compared with Group C (61.02%), the proportions of ALT normalization at week 4 were significantly greater in Group A (84.75%, P = .0029) and Group B (85.71%, P = .0037) respectively. The results from the univariate logistic model showed that the odds of ALT normalized among subjects in Group A were about 3.6 times greater (OR = 3.55, 95% CI: 1.47-8.57, P = .0049) than subjects in Group C. Similar effect was observed among subjects in Group B (OR = 3.83, 95% CI: 1.54-9.55, P = .0039). CONCLUSIONS: This trial provided preliminary evidence that MgIG is an effective and safe treatment for patients with acute DILI.


Asunto(s)
Alanina Transaminasa/sangre , Enfermedad Hepática Inducida por Sustancias y Drogas/tratamiento farmacológico , Saponinas/administración & dosificación , Triterpenos/administración & dosificación , Adulto , Enfermedad Hepática Inducida por Sustancias y Drogas/sangre , China , Método Doble Ciego , Femenino , Humanos , Inyecciones Intravenosas , Hígado/efectos de los fármacos , Hígado/patología , Modelos Logísticos , Masculino , Persona de Mediana Edad , Saponinas/efectos adversos , Triterpenos/efectos adversos , Adulto Joven
7.
Acta Pharmacol Sin ; 40(7): 879-894, 2019 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-30568253

RESUMEN

Increasing evidence has demonstrated that excessive fructose intake induces liver fibrosis. Epithelial-mesenchymal transition (EMT) driven by transforming growth factor-ß1 (TGF-ß1)/mothers against decapentaplegic homolog (Smad) signaling activation promotes the occurrence and development of liver fibrosis. Magnesium isoglycyrrhizinate is clinically used as a hepatoprotective agent to treat liver fibrosis, but its underlying molecular mechanism has not been identified. Using a rat model, we found that high fructose intake reduced microRNA (miR)-375-3p expression and activated the janus-activating kinase 2 (JAK2)/signal transducer and activator of transcription 3 (STAT3) cascade and TGF-ß1/Smad signaling, which is consistent with the EMT and liver fibrosis. To further verify these observations, BRL-3A cells and/or primary rat hepatocytes were exposed to high fructose and/or transfected with a miR-375-3p mimic or inhibitor or treated with a JAK2 inhibitor, and we found that the low expression of miR-375-3p could induce the JAK2/STAT3 pathway to activate TGF-ß1/Smad signaling and promote the EMT. Magnesium isoglycyrrhizinate was found to ameliorate high fructose-induced EMT and liver fibrosis in rats. More importantly, magnesium isoglycyrrhizinate increased miR-375-3p expression to suppress the JAK2/STAT3 pathway and TGF-ß1/Smad signaling in these animal and cell models. This study provides evidence showing that magnesium isoglycyrrhizinate attenuates liver fibrosis associated with a high fructose diet.


Asunto(s)
Cirrosis Hepática/tratamiento farmacológico , MicroARNs/metabolismo , Saponinas/uso terapéutico , Transducción de Señal/efectos de los fármacos , Triterpenos/uso terapéutico , Animales , Línea Celular , Transición Epitelial-Mesenquimal/efectos de los fármacos , Fructosa , Janus Quinasa 2/metabolismo , Cirrosis Hepática/inducido químicamente , Masculino , Ratas Sprague-Dawley , Factor de Transcripción STAT3/metabolismo , Saponinas/farmacología , Proteínas Smad Reguladas por Receptores/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Triterpenos/farmacología
8.
Zhonghua Nan Ke Xue ; 25(5): 392-398, 2019 May.
Artículo en Zh | MEDLINE | ID: mdl-32216222

RESUMEN

OBJECTIVE: To investigate the influence of subchronic exposure to low-dose subchronic nano-nickel oxide (NNO) on the reproductive function of male rats and embryonic development of the pregnant rats. METHODS: Fifty normal healthy male SD rats weighing 180-220 g were randomly divided into five groups of equal number, negative control, 4 mg/ml micro-nickel oxide (MNO), and 0.16, 0.8 and 4 mg/ml NNO, those of the latter four groups exposed to MNO or NNO by non-contact intratracheal instillation once every 3 days for 60 days, and then all mated with normal adult female rats in the ratio of 1∶2. After the female animals were confirmed to be pregnant, the males were sacrificed and the weights of the body, testis and epididymis obtained, followed by calculation of the visceral coefficients, determination of epididymal sperm concentration and viability and the nickel contents in the blood and semen by atomic fluorescence spectrometry. The female rats were killed on the 20th day of gestation for counting of the implanted fertilized eggs and live, dead and resorbed fetuses. RESULTS: After 60 days of exposure, the rats of the NNO groups showed no statistically significant differences from those of the negative control and MNO groups in the weights of the body, testis and epididymis or visceral coefficients. Compared with the negative control group, the animals of the 0.8 and 4 mg/ml NNO groups exhibited markedly decreased sperm concentration (ï¼»9.36 ± 0.98ï¼½ vs ï¼»7.49 ± 1.46ï¼½ and ï¼»6.30 ± 1.36ï¼½ ×106/ml, P < 0.05) and viable sperm (ï¼»85.35 ± 9.16ï¼½% vs ï¼»68.26 ± 16.63ï¼½% and ï¼»65.88 ± 14.68ï¼½ %, P < 0.05), increased morphologically abnormal sperm (ï¼»8.30 ± 2.47ï¼½% vs ï¼»13.99 ± 4.87ï¼½% and ï¼»15.38 ± 8.86ï¼½ %, P < 0.05), and elevated rate of dead and resorbed fetuses (1.18% vs 6.89% and 7.37%, P < 0.05), blood nickel content (ï¼»0.13 ± 0.16ï¼½ vs ï¼»0.52 ± 0.34ï¼½ and ï¼»0.82 ± 0.44ï¼½ mg/L, P < 0.05) and semen nickel content (ï¼»0.08 ± 0.13ï¼½ vs ï¼»0.35 ± 0.23ï¼½ and ï¼»0.63 ± 0.61ï¼½ mg/L, P < 0.05). The nickel level in the semen was correlated significantly with that in the blood (r = 0.912, P <0.01), negatively with the rate of viable sperm (r = -0.879, P <0.01) and positively with the percentage of morphologically abnormal sperm (r = -0.898, P <0.01). CONCLUSIONS: Sixty-day exposure to nano-nickel oxide at 0.8 and 4 mg/ml can produce reproductive toxicity in male rats and result in fetal abnormality in the females, while that at 0.16 mg/ml has no significant toxic effect on the reproductive function of the males.


Asunto(s)
Epidídimo/fisiopatología , Nanopartículas del Metal/toxicidad , Níquel/toxicidad , Efectos Tardíos de la Exposición Prenatal/patología , Testículo/fisiopatología , Animales , Relación Dosis-Respuesta a Droga , Epidídimo/efectos de los fármacos , Femenino , Masculino , Tamaño de los Órganos , Embarazo , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley , Motilidad Espermática , Espermatozoides/patología , Testículo/efectos de los fármacos , Pruebas de Toxicidad Subcrónica
9.
Arterioscler Thromb Vasc Biol ; 36(2): 253-5, 2016 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-26695502

RESUMEN

OBJECTIVE: ATP-binding cassette transporter G1 (ABCG1) mediates cholesterol efflux to lipidated lipoproteins. Conflicting data about cellular localization of ABCG1 and its effect on cholesterol efflux have been reported. Here, we investigated the underlying mechanisms for these different observations. APPROACH AND RESULTS: Confocal microscopy and biotinylation were used to assess cell surface localization of ABCG1. We found that mouse ABCG1 (mABCG1) used in one previous study has a substitution of Leu to Pro at position 550 (mG1-L550P). When the corresponding Leu at position 562 in human ABCG1 (hABCG1) was mutated to Pro (hG1-L562P), the mutant hABCG1, like mG1-L550P, mainly resided intracellularly, whereas wild-type mABCG1 and hABCG1 were localized on the plasma membrane. However, replacement of this Leu with Pro had no significant effect on mABCG1- and hABCG1-mediated cholesterol efflux. CONCLUSIONS: Leu at position 550/562 in mABCG1/hABCG1 is critical for their plasma membrane localization but not for ABCG1-mediated cholesterol efflux. Our findings indicate that the substitution of Leu to Pro at position 550 in mABCG1 may contribute to the non-cell surface localization of mABCG1 observed in the previous study.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Membrana Celular/metabolismo , Colesterol/metabolismo , Lipoproteínas/metabolismo , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1 , Transportadoras de Casetes de Unión a ATP/genética , Sustitución de Aminoácidos , Animales , Genotipo , Células HEK293 , Humanos , Leucina , Lipoproteínas/genética , Ratones , Microscopía Confocal , Mutación , Fenotipo , Transfección
11.
Biochim Biophys Acta ; 1841(6): 847-58, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24576892

RESUMEN

ATP-binding cassette transporter G1 (ABCG1) plays an important role in macrophage reverse cholesterol transport in vivo by promoting cholesterol efflux onto lipidated apoA-I. However, the underlying mechanism is unclear. Here, we found that ABCG1 co-immunoprecipitated with caveolin-1 (CAV1) but not with flotillin-1 and -2. Knockdown of CAV1 expression using siRNAs significantly reduced ABCG1-mediated cholesterol efflux without detectable effect on ABCA1-mediated cholesterol efflux. Disruption of the putative CAV1 binding site in ABCG1, through replacement of tyrosine residues at positions 487 and 489 or at positions 494 and 495 with alanine (Y487AY489A and Y494AY495A), impaired the interaction of ABCG1 with CAV1 and significantly decreased ABCG1-mediated cholesterol efflux. The substitution of Tyr494 and Tyr495 with Phe or Trp that resulted in an intact CAV1 binding site had no effect. Furthermore, Y494AY495A affected trafficking of ABCG1 to the cell surface. The mutant protein is mainly located intracellularly. Finally, we found that CAV1 co-immunoprecipitated with ABCG1 and regulated cholesterol efflux to reconstituted HDL in THP-1-derived macrophages upon the liver X receptor agonist treatment. These findings indicate that CAV1 interacts with ABCG1 and regulates ABCG1-mediated cholesterol efflux.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Caveolina 1/metabolismo , Colesterol/metabolismo , Unión Proteica/genética , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1 , Transportadoras de Casetes de Unión a ATP/genética , Sitios de Unión , Transporte Biológico/genética , Caveolina 1/antagonistas & inhibidores , Caveolina 1/genética , Colesterol/genética , Humanos , Receptores X del Hígado , Proteínas de la Membrana/metabolismo , Receptores Nucleares Huérfanos/antagonistas & inhibidores , ARN Interferente Pequeño
12.
Biochim Biophys Acta ; 1831(6): 1067-78, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23388354

RESUMEN

ATP-binding cassette transporter G1 (ABCG1) mediates cholesterol efflux onto lipidated apolipoprotein A-I and HDL and plays a role in various important physiological functions. However, the mechanism by which ABCG1 mediates cholesterol translocation is unclear. Protein palmitoylation regulates many functions of proteins such as ABCA1. Here we investigated if ABCG1 is palmitoylated and the subsequent effects on ABCG1-mediated cholesterol efflux. We demonstrated that ABCG1 is palmitoylated in both human embryonic kidney 293 cells and in mouse macrophage, J774. Five cysteine residues located at positions 26, 150, 311, 390 and 402 in the NH2-terminal cytoplasmic region of ABCG1 were palmitoylated. Removal of palmitoylation at Cys311 by mutating the residue to Ala (C311A) or Ser significantly decreased ABCG1-mediated cholesterol efflux. On the other hand, removal of palmitoylation at sites 26, 150, 390 and 402 had no significant effect. We further demonstrated that mutations of Cys311 affected ABCG1 trafficking from the endoplasmic reticulum. Therefore, our data suggest that palmitoylation plays a critical role in ABCG1-mediated cholesterol efflux through the regulation of trafficking.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/metabolismo , Riñón/metabolismo , Lipoproteínas/metabolismo , Lipoilación , Macrófagos/metabolismo , Procesamiento Proteico-Postraduccional , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1 , Transportadoras de Casetes de Unión a ATP/genética , Adenosina Trifosfato/metabolismo , Animales , Western Blotting , Células Cultivadas , Colesterol/metabolismo , Cisteína/química , Cisteína/genética , Cisteína/metabolismo , Retículo Endoplásmico/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Riñón/citología , Lipoproteínas/genética , Macrófagos/citología , Ratones , Mutagénesis Sitio-Dirigida , Mutación/genética , Transporte de Proteínas
13.
J Lipid Res ; 54(12): 3345-57, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24103783

RESUMEN

Proprotein convertase subtilisin kexin-like 9 (PCSK9) promotes the degradation of low density lipoprotein receptor (LDLR) and plays an important role in regulating plasma LDL-cholesterol levels. We have shown that the epidermal growth factor precursor homology domain A (EGF-A) of the LDLR is critical for PCSK9 binding at the cell surface (pH 7.4). Here, we further characterized the role of EGF-A in binding of PCSK9 to the LDLR. We found that PCSK9 efficiently bound to the LDLR but not to other LDLR family members. Replacement of EGF-A in the very low density lipoprotein receptor (VLDLR) with EGF-A of the LDLR promoted the degradation of the mutant VLDLR induced by PCSK9. Furthermore, we found that PCSK9 bound to recombinant EGF-A in a pH-dependent manner with stronger binding at pH 6.0. We also identified amino acid residues in EGF-A of the LDLR important for PCSK9 binding. Mutations G293H, D299V, L318D, and L318H reduced PCSK9 binding to the LDLR at neutral pH without effect at pH 6.0, while mutations R329P and E332G reduced PCSK9 binding at both pH values. Thus, our findings reveal that EGF-A of the LDLR is critical for PCSK9 binding at the cell surface (neutral pH) and at the acidic endosomal environment (pH 6.0), but different determinants contribute to efficient PCSK9 binding in different pH environments.


Asunto(s)
Factor de Crecimiento Epidérmico/química , Proproteína Convertasas/metabolismo , Precursores de Proteínas/química , Receptores de LDL/química , Receptores de LDL/metabolismo , Serina Endopeptidasas/metabolismo , Secuencia de Aminoácidos , Animales , Células COS , Chlorocebus aethiops , Secuencia Conservada , Humanos , Concentración de Iones de Hidrógeno , Datos de Secuencia Molecular , Mutación , Proproteína Convertasa 9 , Unión Proteica , Estructura Terciaria de Proteína , Proteolisis , Receptores de LDL/genética
14.
Biochemistry ; 52(52): 9497-509, 2013 Dec 31.
Artículo en Inglés | MEDLINE | ID: mdl-24320932

RESUMEN

ATP-binding cassette transporter G1 (ABCG1) mediates cholesterol and oxysterol efflux onto lipidated lipoproteins and plays an important role in macrophage reverse cholesterol transport. Here, we identified a highly conserved sequence present in the five ABCG transporter family members. The conserved sequence is located between the nucleotide binding domain and the transmembrane domain and contains five amino acid residues from Asn at position 316 to Phe at position 320 in ABCG1 (NPADF). We found that cells expressing mutant ABCG1, in which Asn316, Pro317, Asp319, and Phe320 in the conserved sequence were replaced with Ala simultaneously, showed impaired cholesterol efflux activity compared with wild type ABCG1-expressing cells. A more detailed mutagenesis study revealed that mutation of Asn316 or Phe 320 to Ala significantly reduced cellular cholesterol and 7-ketocholesterol efflux conferred by ABCG1, whereas replacement of Pro317 or Asp319 with Ala had no detectable effect. To confirm the important role of Asn316 and Phe320, we mutated Asn316 to Asp (N316D) and Gln (N316Q), and Phe320 to Ile (F320I) and Tyr (F320Y). The mutant F320Y showed the same phenotype as wild type ABCG1. However, the efflux of cholesterol and 7-ketocholesterol was reduced in cells expressing ABCG1 mutant N316D, N316Q, or F320I compared with wild type ABCG1. Further, mutations N316Q and F320I impaired ABCG1 trafficking while having no marked effect on the stability and oligomerization of ABCG1. The mutant N316Q and F320I could not be transported to the cell surface efficiently. Instead, the mutant proteins were mainly localized intracellularly. Thus, these findings indicate that the two highly conserved amino acid residues, Asn and Phe, play an important role in ABCG1-dependent export of cellular cholesterol, mainly through the regulation of ABCG1 trafficking.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/química , Transportadoras de Casetes de Unión a ATP/metabolismo , Secuencia Conservada , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 1 , Transportadoras de Casetes de Unión a ATP/genética , Adenosina Trifosfato/metabolismo , Secuencia de Aminoácidos , Transporte Biológico , Colesterol/metabolismo , Células Hep G2 , Humanos , Datos de Secuencia Molecular , Familia de Multigenes , Estabilidad Proteica , Estructura Terciaria de Proteína , Transporte de Proteínas , Alineación de Secuencia
15.
IUBMB Life ; 2013 Apr 27.
Artículo en Inglés | MEDLINE | ID: mdl-23625363

RESUMEN

Cholesterol, a major component of mammalian cell membranes, plays important structural and functional roles. However, accumulation of excessive cholesterol is toxic to cells. Aberrant cholesterol trafficking and accumulation is the molecular basis for many diseases, such as atherosclerotic cardiovascular disease and Tangier's disease. Accumulation of excessive cholesterol is also believed to contribute to the early onset of Alzheimer's disease. Thus, cellular cholesterol homeostasis is tightly regulated by uptake, de novo synthesis, and efflux. Any surplus of cholesterol must either be stored in the cytosol in the form of esters or released from the cell. Recently, several ATP-binding cassette (ABC) transporters, such as ABCA1, ABCG1, ABCG5, and ABCG8 have been shown to play important roles in the regulation of cellular cholesterol homeostasis by mediating cholesterol efflux. Mutations in ABC transporters are associated with several human diseases. In this review, we discuss the physiological roles of ABC transporters and the underlying mechanisms by which they mediate cholesterol translocation. © 2013 IUBMB Life, 2013.

16.
IUBMB Life ; 65(6): 505-12, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23983199

RESUMEN

Cholesterol, a major component of mammalian cell membranes, plays important structural and functional roles. However, accumulation of excessive cholesterol is toxic to cells. Aberrant cholesterol trafficking and accumulation is the molecular basis for many diseases, such as atherosclerotic cardiovascular disease and Tangier's disease. Accumulation of excessive cholesterol is also believed to contribute to the early onset of Alzheimer's disease. Thus, cellular cholesterol homeostasis is tightly regulated by uptake, de novo synthesis, and efflux. Any surplus of cholesterol must either be stored in the cytosol in the form of esters or released from the cell. Recently, several ATP-binding cassette (ABC) transporters, such as ABCA1, ABCG1, ABCG5, and ABCG8 have been shown to play important roles in the regulation of cellular cholesterol homeostasis by mediating cholesterol efflux. Mutations in ABC transporters are associated with several human diseases. In this review, we discuss the physiological roles of ABC transporters and the underlying mechanisms by which they mediate cholesterol translocation.


Asunto(s)
Transportadoras de Casetes de Unión a ATP/fisiología , Colesterol/metabolismo , Animales , Transporte Biológico , Homeostasis , Humanos , Esteroles/metabolismo
17.
Sheng Li Ke Xue Jin Zhan ; 44(2): 99-104, 2013 Apr.
Artículo en Zh | MEDLINE | ID: mdl-23847919

RESUMEN

Diabetes is a group of systematic metabolic diseases. In addition to cytochromes P450s, some ATP-binding cassette drug transporters are also under diabetic conditions. Breast cancer resistance protein (BCRP) , an important member of ATP-binding cassette drug transporters, expressed in most of the tissues, is thought to play an important protective role in normal tissues. It was reported that BCRP function and expression were impaired in intestine of diabetes rats, which affects the absorption and efflux of some drugs. Therefore, the alteration of BCRP expression and activity could be of critical importance for the treatment of diabetes mellitus. In this article, we reviewed the relationship between diabetes and


Asunto(s)
Transportadoras de Casetes de Unión a ATP/fisiología , Diabetes Mellitus/metabolismo , Proteínas de Neoplasias/fisiología , Transportador de Casetes de Unión a ATP, Subfamilia G, Miembro 2 , Animales , Diabetes Mellitus/fisiopatología , Humanos , Insulina/sangre , Interleucina-6/sangre , Ratas , Factor de Necrosis Tumoral alfa/sangre
18.
J Mol Cell Biol ; 14(9)2023 02 07.
Artículo en Inglés | MEDLINE | ID: mdl-36574593

RESUMEN

Surfeit 4 is a polytopic transmembrane protein that primarily resides in the endoplasmic reticulum (ER) membrane. It is ubiquitously expressed and functions as a cargo receptor, mediating cargo transport from the ER to the Golgi apparatus via the canonical coat protein complex II (COPII)-coated vesicles or specific vesicles. It also participates in ER-Golgi protein trafficking through a tubular network. Meanwhile, it facilitates retrograde transportation of cargos from the Golgi apparatus to the ER through COPI-coated vesicles. Surf4 can selectively mediate export of diverse cargos, such as PCSK9 very low-density lipoprotein (VLDL), progranulin, α1-antitrypsin, STING, proinsulin, and erythropoietin. It has been implicated in facilitating VLDL secretion, promoting cell proliferation and migration, and increasing replication of positive-strand RNA viruses. Therefore, Surf4 plays a crucial role in various physiological and pathophysiological processes and emerges as a promising therapeutic target. However, the molecular mechanisms by which Surf4 selectively sorts diverse cargos for ER-Golgi protein trafficking remain elusive. Here, we summarize the most recent advances in Surf4, focusing on its role in lipid metabolism.


Asunto(s)
Metabolismo de los Lípidos , Proteínas de la Membrana , Humanos , Aparato de Golgi/metabolismo , Proteínas de la Membrana/metabolismo , Proproteína Convertasa 9/metabolismo , Transporte de Proteínas/genética , Transporte de Proteínas/fisiología
20.
Matrix Biol ; 122: 10-17, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37598898

RESUMEN

Membrane-type I metalloproteinase (MT1-MMP/MMP14) plays a key role in various pathophysiological processes, indicating an unaddressed need for a targeted therapeutic approach. However, mice genetically deficient in Mmp14 show severe defects in development and growth. To investigate the possibility of MT1-MMP inhibition as a safe treatment in adults, we generated global Mmp14 tamoxifen-induced conditional knockout (Mmp14kd) mice and found that MT1-MMP deficiency in adult mice resulted in severe inflammatory arthritis. Mmp14kd mice started to show noticeably swollen joints two weeks after tamoxifen administration, which progressed rapidly. Mmp14kd mice reached a humane endpoint 6 to 8 weeks after tamoxifen administration due to severe arthritis. Plasma TNF-α levels were also significantly increased in Mmp14kd mice. Detailed analysis revealed chondrocyte hypertrophy, synovial fibrosis, and subchondral bone remodeling in the joints of Mmp14kd mice. However, global conditional knockout of MT1-MMP in adult mice did not affect body weight, blood glucose, or plasma cholesterol and triglyceride levels. Furthermore, we observed substantial expression of MT1-MMP in the articular cartilage of patients with osteoarthritis. We then developed chondrocyte-specific Mmp14 tamoxifen-induced conditional knockout (Mmp14chkd) mice. Chondrocyte MT1-MMP deficiency in adult mice also caused apparent chondrocyte hypertrophy. However, Mmp14chkd mice did not exhibit synovial hyperplasia or noticeable arthritis, suggesting that chondrocyte MT1-MMP is not solely responsible for the onset of severe arthritis observed in Mmp14kd mice. Our findings also suggest that highly cell-type specific inhibition of MT1-MMP is required for its potential therapeutic use.


Asunto(s)
Cartílago Articular , Osteoartritis , Animales , Ratones , Glucemia , Peso Corporal , Metaloproteinasa 14 de la Matriz/genética , Osteoartritis/inducido químicamente , Osteoartritis/genética
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA