Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 170
Filtrar
1.
J Virol ; 96(15): e0056122, 2022 08 10.
Artigo em Inglês | MEDLINE | ID: mdl-35867561

RESUMO

Enterovirus A71 (EV-A71) is a human pathogen that causes hand, foot, and mouth disease, which can progress to severe neurological disease. EV-A71 infects humans via the human scavenger receptor B2 (hSCARB2). It can also infect neonatal mice experimentally. Wild-type (WT) EV-A71 strains replicate primarily in the muscle of neonatal mice; however, susceptibility lasts only for a week after birth. Mouse-adapted (MA) strains, which can be obtained by serial passages in neonatal mice, are capable of infecting both muscle and neurons of the central nervous system. It is not clear how the host range and tropism of EV-A71 are regulated and why neonatal mice lose their susceptibility during development. We hypothesized that EV-A71 infection in neonatal mice is mediated by mouse Scarb2 (mScarb2) protein. Rhabdomyosarcoma (RD) cells expressing mScarb2 were prepared. Both WT and MA strains infected mScarb2-expressing cells, but the infection efficiency of the WT strain was much lower than that of the MA strain. Infection by WT and MA strains in vivo was abolished completely in Scarb2-/- mice. Scarb2+/- mice, in which Scarb2 expression was approximately half of that in Scarb2+/+ mice, showed a milder pathology than Scarb2+/+ mice after infection with the WT strain. The Scarb2 expression level in muscle decreased with aging, which was consistent with the reduced susceptibility of aged mice to infection. These results indicated that EV-A71 infection is mediated by mScarb2 and that the severity of the disease, the spread of virus, and the susceptibility period are modulated by mScarb2 expression. IMPORTANCE EV-A71 infects humans naturally but can also infect neonatal mice. The tissue tropism and severity of EV-A71 disease are determined by several factors, among which the virus receptor is thought to be important. We show that EV-A71 can infect neonatal mice using mScarb2. However, the infection efficiency of WT strains via mScarb2 is so low that an elevated virus-receptor interaction associated with mouse adaptation mutation and decrease in mScarb2 expression level during development modulate the severity of the disease, the spread of virus, and the susceptibility period in the artificial neonatal mice model.


Assuntos
Antígenos CD36 , Enterovirus Humano A , Proteínas de Membrana Lisossomal , Receptores Virais , Animais , Animais Recém-Nascidos/metabolismo , Animais Recém-Nascidos/virologia , Antígenos CD36/biossíntese , Antígenos CD36/metabolismo , Modelos Animais de Doenças , Suscetibilidade a Doenças , Enterovirus Humano A/metabolismo , Enterovirus Humano A/patogenicidade , Doença de Mão, Pé e Boca/metabolismo , Doença de Mão, Pé e Boca/transmissão , Doença de Mão, Pé e Boca/virologia , Especificidade de Hospedeiro , Humanos , Proteínas de Membrana Lisossomal/biossíntese , Proteínas de Membrana Lisossomal/metabolismo , Camundongos , Receptores Virais/biossíntese , Receptores Virais/metabolismo , Tropismo Viral , Virulência
2.
JCI Insight ; 6(17)2021 09 08.
Artigo em Inglês | MEDLINE | ID: mdl-34314388

RESUMO

The mechanism controlling long-chain fatty acid (LCFA) mobilization from adipose tissue is not well understood. Here, we investigated how the LCFA transporter CD36 regulates this process. By using tissue-specific KO mouse models, we showed that CD36 in adipocytes and endothelial cells mediated both LCFA deposition into and release from adipose tissue. We demonstrated the role of adipocytic and endothelial CD36 in promoting tumor growth and chemoresistance conferred by adipose tissue-derived LCFAs. We showed that dynamic cysteine S-acylation of CD36 in adipocytes, endothelial cells, and cancer cells mediated intercellular LCFA transport. We demonstrated that lipolysis induction in adipocytes triggered CD36 deacylation and deglycosylation, as well as its dissociation from interacting proteins, prohibitin-1 (PHB) and annexin 2 (ANX2). Our data indicate that lipolysis triggers caveolar endocytosis and translocation of CD36 from the cell membrane to lipid droplets. This study suggests a mechanism for both outside-in and inside-out cellular LCFA transport regulated by CD36 S-acylation and its interactions with PHB and ANX2.


Assuntos
Adipócitos/metabolismo , Antígenos CD36/genética , DNA/genética , Ácidos Graxos/metabolismo , Regulação da Expressão Gênica , Doenças Metabólicas/genética , Processamento de Proteína Pós-Traducional , Adipócitos/patologia , Tecido Adiposo/metabolismo , Animais , Animais Geneticamente Modificados , Transporte Biológico , Antígenos CD36/biossíntese , Membrana Celular/metabolismo , Células Cultivadas , DNA/metabolismo , Modelos Animais de Doenças , Lipólise , Doenças Metabólicas/metabolismo , Doenças Metabólicas/patologia , Camundongos , Camundongos Endogâmicos C57BL
3.
Iran J Allergy Asthma Immunol ; 19(1): 45-55, 2020 Feb 01.
Artigo em Inglês | MEDLINE | ID: mdl-32245320

RESUMO

Cigarette smoking and opium use are risk factors for coronary artery disease (CAD). It has been known that scavenger receptors such as CD36 and CD68 play critical roles in the pathogenesis of CAD. CD9, as a member of the tetraspanin, has been shown to interact with scavenger receptors. The aim of this study was to investigate the effects of these risk factors on expression levels of CD9, CD36, and CD68 on the THP-1 cell line. The THP-1 cell line treated with cigarette smoke extract (CSE( and opium, both individually and combinatory, in 24 h incubation. The protein and mRNA levels of CD9, CD36, and CD68 were evaluated by flow cytometry and quantitative reverse transcription-Polymerase Chain Reaction (qRT-PCR) techniques, respectively. CD36 and CD68 mRNA and protein expression levels were significantly increased in the cells treated with cigarette smoke extract compared to the control (p<0.001 in mRNA expression levels and p=0.016 and p=0.012 in protein expression levels, respectively). The CSE increased the level of CD9 protein expression compared to the control group (p=0.041) on the human macrophage cell line THP-1. No significant differences were observed in the CD9, CD36, and CD68 gene expression and at the protein levels between opium-treated THP-1 cells and controls. In conclusion, cigarettes by increasing the levels of CD36, CD68, and CD9 can be a risk factor in the development of many inflammatory diseases, including cardiovascular diseases, chronic obstructive pulmonary disease (COPD) and lung carcinoma.


Assuntos
Macrófagos/efeitos dos fármacos , Macrófagos/metabolismo , Nicotiana/toxicidade , Ópio/toxicidade , Extratos Vegetais/toxicidade , Fumaça/efeitos adversos , Antígenos CD/biossíntese , Antígenos CD/efeitos dos fármacos , Antígenos de Diferenciação Mielomonocítica/biossíntese , Antígenos de Diferenciação Mielomonocítica/efeitos dos fármacos , Antígenos CD36/biossíntese , Antígenos CD36/efeitos dos fármacos , Humanos , Fumar/efeitos adversos , Células THP-1 , Tetraspanina 29/biossíntese , Tetraspanina 29/efeitos dos fármacos , Produtos do Tabaco/efeitos adversos
4.
Respirology ; 25(6): 620-628, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-31542893

RESUMO

BACKGROUND AND OBJECTIVE: E-cigarettes are often marketed and thought of as emitting harmless vapour; however, verification of their safety for non-smokers is scarce. We have previously shown that E-cigarettes cause decreased phagocytosis of bacteria by macrophages via reductions in surface bacterial recognition receptors. This study assessed the effect of E-cigarette constituents, 3 E-liquid apple flavours, nicotine, vegetable glycerine and propylene glycol, on bronchial epithelial cell viability, apoptosis and cytokine secretion and macrophage phagocytosis of apoptotic airway cells and phagocytic recognition molecules. METHODS: Cell necrosis and apoptosis were measured by Sytox Green stain and Annexin V. Efferocytosis was measured by internalization of pHrodo Green labelled apoptotic airway cells by macrophages. Expression of macrophage cell surface apoptotic cell receptors was measured by flow cytometry. Cytokine release by E-cigarette-exposed airway cells was measured by cytokine bead array. RESULTS: E-cigarette vapour increased primary bronchial epithelial necrosis and apoptosis. E-cigarette vapour reduced efferocytosis (lowest flavour 12.1%) versus control (20.2%, P = 0.032). The efferocytosis receptor CD44 was reduced by one flavour (MFI 1863 vs 2332 control, P = 0.016) and all components reduced expression of CD36, including the glycol bases (MFI 1067-12 274 vs 1415 control). Reduced secretion of TNF-α, IL-6, IP-10, MIP-1α and MIP-1ß was observed for all flavour variants. CONCLUSION: E-cigarettes can cause bronchial epithelial apoptosis and macrophage efferocytosis dysfunction via reduced expression of apoptotic cell recognition receptors. These data further show that E-cigarettes should not be considered harmless to non-smokers and their effects may go far beyond cytotoxicity to cells.


Assuntos
Sistemas Eletrônicos de Liberação de Nicotina , Células Epiteliais/efeitos dos fármacos , Glicerol/toxicidade , Nicotina/toxicidade , Propilenoglicol/toxicidade , Mucosa Respiratória/fisiopatologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Apoptose/efeitos dos fármacos , Brônquios/fisiopatologia , Antígenos CD36/biossíntese , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Quimiocina CXCL10/metabolismo , Células Epiteliais/metabolismo , Humanos , Receptores de Hialuronatos/biossíntese , Interleucina-6/metabolismo , Macrófagos/imunologia , Necrose/induzido quimicamente , Fagocitose/efeitos dos fármacos , Receptores de Superfície Celular/efeitos dos fármacos , Mucosa Respiratória/efeitos dos fármacos , Produtos do Tabaco , Fator de Necrose Tumoral alfa/metabolismo
5.
Cells ; 8(10)2019 10 22.
Artigo em Inglês | MEDLINE | ID: mdl-31652636

RESUMO

MicroRNA-29 (miR-29) has been shown to play a critical role in reducing inflammation and fibrosis following liver injury. Non-alcoholic fatty liver disease (NAFLD) occurs when fat is deposited (steatosis) in the liver due to causes other than excessive alcohol use and is associated with liver fibrosis. In this study, we asked whether miR-29a could reduce experimental high fat diet (HFD)-induced obesity and liver fibrosis in mice. We performed systematical expression analyses of miR-29a transgenic mice (miR-29aTg mice) and wild-type littermates subjected to HFD-induced NAFLD. The results demonstrated that increased miR-29a not only alleviated HFD-induced body weight gain but also subcutaneous, visceral, and intestinal fat accumulation and hepatocellular steatosis in mice. Furthermore, hepatic tissue in the miR-29aTg mice displayed a weak fibrotic matrix concomitant with low fibrotic collagen1α1 expression within the affected tissues compared to the wild-type (WT) mice fed the HFD diet. Increased miR-29a signaling also resulted in the downregulation of expression of the epithelial mesenchymal transition-executing transcription factor snail, mesenchymal markers vimentin, and such pro-inflammation markers as il6 and mcp1 within the liver tissue. Meanwhile, miR-29aTg-HFD mice exhibited significantly lower levels of peroxisome proliferator-activated receptor γ (PPARγ), mitochondrial transcription factor A TFAM, and mitochondria DNA content in the liver than the WT-HFD mice. An in vitro luciferase reporter assay further confirmed that miR-29a mimic transfection reduced fatty acid translocase CD36 expression in HepG2 cells. Conclusion: Our data provide new insights that miR-29a can improve HDF-induced obesity, hepatocellular steatosis, and fibrosis, as well as highlight the role of miR-29a in regulation of NAFLD.


Assuntos
Antígenos CD36/biossíntese , Gorduras na Dieta/efeitos adversos , Cirrose Hepática , MicroRNAs/biossíntese , Hepatopatia Gordurosa não Alcoólica , Animais , Antígenos CD36/genética , Gorduras na Dieta/farmacologia , Células Hep G2 , Humanos , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/genética , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Camundongos , Camundongos Transgênicos , MicroRNAs/genética , Hepatopatia Gordurosa não Alcoólica/induzido quimicamente , Hepatopatia Gordurosa não Alcoólica/genética , Hepatopatia Gordurosa não Alcoólica/metabolismo , Hepatopatia Gordurosa não Alcoólica/patologia
6.
J Exp Clin Cancer Res ; 38(1): 52, 2019 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-30717785

RESUMO

BACKGROUND: Gastric cancer (GC) has a clear predilection for metastasis toward the omentum which is primarily composed of adipose tissue, indicating that fatty acids may contribute to this phenomenon. However their function remains poorly understood in GC. In this study, we investigated the role of palmitate acid (PA) and its cellular receptor CD36 in the progression of GC. METHODS: Immunohistochemical (IHC) staining was performed to detect CD36 expression in GC tissues and its clinical significance was determined statistically. CD36 over-expression and knock-down expression cell models were developed and tested in vitro. Wound-healing assays, migration assays, and invasion assays were performed and peritoneal implants into nude mice were done to assess the biological effects of PA and CD36. The underlying mechanisms were investigated using western blot, immunofluorescence (IF), quantitative real-time PCR (qRT-PCR) and antibody blocking assays. RESULTS: PA promoted the metastasis of GC by phosphorylation of AKT, which facilitated the nuclear localization of ß-catenin through inactivation of GSK-3ß via phosphorylation. This tumor-promoting effect of PA was mediated by CD36, a cell surface receptor of fatty acids (FAs). The higher the CD36 expression levels in GC tissues correlated with the poorer the prognosis of patients according to the TCGA database, the GEO database and our own clinical data. CONCLUSIONS: Our experiments established CD36 as a key mediator of FA-induced metastasis of GC via the AKT/GSK-3ß/ß-catenin signaling pathway. CD36 might, therefore, constitute a potential therapeutic target for clinical intervention in GC.


Assuntos
Antígenos CD36/metabolismo , Glicogênio Sintase Quinase 3 beta/metabolismo , Ácido Palmítico/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Neoplasias Gástricas/metabolismo , Neoplasias Gástricas/patologia , beta Catenina/metabolismo , Animais , Antígenos CD36/biossíntese , Linhagem Celular Tumoral , Movimento Celular/efeitos dos fármacos , Feminino , Humanos , Estimativa de Kaplan-Meier , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Terapia de Alvo Molecular , Invasividade Neoplásica , Metástase Neoplásica , Estadiamento de Neoplasias , Ácido Palmítico/farmacologia
7.
Theranostics ; 8(19): 5452-5468, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30555557

RESUMO

Omental metastasis occurs frequently in gastric cancer (GC) and is considered one of the major causes of gastric cancer-related mortality. Recent research indicated that omental adipocytes might mediate this metastatic predilection. Phosphatidylinositol transfer protein, cytoplasmic 1 (PITPNC1) was identified to have a crucial role in metastasis. However, whether PITPNC1 participates in the interaction between adipocytes and GC omental metastasis is unclear. Methods: We profiled and analyzed the expression of PITPNC1 through analysis of the TCGA database as well as immunohistochemistry staining using matched GC tissues, adjacent normal gastric mucosa tissues (ANTs), and omental metastatic tissues. The regulation of PITPNC1 by adipocytes was explored by co-culture systems. By using both PITPNC1 overexpression and silencing methods, the role of PITPNC1 in anoikis resistance and metastasis was determined through in vitro and in vivo experiments. Results: PITPNC1 was expressed at higher rates in GC tissues than in ANTs; notably, it was higher in omental metastatic lesions. Elevated expression of PITPNC1 predicted higher rates of omental metastasis and a poor prognosis. PITPNC1 promoted anoikis resistance through fatty acid metabolism by upregulating CD36 and CPT1B expression. Further, PITPNC1 was elevated by adipocytes and facilitated GC omental metastasis. Lastly, in vivo studies showed that PITPNC1 was a therapeutic indicator of fatty acid oxidation (FAO) inhibition. Conclusion: Elevated expression of PITPNC1 in GC is correlated with an advanced clinical stage and a poor prognosis. PITPNC1 promotes anoikis resistance through enhanced FAO, which is regulated by omental adipocytes and consequently facilitates GC omental metastasis. Targeting PITPNC1 might present a promising strategy to treat omental metastasis.


Assuntos
Adipócitos/patologia , Ácidos Graxos/metabolismo , Proteínas de Membrana Transportadoras/metabolismo , Neoplasias Peritoneais/fisiopatologia , Neoplasias Peritoneais/secundário , Neoplasias Gástricas/patologia , Neoplasias Gástricas/fisiopatologia , Adenocarcinoma/patologia , Adenocarcinoma/fisiopatologia , Animais , Anoikis , Antígenos CD36/biossíntese , Carnitina O-Palmitoiltransferase/biossíntese , Linhagem Celular Tumoral , Sobrevivência Celular , Técnicas de Cocultura , Modelos Animais de Doenças , Expressão Gênica , Inativação Gênica , Humanos , Imuno-Histoquímica , Proteínas de Membrana Transportadoras/análise , Proteínas de Membrana Transportadoras/genética , Camundongos Nus , Modelos Teóricos , Regulação para Cima
8.
Biomed Res Int ; 2017: 1414070, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28497039

RESUMO

Diabetic kidney disease (DKD) appears to be closely related to lipid deposition in kidney. The aim of this study was to determine whether high glucose (HG) exacerbated lipid deposition by increasing CD36 expression via AKT-PPARγ signaling pathway. Our results showed that HG activated AKT signaling pathway, followed by an increase in PPARγ that induced CD36 overexpression, ultimately causing lipid deposition in HK-2 cells. We also found that inhibition of AKT-PPARγ signaling pathway or knockdown of CD36 could reduce HG-induced lipid accumulation in HK-2 cells. These results indicated that AKT-PPARγ signaling pathway mediated HG-induced lipid deposition by upregulating CD36 expression in HK-2 cells and that inhibition of AKT-PPARγ signaling pathway had the potential beneficial effects of reducing lipid deposition in diabetic kidney.


Assuntos
Antígenos CD36/biossíntese , Glucose/farmacologia , Túbulos Renais/metabolismo , Metabolismo dos Lipídeos/efeitos dos fármacos , PPAR gama/biossíntese , Transdução de Sinais/efeitos dos fármacos , Regulação para Cima/efeitos dos fármacos , Linhagem Celular , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Humanos , Túbulos Renais/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo
9.
Biochem Biophys Res Commun ; 488(3): 477-482, 2017 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-28522296

RESUMO

In obese patients, free fatty acids ectopically accumulated in non-adipose tissues cause cell death. Saturated fatty acids are more deleterious to non-adipose cells, and supplementation with monounsaturated fatty acids has been proposed to rescue cells from saturated fatty acid-induced cytotoxicity; however, the mechanisms are not well understood. To understand the cytoprotective role of monounsaturated fatty acids in lipotoxic cell death of macrophages, we investigated the antagonizing effect of oleate and the underlying mechanisms in palmitate-treated RAW264.7 cells. Palmitate strongly induced apoptosis in macrophages by increasing CD36 expression, which was identified to mediate both endoplasmic reticulum stress and the generation of reactive oxygen species. Co-treatment with oleate significantly reduced CD36 expression and its downstream signaling pathways of apoptosis in palmitate-treated cells. These findings provide a novel mechanism by which oleate protects macrophages from palmitate-induced lipotoxicity.


Assuntos
Apoptose/efeitos dos fármacos , Antígenos CD36/biossíntese , Regulação para Baixo/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Ácido Oleico/farmacologia , Palmitatos/farmacologia , Animais , Antígenos CD36/genética , Antígenos CD36/metabolismo , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Macrófagos/metabolismo , Camundongos , Células RAW 264.7 , Espécies Reativas de Oxigênio/metabolismo , Relação Estrutura-Atividade
10.
J Biol Chem ; 291(33): 16977-89, 2016 08 12.
Artigo em Inglês | MEDLINE | ID: mdl-27358406

RESUMO

Macrophage CD36 binds and internalizes oxidized low density lipoprotein (oxLDL) to facilitate foam cell formation. CD36 expression is activated by peroxisome proliferator-activated receptor γ (PPARγ). Tamoxifen, an anti-breast cancer medicine, has demonstrated pleiotropic functions including cardioprotection with unfully elucidated mechanisms. In this study, we determined that treatment of ApoE-deficient mice with tamoxifen reduced atherosclerosis, which was associated with decreased CD36 and PPARγ expression in lesion areas. At the cellular level, we observed that tamoxifen inhibited CD36 protein expression in human THP-1 monocytes, THP-1/PMA macrophages, and human blood monocyte-derived macrophages. Associated with decreased CD36 protein expression, tamoxifen reduced cellular oxLDL accumulation in a CD36-dependent manner. At the transcriptional level, tamoxifen decreased CD36 mRNA expression, promoter activity, and the binding of the PPARγ response element in CD36 promoter to PPARγ protein. Tamoxifen blocked ligand-induced PPARγ nuclear translocation and CD36 expression, but it increased PPARγ phosphorylation, which was due to that tamoxifen-activated ERK1/2. Furthermore, deficiency of PPARγ expression in macrophages abolished the inhibitory effect of tamoxifen on CD36 expression or cellular oxLDL accumulation both in vitro and in vivo Taken together, our study demonstrates that tamoxifen inhibits CD36 expression and cellular oxLDL accumulation by inactivating the PPARγ signaling pathway, and the inhibition of macrophage CD36 expression can be attributed to the anti-atherogenic properties of tamoxifen.


Assuntos
Antígenos CD36/biossíntese , Regulação da Expressão Gênica/efeitos dos fármacos , Lipoproteínas LDL/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Macrófagos/metabolismo , PPAR gama/metabolismo , Tamoxifeno/farmacologia , Animais , Apolipoproteínas E/deficiência , Antígenos CD36/genética , Linhagem Celular , Feminino , Regulação da Expressão Gênica/genética , Humanos , Lipoproteínas LDL/genética , Sistema de Sinalização das MAP Quinases/genética , Masculino , Camundongos , Camundongos Knockout , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , PPAR gama/genética , Fosforilação/efeitos dos fármacos , Fosforilação/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética
11.
Mol Med Rep ; 13(2): 1533-40, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26707062

RESUMO

ATP-binding cassette transporters A1 (ABCA1) and G1 (ABCG1), and macrophage scavenger receptor, cluster of differentiation (CD)36, function as key mediators of cholesterol efflux and influx from macrophages. In addition, they are associated with foam cell formation and the development of atherosclerosis (AS). The aim of the present study was to investigate the effects of extracellular signal-regulated kinases 1/2 (ERK1/2) inhibition on lipid balance in oxidized-low-density lipoprotein (Ox-LDL)-stimulated rat macrophages, and to examine the role of ERK1/2 inhibitors in AS. Rat peritoneal macrophages were treated with Ox-LDL alone or in combination with an ERK1/2 inhibitor, U0126, and untreated cells served as controls. Ox-LDL-induced lipid accumulation was detected by DiI fluorescence and oil red O staining. In addition, the mRNA and protein expression levels of ABCA1, ABCG1 and CD36 were determined using polymerase chain reaction and western blotting, respectively. Treatment with Ox-LDL significantly increased lipid accumulation and upregulated the mRNA and protein expression levels of ABCA1, ABCG1 and CD36 in macrophages. The addition of U0126 resulted in a marked reduction of lipid deposition, upregulation of ABCA1/G1 expression and suppression of CD36 expression in Ox-LDL-stimulated macrophages. The results of the present study indicated a novel association between ERK1/2 signaling and lipid metabolism, thus suggesting that inhibition of ERK1/2 may be considered a promising therapeutic strategy against AS.


Assuntos
Transportador 1 de Cassete de Ligação de ATP/biossíntese , Transportadores de Cassetes de Ligação de ATP/biossíntese , Antígenos CD36/biossíntese , Macrófagos/metabolismo , Transportador 1 de Cassete de Ligação de ATP/genética , Membro 1 da Subfamília G de Transportadores de Cassetes de Ligação de ATP , Transportadores de Cassetes de Ligação de ATP/genética , Animais , Butadienos/administração & dosagem , Antígenos CD36/genética , Colesterol/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Metabolismo dos Lipídeos/efeitos dos fármacos , Lipoproteínas LDL/administração & dosagem , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Macrófagos/efeitos dos fármacos , Nitrilas/administração & dosagem , RNA Mensageiro/biossíntese , Ratos
12.
J Neurosci ; 35(44): 14783-93, 2015 Nov 04.
Artigo em Inglês | MEDLINE | ID: mdl-26538649

RESUMO

The scavenger receptor CD36 is a critical factor initiating ischemic brain injury, but the cell type(s) expressing CD36 and responsible for its harmful effects remain unknown. Using bone marrow (BM) chimeras subjected to transient middle cerebral artery occlusion, we found that CD36(-/-) mice transplanted with wild-type (WT) BM (WT→CD36(-/-)) have smaller infarcts (-67%), comparable with those of mice lacking CD36 both in brain and hematogenous cells (CD36(-/-) →CD36(-/-); - 72%). Conversely, WT mice receiving CD36(-/-) BM (CD36(-/-) →WT) have infarcts similar to WT→WT mice, suggesting that CD36 in the host brain (i.e., in microglia and endothelial cells), and not in hematogenous cells is involved in the damage. As anticipated, postischemic neutrophil infiltration in CD36(-/-) →CD36(-/-) mice was attenuated. Surprisingly, however, in WT→CD36(-/-) mice, in which infarcts were small, neutrophil infiltration was large and similar to that of CD36(-/-) →WT mice, in which infarcts were not reduced. Postischemic neutrophil free radical production was attenuated in WT→CD36(-/-) mice compared with CD36(-/-) →WT mice, whereas expression of the neutrophil activator colony-stimulating factor 3 (CSF3) was suppressed in CD36(-/-) cerebral endothelial cells, but not microglia. In CD36(-/-) cerebral endothelial cultures exposed to extracts from stroke brains, the upregulation of CSF3, but not neutrophil attractant chemokines, was suppressed. Intracerebroventricular administration of CSF3, 24 h after stroke, reconstituted neutrophil radical production and increased infarct volume in WT→CD36(-/-) mice. The findings identify endothelial cells as a key player in the deleterious effects of CD36 in stroke, and unveil a novel role of endothelial CD36 in enabling neutrophil neurotoxicity through CSF3. SIGNIFICANCE STATEMENT: Ischemic stroke is a leading cause of death and disability worldwide with limited therapeutic options. The inflammatory response initiated by cerebral ischemia-reperfusion contributes to ischemic brain injury and is a potential therapeutic target. Here we report that CD36, an innate immunity receptor involved in the initiation of postischemic inflammation, is a previously unrecognized regulator of neutrophil cytotoxicity. The effect is mediated by endothelial CD36 via upregulation of the neutrophil activator CSF3 in cerebral endothelial cells. Therefore, approaches to modulate cerebral endothelial CD36 signaling or to neutralize CSF3 may provide novel therapeutic opportunities to ameliorate postischemic inflammatory injury.


Assuntos
Lesões Encefálicas/metabolismo , Isquemia Encefálica/metabolismo , Antígenos CD36/biossíntese , Ativação de Neutrófilo/fisiologia , Receptores de Fator Estimulador de Colônias/biossíntese , Animais , Lesões Encefálicas/patologia , Isquemia Encefálica/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Técnicas de Cultura de Órgãos
13.
J Biol Chem ; 290(51): 30306-20, 2015 12 18.
Artigo em Inglês | MEDLINE | ID: mdl-26504087

RESUMO

Previously, we have demonstrated that 15(S)-hydroxyeicosatetranoic acid (15(S)-HETE) induces CD36 expression involving STAT1. Many studies have shown that peroxisome proliferator-activated receptor (PPAR)-γ mediates CD36 expression. Therefore, we asked the question whether these transcriptional factors interact with each other in the regulation of CD36 expression by 15(S)-HETE. Here, we show that STAT1 interacts with PPARγ in the induction of CD36 expression and foam cell formation by 15(S)-HETE. In addition, using molecular biological approaches such as EMSA, supershift EMSA, ChIP, re-ChIP, and promoter-reporter gene assays, we demonstrate that the STAT1 and PPARγ complex binds to the STAT-binding site at -107 nucleotides in the CD36 promoter and enhances its activity. Furthermore, the interaction of STAT1 with PPARγ depends on STAT1 acetylation, which is mediated by p300. In addition, our findings show that reactive oxygen species-dependent Syk and Pyk2 stimulation is required for p300 tyrosine phosphorylation and activation. Together, these results demonstrate that an interaction between STAT1, p300, and peroxisome proliferator-activated receptor-γ is required for 15(S)-HETE-induced CD36 expression, oxidized low density lipoprotein uptake, and foam cell formation, critical events underlying the pathogenesis of atherosclerosis.


Assuntos
Antígenos CD36/biossíntese , Proteína p300 Associada a E1A/metabolismo , Células Espumosas/metabolismo , Regulação da Expressão Gênica , PPAR gama/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Fator de Transcrição STAT1/metabolismo , Aterosclerose/metabolismo , Aterosclerose/patologia , Linhagem Celular Tumoral , Células Espumosas/patologia , Humanos , Ácidos Hidroxieicosatetraenoicos/farmacologia , Elementos de Resposta
14.
Tissue Eng Part A ; 21(11-12): 1837-47, 2015 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-25781458

RESUMO

To improve treatment of obesity, a contributing factor to multiple systemic and metabolic diseases, a better understanding of metabolic state and environmental stress at the cellular level is essential. This work presents development of a three-dimensional (3D) in vitro model of adipose tissue displaying induced lipid accumulation as a function of fatty acid supplementation that, subsequently, investigates cellular responses to a pro-inflammatory stimulus, thereby recapitulating key stages of obesity progression. Three-dimensional spheroid organization of adipose cells was induced by culturing 3T3-L1 mouse preadipocytes on an elastin-like polypeptide-polyethyleneimine (ELP-PEI)-coated surface. Results indicate a more differentiated phenotype in 3D spheroid cultures relative to two-dimensional (2D) monolayer analogues based on triglyceride accumulation, CD36 and CD40 protein expression, and peroxisome proliferator-activated receptor-γ (PPAR-γ) and adiponectin mRNA expression. The 3T3-L1 adipocyte spheroid model was then used to test the effects of a pro-inflammatory microenvironment, namely maturation in the presence of elevated fatty acid levels followed by acute exposure to tumor necrosis factor alpha (TNF-α). Under these conditions, we demonstrate that metabolic function was reduced across all cultures exposed to TNF-α, especially so when pre-exposed to linoleic acid. Further, in response to TNF-α, enhanced lipolysis, monitored as increased extracellular glycerol and fatty acids levels, was observed in adipocytes cultured in the presence of exogenous fatty acids. Taken together, our 3D spheroid model showed enhanced adipogenic differentiation and presents a platform for elucidating the key phenotypic responses that occur in pro-inflammatory microenvironments that characterize obesogenic states.


Assuntos
Adipócitos/metabolismo , Técnicas de Cultura de Células , Inflamação/patologia , Esferoides Celulares , Células 3T3-L1 , Adipócitos/efeitos dos fármacos , Adipogenia/efeitos dos fármacos , Adiponectina/biossíntese , Adiponectina/genética , Animais , Antígenos CD36/biossíntese , Antígenos CD36/genética , Antígenos CD40/biossíntese , Antígenos CD40/genética , Microambiente Celular , Gorduras na Dieta/farmacologia , Ácidos Graxos/farmacologia , Glicerol/metabolismo , Técnicas In Vitro , Inflamação/metabolismo , Ácido Linoleico/farmacologia , Lipólise , Camundongos , Obesidade , PPAR gama/biossíntese , PPAR gama/genética , Peptídeos , Fenótipo , Polietilenoimina , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Triglicerídeos/metabolismo , Fator de Necrose Tumoral alfa/farmacologia
15.
Int J Oncol ; 46(2): 764-74, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25405950

RESUMO

Paeonol, a potent antioxidant isolated from cortex moutan, possesses athero­protective activity, yet the detailed mechanisms are not fully investigated. This study was conducted to explore the role of paeonol and its underlying mechanisms in RAW264.7 macrophages and apolipoprotein E­deficient (ApoE(­/­)) mice. Paeonol treatment significantly attenuated intracellular lipid accumulation in macrophages, which may be the result of decreased oxidized low­density lipoprotein (ox­LDL) uptake and increased cholesterol efflux. Additionally, paeonol markedly inhibited the mRNA and protein expression of the cluster of differentiation 36 (CD36) by decreasing nuclear translocation of c­Jun [a subunit of activator protein­1 (AP­1)]. Moreover, paeonol upregulated the protein stability of ATP­binding cassette transporter A1 (ABCA1) by inhibiting calpain activity, while ABCA1 mRNA expression was not altered. Furthermore, small hairpin RNA (shRNA) targeting haem oxygenase­1 (HO­1) inhibited the paeonol­mediated beneficial effects on the expression of c­Jun, CD36, ABCA1, calpain activity and lipid accumulation in macrophages. Accordingly, paeonol retarded the progress of atherosclerosis in ApoE(­/­) mice and modulated the expression of CD36 and ABCA1 in aortas similarly to that observed in macrophages. These results indicate that paeonol provides protective effects on foam cell formation by a novel HO­1­dependent mediation of cholesterol efflux and lipid accumulation in macrophages.


Assuntos
Transportador 1 de Cassete de Ligação de ATP/biossíntese , Acetofenonas/administração & dosagem , Aterosclerose/tratamento farmacológico , Antígenos CD36/biossíntese , Transportador 1 de Cassete de Ligação de ATP/genética , Animais , Aorta/efeitos dos fármacos , Aterosclerose/genética , Aterosclerose/patologia , Antígenos CD36/genética , Calpaína/biossíntese , Linhagem Celular , Regulação da Expressão Gênica no Desenvolvimento/efeitos dos fármacos , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Humanos , Metabolismo dos Lipídeos/efeitos dos fármacos , Lipoproteínas LDL/genética , Lipoproteínas LDL/metabolismo , Macrófagos/efeitos dos fármacos , Camundongos , Camundongos Knockout
16.
Mycopathologia ; 179(1-2): 11-20, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25179349

RESUMO

Trichosporon asahii is the major cause of invasive trichosporonosis, but little is known about the host immune response to this pathogen. In this study, the early transcriptional response of human monocyte-like THP-1 cells to T. asahii infection was evaluated using cDNA microarray and 1,315 differentially expressed genes were identified. The up-regulated genes were mostly involved in both innate and adaptive immune responses, as well as apoptosis and anti-apoptosis processes. Genes encoding the pro-inflammatory cytokines TNF-α, IL-1ß, IL18 and IL-23α, along with the both C-C motif and C-X-C motif chemokines were strongly up-regulated, suggesting that THP-1 cells can mount a powerful inflammatory response to T. asahii infection. Genes encoding pattern recognition receptors were found up-regulated, such as dendritic cell-specific intercellular adhesion molecule 3-grabbing nonintegrin, cluster of differentiation 36 and the long pentraxin 3. Genes encoding members of the dual-spasticity phosphates family were up-regulated, and these genes were considered as a negative feedback mechanism to prevent excessive inflammatory response. The down-regulated genes in T. asahii-infected THP-1 cells were predominantly associated with cell cycle, mitosis, cell division and DNA repair. Thus, our study defines the early transcriptional response of monocyte-like THP-1 cells to T. asahii infection and provides a foundation for further investigations into the pathogenesis of T. asahii infection.


Assuntos
Interações Hospedeiro-Patógeno/imunologia , Monócitos/imunologia , Trichosporon/imunologia , Tricosporonose/imunologia , Antígenos CD/biossíntese , Proteína C-Reativa/biossíntese , Antígenos CD36/biossíntese , Moléculas de Adesão Celular/biossíntese , Linhagem Celular Tumoral , DNA Fúngico/genética , Interações Hospedeiro-Patógeno/genética , Humanos , Interleucina-18/biossíntese , Interleucina-1beta/biossíntese , Subunidade p19 da Interleucina-23/biossíntese , Análise em Microsséries , Mitose/genética , Componente Amiloide P Sérico/biossíntese , Transcrição Gênica/genética , Tricosporonose/microbiologia , Fator de Necrose Tumoral alfa/biossíntese
17.
Cancer Res ; 74(18): 5032-5044, 2014 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-25172842

RESUMO

Telomere malfunction and other types of DNA damage induce an activin A-dependent stress response in mortal nontumorigenic human mammary epithelial cells that subsequently induces desmoplastic-like phenotypes in neighboring fibroblasts. Some characteristics of this fibroblast/stromal response, such as reduced adipocytes and increased extracellular matrix content, are observed not only in tumor tissues but also in disease-free breast tissues at high risk for developing cancer, especially high mammographic density tissues. We found that these phenotypes are induced by repression of the fatty acid translocase CD36, which is seen in desmoplastic and disease-free high mammographic density tissues. In this study, we show that epithelial cells from high mammographic density tissues have more DNA damage signaling, shorter telomeres, increased activin A secretion and an altered DNA damage response compared with epithelial cells from low mammographic density tissues. Strikingly, both telomere malfunction and activin A expression in epithelial cells can repress CD36 expression in adjacent fibroblasts. These results provide new insights into how high mammographic density arises and why it is associated with breast cancer risk, with implications for the definition of novel invention targets (e.g., activin A and CD36) to prevent breast cancer.


Assuntos
Neoplasias da Mama/patologia , Glândulas Mamárias Humanas/anormalidades , Glândulas Mamárias Humanas/patologia , Densidade da Mama , Neoplasias da Mama/genética , Antígenos CD36/biossíntese , Dano ao DNA , Células Epiteliais/patologia , Feminino , Humanos , Fenótipo , Transdução de Sinais
18.
Pharmazie ; 69(6): 473-6, 2014 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-24974585

RESUMO

Intermedin (IMD) exerts a potent function in preventing atherosclerosis, while the mechanism remains unclear. Here we investigated the potential molecular mechanism responsible for the protective function of IMD in preventing foam cell formation in RAW264.7 cells. In our present study, IMD significantly inhibited intracellular cholesterol accumulation. Additionally, IMD dose-dependently down-regulated CD36 expression, which was confirmed by real-time quantitative reverse transcription-PCR and Western blot analysis. Our data suggest that IMD could inhibit the formation of foam cells through, at least partly, a CD36-dependent mechanism. This study suggests that IMD may be a therapeutic candidate for treating atherosclerosis.


Assuntos
Antígenos CD36/efeitos dos fármacos , Lipoproteínas LDL/metabolismo , Hormônios Peptídicos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Animais , Compostos Azo , Western Blotting , Antígenos CD36/biossíntese , Antígenos CD36/fisiologia , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Cromatografia Líquida de Alta Pressão , Primers do DNA , Relação Dose-Resposta a Droga , Camundongos , Oxirredução , Reação em Cadeia da Polimerase em Tempo Real
19.
Aging (Albany NY) ; 6(4): 281-95, 2014 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-24751397

RESUMO

CD36 has been associated with obesity and diabetes in human liver diseases, however, its role in age-associated nonalcoholic fatty liver disease (NAFLD) is unknown. Therefore, liver biopsies were collected from individuals with histologically normal livers (n=30), and from patients diagnosed with simple steatosis (NAS; n=26). Patients were divided into two groups according to age and liver biopsy samples were immunostained for CD36. NAFLD parameters were examined in young (12-week) and middle-aged (52-week) C57BL/6J mice, some fed with chow-diet and some fed with low-fat (LFD; 10% kcal fat) or high-fat diet (HFD; 60% kcal fat) for 12-weeks. CD36 expression was positively associated with age in individuals with normal livers but not in NAS patients. However, CD36 was predominantly located at the plasma membrane of hepatocytes in aged NAS patients as compared to young. In chow-fed mice, aging, despite an increase in hepatic CD36 expression, was not associated with the development of NAFLD. However, middle-aged mice did exhibit the development of HFD-induced NAFLD, mediated by an increase of CD36 on the membrane. Enhanced CD36-mediated hepatic fat uptake may contribute to an accelerated progression of NAFLD in mice and humans. Therapies to prevent the increase in CD36 expression and/or CD36 from anchoring at the membrane may prevent the development of NAFLD.


Assuntos
Antígenos CD36/biossíntese , Hepatócitos/metabolismo , Fígado/metabolismo , Hepatopatia Gordurosa não Alcoólica/metabolismo , Adulto , Idoso , Envelhecimento , Animais , Membrana Celular/metabolismo , Feminino , Humanos , Immunoblotting , Imuno-Histoquímica , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Reação em Cadeia da Polimerase em Tempo Real , Adulto Jovem
20.
Biochem Biophys Res Commun ; 447(1): 57-63, 2014 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-24685479

RESUMO

Rapamycin, a mammalian target of rapamycin (mTOR)-specific inhibitor, has the effect of anti-lipid deposition on non-alcoholic fatty liver disease (NAFLD), but the mechanisms with which rapamycin alleviates hepatic steatosis are not fully disclosed. CD36 is known to facilitate long-chain fatty acid uptake and contribute to NAFLD progression. Hepatic CD36 expression is closely associated with hepatic steatosis, while mTOR pathway is involved in CD36 translational control. This study was undertaken to investigate whether rapamycin alleviates hepatic steatosis via the inhibition of mTOR pathway-dependent CD36 translation. Human hepatoblastoma HepG2 cells were treated with palmitate and C57BL/6J mice were fed with high fat diet (HFD) to induce hepatic steatosis. Hepatic CD36 protein expression was significantly increased with lipid accumulation in palmitate-treated HepG2 cells or HFD-fed C57BL/6J mice. Rapamycin reduced hepatic steatosis and CD36 protein expression, but it had no influence on CD36 mRNA expression. Rapamycin had no effect on CD36 protein stability, but it significantly decreased CD36 translational efficiency. We further confirmed that rapamycin inhibited the phosphorylation of mTOR and its downstream translational regulators including p70 ribosomal protein S6 kinase (p70S6K), eukaryotic initiation factor 4E-binding protein 1 (4E-BP1), and eukaryotic initiation factor 4E (eIF4E). This study demonstrates that rapamycin inhibits hepatic CD36 translational efficiency through the mTOR pathway, resulting in reduction of CD36 protein expression and alleviation of hepatic steatosis.


Assuntos
Antígenos CD36/biossíntese , Fígado Gorduroso/tratamento farmacológico , Fígado/efeitos dos fármacos , Sirolimo/farmacologia , Animais , Dieta Hiperlipídica , Células Hep G2 , Humanos , Fígado/metabolismo , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Hepatopatia Gordurosa não Alcoólica , Palmitatos/farmacologia , Sirolimo/uso terapêutico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA