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.
Theranostics ; 12(3): 1440-1458, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35154499

RESUMO

Rationale: Impairment in lymphatic transport is associated with the onset and progression of atherosclerosis in animal models. The downregulation of low-density-lipoprotein receptor (LDLR) expression, rather than increased circulating cholesterol level per se, is involved in early atherosclerosis-related lymphatic dysfunction. Enhancing lymphatic function in Ldlr-/- mice with a mutant form of VEGF-C (VEGF-C 152s), a selective VEGFR-3 agonist, successfully delayed atherosclerotic plaque onset when mice were subsequently fed a high-fat diet. However, the specific mechanisms by which LDLR protects against lymphatic function impairment is unknown. Methods and results: We have thus injected wild-type and Pcsk9-/- mice with an adeno-associated virus type 1 expressing a shRNA for silencing Ldlr in vivo. We herein report that lymphatic contractility is reduced upon Ldlr dowregulation in wild-type mice only. Our in vitro experiments reveal that a decrease in LDLR expression at the mRNA level reduces the chromosome duplication phase and the protein expression of VEGFR-3, a membrane-bound key lymphatic marker. Furthermore, it also significantly reduced the levels of 18 lipid subclasses, including key constituents of lipid rafts as well as the transcription of several genes involved in cholesterol biosynthesis and cellular and metabolic processes. Exogenous PCSK9 only reduces lymphatic endothelial-LDLR at the protein level and does not affect lymphatic endothelial cell integrity. This puts forward that PCSK9 may act upon lymphatic muscle cells to mediate its effect on lymphatic contraction capacity in vivo. Conclusion: Our results suggest that treatments that specifically palliate the down regulation of LDLR mRNA in lymphatic endothelial cells preserve the integrity of the lymphatic endothelium and sustain lymphatic function, a prerequisite player in atherosclerosis.


Assuntos
Aterosclerose , Hiperlipidemias , Animais , Aterosclerose/genética , Aterosclerose/metabolismo , Colesterol/metabolismo , Regulação para Baixo , Células Endoteliais/metabolismo , Hiperlipidemias/metabolismo , Lipídeos , Lipoproteínas LDL/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pró-Proteína Convertase 9/genética , Pró-Proteína Convertase 9/metabolismo , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator C de Crescimento do Endotélio Vascular/genética , Fator C de Crescimento do Endotélio Vascular/metabolismo , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/genética , Receptor 3 de Fatores de Crescimento do Endotélio Vascular/metabolismo
2.
Physiol Rep ; 9(3): e14721, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-33527668

RESUMO

BACKGROUND: LDL-cholesterol lowering variants that upregulate receptor uptake of LDL, such as in PCSK9 and HMGCR, are associated with diabetes via unclear mechanisms. Activation of the NLRP3 inflammasome/interleukin-1 beta (IL-1ß) pathway promotes white adipose tissue (WAT) dysfunction and type 2 diabetes (T2D) and is regulated by LDL receptors (LDLR and CD36). We hypothesized that: (a) normocholesterolemic subjects with lower plasma PCSK9, identifying those with higher WAT surface-expression of LDLR and CD36, have higher activation of WAT NLRP3 inflammasome and T2D risk factors, and; (b) LDL upregulate adipocyte NLRP3 inflammasome and inhibit adipocyte function. METHODOLOGY: Post hoc analysis was conducted in 27 overweight/ obese subjects with normal plasma LDL-C and measures of disposition index (DI during Botnia clamps) and postprandial fat metabolism. WAT was assessed for surface-expression of LDLR and CD36 (immunohistochemistry), protein expression (immunoblot), IL-1ß secretion (AlphaLISA), and function (3 H-triolein storage). RESULTS: Compared to subjects with higher than median plasma PCSK9, subjects with lower PCSK9 had higher WAT surface-expression of LDLR (+81%) and CD36 (+36%), WAT IL-1ß secretion (+284%), plasma IL-1 receptor-antagonist (+85%), and postprandial hypertriglyceridemia, and lower WAT pro-IL-1ß protein (-66%), WAT function (-62%), and DI (-28%), without group-differences in body composition, energy intake or expenditure. Adjusting for WAT LDLR or CD36 eliminated group-differences in WAT function, DI, and postprandial hypertriglyceridemia. Native LDL inhibited Simpson-Golabi Behmel-syndrome (SGBS) adipocyte differentiation and function and increased inflammation. CONCLUSION: Normocholesterolemic subjects with lower plasma PCSK9 and higher WAT surface-expression of LDLR and CD36 have higher WAT NLRP3 inflammasome activation and T2D risk factors. This may be due to LDL-induced inhibition of adipocyte function.


Assuntos
Tecido Adiposo Branco/metabolismo , Antígenos CD36/metabolismo , Colesterol/sangue , Inflamassomos/metabolismo , Proteína 3 que Contém Domínio de Pirina da Família NLR/metabolismo , Obesidade/sangue , Pró-Proteína Convertase 9/sangue , Receptores de LDL/metabolismo , Adipócitos Brancos/imunologia , Adipócitos Brancos/metabolismo , Adipogenia , Tecido Adiposo Branco/imunologia , Idoso , Biomarcadores/sangue , Células Cultivadas , Diabetes Mellitus Tipo 2/etiologia , Regulação para Baixo , Feminino , Humanos , Interleucina-1beta/metabolismo , Masculino , Pessoa de Meia-Idade , Obesidade/complicações , Obesidade/enzimologia , Obesidade/imunologia , Medição de Risco , Fatores de Risco
3.
PLoS One ; 11(6): e0157230, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27280970

RESUMO

PCSK9 is a secreted ligand and negative post-translational regulator of low-density lipoprotein receptor (LDLR) in hepatocytes. Gain-of-function (GOF) or loss-of-function (LOF) mutations in PCSK9 are directly correlated with high or low plasma LDL-cholesterol levels, respectively. Therefore, PCSK9 is a prevailing lipid-lowering target to prevent coronary heart diseases and stroke. Herein, we fused monomeric fluorescent proteins to PCSK9 and LDLR to visualize their intra- and extracellular trafficking dynamics by live confocal microscopy. Fluorescence recovery after photobleaching (FRAP) showed that PCSK9 LOF R46L mutant and GOF mutations S127R and D129G, but not the LDLR high-affinity mutant D374Y, significantly accelerate PCSK9 exit from the endoplasmic reticulum (ER). Quantitative analysis of inverse FRAP revealed that only R46L presented a much slower trafficking from the trans-Golgi network (TGN) to the plasma membrane and a lower mobile fraction likely suggesting accumulation or delayed exit at the TGN as an underlying mechanism. While not primarily involved in LDLR binding, PCSK9 C-terminal domain (CTD) was found to be essential to induce LDLR degradation both upon its overexpression in cells or via the extracellular pathway. Our data revealed that PCSK9 CTD is required for the localization of PCSK9 at the TGN and increases its LDLR-mediated endocytosis. Interestingly, intracellular lysosomal targeting of PCSK9-ΔCTD was able to rescue its capacity to induce LDLR degradation emphasizing a role of the CTD in the sorting of PCSK9-LDLR complex towards late endocytic compartments. Finally, we validated our dual fluorescence system as a cell based-assay by preventing PCSK9 internalization using a PCSK9-LDLR blocking antibody, which may be expended to identify protein, peptide or small molecule inhibitors of PCSK9.


Assuntos
Mutação , Pró-Proteína Convertase 9/metabolismo , Proteólise , Receptores de LDL/metabolismo , Células Hep G2 , Humanos , Inibidores de PCSK9 , Domínios Proteicos , Transporte Proteico , Receptores de LDL/genética
4.
Arterioscler Thromb Vasc Biol ; 35(12): 2517-25, 2015 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-26494228

RESUMO

OBJECTIVE: Proprotein convertase subtilisin/kexin type 9 (PCSK9) promotes the degradation of the low-density lipoprotein receptor thereby elevating plasma low-density lipoprotein cholesterol levels and the risk of coronary heart disease. Thus, the use of PCSK9 inhibitors holds great promise to prevent heart disease. Previous work found that PCSK9 is involved in triglyceride metabolism, independently of its action on low-density lipoprotein receptor, and that other yet unidentified receptors could mediate this effect. Therefore, we assessed whether PCSK9 enhances the degradation of CD36, a major receptor involved in transport of long-chain fatty acids and triglyceride storage. APPROACH AND RESULTS: Overexpressed or recombinant PCSK9 induced CD36 degradation in cell lines and primary adipocytes and reduced the uptake of the palmitate analog Bodipy FL C16 and oxidized low-density lipoprotein in 3T3-L1 adipocytes and hepatic HepG2 cells, respectively. Surface plasmon resonance, coimmunoprecipitation, confocal immunofluorescence microscopy, and protein degradation pathway inhibitors revealed that PCSK9 directly interacts with CD36 and targets the receptor to lysosomes through a mechanism involving the proteasome. Importantly, the level of CD36 protein was increased by >3-fold upon small interfering RNA knockdown of endogenous PCSK9 in hepatic cells and similarly increased in the liver and visceral adipose tissue of Pcsk9(-/-) mice. In Pcsk9(-/-) mice, increased hepatic CD36 was correlated with an amplified uptake of fatty acid and accumulation of triglycerides and lipid droplets. CONCLUSIONS: Our results demonstrate an important role of PCSK9 in modulating the function of CD36 and triglyceride metabolism. PCSK9-mediated CD36 degradation may serve to limit fatty acid uptake and triglyceride accumulation in tissues, such as the liver.


Assuntos
Adipócitos/enzimologia , Antígenos CD36/metabolismo , Ácidos Graxos/metabolismo , Gordura Intra-Abdominal/enzimologia , Fígado/enzimologia , Pró-Proteína Convertases/metabolismo , Serina Endopeptidases/metabolismo , Triglicerídeos/metabolismo , Células 3T3-L1 , Adipócitos/efeitos dos fármacos , Animais , Compostos de Boro/metabolismo , Antígenos CD36/genética , Feminino , Células HEK293 , Células Hep G2 , Humanos , Gordura Intra-Abdominal/efeitos dos fármacos , Lipoproteínas LDL/metabolismo , Fígado/efeitos dos fármacos , Lisossomos/enzimologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ácidos Palmíticos/metabolismo , Pró-Proteína Convertase 9 , Pró-Proteína Convertases/deficiência , Pró-Proteína Convertases/genética , Complexo de Endopeptidases do Proteassoma/metabolismo , Inibidores de Proteassoma/farmacologia , Proteólise , Interferência de RNA , Serina Endopeptidases/deficiência , Serina Endopeptidases/genética , Fatores de Tempo , Transfecção
5.
J Biol Chem ; 289(27): 18736-51, 2014 Jul 04.
Artigo em Inglês | MEDLINE | ID: mdl-24855646

RESUMO

DNA methylation and histone acetylation inhibitors are widely used to study the role of epigenetic marks in the regulation of gene expression. In addition, several of these molecules are being tested in clinical trials or already in use in the clinic. Antimetabolites, such as the DNA-hypomethylating agent 5-azacytidine (5-AzaC), have been shown to lower malignant progression to acute myeloid leukemia and to prolong survival in patients with myelodysplastic syndromes. Here we examined the effects of DNA methylation inhibitors on the expression of lipid biosynthetic and uptake genes. Our data demonstrate that, independently of DNA methylation, 5-AzaC selectively and very potently reduces expression of key genes involved in cholesterol and lipid metabolism (e.g. PCSK9, HMGCR, and FASN) in all tested cell lines and in vivo in mouse liver. Treatment with 5-AzaC disturbed subcellular cholesterol homeostasis, thereby impeding activation of sterol regulatory element-binding proteins (key regulators of lipid metabolism). Through inhibition of UMP synthase, 5-AzaC also strongly induced expression of 1-acylglycerol-3-phosphate O-acyltransferase 9 (AGPAT9) and promoted triacylglycerol synthesis and cytosolic lipid droplet formation. Remarkably, complete reversal was obtained by the co-addition of either UMP or cytidine. Therefore, this study provides the first evidence that inhibition of the de novo pyrimidine synthesis by 5-AzaC disturbs cholesterol and lipid homeostasis, probably through the glycerolipid biosynthesis pathway, which may contribute mechanistically to its beneficial cytostatic properties.


Assuntos
Azacitidina/farmacologia , Colesterol/metabolismo , Epigênese Genética/efeitos dos fármacos , Animais , Linhagem Celular Tumoral , Núcleo Celular/efeitos dos fármacos , Núcleo Celular/metabolismo , Cricetinae , Metilação de DNA/efeitos dos fármacos , Homeostase/efeitos dos fármacos , Humanos , Fígado/efeitos dos fármacos , Fígado/metabolismo , Masculino , Camundongos , Pirimidinas/biossíntese , Proteína de Ligação a Elemento Regulador de Esterol 2/genética
6.
PLoS One ; 4(11): e7728, 2009 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-19888469

RESUMO

The peroxisome proliferator-activator receptor PPARgamma plays an essential role in vascular biology, modulating macrophage function and atherosclerosis progression. Recently, we have described the beneficial effect of combined activation of the ghrelin/GHS-R1a receptor and the scavenger receptor CD36 to induce macrophage cholesterol release through transcriptional activation of PPARgamma. Although the interplay between CD36 and PPARgamma in atherogenesis is well recognized, the contribution of the ghrelin receptor to regulate PPARgamma remains unknown. Here, we demonstrate that ghrelin triggers PPARgamma activation through a concerted signaling cascade involving Erk1/2 and Akt kinases, resulting in enhanced expression of downstream effectors LXRalpha and ABC sterol transporters in human macrophages. These effects were associated with enhanced PPARgamma phosphorylation independently of the inhibitory conserved serine-84. Src tyrosine kinase Fyn was identified as being recruited to GHS-R1a in response to ghrelin, but failure of activated Fyn to enhance PPARgamma Ser-84 specific phosphorylation relied on the concomitant recruitment of docking protein Dok-1, which prevented optimal activation of the Erk1/2 pathway. Also, substitution of Ser-84 preserved the ghrelin-induced PPARgamma activity and responsiveness to Src inhibition, supporting a mechanism independent of Ser-84 in PPARgamma response to ghrelin. Consistent with this, we found that ghrelin promoted the PI3-K/Akt pathway in a Galphaq-dependent manner, resulting in Akt recruitment to PPARgamma, enhanced PPARgamma phosphorylation and activation independently of Ser-84, and increased expression of LXRalpha and ABCA1/G1. Collectively, these results illustrate a complex interplay involving Fyn/Dok-1/Erk and Galphaq/PI3-K/Akt pathways to transduce in a concerted manner responsiveness of PPARgamma to ghrelin in macrophages.


Assuntos
Regulação Enzimológica da Expressão Gênica , Grelina/metabolismo , Macrófagos/metabolismo , PPAR gama/metabolismo , Antígenos CD36/biossíntese , Relação Dose-Resposta a Droga , Receptores ErbB/metabolismo , Humanos , Modelos Biológicos , Fosforilação , Regiões Promotoras Genéticas , Estrutura Terciária de Proteína , Proteínas Proto-Oncogênicas c-fyn/metabolismo , Transdução de Sinais , Ativação Transcricional
7.
PPAR Res ; 2008: 364784, 2008.
Artigo em Inglês | MEDLINE | ID: mdl-18288286

RESUMO

Investigating the metabolic functions of the nuclear receptor peroxisome proliferator-activated receptor gamma (PPARgamma) has been extremely rewarding over the past years. Uncovering the biologic roles of PPARgamma and its mechanism of action has greatly advanced our understanding of the transcriptional control of lipid and glucose metabolism, and compounds such as thiazolidinediones which directly regulate PPARgamma have proven to exhibit potent insulin-sensitizer effects in the treatment of diabetes. We review here recent advances on the emerging role of growth hormone releasing peptides in regulating PPARgamma through interaction with scavenger receptor CD36 and ghrelin GHS-R1a receptor. With the impact that these peptides exert on the metabolic pathways involved in lipid metabolism and energy homeostasis, it is hoped that the development of novel approaches in the regulation of PPAR functions will bring additional therapeutic possibilities to face problems related to metabolic diseases.

8.
Endocrinology ; 148(3): 1009-18, 2007 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-17138655

RESUMO

Whereas the uptake of oxidized lipoproteins by scavenger receptor CD36 in macrophages has been associated with foam cell formation and atherogenesis, little is known about the role of CD36 in regulating lipid metabolism in adipocytes. Here we report that treatment of 3T3-L1 adipocytes with hexarelin, a GH-releasing peptide that interacts with CD36, resulted in a depletion of intracellular lipid content with no significant change in CD36 expression. Microarray analysis revealed an increased pattern in several genes involved in fatty acid mobilization toward the mitochondrial oxidative phosphorylation process in response to hexarelin. Interestingly, many of these up-regulated genes are known targets of peroxisomal proliferator-activated receptor (PPAR)-gamma, such as FATP, CPT-1, and F(1)-ATPase, suggesting that adipocyte response to hexarelin may involve PPARgamma activation. Expression studies also indicate an increase in thermogenic markers PPARgamma coactivator 1alpha and uncoupling protein-1, which are normally expressed in brown adipocytes. Electron microscopy of hexarelin-treated 3T3-L1 adipocytes showed an intense and highly organized cristae formation that spans the entire width of mitochondria, compared with untreated cells, and cytochrome c oxidase activity was enhanced by hexarelin, two features characteristic of highly oxidative tissues. A similar mitochondrial phenotype was detected in epididymal white fat of mice treated with hexarelin, along with an increased expression of thermogenic markers that was lost in treated CD36-null mice, suggesting that the ability of hexarelin to promote a brown fat-like phenotype also occurs in vivo and is dependent on CD36. These results provide a potential role for CD36 to impact the overall metabolic activity of fat usage and mitochondrial biogenesis in adipocytes.


Assuntos
Adipócitos Brancos/efeitos dos fármacos , Antígenos CD36/fisiologia , Mitocôndrias/efeitos dos fármacos , Oligopeptídeos/farmacologia , Células 3T3-L1 , Adipócitos Brancos/química , Adipócitos Brancos/metabolismo , Adipócitos Brancos/ultraestrutura , Animais , Antígenos CD36/genética , Complexo IV da Cadeia de Transporte de Elétrons/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Metabolismo dos Lipídeos/efeitos dos fármacos , Lipídeos/análise , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Mitocôndrias/fisiologia , Análise de Sequência com Séries de Oligonucleotídeos , Oxirredução/efeitos dos fármacos , Receptores Depuradores/fisiologia , Termogênese/efeitos dos fármacos , Termogênese/genética
9.
Mol Endocrinol ; 20(12): 3165-78, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16959872

RESUMO

Macrophages play a central role in the pathogenesis of atherosclerosis by accumulating cholesterol through increased uptake of oxidized low-density lipoproteins by scavenger receptor CD36, leading to foam cell formation. Here we demonstrate the ability of hexarelin, a GH-releasing peptide, to enhance the expression of ATP-binding cassette A1 and G1 transporters and cholesterol efflux in macrophages. These effects were associated with a transcriptional activation of nuclear receptor peroxisome proliferator-activated receptor (PPAR)gamma in response to binding of hexarelin to CD36 and GH secretagogue-receptor 1a, the receptor for ghrelin. The hormone binding domain was not required to mediate PPARgamma activation by hexarelin, and phosphorylation of PPARgamma was increased in THP-1 macrophages treated with hexarelin, suggesting that the response to hexarelin may involve PPARgamma activation function-1 activity. However, the activation of PPARgamma by hexarelin did not lead to an increase in CD36 expression, as opposed to liver X receptor (LXR)alpha, suggesting a differential regulation of PPARgamma-targeted genes in response to hexarelin. Chromatin immunoprecipitation assays showed that, in contrast to a PPARgamma agonist, the occupancy of the CD36 promoter by PPARgamma was not increased in THP-1 macrophages treated with hexarelin, whereas the LXRalpha promoter was strongly occupied by PPARgamma in the same conditions. Treatment of apolipoprotein E-null mice maintained on a lipid-rich diet with hexarelin resulted in a significant reduction in atherosclerotic lesions, concomitant with an enhanced expression of PPARgamma and LXRalpha target genes in peritoneal macrophages. The response was strongly impaired in PPARgamma(+/-) macrophages, indicating that PPARgamma was required to mediate the effect of hexarelin. These findings provide a novel mechanism by which the beneficial regulation of PPARgamma and cholesterol metabolism in macrophages could be regulated by CD36 and ghrelin receptor downstream effects.


Assuntos
Transportadores de Cassetes de Ligação de ATP/metabolismo , Colesterol/metabolismo , Lipoproteínas/metabolismo , Macrófagos Peritoneais/efeitos dos fármacos , Oligopeptídeos/farmacologia , PPAR gama/metabolismo , Transportador 1 de Cassete de Ligação de ATP , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Animais , Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/prevenção & controle , Antígenos CD36/genética , Antígenos CD36/metabolismo , Células Cultivadas , Proteínas de Ligação a DNA/genética , Humanos , Receptores X do Fígado , Macrófagos Peritoneais/metabolismo , Camundongos , Camundongos Knockout , Receptores Nucleares Órfãos , Fosforilação , Regiões Promotoras Genéticas , Receptores Citoplasmáticos e Nucleares/genética , Receptores Acoplados a Proteínas G/genética , Receptores de Grelina , Transcrição Gênica , Regulação para Cima
10.
FASEB J ; 19(13): 1869-71, 2005 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-16123174

RESUMO

CD36, a type B scavenger receptor expressed on macrophages, appears to play a major role in fatty streak formation through scavenging oxidatively modified lipoproteins in the arterial wall. We tested the hypothesis that EP 80317, a novel CD36 ligand derived from the growth hormone (GH)-releasing peptide family but devoided of any GH releasing activity, exerts anti-atherosclerotic effects in apolipoprotein E-deficient (apoE-/-) mice fed an atherogenic diet from 6 wk of age. Daily subcutaneous injections of EP 80317 (300 microg/kg) or vehicle were initiated at 6, 10, 12, or 14 wk until death at 18 wk. En face analyses of the entire aortic tree revealed a striking reduction (up to 51%) of lesion areas in EP 80317-treated apoE-/- mice compared with controls. Chronic treatment with EP 80317 (12 wk) is also associated with a 30% decrease in total plasma cholesterol, suggesting potential effects of this drug on cholesterol metabolism at the intestine/hepatic levels. EP 80317 exerts both preventive and curative effects on atherosclerotic lesion progression that were shown to be reversible after cessation of treatment. At the macrophage level, EP 80317 reduced oxidized low density lipoproteins internalization and up-regulated genes involved in cholesterol efflux, including peroxisome proliferator-activated receptor gamma (PPARgamma), liver x receptor alpha (LXRalpha), and the ATP binding cassette (ABC) transporters ABCA1 and ABCG1, supporting a role in regulating peripheral cholesterol trafficking. Importantly, the effects of EP 80317 were shown to be CD36 dependent, inasmuch as no anti-atherosclerotic or hypocholesterolemic effects were observed in apoE/CD36 double-deficient mice. In addition, long-term treatment of apoE/CD36 double-deficient mice with EP 80317 did not modulate the expression of genes of the PPARgamma-LXRalpha-ABC transporters pathway. Our results suggest that EP 80317, as a CD36 ligand, might be a prototype for a novel class of anti-atherosclerotic agents.


Assuntos
Apolipoproteínas E/genética , Aterosclerose/genética , Aterosclerose/patologia , Antígenos CD36/metabolismo , Ligantes , Oligopeptídeos/farmacologia , Transportador 1 de Cassete de Ligação de ATP , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/metabolismo , Animais , Aorta/patologia , Aterosclerose/prevenção & controle , Transporte Biológico , Western Blotting , HDL-Colesterol/metabolismo , Proteínas de Ligação a DNA/metabolismo , Hormônio do Crescimento/química , Metabolismo dos Lipídeos , Lipídeos/química , Lipoproteínas/química , Lipoproteínas/metabolismo , Lipoproteínas LDL/metabolismo , Receptores X do Fígado , Macrófagos/metabolismo , Macrófagos Peritoneais/metabolismo , Masculino , Camundongos , Camundongos Transgênicos , Modelos Biológicos , Modelos Estatísticos , Oligopeptídeos/química , Receptores Nucleares Órfãos , Oxigênio/metabolismo , PPAR gama/metabolismo , Peptídeos/química , RNA Mensageiro/metabolismo , Receptores Citoplasmáticos e Nucleares/metabolismo , Receptores Depuradores Classe A/metabolismo , Transdução de Sinais , Fatores de Tempo , Regulação para Cima
11.
Biochem J ; 382(Pt 2): 417-24, 2004 Sep 01.
Artigo em Inglês | MEDLINE | ID: mdl-15176951

RESUMO

The GHRPs (growth hormone-releasing peptides) are a class of small synthetic peptides known to stimulate GH release through binding of a G-protein-coupled receptor (designated GHS-R). We have found that hexarelin, a hexapeptide member of the GHRPs, binds to another protein identified as CD36, a scavenger receptor that is expressed in various tissues, including monocytes/macrophages and the endothelial microvasculature. CD36 is involved in the endocytosis of oxLDL (oxidized low-density lipoprotein) by macrophages, and in the modulation of angiogenesis elicited by thrombospondin-1 through binding to endothelial cells. To define the binding domain for hexarelin on CD36, covalent photolabelling of CD36 followed by enzymic and chemical degradation of the photoligand-receptor complex was performed. A 8 kDa photolabelled fragment corresponding to the CD36-(Asn132-Glu177) sequence has been identified as the hexarelin-binding site. Chemical cleavage of this fragment with CNBr resulted in the release of the free ligand, suggesting that Met169 is the contact point for the ligand within the receptor binding pocket. We conclude that the binding domain for hexarelin on CD36 overlaps with that for oxLDL, which corresponds to residues Gln155-Lys183 of CD36. Hence hexarelin might interfere with the CD36-mediated uptake of modified lipoproteins by macrophages. This may contribute, at least in part, to the anti-atherosclerotic effect of GHRPs in apolipoprotein E-deficient mice.


Assuntos
Antígenos CD36/química , Oligopeptídeos/metabolismo , Marcadores de Fotoafinidade/metabolismo , Sequência de Aminoácidos , Animais , Sítios de Ligação , Antígenos CD36/biossíntese , Antígenos CD36/metabolismo , Reagentes de Ligações Cruzadas/síntese química , Reagentes de Ligações Cruzadas/metabolismo , Brometo de Cianogênio/metabolismo , Glicosilação , Humanos , Hidrólise , Radioisótopos do Iodo/metabolismo , Rim/citologia , Rim/embriologia , Rim/metabolismo , Lipoproteínas LDL/metabolismo , Metionina/metabolismo , Modelos Estruturais , Dados de Sequência Molecular , Oligopeptídeos/síntese química , Oligopeptídeos/química , Marcadores de Fotoafinidade/síntese química , Ratos , Ratos Sprague-Dawley , Serina Endopeptidases/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...