Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
PLoS Pathog ; 16(5): e1008497, 2020 05.
Artigo em Inglês | MEDLINE | ID: mdl-32453780

RESUMO

Heparan sulfate proteoglycans (HSPGs) are at the forefront of host-microbe interactions. Molecular and cell-based studies suggest that HSPG-pathogen interactions promote pathogenesis by facilitating microbial attachment and invasion of host cells. However, the specific identity of HSPGs, precise mechanisms by which HSPGs promote pathogenesis, and the in vivo relevance of HSPG-pathogen interactions remain to be determined. HSPGs also modulate host responses to tissue injury and inflammation, but functions of HSPGs other than facilitating microbial attachment and internalization are understudied in infectious disease. Here we examined the role of syndecan-1 (Sdc1), a major cell surface HSPG of epithelial cells, in mouse models of Listeria monocytogenes (Lm) infection. We show that Sdc1-/- mice are significantly less susceptible to both intragastric and intravenous Lm infection compared to wild type (Wt) mice. This phenotype is not seen in Sdc3-/- or Sdc4-/- mice, indicating that ablation of Sdc1 causes a specific gain of function that enables mice to resist listeriosis. However, Sdc1 does not support Lm attachment or invasion of host cells, indicating that Sdc1 does not promote pathogenesis as a cell surface Lm receptor. Instead, Sdc1 inhibits the clearance of Lm before the bacterium gains access to its intracellular niche. Large intravascular aggregates of neutrophils and neutrophil extracellular traps (NETs) embedded with antimicrobial compounds are formed in Sdc1-/- livers, which trap and kill Lm. Lm infection induces Sdc1 shedding from the surface of hepatocytes in Wt livers, which is directly associated with the decrease in size of intravascular aggregated NETs. Furthermore, administration of purified Sdc1 ectodomains or DNase inhibits the formation of intravascular aggregated neutrophils and NETs and significantly increases the liver bacterial burden in Sdc1-/- mice. These data indicate that Lm induces Sdc1 shedding to subvert the activity of Sdc1 ectodomains to inhibit its clearance by intravascular aggregated NETs.


Assuntos
Armadilhas Extracelulares/imunologia , Listeria monocytogenes/imunologia , Listeriose/imunologia , Neutrófilos/imunologia , Sindecana-1/imunologia , Animais , Armadilhas Extracelulares/genética , Hepatócitos/imunologia , Hepatócitos/patologia , Listeria monocytogenes/patogenicidade , Listeriose/genética , Listeriose/patologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Neutrófilos/patologia , Domínios Proteicos , Sindecana-1/genética
2.
J Biol Chem ; 290(26): 16157-67, 2015 Jun 26.
Artigo em Inglês | MEDLINE | ID: mdl-25931123

RESUMO

Ablation of syndecan-1 in mice is a gain of function mutation that enables mice to significantly resist infection by several bacterial pathogens. Syndecan-1 shedding is induced by bacterial virulence factors, and inhibition of shedding attenuates bacterial virulence, whereas administration of purified syndecan-1 ectodomain enhances virulence, suggesting that bacteria subvert syndecan-1 ectodomains released by shedding for their pathogenesis. However, the pro-pathogenic functions of syndecan-1 ectodomain have yet to be clearly defined. Here, we examined how syndecan-1 ectodomain enhances Staphylococcus aureus virulence in injured mouse corneas. We found that syndecan-1 ectodomain promotes S. aureus corneal infection in an HS-dependent manner. Surprisingly, we found that this pro-pathogenic activity is dependent on 2-O-sulfated domains in HS, indicating that the effects of syndecan-1 ectodomain are structure-based. Our results also showed that purified syndecan-1 ectodomain and heparan compounds containing 2-O-sulfate motifs inhibit S. aureus killing by antimicrobial factors secreted by degranulated neutrophils, but does not affect intracellular phagocytic killing by neutrophils. Immunodepletion of antimicrobial factors with staphylocidal activities demonstrated that CRAMP, a cationic antimicrobial peptide, is primarily responsible for S. aureus killing among other factors secreted by degranulated neutrophils. Furthermore, we found that purified syndecan-1 ectodomain and heparan compounds containing 2-O-sulfate units potently and specifically inhibit S. aureus killing by synthetic CRAMP. These results provide compelling evidence that a specific subclass of sulfate groups, and not the overall charge of HS, permits syndecan-1 ectodomains to promote S. aureus corneal infection by inhibiting a key arm of neutrophil host defense.


Assuntos
Catelicidinas/imunologia , Doenças da Córnea/imunologia , Neutrófilos/imunologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/fisiologia , Sindecana-1/metabolismo , Animais , Peptídeos Catiônicos Antimicrobianos , Doenças da Córnea/genética , Doenças da Córnea/microbiologia , Humanos , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Neutrófilos/microbiologia , Estrutura Terciária de Proteína , Infecções Estafilocócicas/genética , Infecções Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Staphylococcus aureus/patogenicidade , Sindecana-1/química , Sindecana-1/genética , Virulência
3.
Proteoglycan Res ; 1(3)2023 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-38957622

RESUMO

A large number of bacterial pathogens bind to host extracellular matrix (ECM) components. For example, many Gram-negative and Gram-positive pathogens express binding proteins for fibronectin (FN) on their cell surface. Mutagenesis studies of bacterial FN-binding proteins have demonstrated their importance in pathogenesis in preclinical animal models. However, means to draw on these findings to design therapeutic approaches that specifically target FN-bacteria interactions have not been successful because bacterial pathogens can elaborate several FN-binding proteins and also because FN is an essential protein and likely a nondruggable target. Here we report that select heparan compounds potently inhibit Streptococcus pneumoniae infection of injured corneas in mice. Using intact heparan sulfate (HS) and heparin (HP), heparinase-digested fragments of HS, HP oligosaccharides, and chemically or chemoenzymatically modified heparan compounds, we found that inhibition of S. pneumoniae corneal infection by heparan compounds is not mediated by simple charge effects but by a selective sulfate group. Removal of 2-O-sulfates significantly inhibited the ability of HP to inhibit S. pneumoniae corneal infection, whereas the addition of 2-O-sulfates to heparosan (H) significantly increased H's ability to inhibit bacterial corneal infection. Proximity ligation assays indicated that S. pneumoniae attaches directly to FN fibrils in the corneal epithelial ECM and that HS and HP specifically inhibit this binding interaction in a 2-O-sulfate-dependent manner. These data suggest that heparan compounds containing 2-O-sulfate groups protect against S. pneumoniae corneal infection by inhibiting bacterial attachment to FN fibrils in the subepithelial ECM of injured corneas. Moreover, 2-O-sulfated heparan compounds significantly inhibited corneal infection in immunocompromised hosts, by a clinical keratitis isolate of S. pneumoniae, and also when topically administered in a therapeutic manner. These findings suggest that the administration of nonanticoagulant 2-O-sulfated heparan compounds may represent a plausible approach to the treatment of S. pneumoniae keratitis.

4.
J Biol Chem ; 286(5): 3288-97, 2011 Feb 04.
Artigo em Inglês | MEDLINE | ID: mdl-21127056

RESUMO

Many microbial pathogens subvert cell surface heparan sulfate proteoglycans (HSPGs) to infect host cells in vitro. The significance of HSPG-pathogen interactions in vivo, however, remains to be determined. In this study, we examined the role of syndecan-1, a major cell surface HSPG of epithelial cells, in Staphylococcus aureus corneal infection. We found that syndecan-1 null (Sdc1(-/-)) mice significantly resist S. aureus corneal infection compared with wild type (WT) mice that express abundant syndecan-1 in their corneal epithelium. However, syndecan-1 did not bind to S. aureus, and syndecan-1 was not required for the colonization of cultured corneal epithelial cells by S. aureus, suggesting that syndecan-1 does not mediate S. aureus attachment to corneal tissues in vivo. Instead, S. aureus induced the shedding of syndecan-1 ectodomains from the surface of corneal epithelial cells. Topical administration of purified syndecan-1 ectodomains or heparan sulfate (HS) significantly increased, whereas inhibition of syndecan-1 shedding significantly decreased the bacterial burden in corneal tissues. Furthermore, depletion of neutrophils in the resistant Sdc1(-/-) mice increased the corneal bacterial burden to that of the susceptible WT mice, suggesting that syndecan-1 moderates neutrophils to promote infection. We found that syndecan-1 does not affect the infiltration of neutrophils into the infected cornea but that purified syndecan-1 ectodomain and HS significantly inhibit neutrophil-mediated killing of S. aureus. These data suggest a previously unknown bacterial subversion mechanism where S. aureus exploits the capacity of syndecan-1 ectodomains to inhibit neutrophil-mediated bacterial killing mechanisms in an HS-dependent manner to promote its pathogenesis in the cornea.


Assuntos
Doenças da Córnea/microbiologia , Interações Hospedeiro-Patógeno/imunologia , Neutrófilos/imunologia , Staphylococcus aureus/patogenicidade , Sindecana-1/fisiologia , Animais , Proteoglicanas de Heparan Sulfato/metabolismo , Heparitina Sulfato/metabolismo , Camundongos , Camundongos Knockout , Neutrófilos/microbiologia , Infecções Estafilocócicas/patologia
5.
Methods Mol Biol ; 2303: 605-625, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-34626410

RESUMO

Heparan sulfate proteoglycans (HSPGs) are at the forefront of host-microbe interactions. Cell surface HSPGs are thought to promote infection as attachment and internalization receptors for many bacterial pathogens and as soluble inhibitors of host immunity when released from the cell surface by ectodomain shedding. However, the importance of HSPG-pathogen interactions in vivo has yet to be clearly established. Here we describe several representative methods to study the role of HSPGs in systemic bacterial infections, such as bacteremia and sepsis. The overall experimental strategy is to use mouse models to establish the physiological significance of HSPGs, to determine the identity of HSPGs that specifically promote infection, and to define key structural features of HSPGs that enhance bacterial virulence in systemic infections.


Assuntos
Infecções Bacterianas , Animais , Membrana Celular , Modelos Animais de Doenças , Proteoglicanas de Heparan Sulfato , Heparitina Sulfato , Camundongos , Sepse
6.
Am J Pathol ; 174(2): 509-18, 2009 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-19147831

RESUMO

In pneumonia caused by the bacterium Staphylococcus aureus, the intense inflammatory response that is triggered by this infection can lead to the development of lung injury. Little is known, however, about the impact of specific virulence factors on this inflammatory disorder, which causes both significant mortality and morbidity. In this study, we examined the role of beta-toxin, a neutral sphingomyelinase, in S. aureus-induced lung injury. Our results showed that the central features of lung injury--specifically, increased neutrophilic inflammation, vascular leakage of serum proteins into the lung tissue, and exudation of proteins into the airway--are significantly attenuated in mice infected intranasally with S. aureus deficient in beta-toxin compared with mice infected with S. aureus expressing beta-toxin. In addition, intranasal administration of beta-toxin evoked the characteristic features of lung injury in wild-type mice whereas neutropenic mice were protected from such injury. However, mutant beta-toxin mice deficient in sphingomyelinase activity failed to trigger features of lung injury. Ablation of sphingomyelinase activity also interfered with the ability of beta-toxin to stimulate ectodomain shedding of syndecan-1, a major heparan sulfate proteoglycan found in epithelial cells. Moreover, syndecan-1-null mice were significantly protected from beta-toxin-induced lung injury relative to wild-type mice. These data indicate that S. aureus beta-toxin is a critical virulence factor that induces neutrophil-mediated lung injury through both its sphingomyelinase activity and syndecan-1.


Assuntos
Toxinas Bacterianas/metabolismo , Proteínas Hemolisinas/metabolismo , Pneumopatias/patologia , Lesão Pulmonar/metabolismo , Pneumonia Estafilocócica/patologia , Esfingomielina Fosfodiesterase/metabolismo , Sindecana-1/metabolismo , Fatores de Virulência/metabolismo , Animais , Toxinas Bacterianas/genética , Proteínas Hemolisinas/genética , Pneumopatias/metabolismo , Pneumopatias/microbiologia , Lesão Pulmonar/imunologia , Lesão Pulmonar/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Neutrófilos/imunologia , Pneumonia Estafilocócica/imunologia , Pneumonia Estafilocócica/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/metabolismo , Esfingomielina Fosfodiesterase/genética , Fatores de Virulência/genética
7.
mBio ; 11(6)2020 12 08.
Artigo em Inglês | MEDLINE | ID: mdl-33293379

RESUMO

Subversion of heparan sulfate proteoglycans (HSPGs) is thought to be a common virulence mechanism shared by many microbial pathogens. The prevailing assumption is that pathogens co-opt HSPGs as cell surface attachment receptors or as inhibitors of innate host defense. However, there are few data that clearly support this idea in vivo We found that deletion of syndecan-1 (Sdc1), a major cell surface HSPG of epithelial cells, causes a gain of function in a mouse model of scarified corneal infection, where Sdc1-/- corneas were significantly less susceptible to Streptococcus pneumoniae infection. Administration of excess Sdc1 ectodomains significantly inhibited S. pneumoniae corneal infection, suggesting that Sdc1 promotes infection as a cell surface attachment receptor. However, S. pneumoniae did not interact with Sdc1 and Sdc1 was shed upon S. pneumoniae infection, indicating that Sdc1 does not directly support S. pneumoniae adhesion. Instead, Sdc1 promoted S. pneumoniae adhesion by driving the assembly of fibronectin (FN) fibrils in the corneal basement membrane to which S. pneumoniae attaches when infecting injured corneas. S. pneumoniae specifically bound to corneal FN via PavA, and PavA deletion significantly attenuated S. pneumoniae virulence in the cornea. Excess Sdc1 ectodomains inhibited S. pneumoniae corneal infection by binding to the Hep II domain and interfering with S. pneumoniae PavA binding to FN. These findings reveal a previously unknown virulence mechanism of S. pneumoniae where key extracellular matrix (ECM) interactions and structures that are essential for host cell homeostasis are exploited for bacterial pathogenesis.IMPORTANCE Bacterial pathogens have evolved several ingenious mechanisms to subvert host cell biology for their pathogenesis. Bacterial attachment to the host ECM establishes a niche to grow and is considered one of the critical steps of infection. This pathogenic mechanism entails coordinated assembly of the ECM by the host to form the ECM structure and organization that are specifically recognized by bacteria for their adhesion. We serendipitously discovered that epithelial Sdc1 facilitates the assembly of FN fibrils in the corneal basement membrane and that this normal biological function of Sdc1 has detrimental consequences for the host in S. pneumoniae corneal infection. Our studies suggest that bacterial subversion of the host ECM is more complex than previously appreciated.


Assuntos
Fibronectinas/metabolismo , Interações Hospedeiro-Patógeno , Ceratite/metabolismo , Ceratite/microbiologia , Streptococcus pneumoniae/fisiologia , Sindecana-1/metabolismo , Animais , Aderência Bacteriana , Córnea/metabolismo , Córnea/microbiologia , Córnea/patologia , Modelos Animais de Doenças , Matriz Extracelular/metabolismo , Imunofluorescência , Mutação com Ganho de Função , Expressão Gênica , Proteoglicanas de Heparan Sulfato/genética , Proteoglicanas de Heparan Sulfato/metabolismo , Interações Hospedeiro-Patógeno/genética , Imuno-Histoquímica , Ceratite/patologia , Camundongos , Camundongos Knockout , Sindecana-1/genética
8.
Atherosclerosis ; 193(1): 20-7, 2007 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-17055514

RESUMO

Mulberry (Morus Alba L., family Moraceae) leaf extracts have various biological effects including inhibition of oxidative modification of low-density lipoprotein (LDL), which is the major cause of atherosclerosis. Endothelial dysfunction elicited by oxidized LDL (Ox-LDL) has been implicated in atherogenesis. Lectin-like Ox-LDL receptor-1 (LOX-1), a cell-surface receptor for atherogenic Ox-LDL, appears to mediate Ox-LDL-induced inflammation, which may be crucial in atherogenesis. Previous studies revealed that expression of LOX-1 is highly inducible by proinflammatory stimuli, including tumor necrosis factor-alpha (TNF-alpha), lipopolysaccharide (LPS), and transforming growth factor-beta (TGF-beta). Therefore, we examined whether mulberry leaf aqueous fractions inhibit LOX-1 expression induced by proinflammatory stimuli. Pretreatment of cultured bovine aortic endothelial cells (BAECs) with mulberry leaf aqueous fractions inhibited TNF-alpha- and LPS-induced expression of LOX-1 at both protein and mRNA levels in a time- and concentration-dependent manner. In contrast, mulberry leaf aqueous fractions did not affect TGF-beta-induced LOX-1 expression. Furthermore, mulberry leaf aqueous fractions inhibited TNF-alpha-induced activation of nuclear factor-kappaB (NF-kappaB) and phosphorylation of inhibitory factor of NF-kappaB-alpha (IkappaB-alpha) in a time- and concentration-dependent fashion. Thus, mulberry leaf aqueous fractions suppress TNF-alpha- and LPS-induced LOX-1 gene expression, by inhibiting NF-kappaB activation.


Assuntos
Células Endoteliais/efeitos dos fármacos , Células Endoteliais/metabolismo , Morus/química , NF-kappa B/metabolismo , Receptores Depuradores Classe E/genética , Fator de Necrose Tumoral alfa/farmacologia , Animais , Sequência de Bases , Bovinos , Células Cultivadas , DNA/genética , Expressão Gênica/efeitos dos fármacos , Humanos , Proteínas I-kappa B/metabolismo , Inibidor de NF-kappaB alfa , Estresse Oxidativo , Fosforilação , Extratos Vegetais/farmacologia , Folhas de Planta/química , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Proteínas Recombinantes/farmacologia
9.
J Exp Med ; 214(3): 623-637, 2017 03 06.
Artigo em Inglês | MEDLINE | ID: mdl-28148688

RESUMO

We studied three patients with severe skeletal dysplasia, T cell immunodeficiency, and developmental delay. Whole-exome sequencing revealed homozygous missense mutations affecting exostosin-like 3 (EXTL3), a glycosyltransferase involved in heparan sulfate (HS) biosynthesis. Patient-derived fibroblasts showed abnormal HS composition and altered fibroblast growth factor 2 signaling, which was rescued by overexpression of wild-type EXTL3 cDNA. Interleukin-2-mediated STAT5 phosphorylation in patients' lymphocytes was markedly reduced. Interbreeding of the extl3-mutant zebrafish (box) with Tg(rag2:green fluorescent protein) transgenic zebrafish revealed defective thymopoiesis, which was rescued by injection of wild-type human EXTL3 RNA. Targeted differentiation of patient-derived induced pluripotent stem cells showed a reduced expansion of lymphohematopoietic progenitor cells and defects of thymic epithelial progenitor cell differentiation. These data identify EXTL3 mutations as a novel cause of severe immune deficiency with skeletal dysplasia and developmental delay and underline a crucial role of HS in thymopoiesis and skeletal and brain development.


Assuntos
Doenças do Desenvolvimento Ósseo/etiologia , Deficiências do Desenvolvimento/etiologia , Síndromes de Imunodeficiência/etiologia , Mutação , N-Acetilglucosaminiltransferases/genética , Animais , Pré-Escolar , Feminino , Heparitina Sulfato/fisiologia , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Lactente , Linfócitos/fisiologia , Peixe-Zebra
10.
J Infect Dis ; 198(10): 1529-35, 2008 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-18823272

RESUMO

BACKGROUND: Staphylococcus aureus alpha-toxin is a major virulence factor, but its mechanism of action in vivo is incompletely understood. METHODS: We examined the role of alpha-toxin in S. aureus pneumonia using the mouse model of intranasal lung infection with S. aureus strain 8325-4 (hla(+) S. aureus) and an alpha-toxin-deficient mutant strain made on the 8325-4 background (hla(-) S. aureus). RESULTS: Intranasal infection of mice with hla(-) S. aureus resulted in substantially less lung injury and inflammation, pulmonary edema, and tissue bacterial burden than did infection with hla(+) S. aureus. Furthermore, fewer mice infected with hla(-) S. aureus died of the infection, compared with those infected with hla(+) S. aureus. Levels of the CXC chemokines keratinocyte-derived chemokine and macrophage inflammatory protein-2 were significantly lower in the airways of mice infected with hla(-) S. aureus, and this difference was the result of reduced secretion of newly synthesized chemokines into the airway. Consistent with these data, significantly fewer neutrophils were present in the airways and lungs of mice infected with hla(-) S. aureus, compared with those infected with hla(+) S. aureus. CONCLUSIONS: These data suggest that alpha-toxin enhances virulence by facilitating the generation of CXC chemokine gradients and stimulating chemokine-induced neutrophil influx in S. aureus pneumonia.


Assuntos
Toxinas Bacterianas/metabolismo , Quimiocinas CXC/imunologia , Proteínas Hemolisinas/metabolismo , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , Pneumonia Estafilocócica/imunologia , Staphylococcus aureus/fisiologia , Animais , Toxinas Bacterianas/genética , Quimiocinas/metabolismo , Proteínas Hemolisinas/genética , Camundongos , Camundongos Endogâmicos C57BL , Staphylococcus aureus/genética , Staphylococcus aureus/imunologia , Staphylococcus aureus/patogenicidade
11.
J Biol Chem ; 282(1): 159-67, 2007 Jan 05.
Artigo em Inglês | MEDLINE | ID: mdl-17098735

RESUMO

Several microbial pathogens stimulate the ectodomain shedding of host cell surface proteins to promote their pathogenesis. We reported previously that Pseudomonas aeruginosa and Staphylococcus aureus activate the ectodomain shedding of syndecan-1 and that syndecan-1 shedding promotes P. aeruginosa pathogenesis in mouse models of lung and burned skin infections. However, it remains to be determined whether activation of syndecan-1 shedding is a virulence mechanism broadly used by pathogens. Here we show that Streptococcus pneumoniae stimulates syndecan-1 shedding in cell culture-based assays. S. pneumoniae-induced syndecan-1 shedding was repressed by peptide hydroxamate inhibitors of metalloproteinases but not by inhibitors of intracellular signaling pathways previously found to be essential for syndecan-1 shedding caused by P. aeruginosa, S. aureus, or other shedding agonists. A 170-kDa protein fraction with a peptide hydroxamate-sensitive shedding activity was purified by ammonium sulfate precipitation, DEAE chromatography, and size exclusion chromatography. Mass spectrometry analyses revealed that the 170-kDa fraction is composed of ZmpB and ZmpC, two metalloproteinase virulence factors of S. pneumoniae. Both the purified 170-kDa ZmpB/ZmpC fraction and unfractionated S. pneumoniae culture supernatant generated syndecan-1 ectodomains that are smaller than those released by endogenous shedding. Further, a mutant S. pneumoniae strain deficient in zmpC, but not zmpB, lost its capacity to stimulate syndecan-1 shedding. These data demonstrate that S. pneumoniae directly sheds syndecan-1 ectodomains through the action of ZmpC.


Assuntos
Metaloendopeptidases/química , Streptococcus pneumoniae/metabolismo , Sindecana-1/metabolismo , Fatores de Virulência/química , Animais , Primers do DNA/química , Relação Dose-Resposta a Droga , Elementos Facilitadores Genéticos , Deleção de Genes , Espectrometria de Massas , Metaloendopeptidases/metabolismo , Camundongos , Estrutura Terciária de Proteína , Ovinos , Transdução de Sinais
12.
Biochem Biophys Res Commun ; 323(3): 1116-23, 2004 Oct 22.
Artigo em Inglês | MEDLINE | ID: mdl-15381115

RESUMO

Lectin-like oxidized low-density lipoprotein receptor-1 (LOX-1) is a receptor for oxidized LDL. Peroxisome proliferator-activated receptors (PPARs) are nuclear receptors regulating transcription of various genes. We examined effects of PPAR ligands on LOX-1 expression and their transcriptional regulation in vascular endothelial cells. PPARalpha-specific ligands, such as fenofibrate and WY-14643, but not PPARgamma-specific ligands induced LOX-1 expression. Reduced expression of PPARalpha by antisense oligonucleotides directed to PPARalpha blocked fenofibrate-induced LOX-1 expression. Luciferase reporter gene assays with deletion and point mutations in the LOX-1 promoter revealed that transcriptional activity of LOX-1 gene by fenofibrate was localized in the -114/-106 GC box. Electrophoretic mobility shift assays with the radiolabeled GC box sequence showed inducible bands by PPARalpha ligands, which is competitively suppressed by unlabeled GC box motif and by an antibody to PPARalpha. In conclusion, PPARalpha appears to be one of the key regulators that induce LOX-1 expression, utilizing the GC box as a promoter.


Assuntos
Células Endoteliais/fisiologia , Lipoproteínas LDL/metabolismo , Receptores Ativados por Proliferador de Peroxissomo/metabolismo , Receptores de LDL/metabolismo , Ativação Transcricional/fisiologia , Animais , Composição de Bases/genética , Bovinos , Células Cultivadas , Regulação da Expressão Gênica/fisiologia , Ligantes , Lipoproteínas LDL/genética , Receptores Ativados por Proliferador de Peroxissomo/genética , Regiões Promotoras Genéticas/genética , Receptores de LDL/genética , Receptores de LDL Oxidado , Receptores Depuradores Classe E
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA