Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 83
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Kidney Int ; 103(3): 529-543, 2023 03.
Artículo en Inglés | MEDLINE | ID: mdl-36565808

RESUMEN

Chronic kidney disease (CKD) is a common cause of morbidity in human immunodeficiency virus (HIV)-positive individuals. HIV infection leads to a wide spectrum of kidney cell damage, including tubular epithelial cell (TEC) injury. Among the HIV-1 proteins, the pathologic effects of viral protein R (Vpr) are well established and include DNA damage response, cell cycle arrest, and cell death. Several in vitro studies have unraveled the molecular pathways driving the cytopathic effects of Vpr in tubular epithelial cells. However, the in vivo effects of Vpr on tubular injury and CKD pathogenesis have not been thoroughly investigated. Here, we use a novel inducible tubular epithelial cell-specific Vpr transgenic mouse model to show that Vpr expression leads to progressive tubulointerstitial damage, interstitial inflammation and fibrosis, and tubular cyst development. Importantly, Vpr-expressing tubular epithelial cells displayed significant hypertrophy, aberrant cell division, and atrophy; all reminiscent of tubular injuries observed in human HIV-associated nephropathy (HIVAN). Single-cell RNA sequencing analysis revealed the Vpr-mediated transcriptomic responses in specific tubular subsets and highlighted the potential multifaceted role of p53 in the regulation of cell metabolism, proliferation, and death pathways in Vpr-expressing tubular epithelial cells. Thus, our study demonstrates that HIV Vpr expression in tubular cells is sufficient to induce HIVAN-like tubulointerstitial damage and fibrosis, independent of glomerulosclerosis and proteinuria. Additionally, as this new mouse model develops progressive CKD with diffuse fibrosis and kidney failure, it can serve as a useful tool to examine the mechanisms of kidney disease progression and fibrosis in vivo.


Asunto(s)
Nefropatía Asociada a SIDA , Productos del Gen vpr , Infecciones por VIH , VIH-1 , Insuficiencia Renal Crónica , Animales , Humanos , Ratones , Nefropatía Asociada a SIDA/genética , Modelos Animales de Enfermedad , Productos del Gen vpr/genética , Productos del Gen vpr/metabolismo , Productos del Gen vpr/farmacología , Infecciones por VIH/complicaciones , VIH-1/genética , VIH-1/metabolismo , Proteínas del Virus de la Inmunodeficiencia Humana , Ratones Transgénicos , Insuficiencia Renal Crónica/complicaciones
2.
J Virol ; 96(14): e0062422, 2022 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-35867560

RESUMEN

HIV-1 persistence in different cell types presents the main obstacle to an HIV-1 cure. We have previously shown that the renal epithelium is a site of HIV-1 infection and that the kidney represents a separate viral compartment from blood. Whether renal cells can harbor latent virus that can be reactivated upon treatment with latency reversing agents (LRAs) is unknown. To address this question, we developed an in vitro HIV-1 latency model in renal tubule epithelial (RTE) cells using a dual color HIV-1 reporter virus, R7/E-/GFP/EF1a-mCherry (R7GEmC), and evaluated the effect of LRAs, both as single agents and in combination, on viral reactivation. Our data show that HIV-1 can establish latency in RTE cells early postinfection. While the pool of latently infected cells expanded overtime, the percentage of productively infected cells declined. Following LRA treatment only a small fraction of latently infected cells, both T cells and RTE cells, could be reactivated, and the drug combinations more effective in reactivating HIV transcription in RTE cells differed from those more active in T cells. Our study demonstrates that HIV can establish latency in RTE cells and that current LRAs are only marginally effective in inducing HIV-1 reactivation. This suggests that further study of LRA dynamics in non-T cells may be warranted to assess the suitability of LRAs as a sterilizing cure strategy. IMPORTANCE Anti-retroviral therapy (ART) has dramatically reduced HIV-related morbidity and mortality. Despite this success, a number of challenges remain, including the long-term persistence of multiple, clinically latent viral reservoirs capable of reactivation in the absence of ART. As efforts proceed toward HIV eradication or functional cure, further understanding of the dynamics of HIV-1 replication, establishment of latency and mechanisms of reactivation in reservoirs harboring the virus throughout the body is necessary. HIV-1 can infect renal epithelial cells and the expression of viral genes in those cells contributes to the development of HIV associated nephropathy (HIVAN) in untreated individuals. The significance of our work is in developing the first model of HIV-1 latency in renal epithelial cells. This model enhances our understanding of HIV-1 latency and persistence in the kidney and can be used to screen candidate latency reversing agents.


Asunto(s)
Células Epiteliales , Infecciones por VIH , Riñón , Activación Viral , Latencia del Virus , Linfocitos T CD4-Positivos , Células Cultivadas , Células Epiteliales/virología , VIH-1 , Humanos , Riñón/citología , Riñón/virología
3.
J Virol ; 92(2)2018 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-29093088

RESUMEN

Prior studies have found that HIV, through the Vpr protein, promotes genome reduplication (polyploidy) in infection-surviving epithelial cells within renal tissue. However, the temporal progression and molecular regulation through which Vpr promotes polyploidy have remained unclear. Here we define a sequential progression to Vpr-mediated polyploidy in human renal tubule epithelial cells (RTECs). We found that as in many cell types, Vpr first initiates G2 cell cycle arrest in RTECs. We then identified a previously unreported cascade of Vpr-dependent events that lead to renal cell survival and polyploidy. Specifically, we found that a fraction of G2-arrested RTECs reenter the cell cycle. Following this cell cycle reentry, two distinct outcomes occur. Cells that enter complete mitosis undergo mitotic cell death due to extra centrosomes and aberrant division. Conversely, cells that abort mitosis undergo endoreplication to become polyploid. We further show that multiple small-molecule inhibitors of the phosphatidylinositol 3-kinase-related kinase (PIKK) family, including those that target ATR, ATM, and mTOR, indirectly prevent Vpr-mediated polyploidy by preventing G2 arrest. In contrast, an inhibitor that targets DNA-dependent protein kinase (DNA-PK) specifically blocks the Vpr-mediated transition from G2 arrest to polyploidy. These findings outline a temporal, molecularly regulated path to polyploidy in HIV-positive renal cells.IMPORTANCE Current cure-focused efforts in HIV research aim to elucidate the mechanisms of long-term persistence of HIV in compartments. The kidney is recognized as one such compartment, since viral DNA and mRNA persist in the renal tissues of HIV-positive patients. Further, renal disease is a long-term comorbidity in the setting of HIV. Thus, understanding the regulation and impact of HIV infection on renal cell biology will provide important insights into this unique HIV compartment. Our work identifies mechanisms that distinguish between HIV-positive cell survival and death in a known HIV compartment, as well as pharmacological agents that alter these outcomes.


Asunto(s)
Células Epiteliales/metabolismo , Células Epiteliales/virología , VIH-1/fisiología , Interacciones Huésped-Patógeno , Mitosis , Poliploidía , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/metabolismo , Muerte Celular , Línea Celular , Supervivencia Celular , Proteína Quinasa Activada por ADN/antagonistas & inhibidores , Técnica del Anticuerpo Fluorescente , Puntos de Control de la Fase G2 del Ciclo Celular , Humanos , Túbulos Renales/citología , Túbulos Renales/virología , Microscopía Fluorescente , Modelos Biológicos , Inhibidores de Proteínas Quinasas/farmacología , Transducción de Señal
4.
Mol Ther ; 24(11): 2021-2032, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27455880

RESUMEN

The design of an effective HIV-1 vaccine remains a major challenge. Several vaccine strategies based on viral vectors have been evaluated in preclinical and clinical trials, with largely disappointing results. Integrase defective lentiviral vectors (IDLV) represent a promising vaccine candidate given their ability to induce durable and protective immune responses in mice after a single immunization. Here, we evaluated the immunogenicity of a SIV-based IDLV in nonhuman primates. Six rhesus monkeys were primed intramuscularly with IDLV-Env and boosted with the same vector after 1 year. A single immunization with IDLV-Env induced broad humoral and cellular immune responses that waned over time but were still detectable at 1 year postprime. The boost with IDLV-Env performed at 1 year from the prime induced a remarkable increase in both antibodies and T-cell responses. Antibody binding specificity showed a predominant cross-clade gp120-directed response. Monkeys' sera efficiently blocked anti-V2 and anti-CD4 binding site antibodies, neutralized the tier 1 MW965.26 pseudovirus and mediated antibody-dependent cellular cytotoxicity (ADCC). Durable polyfunctional Env-specific T-cell responses were also elicited. Our study demonstrates that an IDLV-Env-based vaccine induces functional, comprehensive, and durable immune responses in Rhesus macaques. These results support further evaluation of IDLV as a new HIV-1 vaccine delivery platform.


Asunto(s)
Vacunas contra el SIDA/administración & dosificación , Integrasas/deficiencia , Virus de la Inmunodeficiencia de los Simios/genética , Productos del Gen env del Virus de la Inmunodeficiencia Humana/inmunología , Vacunas contra el SIDA/inmunología , Animales , Inmunidad Celular , Inmunización Secundaria , Inyecciones Intramusculares , Macaca mulatta , Linfocitos T/metabolismo , Vacunación/métodos , Productos del Gen env del Virus de la Inmunodeficiencia Humana/genética
5.
Nat Rev Immunol ; 6(6): 447-56, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16724099

RESUMEN

Defensins are small antimicrobial peptides that are produced by leukocytes and epithelial cells, and that have an important role in innate immunity. Recent advances in understanding the mechanisms of the antiviral action(s) of defensins indicate that they have a dual role in antiviral defence, acting directly on the virion and on the host cell. This Review focuses on the antiviral activities and mechanisms of action of mammalian defensins, and on the clinical relevance of these activities. Understanding the complex function of defensins in innate immunity against viral infection has implications for the prevention and treatment of viral disease.


Asunto(s)
Defensinas/fisiología , Inmunidad Innata , Virosis/inmunología , Virosis/prevención & control , Animales , Humanos , Virosis/virología
6.
Circulation ; 128(22): 2351-63, 2013 Nov 26.
Artículo en Inglés | MEDLINE | ID: mdl-24043300

RESUMEN

BACKGROUND: Smooth muscle cell (SMC) migration and proliferation critically influence the clinical course of vascular disease. We tested the effect of the novel small leucine-rich repeat protein podocan on SMC migration and proliferation using a podocan-deficient mouse in combination with a model of arterial injury and aortic explant SMC culture. In addition, we examined the effect of overexpression of the human form of podocan on human SMCs and tested for podocan expression in human atherosclerosis. In all these conditions, we concomitantly evaluated the Wnt-TCF (T-cell factor) pathway. METHODS AND RESULTS: Podocan was strongly and selectively expressed in arteries of wild-type mice after injury. Podocan-deficient mice showed increased arterial lesion formation compared with wild-type littermates in response to injury (P<0.05). Also, SMC proliferation was increased in arteries of podocan-deficient mice compared with wild-type (P<0.05). In vitro, migration and proliferation were increased in podocan-deficient SMCs and were normalized by transfection with the wild-type podocan gene (P<0.05). In addition, upregulation of the Wnt-TCF pathway was found in SMCs of podocan-deficient mice both in vitro and in vivo. On the other hand, podocan overexpression in human SMCs significantly reduced SMC migration and proliferation, inhibiting the Wnt-TCF pathway. Podocan and a Wnt-TCF pathway marker were differently expressed in human coronary restenotic versus primary lesions. CONCLUSIONS: Podocan appears to be a potent negative regulator of the migration and proliferation of both murine and human SMCs. The lack of podocan results in excessive arterial repair and prolonged SMC proliferation, which likely is mediated by the Wnt-TCF pathway.


Asunto(s)
Movimiento Celular/fisiología , Glicoproteínas/genética , Músculo Liso Vascular/patología , Neointima/patología , Neointima/fisiopatología , Placa Aterosclerótica/patología , Adulto , Anciano , Animales , Aorta/patología , Aorta/fisiología , Proliferación Celular , Células Cultivadas , Femenino , Arteria Femoral/lesiones , Arteria Femoral/patología , Arteria Femoral/fisiología , Expresión Génica/fisiología , Glicoproteínas/metabolismo , Humanos , Péptidos y Proteínas de Señalización Intercelular , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Persona de Mediana Edad , Músculo Liso Vascular/fisiología , Placa Aterosclerótica/fisiopatología , Transfección , Vía de Señalización Wnt/fisiología
7.
J Infect Dis ; 208(11): 1756-67, 2013 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-23908485

RESUMEN

The female genital epithelium plays a protective role against invading pathogens; however, sexual transmission of human immunodeficiency virus type 1 (HIV-1) still occurs in healthy women. To model virus-cell interactions in this barrier during sexual transmission, we studied the uptake and infection of ectocervical and endocervical cell lines with cell-free fluorescent protein-expressing recombinant HIV-1 carrying primary transmitted/founder envelope genes. We observed that a subset of both the ectocervical and endocervical epithelial cells become productively infected with cell-free HIV-1 in a CD4-independent manner. In addition, the ability of the semen-derived enhancer of virus infection (SEVI) to enhance virus-epithelial cell interactions was studied. This infection is increased approximately 2-5 fold when inoculation occurs in the presence of SEVI fibrils. Once infected, the epithelial cells are capable of transmitting the virus to target CD4 T cells in coculture in a contact-dependent manner that uses conventional CD4- and coreceptor-dependent entry. The infection of target CD4 T cells only occurs when de novo HIV-1 is produced within the epithelial cells. These findings suggest that a subset of cervical epithelial cells may be actively involved in establishing a systemic HIV infection and should be a target when designing prevention strategies to protect against HIV-1 sexual transmission.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Cuello del Útero/virología , Células Epiteliales/virología , Infecciones por VIH/transmisión , VIH-1/fisiología , Linfocitos T CD4-Positivos/inmunología , Línea Celular , Cuello del Útero/citología , Cuello del Útero/inmunología , Células Epiteliales/citología , Células Epiteliales/inmunología , Femenino , Regulación Viral de la Expresión Génica , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/inmunología , Interacciones Huésped-Patógeno , Humanos , ARN Viral/genética , ARN Viral/aislamiento & purificación , Internalización del Virus
8.
Res Sq ; 2024 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-38659814

RESUMEN

Diverse and rapidly mutating viruses pose challenges to immunogen and vaccine design. In this study, we evaluated the ability of memory B-cells obtained from two independent NHP trials to cross-react with individual HIV-1 vaccine components of two different multivalent immunization strategies. We demonstrated that while an HIV-1 Env multiclade, multivalent immunization regimen resulted in a dominant memory B-cell response that converged toward shared epitopes, in a sequential immunization with clonally-related non-stabilized gp140 HIV-1 Envs followed by SOSIP-stabilized gp140 trimers, the change in immunogen format resulted in repriming of the B-cell response.

9.
Proc Natl Acad Sci U S A ; 107(22): 10178-83, 2010 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-20479248

RESUMEN

Induction of type I interferons (IFN) is a central feature of innate immune responses to microbial pathogens and is mediated via Toll-like receptor (TLR)-dependent and -independent pathways. Prothymosin-alpha (ProTalpha), a small acidic protein produced and released by CD8(+) T cells, inhibits HIV-1, although the mechanism for its antiviral activity was not known. We demonstrate that exogenous ProTalpha acts as a ligand for TLR4 and stimulates type I IFN production to potently suppress HIV-1 after entry into cells. These activities are induced by native and recombinant ProTalpha, retained by an acidic peptide derived from ProTalpha, and lost in the absence of TLR4. Furthermore, we demonstrate that ProTalpha accounts for some of the soluble postintegration HIV-1 inhibitory activity long ascribed to CD8(+) cells. Thus, a protein produced by CD8(+) T cells of the adaptive immune system can exert potent viral suppressive activity through an innate immune response. Understanding the mechanism of IFN induction by ProTalpha may provide therapeutic leads for IFN-sensitive viruses.


Asunto(s)
VIH-1/efectos de los fármacos , Interferón Tipo I/biosíntesis , Precursores de Proteínas/farmacología , Timosina/análogos & derivados , Receptor Toll-Like 4/efectos de los fármacos , Receptor Toll-Like 4/metabolismo , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Secuencia de Aminoácidos , Animales , Fármacos Anti-VIH/inmunología , Fármacos Anti-VIH/farmacología , Linfocitos T CD8-positivos/inmunología , VIH-1/genética , VIH-1/inmunología , VIH-1/fisiología , Humanos , Inmunidad Innata/efectos de los fármacos , Técnicas In Vitro , Interferón Tipo I/genética , Ligandos , Macrófagos/efectos de los fármacos , Macrófagos/inmunología , Macrófagos/virología , Ratones , Ratones Noqueados , Datos de Secuencia Molecular , Factor 88 de Diferenciación Mieloide/inmunología , Precursores de Proteínas/genética , Precursores de Proteínas/inmunología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas Recombinantes/genética , Proteínas Recombinantes/inmunología , Proteínas Recombinantes/farmacología , Homología de Secuencia de Aminoácido , Timosina/genética , Timosina/inmunología , Timosina/farmacología , Factor de Necrosis Tumoral alfa/biosíntesis , Factor de Necrosis Tumoral alfa/genética , Replicación Viral/efectos de los fármacos
10.
Nat Genet ; 34(1): 70-4, 2003 May.
Artículo en Inglés | MEDLINE | ID: mdl-12692554

RESUMEN

WHIM syndrome is an immunodeficiency disease characterized by neutropenia, hypogammaglobulinemia and extensive human papillomavirus (HPV) infection. Despite the peripheral neutropenia, bone marrow aspirates from affected individuals contain abundant mature myeloid cells, a condition termed myelokathexis. The susceptibility to HPV is disproportionate compared with other immunodeficiency conditions, suggesting that the product of the affected gene may be important in the natural control of this infection. We describe here the localization of the gene associated with WHIM syndrome to a region of roughly 12 cM on chromosome 2q21 and the identification of truncating mutations in the cytoplasmic tail domain of the gene encoding chemokine receptor 4 (CXCR4). Haplotype and mutation analyses in a pedigree transmitting myelokathexis as an apparently autosomal recessive trait support genetic heterogeneity for this aspect of the WHIM syndrome phenotype. Lymphoblastoid cell lines carrying a 19-residue truncation mutation show significantly greater calcium flux relative to control cell lines in response to the CXCR4 ligand, SDF-1, consistent with dysregulated signaling by the mutant receptor. The identification of mutations in CXCR4 in individuals with WHIM syndrome represents the first example of aberrant chemokine receptor function causing human disease and suggests that the receptor may be important in cell-mediated immunity to HPV infection.


Asunto(s)
Síndromes de Inmunodeficiencia/genética , Mutación , Receptores CXCR4/genética , Agammaglobulinemia/genética , Linfocitos B/inmunología , Linfocitos B/metabolismo , Secuencia de Bases , Señalización del Calcio , Cromosomas Humanos Par 2/genética , ADN/genética , Análisis Mutacional de ADN , Femenino , Ligamiento Genético , Humanos , Masculino , Modelos Moleculares , Datos de Secuencia Molecular , Neutropenia/genética , Linaje , Receptores CXCR4/química , Verrugas/genética
11.
Retrovirology ; 9: 69, 2012 Aug 22.
Artículo en Inglés | MEDLINE | ID: mdl-22913641

RESUMEN

BACKGROUND: Integrase defective lentiviral vectors (IDLV) represent a promising delivery system for immunization purposes. Human dendritic cells (DC) are the main cell types mediating the immune response and are readily transduced by IDLV, allowing effective triggering of in vitro expansion of antigen-specific primed CD8+ T cells. However, IDLV expression in transduced DC is at lower levels than those of the integrase (IN) competent counterpart, thus requiring further improvement of IDLV for future use in the clinic. RESULTS: In this paper we show that the addition of simian immunodeficiency (SIV)-Vpx protein in the vector preparation greatly improves transduction of human and simian DC, but not of murine DC, thus increasing the ability of transduced DC to act as functional antigen presenting cells, in the absence of integrated vector sequences. Importantly, the presence of SIV-Vpx allows for using lower dose of input IDLV during in vitro transduction, thus further improving the IDLV safety profile. CONCLUSIONS: These results have significant implications for the development of IDLV-based vaccines.


Asunto(s)
Células Dendríticas/inmunología , Vectores Genéticos , Lentivirus/genética , Proteínas Reguladoras y Accesorias Virales/genética , Vacunas Virales/genética , Vacunas Virales/inmunología , Animales , Células Cultivadas , Expresión Génica , Humanos , Macaca fascicularis , Macaca mulatta , Ratones , Ratones Endogámicos BALB C , Transducción Genética
12.
J Immunol ; 184(6): 2814-24, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20147631

RESUMEN

Sexually transmitted infections increase the likelihood of HIV-1 transmission. We investigated the effect of Neisseria gonorrheae (gonococcus [GC]) exposure on HIV replication in primary resting CD4(+) T cells, a major HIV target cell during the early stage of sexual transmission of HIV. GC and TLR2 agonists, such as peptidylglycan (PGN), Pam(3)CSK(4), and Pam(3)C-Lip, a GC-derived synthetic lipopeptide, but not TLR4 agonists including LPS or GC lipooligosaccharide enhanced HIV-1 infection of primary resting CD4(+) T cells after viral entry. Pretreatment of CD4(+) cells with PGN also promoted HIV infection. Anti-TLR2 Abs abolished the HIV enhancing effect of GC and Pam(3)C-Lip, indicating that GC-mediated enhancement of HIV infection of resting CD4(+) T cells was through TLR2. IL-2 was required for TLR2-mediated HIV enhancement. PGN and GC induced cell surface expression of T cell activation markers and HIV coreceptors, CCR5 and CXCR4. The maximal postentry HIV enhancing effect was achieved when PGN was added immediately after viral exposure. Kinetic studies and analysis of HIV DNA products indicated that GC exposure and TLR2 activation enhanced HIV infection at the step of nuclear import. We conclude that GC enhanced HIV infection of primary resting CD4(+) T cells through TLR2 activation, which both increased the susceptibility of primary CD4(+) T cells to HIV infection as well as enhanced HIV-infected CD4(+) T cells at the early stage of HIV life cycle after entry. This study provides a molecular mechanism by which nonulcerative sexually transmitted infections mediate enhancement of HIV infection and has implication for HIV prevention and therapeutics.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/microbiología , VIH-1/inmunología , Neisseria gonorrhoeae/inmunología , Fase de Descanso del Ciclo Celular/inmunología , Receptor Toll-Like 2/metabolismo , Transporte Activo de Núcleo Celular/inmunología , Secuencia de Aminoácidos , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD4-Positivos/virología , Línea Celular , Células Cultivadas , Infecciones por VIH/inmunología , Infecciones por VIH/microbiología , Infecciones por VIH/transmisión , Humanos , Datos de Secuencia Molecular , Receptor Toll-Like 2/agonistas , Receptor Toll-Like 2/fisiología
13.
J Am Soc Nephrol ; 22(3): 496-507, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21335514

RESUMEN

In animal models of HIV-associated nephropathy, the expression of HIV regulatory genes in epithelial cells is sufficient to cause disease, but how the CD4-negative epithelial cells come to express HIV genes is unknown. Here, we co-cultured T cells infected with fluorescently tagged HIV with renal tubular epithelial cells and observed efficient virus transfer between these cells. The quantity of HIV transferred was much greater than that achieved by exposure to large amounts of cell-free virus and occurred without a requirement for CD4 or Env. The transfer required stable cell-cell adhesion, which could be blocked by sulfated polysaccharides or poly-anionic compounds. We found that the internalization of virus could lead to de novo synthesis of viral protein from incoming viral RNAs even in the presence of a reverse transcriptase inhibitor. These results illustrate an interaction between infected T cells and nonimmune cells, supporting the presence of virological synapses between HIV-harboring T cells and renal tubular epithelial cells, allowing viral uptake and gene expression in epithelial cells.


Asunto(s)
Comunicación Celular/fisiología , Células Epiteliales/virología , Regulación Viral de la Expresión Génica/fisiología , VIH-1/fisiología , Túbulos Renales Proximales/virología , Nefropatía Asociada a SIDA/etiología , Nefropatía Asociada a SIDA/patología , Nefropatía Asociada a SIDA/fisiopatología , Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/virología , Adhesión Celular/efectos de los fármacos , Adhesión Celular/fisiología , Comunicación Celular/efectos de los fármacos , Línea Celular , Técnicas de Cocultivo , Células Epiteliales/citología , VIH-1/genética , Humanos , Túbulos Renales Proximales/citología , Polisacáridos/farmacología , Replicación Viral/fisiología
14.
J Am Soc Nephrol ; 22(11): 1991-6, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-21997397

RESUMEN

A chromosome 22q13 locus strongly associates with increased risk for idiopathic focal segmental glomerulosclerosis (FSGS), HIV-1-associated nephropathy (HIVAN), and hypertensive ESRD among individuals of African descent. Although initial studies implicated MYH9, more recent analyses localized the strongest association within the neighboring APOL1 gene. In this replication study, we examined the six top-most associated variants in APOL1 and MYH9 in an independent cohort of African Americans with various nephropathies (44 with FSGS, 21 with HIVAN, 32 with IgA nephropathy, and 74 healthy controls). All six variants associated with FSGS and HIVAN (additive ORs, 1.8 to 3.0; P values 3 × 10(-2) to 5 × 10(-5)) but not with IgA nephropathy. In conditional and haplotype analyses, two APOL1 haplotypes accounted for virtually all of the association with FSGS and HIVAN on chromosome 22q13 (haplotype P value = 5.6 × 10(-8)). To assess the role of MYH9 deficiency in nephropathy, we crossbred Myh9-haploinsufficient mice (Myh9(+/-)) with HIV-1 transgenic mice. Myh9(+/-) mice were healthy and did not demonstrate overt proteinuria or nephropathy, irrespective of the presence of the HIV-1 transgene. These data further support the strong association of genetic variants in APOL1 with susceptibility to FSGS and HIVAN among African Americans.


Asunto(s)
Nefropatía Asociada a SIDA/genética , Apolipoproteínas/genética , Negro o Afroamericano/genética , Glomerulonefritis por IGA/genética , Glomeruloesclerosis Focal y Segmentaria/genética , Lipoproteínas HDL/genética , Nefropatía Asociada a SIDA/etnología , Negro o Afroamericano/estadística & datos numéricos , Animales , Apolipoproteína L1 , Modelos Animales de Enfermedad , Variación Genética , Glomerulonefritis por IGA/etnología , Glomeruloesclerosis Focal y Segmentaria/etnología , Haplotipos , Humanos , Ratones , Ratones Transgénicos , Proteínas Motoras Moleculares/genética , Cadenas Pesadas de Miosina/genética , Miosina Tipo IIA no Muscular/genética , Polimorfismo de Nucleótido Simple , Factores de Riesgo
15.
NPJ Vaccines ; 7(1): 44, 2022 Apr 21.
Artículo en Inglés | MEDLINE | ID: mdl-35449174

RESUMEN

Integrase Defective Lentiviral Vectors (IDLVs) represent an attractive vaccine platform for delivering HIV-1 antigens, given their ability to induce specific and persistent immune responses in both mice and non-human primates (NHPs). Recent advances in HIV-1 immunogen design demonstrated that native-like HIV-1 Envelope (Env) trimers that mimic the structure of virion-associated Env induce neutralization breadth in rabbits and macaques. Here, we describe the development of an IDLV-based HIV-1 vaccine expressing either soluble ConSOSL.UFO.664 or membrane-tethered ConSOSL.UFO.750 native-like Env immunogens with enhanced bNAb epitopes exposure. We show that IDLV can be pseudotyped with properly folded membrane-tethered native-like UFO.750 trimers. After a single IDLV injection in BALB/c mice, IDLV-UFO.750 induced a faster humoral kinetic as well as higher levels of anti-Env IgG compared to IDLV-UFO.664. IDLV-UFO.750 vaccinated cynomolgus macaques developed unusually long-lasting anti-Env IgG antibodies, as underlined by their remarkable half-life both after priming and boost with IDLV. After boosting with recombinant ConM SOSIP.v7 protein, two animals developed neutralization activity against the autologous tier 1B ConS virus mediated by V1/V2 and V3 glycan sites responses. By combining the possibility to display stabilized trimeric Env on the vector particles with the ability to induce sustained humoral responses, IDLVs represent an appropriate strategy for delivering rationally designed antigens to progress towards an effective HIV-1 vaccine.

16.
Hepatology ; 52(2): 612-22, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20683959

RESUMEN

UNLABELLED: Patients coinfected with human immunodeficiency virus (HIV) and hepatitis C virus (HCV) develop more rapid fibrosis than those infected with HCV only. In HIV/HCV-coinfected patients, fibrosis progression correlates with HIV RNA levels, suggesting a direct role of HIV in liver fibrogenesis. Chemokine (C-C motif) receptor 5 (CCR5) and cysteine-X-cysteine receptor 4 (CXCR4), the two major coreceptors required for HIV entry into cells, are expressed on activated hepatic stellate cells (HSCs), the principle fibrogenic cell type in the liver. We therefore examined whether HIV can infect HSCs, explored the potential mechanisms of viral entry, and assessed the impact of infection as reflected by the ability of HSCs to transfer virus to T lymphocytes and elicit a proinflammatory and profibrogenic response. We report that the laboratory-adapted viruses HIV-IIIB (CXCR4-tropic or X4) and HIV-BaL (CCR5-tropic or R5) and primary HIV isolates can infect both a human stellate cell line, LX-2, and primary human HSCs. HIV entry and gene expression in HSCs was confirmed using HIV-green fluorescent protein (GFP) expression viral constructs in the presence or absence of the reverse-transcriptase inhibitor azidothymidine. CD4 expression on a subset of primary HSCs was demonstrated using fluorescence-activated cell sorting and immunofluorescence staining. Blocking experiments in the presence of anti-CD4, anti-CXCR4, and anti-CCR5 revealed that HIV entry into HSCs is predominantly CD4/chemokine coreceptor-independent. HIV infection promoted HSC collagen I expression and secretion of the proinflammatory cytokine monocyte chemoattractant protein-1. Furthermore, infected LX-2 cells were capable of transferring GFP-expressing virus to T lymphocytes in a coculture system. CONCLUSION: Taken together, our results suggest a potential role of HIV in liver fibrosis/inflammation mediated through effects on HSCs. The role of early highly active antiretroviral therapy initiation in patients with HIV/HCV coinfection warrants further investigation.


Asunto(s)
Quimiocina CCL2/biosíntesis , Colágeno Tipo I/biosíntesis , Colágeno Tipo I/metabolismo , Infecciones por VIH/complicaciones , VIH-1/patogenicidad , Células Estrelladas Hepáticas/virología , Hepatitis C/complicaciones , Cirrosis Hepática/virología , Células Cultivadas , Humanos
17.
Nat Med ; 8(5): 522-6, 2002 May.
Artículo en Inglés | MEDLINE | ID: mdl-11984599

RESUMEN

HIV-associated nephropathy is a clinicopathologic entity that includes proteinuria, focal segmental glomerulosclerosis often of the collapsing variant, and microcystic tubulointerstitial disease. Increasing evidence supports a role for HIV-1 infection of renal epithelium in the pathogenesis of HIV-associated nephropathy. Using in situ hybridization, we previously demonstrated HIV-1 gag and nef mRNA in renal epithelial cells of patients with HIV-associated nephropathy. Here, to investigate whether renal epithelial cells were productively infected by HIV-1, we examined renal tissue for the presence of HIV-1 DNA and mRNA by in situ hybridization and PCR, and we molecularly characterized the HIV-1 quasispecies in the renal compartment. Infected renal epithelial cells were removed by laser-capture microdissection from biopsies of two patients, DNA was extracted, and HIV-1 V3-loop or gp120-envelope sequences were amplified from individually dissected cells by nested PCR. Phylogenetic analysis of kidney-derived sequences as well as corresponding sequences from peripheral blood mononuclear cells of the same patients revealed evidence of tissue-specific viral evolution. In phylogenetic trees constructed from V3 and gp120 sequences, kidney-derived sequences formed tissue-specific subclusters within the radiation of blood mononuclear cell-derived viral sequences from both patients. These data, along with the detection of HIV-1-specific proviral DNA and mRNA in tubular epithelium cells, argue strongly for localized replication of HIV-1 in the kidney and the existence of a renal viral reservoir.


Asunto(s)
Infecciones por VIH/complicaciones , VIH-1/fisiología , Enfermedades Renales/virología , Riñón/virología , Urotelio/virología , Replicación Viral , Secuencia de Bases , Biopsia , Cartilla de ADN , Productos del Gen env/genética , Infecciones por VIH/patología , VIH-1/clasificación , VIH-1/genética , Humanos , Riñón/patología , Enfermedades Renales/patología , Datos de Secuencia Molecular , Reacción en Cadena de la Polimerasa , Urotelio/patología
18.
Mol Ther Methods Clin Dev ; 23: 263-275, 2021 Dec 10.
Artículo en Inglés | MEDLINE | ID: mdl-34729374

RESUMEN

Integrase-defective lentiviral vectors (IDLVs) represent an attractive platform for vaccine development as a result of the ability to induce persistent humoral- and cellular-mediated immune responses against the encoded transgene. Compared with the parental integrating vector, the main advantages for using IDLV are the reduced hazard of insertional mutagenesis and the decreased risk for vector mobilization by wild-type viruses. Here we report on the development and use in the mouse immunogenicity model of simian immunodeficiency virus (SIV)-based IDLV containing a long deletion in the U3 region and with the 3' polypurine tract (PPT) removed from the transfer vector for improving safety and/or efficacy. Results show that a safer extended deletion of U3 sequences did not modify integrase-mediated or -independent integration efficiency. Interestingly, 3' PPT deletion impaired integrase-mediated integration but did not reduce illegitimate, integrase-independent integration efficiency, contrary to what was previously reported in the HIV system. Importantly, although the extended deletion in the U3 did not affect expression or immunogenicity from IDLV, deletion of 3' PPT considerably reduced both expression and immunogenicity of IDLV.

19.
J Virol ; 83(22): 11983-8, 2009 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-19726511

RESUMEN

Human immunodeficiency virus (HIV)-associated nephropathy is a significant cause of morbidity and mortality in HIV-infected persons. Vpr-induced cell cycle dysregulation and apoptosis of renal tubular epithelial cells are important components of the pathogenesis of HIV-associated nephropathy (HIVAN). FAT10 is a ubiquitin-like protein that is upregulated in renal tubular epithelial cells in HIVAN. In these studies, we report that Vpr induces increased expression of FAT10 in tubular cells and that inhibition of FAT10 expression prevents Vpr-induced apoptosis in human and murine tubular cells. Moreover, we found that Vpr interacts with FAT10 and that these proteins colocalize at mitochondria. These studies establish FAT10 as a novel mediator of Vpr-induced cell death.


Asunto(s)
Nefropatía Asociada a SIDA/metabolismo , Apoptosis/fisiología , Ubiquitinas/fisiología , Productos del Gen vpr del Virus de la Inmunodeficiencia Humana/fisiología , Nefropatía Asociada a SIDA/fisiopatología , Animales , Ciclo Celular , Línea Celular , VIH-1/fisiología , Humanos , Ratones
20.
J Biomed Biotechnol ; 2010: 534501, 2010.
Artículo en Inglés | MEDLINE | ID: mdl-20508727

RESUMEN

CD8+ T cells are an essential component of an effective host immune response to tumors and viral infections. Genetic immunization is particularly suitable for inducing CTL responses, because the encoded proteins enter the MHC class I processing pathway through either transgene expression or cross-presentation. In order to compare the efficiency and persistence of immune response induced by genetic vaccines, BALB/c mice were immunized either twice intramuscularly with DNA plasmid expressing a codon-optimized HIV-1 gp120 Envelope sequence together with murine GM-CSF sequence or with a single immunization using an integrase defective lentiviral vector (IDLV) expressing the same proteins. Results strongly indicated that the schedule based on IDLV vaccine was more efficient in inducing specific immune response, as evaluated three months after the last immunization by IFNgamma ELISPOT in both splenocytes and bone marrow- (BM-) derived cells, chromium release assay in splenocytes, and antibody detection in sera. In addition, IDLV immunization induced high frequency of polyfunctional CD8+ T cells able to simultaneously produce IFNgamma, TNFalpha, and IL2.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA