Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Mais filtros








Base de dados
Intervalo de ano de publicação
1.
PLoS Genet ; 18(6): e1010261, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-35714152

RESUMO

Genome wide association studies (GWAS) have identified thousands of single nucleotide polymorphisms (SNPs) associated with the risk of common disorders. However, since the large majority of these risk SNPs reside outside gene-coding regions, GWAS generally provide no information about causal mechanisms regarding the specific gene(s) that are affected or the tissue(s) in which these candidate gene(s) exert their effect. The 'gold standard' method for understanding causal genes and their mechanisms of action are laborious basic science studies often involving sophisticated knockin or knockout mouse lines, however, these types of studies are impractical as a high-throughput means to understand the many risk variants that cause complex diseases like coronary artery disease (CAD). As a solution, we developed a streamlined, data-driven informatics pipeline to gain mechanistic insights on complex genetic loci. The pipeline begins by understanding the SNPs in a given locus in terms of their relative location and linkage disequilibrium relationships, and then identifies nearby expression quantitative trait loci (eQTLs) to determine their relative independence and the likely tissues that mediate their disease-causal effects. The pipeline then seeks to understand associations with other disease-relevant genes, disease sub-phenotypes, potential causality (Mendelian randomization), and the regulatory and functional involvement of these genes in gene regulatory co-expression networks (GRNs). Here, we applied this pipeline to understand a cluster of SNPs associated with CAD within and immediately adjacent to the gene encoding HDAC9. Our pipeline demonstrated, and validated, that this locus is causal for CAD by modulation of TWIST1 expression levels in the arterial wall, and by also governing a GRN related to metabolic function in skeletal muscle. Our results reconciled numerous prior studies, and also provided clear evidence that this locus does not govern HDAC9 expression, structure or function. This pipeline should be considered as a powerful and efficient way to understand GWAS risk loci in a manner that better reflects the highly complex nature of genetic risk associated with common disorders.


Assuntos
Doença da Artéria Coronariana , Estudo de Associação Genômica Ampla , Proteína 1 Relacionada a Twist/metabolismo , Animais , Doença da Artéria Coronariana/genética , Predisposição Genética para Doença , Estudo de Associação Genômica Ampla/métodos , Histona Desacetilases/metabolismo , Desequilíbrio de Ligação , Camundongos , Polimorfismo de Nucleotídeo Único , Locos de Características Quantitativas/genética , Proteínas Repressoras/metabolismo
2.
Circ Genom Precis Med ; 15(1): e003365, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34961328

RESUMO

BACKGROUND: Hundreds of candidate genes have been associated with coronary artery disease (CAD) through genome-wide association studies. However, a systematic way to understand the causal mechanism(s) of these genes, and a means to prioritize them for further study, has been lacking. This represents a major roadblock for developing novel disease- and gene-specific therapies for patients with CAD. Recently, powerful integrative genomics analyses pipelines have emerged to identify and prioritize candidate causal genes by integrating tissue/cell-specific gene expression data with genome-wide association study data sets. METHODS: We aimed to develop a comprehensive integrative genomics analyses pipeline for CAD and to provide a prioritized list of causal CAD genes. To this end, we leveraged several complimentary informatics approaches to integrate summary statistics from CAD genome-wide association studies (from UK Biobank and CARDIoGRAMplusC4D) with transcriptomic and expression quantitative trait loci data from 9 cardiometabolic tissue/cell types in the STARNET study (Stockholm-Tartu Atherosclerosis Reverse Network Engineering Task). RESULTS: We identified 162 unique candidate causal CAD genes, which exerted their effect from between one and up to 7 disease-relevant tissues/cell types, including the arterial wall, blood, liver, skeletal muscle, adipose, foam cells, and macrophages. When their causal effect was ranked, the top candidate causal CAD genes were CDKN2B (associated with the 9p21.3 risk locus) and PHACTR1; both exerting their causal effect in the arterial wall. A majority of candidate causal genes were represented in cross-tissue gene regulatory co-expression networks that are involved with CAD, with 22/162 being key drivers in those networks. CONCLUSIONS: We identified and prioritized candidate causal CAD genes, also localizing their tissue(s) of causal effect. These results should serve as a resource and facilitate targeted studies to identify the functional impact of top causal CAD genes.


Assuntos
Aterosclerose , Doença da Artéria Coronariana , Aterosclerose/genética , Doença da Artéria Coronariana/genética , Doença da Artéria Coronariana/metabolismo , Redes Reguladoras de Genes , Estudo de Associação Genômica Ampla/métodos , Genômica/métodos , Humanos , Locos de Características Quantitativas
3.
J Clin Invest ; 131(15)2021 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-34338228

RESUMO

Endothelial-mesenchymal transition (EndMT) is associated with various cardiovascular diseases and in particular with atherosclerosis and plaque instability. However, the molecular pathways that govern EndMT are poorly defined. Specifically, the role of epigenetic factors and histone deacetylases (HDACs) in controlling EndMT and the atherosclerotic plaque phenotype remains unclear. Here, we identified histone deacetylation, specifically that mediated by HDAC9 (a class IIa HDAC), as playing an important role in both EndMT and atherosclerosis. Using in vitro models, we found class IIa HDAC inhibition sustained the expression of endothelial proteins and mitigated the increase in mesenchymal proteins, effectively blocking EndMT. Similarly, ex vivo genetic knockout of Hdac9 in endothelial cells prevented EndMT and preserved a more endothelial-like phenotype. In vivo, atherosclerosis-prone mice with endothelial-specific Hdac9 knockout showed reduced EndMT and significantly reduced plaque area. Furthermore, these mice displayed a more favorable plaque phenotype, with reduced plaque lipid content and increased fibrous cap thickness. Together, these findings indicate that HDAC9 contributes to vascular pathology by promoting EndMT. Our study provides evidence for a pathological link among EndMT, HDAC9, and atherosclerosis and suggests that targeting of HDAC9 may be beneficial for plaque stabilization or slowing the progression of atherosclerotic disease.


Assuntos
Aterosclerose/enzimologia , Endotélio/enzimologia , Histona Desacetilases/metabolismo , Placa Aterosclerótica/enzimologia , Proteínas Repressoras/metabolismo , Animais , Aterosclerose/genética , Aterosclerose/patologia , Endotélio/patologia , Histona Desacetilases/genética , Células Endoteliais da Veia Umbilical Humana , Humanos , Camundongos , Camundongos Knockout para ApoE , Placa Aterosclerótica/genética , Placa Aterosclerótica/patologia , Proteínas Repressoras/genética
4.
Atherosclerosis ; 311: 20-29, 2020 10.
Artigo em Inglês | MEDLINE | ID: mdl-32919281

RESUMO

BACKGROUND AND AIMS: Coronary artery disease (CAD) arises from the interaction of genetic and environmental factors. Although genome-wide association studies (GWAS) have identified multiple risk loci and single nucleotide polymorphisms (SNPs) associated with risk of CAD, they are predominantly located in non-coding or intergenic regions and their mechanisms of effect are largely unknown. Accordingly, our objective was to develop a data-driven informatics pipeline to understand complex CAD risk loci, and to apply this to a poorly understood cluster of SNPs in the vicinity of ZEB2. METHODS: We developed a unique informatics pipeline leveraging a multi-tissue CAD genetics-of-gene-expression dataset, GWAS datasets, and other resources. The pipeline first dissected SNP locations and their linkage disequilibrium relationships, and progressed through analyses of tissue-specific expression quantitative trait loci, and then gene-gene, gene-phenotype, SNP-phenotype relationships. The pipeline concluded by exploring CAD-relevant gene regulatory networks (GRNs). RESULTS: We identified three independent CAD risk SNPs in close proximity to the ZEB2 coding region (rs6740731, rs17678683 and rs2252641/rs1830321). Our pipeline determined that these SNPs likely act in concert via the atherosclerotic arterial wall and adipose tissues, by governing metabolic and lipid functions. In addition, ZEB2 is the top key driver of a liver-specific GRN that is related to lipid levels, metabolic and anthropometric measures, and CAD severity. CONCLUSIONS: Using a novel informatics pipeline, we disclosed the multi-faceted mechanisms of action of the ZEB2-associated CAD risk SNPs. This pipeline can serve as a roadmap to dissect complex SNP-gene-tissue-phenotype relationships and to reveal targets for tissue- and gene-specific therapeutic interventions.


Assuntos
Doença da Artéria Coronariana , Estudo de Associação Genômica Ampla , Doença da Artéria Coronariana/genética , Redes Reguladoras de Genes , Predisposição Genética para Doença , Humanos , Polimorfismo de Nucleotídeo Único , Locos de Características Quantitativas , Homeobox 2 de Ligação a E-box com Dedos de Zinco/genética
5.
Sci Rep ; 9(1): 3582, 2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30837512

RESUMO

APOL1-miR193a axis participates in the preservation of molecular phenotype of differentiated podocytes (DPDs). We examined the hypothesis that APOL1 (G0) preserves, but APOL1 risk alleles (G1 and G2) disrupt APOL1-miR193a axis in DPDs. DPDG0s displayed down-regulation of miR193a, but upregulation of nephrin expression. DPDG1s/G2s exhibited an increase in miR193a and down-regulation of the expression of adherens complex's constituents (CD2AP, nephrin, and dendrin). DPDG0s showed decreased Cathepsin L, enhanced dynamin expressions, and the intact actin cytoskeleton. On the contrary, DPDG1s/G2s displayed an increase in Cathepsin L, but down-regulation of dynamin expressions and disorganization of the actin cytoskeleton. APOL1 silencing enhanced miR193a and Cathepsin L, but down-regulated dynamin expressions. DPDG1s/G2s displayed nuclear import of dendrin, indicating an occurrence of destabilization of adherens complexes in APOL1 risk milieu. These findings suggest that DPDG1s and DPDG2s developed disorganized actin cytoskeleton as a consequence of disrupted APOL1-miR193a axis. Interestingly, docking and co-labeling studies suggested an interaction between APOL1 and CD2AP. APOL1G1/G1 and APOL1G1/G2 transgenic mice displayed nuclear import of dendrin indicating destabilization of adherens complexes in podocytes; moreover, these mice showed a four-fold increase in urinary albumin to creatinine ratio and development of focal segmental glomerular lesions.


Assuntos
Citoesqueleto de Actina/metabolismo , Apolipoproteína L1/metabolismo , Podócitos/citologia , Proteínas Adaptadoras de Transdução de Sinal/metabolismo , Alelos , Animais , Apolipoproteína L1/química , Apolipoproteína L1/genética , Diferenciação Celular , Proteínas do Citoesqueleto/metabolismo , Regulação da Expressão Gênica , Humanos , Camundongos , Modelos Moleculares , Podócitos/metabolismo , Conformação Proteica , Transdução de Sinais
6.
Am J Pathol ; 188(11): 2508-2528, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30201495

RESUMO

Human parietal epithelial cells (PECs) are progenitor cells that sustain podocyte homeostasis. We hypothesized that the lack of apolipoprotein (APO) L1 ensures the PEC phenotype, but its induction initiates PEC transition (expression of podocyte markers). APOL1 expression and down-regulation of miR193a coincided with the expression of podocyte markers during the transition. The induction of APOL1 also stimulated transition markers in human embryonic kidney cells (cells with undetectable APOL1 protein expression). APOL1 silencing in PECs up-regulated miR193a expression, suggesting the possibility of a reciprocal feedback relationship between APOL1 and miR193a. HIV, interferon-γ, and vitamin D receptor agonist down-regulated miR193a expression and induced APOL1 expression along with transition markers in PECs. Luciferase assay suggested a putative interaction between miR193a and APOL1. Since silencing of APOL1 attenuated HIV-, vitamin D receptor agonist-, miR193a inhibitor-, and interferon-γ-induced expression of transition markers, APOL1 appears to be a critical functional constituent of the miR193a- APOL1 axis in PECs. This notion was confirmed by further enhanced expression of PEC markers in APOL1 mRNA-silenced PECs. In vivo studies, glomeruli in patients with HIV, and HIV/APOL1 transgenic mice had foci of PECs expressing synaptopodin, a transition marker. APOL1 likely regulates PEC molecular phenotype through modulation of miR193a expression, and APOL1 and miR193a share a reciprocal feedback relationship.


Assuntos
Nefropatia Associada a AIDS/patologia , Apolipoproteína L1/metabolismo , Células Epiteliais/patologia , Regulação da Expressão Gênica , Glomérulos Renais/patologia , MicroRNAs/genética , Nefropatia Associada a AIDS/metabolismo , Nefropatia Associada a AIDS/virologia , Animais , Apolipoproteína L1/genética , Estudos de Casos e Controles , Células Epiteliais/metabolismo , Células HEK293 , Células Hep G2 , Humanos , Glomérulos Renais/metabolismo , Camundongos , Camundongos Transgênicos
7.
Exp Mol Pathol ; 102(1): 97-105, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-28069388

RESUMO

Vitamin D receptor (VDR) deficient status has been shown to be associated with the activation of renin angiotensin system (RAS). We hypothesized that lack of VDR would enhance p53 expression in podocytes through down regulation of SIRT1; the former would enhance the transcription of angiotensinogen (Agt) and angiotensinogen II type 1 receptor (AT1R) leading to the activation of RAS. Renal tissues of VDR mutant (M) mice displayed increased expression of p53, Agt, renin, and AT1R. In vitro studies, VDR knockout podocytes not only displayed up regulation p53 but also displayed enhanced expression of Agt, renin and AT1R. VDR deficient podocytes also displayed an increase in mRNA expression for p53, Agt, renin, and AT1R. Interestingly, renal tissues of VDR-M as well as VDR heterozygous (h) mice displayed attenuated expression of deacetylase SIRT1. Renal tissues of VDR-M mice showed acetylation of p53 at lysine (K) 382 residues inferring that enhanced p53 expression in renal tissues could be the result of ongoing acetylation, a consequence of SIRT1 deficient state. Notably, podocytes lacking SIRT1 not only showed acetylation of p53 at lysine (K) 382 residues but also displayed enhanced p53 expression. Either silencing of SIRT1/VDR or treatment with high glucose enhanced podocyte PPAR-y expression, whereas, immunoprecipitation (IP) of their lysates with anti-retinoid X receptor (RXR) antibody revealed presence of PPAR-y. It appears that either the deficit of SIRT1 has de-repressed expression of PPAR-y or enhanced podocyte expression of PPAR-y (in the absence of VDR) has contributed to the down regulation of SIRT1.


Assuntos
Podócitos/metabolismo , Receptores de Calcitriol/genética , Sistema Renina-Angiotensina/genética , Sirtuína 1/genética , Acetilação , Angiotensinogênio/genética , Angiotensinogênio/metabolismo , Animais , Western Blotting , Células Cultivadas , Humanos , Rim/citologia , Rim/metabolismo , Lisina/genética , Lisina/metabolismo , Camundongos Knockout , Modelos Genéticos , Podócitos/citologia , Interferência de RNA , Receptor Tipo 1 de Angiotensina/genética , Receptor Tipo 1 de Angiotensina/metabolismo , Receptores de Calcitriol/deficiência , Renina/genética , Renina/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sirtuína 1/metabolismo , Proteína Supressora de Tumor p53/genética , Proteína Supressora de Tumor p53/metabolismo
8.
Front Microbiol ; 6: 788, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26347716

RESUMO

Illicit drug abuse is highly prevalent and serves as a powerful co-factor for HIV exacerbation. Epigenetic alterations in drug abuse and HIV infection determine expression of several critical genes such as vitamin D receptor (VDR), which participates in proliferation, differentiation, cell death under both physiological and pathological conditions. On that account, active vitamin D, the ligand of VDR, is used as an adjuvant therapy to control infection, slow down progression of chronic kidney diseases, and cancer chemotherapy. Interestingly, vitamin D may not be able to augment VDR expression optimally in several instances where epigenetic contributes to down regulation of VDR; however, reversal of epigenetic corruption either by demethylating agents (DACs) or histone deacetylase (HDAC) inhibitors would be able to maximize expression of VDR in these instances.

9.
J Mol Biol ; 427(20): 3201-3215, 2015 Oct 09.
Artigo em Inglês | MEDLINE | ID: mdl-26210663

RESUMO

HIV (human immunodeficiency virus) has been reported to induce podocyte injury through down regulation of vitamin D receptor (VDR) and activation of renin angiotensin system; however, the involved mechanism is not clear. Since HIV has been reported to modulate gene expression via epigenetic phenomena, we asked whether epigenetic factors contribute to down regulation of VDR. Kidney cells in HIV transgenic mice and HIV-infected podocytes (HIV/HPs) displayed enhanced expression of SNAIL, a repressor of VDR. To elucidate the mechanism, we studied the effect of HIV on expression of molecules involved in SNAIL repressor complex formation and demonstrated that HIV enhances expression of the histone deacetylase HDAC1 and DNA methyl transferases DNMT3b and DNMT1. 293T cells, when stably transfected with SNAIL (SNAIL/293T), displayed suppressed transcription and translation of VDR. In SNAIL/293T cells, co-immunoprecipitation studies revealed the association of HDAC1, DNMT3b, DNMT1, and mSin3A with SNAIL. Chromatin immunoprecipitation experiments confirmed the presence of the SNAIL repressor complex at the VDR promoter. Consistent with the enhanced DNA methyl transferase expression in HIV/HPs, there was an increased CpG methylation at the VDR promoter. Chromatin immunoprecipitation assay confirmed occurrence of H3K4 trimethylation on SNAIL promoter. Neither a VDR agonist (VDA) nor an HDAC inhibitor (HDACI) nor a demethylating agent (DAC) individually could optimally up regulate VDR in HIV milieu. However, VDA and HDACI when combined were successful in de-repressing VDR expression. Our findings demonstrate that SNAIL recruits multiple chromatin enzymes to form a repressor complex in HIV milieu that down regulates VDR expression.


Assuntos
Metilação de DNA/genética , Podócitos/metabolismo , Receptores de Calcitriol/biossíntese , Fatores de Transcrição/metabolismo , Animais , Linhagem Celular , Imunoprecipitação da Cromatina , DNA (Citosina-5-)-Metiltransferase 1 , DNA (Citosina-5-)-Metiltransferases/biossíntese , Ativação Enzimática , Células HEK293 , HIV/genética , Infecções por HIV/genética , Histona Desacetilase 1/biossíntese , Histonas/metabolismo , Humanos , Rim/citologia , Camundongos , Camundongos Transgênicos , Podócitos/virologia , Regiões Promotoras Genéticas , Interferência de RNA , RNA Interferente Pequeno , Receptores de Calcitriol/metabolismo , Sistema Renina-Angiotensina/fisiologia , Proteínas Repressoras/biossíntese , Complexo Correpressor Histona Desacetilase e Sin3 , Fatores de Transcrição da Família Snail , Fatores de Transcrição/biossíntese , Fatores de Transcrição/genética , DNA Metiltransferase 3B
10.
Am J Physiol Renal Physiol ; 309(3): F189-203, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26084932

RESUMO

ANG II type 1 receptor blockade (AT1R-BLK) is used extensively to slow down the progression of proteinuric kidney diseases. We hypothesized that AT1R-BLK provides podocyte protection through regulation of silencing mediator of retinoic acid and thyroid hormone receptor (SMRT) and vitamin D receptor (VDR) expression under adverse milieus such as high glucose and human immunodeficiency virus infection. Both AT1R-BLK and VDR agonists (VDAs) stimulated VDR complex formation that differed not only in their composition but also in their functionality. AT1R-BLK-induced VDR complexes contained predominantly unliganded VDR, SMRT, and phosphorylated histone deacetylase 3, whereas VDA-VDR complexes were constituted by liganded VDR and CREB-binding protein/p300. AT1R-BLK-induced complexes attenuated podocyte acetyl-histone 3 levels as well as cytochrome P-450 family 24A1 expression, thus indicating their deacetylating and repressive properties. On the other hand, VDA-VDR complexes not only increased podocyte acetyl-histone 3 levels but also enhanced cytochrome P-450 family 24A1 expression, thus suggesting their acetylating and gene activation properties. AT1R-BLK- induced podocyte SMRT inhibited expression of the proapoptotic gene BAX through downregulation of Wip1 and phosphorylation of checkpoint kinase 2 in high-glucose milieu. Since SMRT-depleted podocytes lacked AT1R-BLK-mediated protection against DNA damage, it appears that SMRT is necessary for DNA repairs during AT1R-BLK. We conclude that AT1R-BLK provides podocyte protection in adverse milieus predominantly through SMRT expression and partly through unliganded VDR expression in 1,25(OH)2D-deficient states; on the other hand, AT1R-BLK contributes to liganded VDR expression in 1,25(OH)2D-sufficient states.


Assuntos
Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Correpressor 2 de Receptor Nuclear/fisiologia , Acetilação , Proteínas Reguladoras de Apoptose/biossíntese , Proteínas Correpressoras/efeitos dos fármacos , Dano ao DNA , Relação Dose-Resposta a Droga , Histonas/metabolismo , Humanos , Losartan/farmacologia , Podócitos/efeitos dos fármacos , Podócitos/enzimologia , Complexo de Endopeptidases do Proteassoma/efeitos dos fármacos , Substâncias Protetoras/farmacologia , Receptores de Calcitriol/efeitos dos fármacos , Vitamina D3 24-Hidroxilase/biossíntese , Vitamina D3 24-Hidroxilase/metabolismo
11.
J Leukoc Biol ; 96(4): 601-9, 2014 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-24970860

RESUMO

HIV is known to subvert cellular machinery to enhance its replication. Recently, HIV has been reported to enhance TC renin expression. We hypothesized that HIV induces and maintains high renin expression to promote its own replication in TCs. Renin enhanced HIV replication in TCs in a dose-dependent manner. (P)RR-deficient TCs, as well as those lacking renin, displayed attenuated NF-κB activity and HIV replication. TCs treated with renin and Hpr displayed activation of the (P)RR-PLZF protein signaling cascade. Renin, HIV, and Hpr activated the PI3K pathway. Both renin and Hpr cleaved Agt (a renin substrate) to Ang I and also cleaved Gag polyproteins (protease substrate) to p24. Furthermore, aliskiren, a renin inhibitor, reduced renin- and Hpr-induced cleavage of Agt and Gag polyproteins. These findings indicate that renin contributes to HIV replication in TCs via the (P)RR-PLZF signaling cascade and through cleavage of the Gag polyproteins.


Assuntos
HIV-1/efeitos dos fármacos , HIV-1/fisiologia , Renina/farmacologia , Linfócitos T/metabolismo , Linfócitos T/virologia , Replicação Viral/efeitos dos fármacos , Humanos , Fatores de Transcrição Kruppel-Like/metabolismo , Modelos Biológicos , NF-kappa B/metabolismo , Proteína com Dedos de Zinco da Leucemia Promielocítica , Proteólise/efeitos dos fármacos , Receptores de Superfície Celular/deficiência , Receptores de Superfície Celular/metabolismo , Renina/metabolismo , Transdução de Sinais , ATPases Vacuolares Próton-Translocadoras/deficiência , ATPases Vacuolares Próton-Translocadoras/metabolismo
12.
J Leukoc Biol ; 94(4): 769-77, 2013 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-23911867

RESUMO

It is well known that patients with HIV are prone to diabetes mellitus because of the side effects of HARRT. However, whether high glucose affects the HIV infection of T cells is not clear. Recent studies have shown that upregulation of GLUT-1 renders T cells susceptible to HIV infection. We hypothesized that hyperglycemia has the potential to increase HIV infection by enhancing its entry into immune cells. The effect of high glucose on HIV entry into T cells (Jurkat cells and PBMCs) and the mechanisms involved were investigated. High glucose significantly enhanced HIV entry, which was associated with increased T-cell expression of CXCR4. However, T cells with silenced HIF-1α displayed attenuated expression of CXCR4, whereas T cells with silenced CXCR4 showed decreased HIV entry in a high-glucose milieu. On the one hand, high glucose stimulated T-cell ROS generation, and H(2)O(2) at low concentrations enhanced the entry of HIV into T cells. On the other hand, inhibition of ROS not only attenuated high-glucose-mediated T-cell expression of CXCR4 and HIF-1α but also mitigated T-cell HIV entry in a high-glucose milieu. In our study, high glucose enhanced HIV entry into T cells by increasing expression of CXCR4 and HIF-1α.


Assuntos
Glucose/farmacologia , Receptores CXCR4/genética , Linfócitos T/metabolismo , Linfócitos T/virologia , Regulação para Cima/efeitos dos fármacos , Internalização do Vírus/efeitos dos fármacos , Membrana Celular/efeitos dos fármacos , Membrana Celular/metabolismo , Inativação Gênica/efeitos dos fármacos , Humanos , Peróxido de Hidrogênio/farmacologia , Subunidade alfa do Fator 1 Induzível por Hipóxia/genética , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Células Jurkat , Modelos Biológicos , Estresse Oxidativo/efeitos dos fármacos , Estresse Oxidativo/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Receptores CXCR4/metabolismo , Linfócitos T/efeitos dos fármacos
13.
Am J Physiol Renal Physiol ; 305(3): F343-54, 2013 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-23678040

RESUMO

Oxidative stress has been implicated to contribute to HIV-induced kidney cell injury; however, the role of p53, a modulator of oxidative stress, has not been evaluated in the development of HIV-associated nephropathy (HIVAN). We hypothesized that mammalian target of rapamycin (mTOR) may be critical for the induction of p53-mediated oxidative kidney cell injury in HIVAN. To test our hypothesis, we evaluated the effect of an mTOR inhibitor, rapamycin, on kidney cell p53 expression, downstream signaling, and kidney cell injury in both in vivo and in vitro studies. Inhibition of the mTOR pathway resulted in downregulation of renal tissue p53 expression, associated downstream signaling, and decreased number of sclerosed glomeruli, tubular microcysts, and apoptosed and 8-hydroxy deoxyguanosine (8-OHdG)-positive (+ve) cells in Tg26 mice. mTOR inhibition not only attenuated kidney cell expression of p66ShcA and phospho-p66ShcA but also reactivated the redox-sensitive stress response program in the form of enhanced expression of manganese superoxide dismutase (MnSOD) and catalase. In in vitro studies, the mTOR inhibitor also provided protection against HIV-induced podocyte apoptosis. Moreover, mTOR inhibition downregulated HIV-induced podocyte (HP/HIV) p53 expression. Since HP/HIV silenced for mTOR displayed a lack of expression of p53 as well as attenuated podocyte apoptosis, this suggests that mTOR is critical for kidney cell p53 activation and associated oxidative kidney cell injury in the HIV milieu.


Assuntos
Nefropatia Associada a AIDS/patologia , Injúria Renal Aguda/etiologia , Injúria Renal Aguda/patologia , Infecções por HIV/complicações , Infecções por HIV/patologia , Estresse Oxidativo/fisiologia , Serina-Treonina Quinases TOR/fisiologia , Proteína Supressora de Tumor p53/fisiologia , 8-Hidroxi-2'-Desoxiguanosina , Animais , Apoptose/fisiologia , Catalase/metabolismo , Desoxiguanosina/análogos & derivados , Desoxiguanosina/metabolismo , Inativação Gênica , Humanos , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Camundongos , Camundongos Transgênicos , Podócitos/patologia , Ratos , Reação em Cadeia da Polimerase em Tempo Real , Transdução de Sinais/fisiologia , Superóxido Dismutase/metabolismo
14.
PLoS One ; 8(3): e55748, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23555556

RESUMO

Morphine has been reported to accelerate the progression of chronic kidney disease. However, whether morphine affects slit diaphragm (SD), the major constituent of glomerular filtration barrier, is still unclear. In the present study, we examined the effect of morphine on glomerular filtration barrier in general and podocyte integrity in particular. Mice were administered either normal saline or morphine for 72 h, then urine samples were collected and kidneys were subsequently isolated for immunohistochemical studies and Western blot. For in vitro studies, human podocytes were treated with morphine and then probed for the molecular markers of slit diaphragm. Morphine-receiving mice displayed a significant increase in albuminuria and showed effacement of podocyte foot processes. In both in vivo and in vitro studies, the expression of synaptopodin, a molecular marker for podocyte integrity, and the slit diaphragm constituting molecules (SDCM), such as nephrin, podocin, and CD2-associated protein (CD2AP), were decreased in morphine-treated podocytes. In vitro studies indicated that morphine modulated podocyte expression of SDCM through opiate mu (MOR) and kappa (KOR) receptors. Since morphine also enhanced podocyte oxidative stress, the latter seems to contribute to decreased SDCM expression. In addition, AKT, p38, and JNK pathways were involved in morphine-induced down regulation of SDCM in human podocytes. These findings demonstrate that morphine has the potential to alter the glomerular filtration barrier by compromising the integrity of podocytes.


Assuntos
Albuminúria/metabolismo , Morfina/efeitos adversos , Entorpecentes/efeitos adversos , Podócitos/metabolismo , Proteínas Adaptadoras de Transdução de Sinal/biossíntese , Albuminúria/induzido quimicamente , Albuminúria/patologia , Animais , Linhagem Celular Transformada , Proteínas do Citoesqueleto/biossíntese , Regulação da Expressão Gênica/efeitos dos fármacos , Taxa de Filtração Glomerular/efeitos dos fármacos , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/biossíntese , MAP Quinase Quinase 4/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Proteínas de Membrana/biossíntese , Camundongos , Morfina/farmacologia , Entorpecentes/farmacologia , Podócitos/patologia , Proteínas Proto-Oncogênicas c-akt/metabolismo , Receptores Opioides kappa/biossíntese , Receptores Opioides mu/biossíntese , Insuficiência Renal Crônica/metabolismo , Insuficiência Renal Crônica/patologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
15.
Am J Physiol Renal Physiol ; 304(11): F1347-57, 2013 Jun 01.
Artigo em Inglês | MEDLINE | ID: mdl-23467424

RESUMO

Alterations in the podocyte actin cytoskeleton have been implicated in the development of proteinuric kidney diseases. In the present study, we evaluated the effect of HIV on the podocyte actin cytoskeleton and the mechanism involved. We hypothesized that HIV may be compromising the actin cytoskeleton via downregulation of the vitamin D receptor (VDR) of conditionally immortalized differentiated human podocytes (CIDHPs). HIV-transduced podocytes (HIV/CIDHPs) not only displayed downregulation of VDR but also showed activation of the renin-angiotensin system (RAS) in the form of enhanced expression of renin and increased production of ANG II. Moreover, CIDHPs lacking VDR displayed enhanced ANG II production, and treatment of HIV/CIDHPs with EB1089 (vitamin D3; VD) attenuated ANG II production. HIV/CIDHPs as well as ANG II-treated CIDHPs exhibited enhanced expression of cathepsin (CTS) L. Additionally, losartan (an ANG II type I receptor blocker) inhibited both HIV- and ANG II-induced podocyte cathepsin L expression. Furthermore, VD downregulated HIV-induced podocyte CTSL expression. Both losartan and free radical scavengers attenuated HIV- and ANG II-induced podocyte reactive oxygen species (ROS) generation. HIV also led to cytosolic CTSL accumulation through enhancement of podocyte lysosomal membrane permeabilization; on the other hand, VD, losartan, and superoxide dismutase (SOD) attenuated HIV-induced enhanced podocyte cytosolic CTSL accumulation. Morphological evaluation of HIV/CIDHPs revealed sparse actin filaments and attenuated expression of dynamin. Interestingly, podocytes lacking CTSL displayed enhanced dynamin expression, and HIV/CIDHPs expressing CTSL exhibited downregulation of dynamin. These findings indicate that HIV-induced downregulation of podocyte VDR and associated RAS activation and cytosolic CTSL accumulation compromised the actin cytoskeleton.


Assuntos
Actinas , Citoesqueleto/ultraestrutura , Regulação para Baixo , HIV-1/fisiologia , Podócitos/virologia , Receptores de Calcitriol/genética , Nefropatia Associada a AIDS/virologia , Angiotensina II/biossíntese , Catepsina L/genética , Expressão Gênica , Infecções por HIV/patologia , Infecções por HIV/fisiopatologia , HIV-1/genética , Humanos , Estresse Oxidativo , Podócitos/ultraestrutura , Receptores de Calcitriol/fisiologia , Sistema Renina-Angiotensina/fisiologia , Transdução Genética , Transfecção
16.
J Leukoc Biol ; 93(4): 623-31, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23390308

RESUMO

Epigenetics contributes to the development of variety of diseases by modulation of gene expression. We evaluated the effect of HIV-induced VDR methylation on loss of TCs. HIV/TC displayed enhanced VDR-CpG methylation and increased expression of Dnmt3b but attenuated expression of VDR. A demethylating agent, AZA, inhibited this effect of HIV. HIV/TC also displayed the activation of the RAS, which was reversed by EB (a VDA). Further, HIV/TCs displayed enhanced generation of ROS and induction of DSBs but attenuated DNA repair response. However, in the presence of AZA, EB, LOS (a RAS blocker), Cat, and tempol (free radical scavengers), HIV-induced TC ROS generation and induction of DSBs were attenuated but associated with enhanced DNA repair. Additionally, AZA, EB, and LOS provided protection against HIV-induced TC apoptosis. These findings suggested that HIV-induced TC apoptosis was mediated through ROS generation in response to HIV-induced VDR methylation and associated activation of the RAS.


Assuntos
HIV-1/fisiologia , Receptores de Calcitriol/genética , Linfócitos T/virologia , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Antimetabólitos Antineoplásicos/farmacologia , Apoptose , Azacitidina/farmacologia , Calcitriol/análogos & derivados , Calcitriol/farmacologia , Catalase/antagonistas & inibidores , Catalase/genética , Catalase/metabolismo , Células Cultivadas , Ilhas de CpG , DNA (Citosina-5-)-Metiltransferases/antagonistas & inibidores , DNA (Citosina-5-)-Metiltransferases/genética , DNA (Citosina-5-)-Metiltransferases/metabolismo , Metilação de DNA , Reparo do DNA , Inibidores Enzimáticos/farmacologia , Epigênese Genética , Regulação Neoplásica da Expressão Gênica , Humanos , Losartan/farmacologia , Contagem de Linfócitos , Espécies Reativas de Oxigênio/metabolismo , Receptores de Calcitriol/antagonistas & inibidores , Receptores de Calcitriol/metabolismo , Linfócitos T/efeitos dos fármacos , Linfócitos T/metabolismo , Linfócitos T/patologia , Proteínas ras/antagonistas & inibidores , Proteínas ras/genética , Proteínas ras/metabolismo , DNA Metiltransferase 3B
17.
J Neuroimmune Pharmacol ; 8(1): 251-61, 2013 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-23054367

RESUMO

Ethanol has been demonstrated to cause T cell apoptosis. In the present study, we evaluated the role of VDR and the renin angiotensin system (RAS) in oxidative stress-induced T cell apoptosis. Ethanol-treated human T cells displayed down regulation of vitamin D receptor (VDR) and the activation of the RAS in the form of enhanced T cell renin expression and angiotensin II (Ang II) production. The silencing of VDR with siRNA displayed the activation of the RAS, and activation of the VDR resulted in the down regulation of the RAS. It suggested that ethanol-induced T cell RAS activation was dependent on the VDR status. T cell ROS generation by ethanol was found to be dose dependent. Conversely, ethanol-induced ROS generation was inhibited if VDR was activated or Ang II was blocked by an angiotensin II type 1 (AT1) receptor blocker (Losartan). Furthermore, it was observed that ethanol not only induced double strand breaks in T cells but also attenuated DNA repair response, whereas, VDR activation inhibited ethanol-induced double strand breaks and also enhanced DNA repairs. Since free radical scavengers inhibited ethanol-induced DNA damage, it would indicate that ethanol-induced DNA damage was mediated through ROS generation. These findings indicated that ethanol-induced T cell apoptosis was mediated through ROS generation in response to ethanol-induced down regulation of VDR and associated activation of the RAS.


Assuntos
Apoptose/efeitos dos fármacos , Depressores do Sistema Nervoso Central/farmacologia , Etanol/farmacologia , Receptores de Calcitriol/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos , Angiotensina II/metabolismo , Bloqueadores do Receptor Tipo 1 de Angiotensina II/farmacologia , Anexina A5/metabolismo , Western Blotting , Sobrevivência Celular/efeitos dos fármacos , Quebras de DNA de Cadeia Dupla/efeitos dos fármacos , Reparo do DNA , Ensaio de Imunoadsorção Enzimática , Imunofluorescência , Humanos , Marcação In Situ das Extremidades Cortadas , Células Jurkat , Cinética , Losartan/farmacologia , Espécies Reativas de Oxigênio/metabolismo , Sistema Renina-Angiotensina/efeitos dos fármacos , Sistema Renina-Angiotensina/fisiologia
18.
Am J Physiol Cell Physiol ; 303(6): C607-15, 2012 Sep 15.
Artigo em Inglês | MEDLINE | ID: mdl-22763121

RESUMO

Opiates have been reported to induce T cell loss. We evaluated the role of vitamin D receptor (VDR) and the activation of the renin-angiotensin system (RAS) in morphine-induced T cell loss. Morphine-treated human T cells displayed downregulation of VDR and the activation of the RAS. On the other hand, a VDR agonist (EB1089) enhanced T cell VDR expression both under basal and morphine-stimulated states. Since T cells with silenced VDR displayed the activation of the RAS, whereas activation of the VDR was associated with downregulation of the RAS, it appears that morphine-induced T cell RAS activation was dependent on the VDR status. Morphine enhanced reactive oxygen species (ROS) generation in a dose-dependent manner. Naltrexone (an opiate receptor antagonist) inhibited morphine-induced ROS generation and thus, suggested the role of opiate receptors in T cell ROS generation. The activation of VDR as well as blockade of ANG II (by losartan, an AT(1) receptor blocker) also inhibited morphine-induced T cell ROS generation. Morphine not only induced double-strand breaks (DSBs) in T cells but also attenuated DNA repair response, whereas activation of VDR not only inhibited morphine-induced DSBs but also enhanced DNA repair. Morphine promoted T cell apoptosis; however, this effect of morphine was inhibited by blockade of opiate receptors, activation of the VDR, and blockade of the RAS. These findings indicate that morphine-induced T cell apoptosis is mediated through ROS generation in response to morphine-induced downregulation of VDR and associated activation of the RAS.


Assuntos
Apoptose/fisiologia , Regulação para Baixo/fisiologia , Morfina/farmacologia , Receptores de Calcitriol/metabolismo , Sistema Renina-Angiotensina/fisiologia , Linfócitos T/fisiologia , Apoptose/efeitos dos fármacos , Regulação para Baixo/efeitos dos fármacos , Humanos , Células Jurkat , Leucócitos Mononucleares/efeitos dos fármacos , Leucócitos Mononucleares/fisiologia , Morfina/antagonistas & inibidores , Espécies Reativas de Oxigênio/metabolismo , Sistema Renina-Angiotensina/efeitos dos fármacos , Linfócitos T/efeitos dos fármacos
19.
Am J Physiol Renal Physiol ; 303(5): F711-20, 2012 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-22718888

RESUMO

In the present study, we evaluated the effect of inhibition of renin activity (aliskiren) on the progression of renal lesions in two different mouse models (Vpr and Tg26) of human immunodeficiency virus (HIV)-associated nephropathy (HIVAN). In protocol A, Vpr mice were fed either water (C-VprA) or doxycycline [Doxy (D-VprA)] in their drinking water for 6 wk. In protocols B and C, Vpr mice received either normal saline (C-VprB/C), Doxy + normal saline (D-VprB/C), or Doxy + aliskiren (AD-VprB/C) for 6 wk (protocol B) or 12 wk (protocol C). In protocols D and E, Vpr mice were fed Doxy for 6 wk followed by kidney biopsy. Subsequently, half of the mice were administered either normal saline (D-VprD/E) or aliskiren (AD-VprD/E) for 4 wk (protocol D) or 8 (protocol E) wk. All D-VprA mice showed renal lesions in the form of focal segmental glomerular sclerosis and dilatation of tubules. In protocols B and C, aliskiren diminished both progression of renal lesions and proteinuria. In protocol C, aliskiren also diminished (P < 0.01) the rise in blood urea. In all groups, Doxy-treated mice displayed increased serum ANG I levels (the product of plasma renin activity); on the other hand, all aliskiren-treated mice displayed diminished serum ANG I levels. Renal tissues of D-VprC displayed increased ANG II content; however, aliskiren attenuated renal tissue ANG II production in AD-VprC. In protocol D, AD-VprD showed a 24.2% increase in the number of sclerosed glomeruli compared with 139.2% increase in sclerosed glomeruli in D-VprD (P < 0.01) from their baseline. The attenuating effect of aliskiren on the progression of renal lesions continued in AD-VprE. Aliskiren also diminished blood pressure, proteinuria, and progression of renal lesions in Tg26 mice. These findings indicate that inhibition of renin activity has a potential to slow down the progression of HIVAN.


Assuntos
Nefropatia Associada a AIDS/tratamento farmacológico , Renina/antagonistas & inibidores , Nefropatia Associada a AIDS/patologia , Amidas/uso terapêutico , Animais , Biópsia , Progressão da Doença , Doxiciclina/administração & dosagem , Fumaratos/uso terapêutico , Glomerulosclerose Segmentar e Focal/patologia , Rim/patologia , Camundongos , Camundongos Transgênicos , Sistema Renina-Angiotensina/fisiologia
20.
Nephrology (Carlton) ; 16(4): 383-8, 2011 May.
Artigo em Inglês | MEDLINE | ID: mdl-21114569

RESUMO

AIM: Whether or not completing the hepatitis B vaccination in patients who have undergone kidney transplantation in the middle of incomplete vaccination schedule leads to development of protective antibody titres is not known. This study was designed to determine whether the strategy of completing hepatitis B virus (HBV) vaccination after transplantation is efficacious. METHODS: Sixty-four end-stage renal disease (ESRD) patients were screened for hepatitis B surface antigen (HBsAg), antibodies to hepatitis B surface antigen (anti-HBs), hepatitis B e-antigen (HBeAg) and HBV DNA. HBsAg negative patients received four doses of 40 µg recombinant HBV vaccine. Schedule was continued in after transplantation period if it was incomplete before transplant. Anti-Hbs titres were evaluated at 1, 3, 6, 9 and 12 months. RESULTS: Past HBV infection was noted in 12 patients: 10 by serology plus viraemia and two by viraemia alone. Of the 46 patients without current or past HBV infection who had received at least two doses of the vaccine before transplant, 17 each had received two and three doses and 12 had completed the schedule. Seventeen (37%) exhibited protective titres. Patients who had completed vaccination were more likely to have protective titres than those incompletely vaccinated (P = 0.02). Five patients responded to post-transplant vaccination. CONCLUSION: Partially vaccinated patients do not mount an adequate antibody response despite continued vaccination in the post-transplant period, whereas complete vaccination provides protection in 60%. The present study data highlights the need of administration of a full schedule of HBV vaccination before kidney transplantation. Nucleic acid-based tests can identify occult HBV infection.


Assuntos
Vacinas contra Hepatite B/administração & dosagem , Hepatite B/prevenção & controle , Falência Renal Crônica/terapia , Transplante de Rim , Diálise Renal , Adulto , Biomarcadores/sangue , DNA Viral/sangue , Feminino , Hepatite B/diagnóstico , Hepatite B/epidemiologia , Anticorpos Anti-Hepatite B/sangue , Antígenos de Superfície da Hepatite B/sangue , Antígenos E da Hepatite B/sangue , Vírus da Hepatite B/genética , Vírus da Hepatite B/imunologia , Humanos , Esquemas de Imunização , Índia/epidemiologia , Falência Renal Crônica/epidemiologia , Transplante de Rim/efeitos adversos , Masculino , Pessoa de Meia-Idade , Diálise Renal/efeitos adversos , Fatores de Tempo , Resultado do Tratamento , Vacinação , Carga Viral , Adulto Jovem
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA