Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Mais filtros











Base de dados
Intervalo de ano de publicação
1.
Viruses ; 11(7)2019 06 27.
Artigo em Inglês | MEDLINE | ID: mdl-31252545

RESUMO

BACKGROUND: BK polyomavirus (BKV) reactivates from latency after immunosuppression in renal transplant patients, resulting in BKV-associated nephropathy (BKVAN). BKVAN has emerged as an important cause of graft dysfunction and graft loss among transplant patients. BKV infection in kidney transplant patients has increased over recent decades which correlates with the use of more potent immunosuppressive therapies. BKV infection of the Glomerular Vascular Unit (GVU) consisting of podocytes, mesangial cells, and glomerular endothelial cells could lead to glomerular inflammation and contribute to renal fibrosis. The effects of BKV on GVU infectivity have not been reported. METHODS: We infected GVU cells with the Dunlop strain of BKV. Viral infectivity was analyzed by microscopy, immunofluorescence, Western blot analysis, and quantitative RT-PCR (qRT-PCR). The expression of specific proinflammatory cytokines induced by BKV was analyzed by qRT-PCR. RESULTS: BKV infection of podocytes, mesangial cells, and glomerular endothelial cells was confirmed by qRT-PCR and positive staining with antibodies to the BKV VP1 major capsid protein, or the SV40 Large T-Antigen. The increased transcriptional expression of interferon gamma-induced protein 10 (CXCL10/IP-10) and interferon beta (IFNß) was detected in podocytes and mesangial cells at 96 h post-infection. CONCLUSIONS: All cellular components of the GVU are permissive for BKV replication. Cytopathic effects induced by BKV in podocytes and glomerular endothelial cells and the expression of CXCL10 and IFNß genes by podocytes and mesangial cells may together contribute to glomerular inflammation and cytopathology in BKVAN.


Assuntos
Vírus BK/fisiologia , Nefropatias/virologia , Glomérulos Renais/virologia , Infecções por Polyomavirus/virologia , Complicações Pós-Operatórias/virologia , Vírus BK/genética , Citocinas/genética , Citocinas/imunologia , Humanos , Nefropatias/genética , Nefropatias/imunologia , Glomérulos Renais/imunologia , Transplante de Rim/efeitos adversos , Infecções por Polyomavirus/genética , Infecções por Polyomavirus/imunologia , Complicações Pós-Operatórias/etiologia , Complicações Pós-Operatórias/genética , Complicações Pós-Operatórias/imunologia , Ativação Viral , Replicação Viral
2.
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
3.
Virology ; 519: 77-85, 2018 06.
Artigo em Inglês | MEDLINE | ID: mdl-29679791

RESUMO

BACKGROUND: Zika virus (ZIKV) infection has been associated with microcephaly in infants. Currently there is no treatment or vaccine. Here we explore the use of a morpholino oligonucleotide targeted to the 5' untranslated region (5'-UTR) of the ZIKV RNA to prevent ZIKV replication. METHODS: Morpholino DWK-1 inhibition of ZIKV replication in human glomerular podocytes was examined by qRT-PCR, reduction in ZIKV genome copy number, western blot analysis, immunofluorescence and proinflammatory cytokine gene expression. RESULTS: Podocytes pretreated with DWK-1 showed reduced levels of both viral mRNA and ZIKV E protein expression compared to controls. We observed suppression in proinflammatory gene expression for IFN-ß (interferon ß) RANTES (regulated on activation, normal T cell expressed and secreted), MIP-1α (macrophage inflammatory protein-1α), TNF-α (tumor necrosis factor-α) and IL1-α (interleukin 1-α) in ZIKV-infected podocytes pretreated with DWK-1. CONCLUSIONS: Morpholino DWK-1 targeting the ZIKV 5'-UTR effectively inhibits ZIKV replication and suppresses ZIKV-induced proinflammatory gene expression.


Assuntos
Regiões 5' não Traduzidas/efeitos dos fármacos , Morfolinos/farmacologia , Replicação Viral/efeitos dos fármacos , Zika virus/efeitos dos fármacos , Quimiocina CCL5/genética , Citocinas/genética , Imunofluorescência , Humanos , Interferon beta/genética , Podócitos/virologia , Reação em Cadeia da Polimerase em Tempo Real , Zika virus/genética , Zika virus/fisiologia
4.
Am J Physiol Renal Physiol ; 314(5): F832-F843, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29357419

RESUMO

The loss of podocyte (PD) molecular phenotype is an important feature of diabetic podocytopathy. We hypothesized that high glucose (HG) induces dedifferentiation in differentiated podocytes (DPDs) through alterations in the apolipoprotein (APO) L1-microRNA (miR) 193a axis. HG-induced DPD dedifferentiation manifested in the form of downregulation of Wilms' tumor 1 (WT1) and upregulation of paired box 2 (PAX2) expression. WT1-silenced DPDs displayed enhanced expression of PAX2. Immunoprecipitation of DPD cellular lysates with anti-WT1 antibody revealed formation of WT1 repressor complexes containing Polycomb group proteins, enhancer of zeste homolog 2, menin, and DNA methyltransferase (DNMT1), whereas silencing of either WT1 or DNMT1 disrupted this complex with enhanced expression of PAX2. HG-induced DPD dedifferentiation was associated with a higher expression of miR193a, whereas inhibition of miR193a prevented DPD dedifferentiation in HG milieu. HG downregulated DPD expression of APOL1. miR193a-overexpressing DPDs displayed downregulation of APOL1 and enhanced expression of dedifferentiating markers; conversely, silencing of miR193a enhanced the expression of APOL1 and preserved DPD phenotype. Moreover, stably APOL1G0-overexpressing DPDs displayed the enhanced expression of WT1 but attenuated expression of miR193a; nonetheless, silencing of APOL1 reversed these effects. Since silencing of APOL1 enhanced miR193a expression as well as dedifferentiation in DPDs, it appears that downregulation of APOL1 contributed to dedifferentiation of DPDs through enhanced miR193a expression in HG milieu. Vitamin D receptor agonist downregulated miR193a, upregulated APOL1 expression, and prevented dedifferentiation of DPDs in HG milieu. These findings suggest that modulation of the APOL1-miR193a axis carries a potential to preserve DPD molecular phenotype in HG milieu.


Assuntos
Apolipoproteína L1/metabolismo , Desdiferenciação Celular/efeitos dos fármacos , Glucose/toxicidade , MicroRNAs/metabolismo , Podócitos/efeitos dos fármacos , Apolipoproteína L1/genética , Calcitriol/análogos & derivados , Calcitriol/farmacologia , Linhagem Celular Transformada , DNA (Citosina-5-)-Metiltransferase 1/genética , DNA (Citosina-5-)-Metiltransferase 1/metabolismo , Proteína Potenciadora do Homólogo 2 de Zeste/genética , Proteína Potenciadora do Homólogo 2 de Zeste/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , MicroRNAs/genética , Fator de Transcrição PAX2/genética , Fator de Transcrição PAX2/metabolismo , Fenótipo , Podócitos/metabolismo , Podócitos/patologia , Proteínas do Grupo Polycomb/genética , Proteínas do Grupo Polycomb/metabolismo , Receptores de Calcitriol/agonistas , Receptores de Calcitriol/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas WT1/genética , Proteínas WT1/metabolismo
5.
PLoS Pathog ; 11(5): e1004864, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-26020637

RESUMO

Quiescent CD4+ T cells restrict human immunodeficiency virus type 1 (HIV-1) infection at early steps of virus replication. Low levels of both deoxyribonucleotide triphosphates (dNTPs) and the biosynthetic enzymes required for their de novo synthesis provide one barrier to infection. CD4+ T cell activation induces metabolic reprogramming that reverses this block and facilitates HIV-1 replication. Here, we show that phospholipase D1 (PLD1) links T cell activation signals to increased HIV-1 permissivity by triggering a c-Myc-dependent transcriptional program that coordinates glucose uptake and nucleotide biosynthesis. Decreasing PLD1 activity pharmacologically or by RNA interference diminished c-Myc-dependent expression during T cell activation at the RNA and protein levels. PLD1 inhibition of HIV-1 infection was partially rescued by adding exogenous deoxyribonucleosides that bypass the need for de novo dNTP synthesis. Moreover, the data indicate that low dNTP levels that impact HIV-1 restriction involve decreased synthesis, and not only increased catabolism of these nucleotides. These findings uncover a unique mechanism of action for PLD1 inhibitors and support their further development as part of a therapeutic combination for HIV-1 and other viral infections dependent on host nucleotide biosynthesis.


Assuntos
Linfócitos T CD4-Positivos/virologia , Desoxirribonucleotídeos/metabolismo , Infecções por HIV/virologia , HIV-1/fisiologia , Fosfolipase D/metabolismo , Proteínas Proto-Oncogênicas c-myc/metabolismo , Replicação Viral , Apoptose , Western Blotting , Linfócitos T CD4-Positivos/metabolismo , Proliferação de Células , Células Cultivadas , Replicação do DNA , Infecções por HIV/imunologia , Infecções por HIV/metabolismo , Humanos , Ativação Linfocitária , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Reação em Cadeia da Polimerase Via Transcriptase Reversa
6.
J Virol ; 88(1): 592-603, 2014 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-24173214

RESUMO

Apolipoprotein L1 (APOL1) is a major component of the human innate immune response against African trypanosomes. Although the mechanism of the trypanolytic activity of circulating APOL1 has been recently clarified, the intracellular function(s) of APOL1 in human cells remains poorly defined. Like that of many genes linked to host immunity, APOL1 expression is induced by proinflammatory cytokines gamma interferon (IFN-γ) and tumor necrosis factor alpha (TNF-α). Additionally, IFN-γ-polarized macrophages that potently restrict HIV-1 replication express APOL1, which suggests that APOL1 may contribute to HIV-1 suppression. Here, we report that APOL1 inhibits HIV-1 replication by multiple mechanisms. We found that APOL1 protein targeted HIV-1 Gag for degradation by the endolysosomal pathway. Interestingly, we found that APOL1 stimulated both endocytosis and lysosomal biogenesis by promoting nuclear localization of transcription factor EB (TFEB) and expression of TFEB target genes. Moreover, we demonstrated that APOL1 depletes cellular viral accessory protein Vif, which counteracts the host restriction factor APOBEC3G, via a pathway involving degradation of Vif in lysosomes and by secretion of Vif in microvesicles. As a result of Vif depletion by APOL1, APOBEC3G was not degraded and reduced infectivity of progeny virions. In support of this model, we also showed that endogenous expression of APOL1 in differentiated U937 monocytic cells stimulated with IFN-γ resulted in a reduced production of virus particles. This finding supports the hypothesis that induction of APOL1 contributes to HIV-1 suppression in differentiated monocytes. Deciphering the precise mechanism of APOL1-mediated HIV-1 restriction may facilitate the design of unique therapeutics to target HIV-1 replication.


Assuntos
Apolipoproteínas/fisiologia , Infecções por HIV/imunologia , HIV-1/isolamento & purificação , Imunidade Inata , Lipoproteínas HDL/fisiologia , Apolipoproteína L1 , Diferenciação Celular , Linhagem Celular , Endocitose , Citometria de Fluxo , Produtos do Gene gag/imunologia , HIV-1/fisiologia , Humanos , Interferons/imunologia , Macrófagos/imunologia , Replicação Viral
7.
Virology ; 400(1): 68-75, 2010 Apr 25.
Artigo em Inglês | MEDLINE | ID: mdl-20153011

RESUMO

Human cytidine deaminases, including APOBEC3G (A3G) and A3F, are part of a cellular defense system against retroviruses and retroelements including non-LTR retrotransposons LINE-1 (L1) and Alu. Expression of cellular A3 proteins is sufficient for inhibition of L1 and Alu retrotransposition, but the effect of A3 proteins transferred in exosomes on retroelement mobilization is unknown. Here, we demonstrate for the first time that exosomes secreted by CD4(+)H9 T cells and mature monocyte-derived dendritic cells encapsidate A3G and A3F and inhibit L1 and Alu retrotransposition. A3G is the major contributor to the inhibitory activity of exosomes, however, the contribution of A3F in H9 exosomes cannot be excluded. Additionally, we show that exosomes encapsidate mRNAs coding for A3 proteins. A3G mRNA, and less so A3F, was enriched in exosomes secreted by H9 cells. Exosomal A3G mRNA was functional in vitro. Whether exosomes inhibit retrotransposons in vivo requires further investigation.


Assuntos
Elementos Alu , Citidina Desaminase/metabolismo , Citosina Desaminase/metabolismo , Exossomos/enzimologia , Exossomos/genética , Elementos Nucleotídeos Longos e Dispersos , Desaminase APOBEC-3G , Linfócitos T CD4-Positivos/metabolismo , Linhagem Celular , Células Dendríticas/metabolismo , Humanos , Plasmídeos/genética , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Retroviridae/genética
8.
J Neurovirol ; 11(3): 281-91, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-16036808

RESUMO

Cocaine is a suspected cofactor in human immunodeficiency virus (HIV)-associated dementia but cocaine's effects are not clear. Herein the authors describe investigations of the mechanisms by which cocaine increases HIV-1 invasion through brain microvascular endothelial cells (BMVECs). Cocaine binds to a site on BMVECs, which is not a biogenic amine transporter, a binding site for estrogen, or a muscarinic receptor and for which benztropine and tamoxifen have the highest affinity. Cocaine treatment of BMVECs disrupts intercellular junctions and induces cell ruffling, which could account for their increased permeability and decreased electrical resistance. HIV-1 enters BMVECs by macropinocytosis and is transported to lysosomes and inactivated. In cocaine-treated BMVECs, the virus enters and persists in large cytoplasmic "lakes." Cocaine exposure of BMVECs up-regulates transcription of genes important in cytoskeleton organization, signal transduction, cell swelling, vesicular trafficking, and cell adhesion. The toxicity of cocaine for the blood-brain barrier may lead to increased virus neuroinvasion and neurovascular complications of cocaine abuse.


Assuntos
Cocaína/farmacologia , HIV-1/fisiologia , Barreira Hematoencefálica/virologia , Encéfalo/irrigação sanguínea , Células Cultivadas , Cocaína/metabolismo , Relação Dose-Resposta a Droga , Impedância Elétrica , Endotélio Vascular/fisiologia , Endotélio Vascular/virologia , Humanos , Permeabilidade , Pinocitose , Receptores de Estrogênio/metabolismo , Receptores Muscarínicos/metabolismo , Transdução de Sinais , Fatores de Tempo , Replicação Viral/efeitos dos fármacos
9.
Cardiovasc Toxicol ; 4(2): 97-107, 2004.
Artigo em Inglês | MEDLINE | ID: mdl-15371627

RESUMO

We examined heart tissues of AIDS patients with or without HIV cardiomyopathy (HIVCM) by immunohistochemistry, in situ polymerase chain reaction, in situ riboprobe hybridization, and the TUNEL technique for apoptosis. In HIVCM tissues, only inflammatory cells, but not endothelial cells or cardiomyocytes, displayed HIV-1 DNA and RNA. However, macrophages, lymphocytes, and--in a patchy fashion--cardiomyocytes and endothelial cells exhibited virus envelope protein gp120. Macrophages infiltrated the myocardium in a perivascular fashion and expressed tumor necrosis factor family ligands; adjacent cardiomyocytes suffered apoptosis. In vitro HIV-1 strongly invaded neonatal rat ventricular myocytes (NRVMs) and coronary artery endothelial cells (CAECs) and induced microvilli but did not replicate. HIV-1, gp120, or Tat induced Erk 1/2 phosphorylation, activation of caspase-3, and apoptosis of NRVMs and CAECs; all of these were inhibited by a MAPK/ERK-kinase (MEK) inhibitor U0126. The pathogenesis of HIVCM involves HIV-1 replication in inflammatory cells and induction of cardiomyocyte apoptosis by (1) the extrinsic pathway through apoptotic ligands and (2) the intrinsic pathway through direct virus entry and gp120- and Tat-proapoptotic signaling.


Assuntos
Apoptose , Cardiomiopatias/etiologia , Citocinas/fisiologia , Produtos do Gene tat/fisiologia , Proteína gp120 do Envelope de HIV/fisiologia , Infecções por HIV/complicações , HIV-1 , Miócitos Cardíacos/patologia , Animais , Animais Recém-Nascidos , Cardiomiopatias/metabolismo , Cardiomiopatias/patologia , Caspase 3 , Caspases/metabolismo , Células Cultivadas , Vasos Coronários/metabolismo , Vasos Coronários/patologia , Vasos Coronários/ultraestrutura , DNA Viral/metabolismo , Células Endoteliais/metabolismo , Células Endoteliais/patologia , Células Endoteliais/ultraestrutura , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Humanos , Imuno-Histoquímica , Hibridização In Situ , Marcação In Situ das Extremidades Cortadas , Macrófagos/virologia , Miócitos Cardíacos/metabolismo , Miócitos Cardíacos/ultraestrutura , Fosforilação , Reação em Cadeia da Polimerase , RNA Viral/metabolismo , Ratos , Transdução de Sinais , Produtos do Gene tat do Vírus da Imunodeficiência Humana
10.
J Biol Chem ; 279(33): 34083-6, 2004 Aug 13.
Artigo em Inglês | MEDLINE | ID: mdl-15215254

RESUMO

APOBEC3G belongs to the family of cellular cytidine deaminase-editing enzymes with a potent antiretroviral activity, which is counteracted by the Vif protein expressed by lentiviruses. Antiretroviral activity of APOBEC3G requires its packaging into assembling virions, presumably to ensure its close association with nascent retroviral cDNA. Here, we demonstrate that APOBEC3G is encapsidated through a direct interaction with the HIV-1 Gag polyprotein which likely takes place on the membranes of the multivesicular bodies (MVB)/late endosomal compartments. This interaction is mediated by the Gag nucleocapsid protein NC, and the N-terminal part of NC is most critical for this interaction. Binding to the NC domain would ensure that APOBEC3G will be concentrated in the viral core of mature HIV-1, in close proximity to the reverse transcription complex.


Assuntos
Produtos do Gene gag/metabolismo , HIV-1/metabolismo , Proteínas do Nucleocapsídeo/metabolismo , Proteínas/química , Desaminase APOBEC-3G , Antirretrovirais/farmacologia , Western Blotting , Linhagem Celular , Citidina Desaminase , DNA Complementar/metabolismo , Endossomos/metabolismo , Produtos do Gene vif/metabolismo , Glutationa Transferase/metabolismo , Humanos , Lentivirus/genética , Lipídeos/química , Microdomínios da Membrana , Microscopia Confocal , Microscopia de Fluorescência , Nucleocapsídeo/metabolismo , Nucleosídeo Desaminases , Testes de Precipitina , Ligação Proteica , Estrutura Terciária de Proteína , RNA/metabolismo , Proteínas Recombinantes/química , Proteínas Repressoras , Retroviridae/genética , Transcrição Gênica , Transfecção , Montagem de Vírus , Replicação Viral , Produtos do Gene vif do Vírus da Imunodeficiência Humana
11.
Proc Natl Acad Sci U S A ; 99(22): 14386-91, 2002 Oct 29.
Artigo em Inglês | MEDLINE | ID: mdl-12379743

RESUMO

We investigated 18 AIDS hearts (5 with and 13 without cardiomyopathy) by using immunocytochemistry and computerized image analysis regarding the roles of HIV-1 proteins and tumor necrosis factor ligands in HIV cardiomyopathy (HIVCM). HIVCM and cardiomyocyte apoptosis were significantly related to each other and to the expression by inflammatory cells of gp120 and tumor necrosis factor-alpha. In HIVCM heart, active caspase 9, a component of the mitochondrion-controlled apoptotic pathway, and the elements of the death receptor-mediated pathway, tumor necrosis factor-alpha and Fas ligand, were expressed strongly on macrophages and weakly on cardiomyocytes. HIVCM showed significantly greater macrophage infiltration and cardiomyocyte apoptosis rate compared with non-HIVCM. HIV-1 entered cultured neonatal rat ventricular myocytes by macropinocytosis but did not replicate. HIV-1- or gp120-induced apoptosis of rat myocytes through a mitochondrion-controlled pathway, which was inhibited by heparin, AOP-RANTES, or pertussis toxin, suggesting that cardiomyocyte apoptosis is induced by signaling through chemokine receptors. In conclusion, in patients with HIVCM, cardiomyocytes die through both mitochondrion- and death receptor-controlled apoptotic pathways.


Assuntos
Síndrome da Imunodeficiência Adquirida/metabolismo , Apoptose , Cardiomiopatias/metabolismo , Quimiocina CCL5/análogos & derivados , HIV-1/metabolismo , Glicoproteínas de Membrana/metabolismo , Mitocôndrias/metabolismo , Miocárdio/citologia , Fator de Necrose Tumoral alfa/metabolismo , Síndrome da Imunodeficiência Adquirida/complicações , Síndrome da Imunodeficiência Adquirida/patologia , Animais , Cardiomiopatias/complicações , Cardiomiopatias/patologia , Células Cultivadas , Quimiocina CCL5/metabolismo , Colesterol/metabolismo , Proteína Ligante Fas , Gangliosídeo G(M1)/metabolismo , Proteína gp120 do Envelope de HIV/metabolismo , Heparina/metabolismo , Humanos , Macrófagos , Pinocitose , Ratos , Ratos Sprague-Dawley
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA