Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 27
Filtrar
1.
Retrovirology ; 21(1): 14, 2024 Jul 17.
Artículo en Inglés | MEDLINE | ID: mdl-39014486

RESUMEN

BACKGROUND: Human T cell lymphotropic virus type 1 (HTLV-1) infection remains a largely neglected public health problem, particularly in resource-poor areas with high burden of communicable and non-communicable diseases, such as some remote populations in Central Australia where an estimated 37% of adults are infected with HTLV-1. Most of our understanding of HTLV-1 infection comes from studies of the globally spread subtype-A (HTLV-1a), with few molecular studies reported with the Austral-Melanesian subtype-C (HTLV-1c) predominant in the Indo-Pacific and Oceania regions. RESULTS: Using a primer walking strategy and direct sequencing, we constructed HTLV-1c genomic consensus sequences from 22 First Nations participants living with HTLV-1c in Central Australia. Phylogenetic and pairwise analysis of this subtype-C proviral gDNA showed higher levels of genomic divergence in comparison to previously published HTLV-1a genomes. While the overall genomic homology between subtypes was 92.5%, the lowest nucleotide and amino acid sequence identity occurred near the 3' end of the proviral genome coding regulatory genes, especially overlapping hbz (85.37%, 77.46%, respectively) and orf-I product p12 (82.00%, 70.30%, respectively). Strikingly, the HTLV-1c genomic consensus sequences uniformly showed a defective translation start codon for the immune regulatory proteins p12/p8 encoded by the HTLV-1A orf-I. Deletions in the proviral genome were detected in many subjects, particularly in the structural gag, pol and env genes. Similarly, using a droplet digital PCR assay measuring the copies of gag and tax per reference host genome, we quantitatively confirmed that provirus retains the tax gene region at higher levels than gag. CONCLUSIONS: Our genomic analysis of HTLV-1c in Central Australia in conjunction with earlier Melanesian HTLV-1c sequences, elucidate substantial differences with respect to the globally spread HTLV-1a. Future studies should address the impact these genomic differences have on infection and the regionally distinctive frequency of associated pulmonary disease. Understanding the host and virus subtype factors which contribute to the differential morbidity observed, is crucial for the development of much needed therapeutics and vaccine strategies against this highly endemic infection in remote First Nations communities in Central Australia.


Asunto(s)
Infecciones por HTLV-I , Virus Linfotrópico T Tipo 1 Humano , Filogenia , Proteínas de los Retroviridae , Virus Linfotrópico T Tipo 1 Humano/genética , Virus Linfotrópico T Tipo 1 Humano/clasificación , Humanos , Infecciones por HTLV-I/virología , Australia , Proteínas de los Retroviridae/genética , Variación Genética , Adulto , Genoma Viral , Proteínas Reguladoras y Accesorias Virales/genética , Análisis de Secuencia de ADN , Masculino , Femenino , Persona de Mediana Edad , ADN Viral/genética , Proteínas Virales/genética , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico
3.
Viruses ; 14(12)2022 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-36560624

RESUMEN

SARS-CoV-2 is the causative agent of the COVID-19 pandemic. Vaccination, supported by social and public health measures, has proven efficacious for reducing disease severity and virus spread. However, the emergence of highly transmissible viral variants that escape prior immunity highlights the need for additional mitigation approaches. Heparin binds the SARS-CoV-2 spike protein and can inhibit virus entry and replication in susceptible human cell lines and bronchial epithelial cells. Primary infection predominantly occurs via the nasal epithelium, but the nasal cell biology of SARS-CoV-2 is not well studied. We hypothesized that prophylactic intranasal administration of heparin may provide strain-agnostic protection for household contacts or those in high-risk settings against SARS-CoV-2 infection. Therefore, we investigated the ability of heparin to inhibit SARS-CoV-2 infection and replication in differentiated human nasal epithelial cells and showed that prolonged exposure to heparin inhibits virus infection. Furthermore, we establish a method for PCR detection of SARS-CoV-2 viral genomes in heparin-treated samples that can be adapted for the detection of viruses in clinical studies.


Asunto(s)
Células Epiteliales , Heparina , SARS-CoV-2 , Replicación Viral , Humanos , COVID-19 , Células Epiteliales/virología , Heparina/farmacología , Pandemias , SARS-CoV-2/efectos de los fármacos , SARS-CoV-2/fisiología , Glicoproteína de la Espiga del Coronavirus/metabolismo , Replicación Viral/efectos de los fármacos
4.
Int J Mol Sci ; 23(2)2022 Jan 13.
Artículo en Inglés | MEDLINE | ID: mdl-35055020

RESUMEN

The global urgency to uncover medical countermeasures to combat the COVID-19 pandemic caused by the severe acute respiratory syndrome-coronavirus 2 (SARS-CoV-2) has revealed an unmet need for robust tissue culture models that faithfully recapitulate key features of human tissues and disease. Infection of the nose is considered the dominant initial site for SARS-CoV-2 infection and models that replicate this entry portal offer the greatest potential for examining and demonstrating the effectiveness of countermeasures designed to prevent or manage this highly communicable disease. Here, we test an air-liquid-interface (ALI) differentiated human nasal epithelium (HNE) culture system as a model of authentic SARS-CoV-2 infection. Progenitor cells (basal cells) were isolated from nasal turbinate brushings, expanded under conditionally reprogrammed cell (CRC) culture conditions and differentiated at ALI. Differentiated cells were inoculated with different SARS-CoV-2 clinical isolates. Infectious virus release into apical washes was determined by TCID50, while infected cells were visualized by immunofluorescence and confocal microscopy. We demonstrate robust, reproducible SARS-CoV-2 infection of ALI-HNE established from different donors. Viral entry and release occurred from the apical surface, and infection was primarily observed in ciliated cells. In contrast to the ancestral clinical isolate, the Delta variant caused considerable cell damage. Successful establishment of ALI-HNE is donor dependent. ALI-HNE recapitulate key features of human SARS-CoV-2 infection of the nose and can serve as a pre-clinical model without the need for invasive collection of human respiratory tissue samples.


Asunto(s)
COVID-19/virología , Mucosa Nasal/citología , Mucosa Nasal/virología , Técnicas de Cultivo de Tejidos/métodos , Adolescente , Adulto , Enzima Convertidora de Angiotensina 2/metabolismo , Técnicas de Cultivo de Célula , Diferenciación Celular , Células Epiteliales/citología , Células Epiteliales/virología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Modelos Biológicos , SARS-CoV-2 , Internalización del Virus
5.
Lancet Infect Dis ; 21(1): e2-e10, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32986997

RESUMEN

Human T-cell lymphotropic virus type-1 (HTLV-1) has a large global burden and in some key communities, such as Indigenous Australians living in remote areas, greater than 45% of people are infected. Despite HTLV-1 causing serious malignancy and myelopathic paraparesis, and a significant association with a range of inflammatory comorbidities and secondary infections that shorten lifespan, few biomedical interventions are available. HTLV-1 starkly contrasts with other blood-borne sexually transmitted viral infections, such as, HIV, hepatitis B virus, and hepatitis C virus, with no antiviral treatments that reduce virus-infected cells, no rapid diagnostics or biomarker assays suitable for use in remote settings, and no effective vaccine. We review how the replication strategies and molecular properties of HTLV-1 establish a long-term stealthy viral pathogenesis through a fine-tuned balance of persistence, immune cell dysfunction, and proliferation of proviral infected cells that collectively present robust barriers to treatment and prevention. An understanding of the nature of the HTLV-1 provirus and opposing actions of viral-coded negative-sense HBZ and positive-sense regulatory proteins Tax, p12 and its cleaved product p8, and p30, is needed to improve the biomedical tools for preventing transmission and improving the long-term health of people with this lifelong infection.


Asunto(s)
Regulación Viral de la Expresión Génica , Infecciones por HTLV-I/epidemiología , Infecciones por HTLV-I/patología , Interacciones Huésped-Patógeno , Virus Linfotrópico T Tipo 1 Humano/genética , Virus Linfotrópico T Tipo 1 Humano/patogenicidad , Linfocitos T/patología , Australia/epidemiología , Humanos , Recurrencia
6.
Retrovirology ; 17(1): 11, 2020 05 12.
Artículo en Inglés | MEDLINE | ID: mdl-32398094

RESUMEN

Human T cell leukemia virus type-1 (HTLV-1) was the first retrovirus found to cause cancer in humans, but the mechanisms that drive the development of leukemia and other diseases associated with HTLV-1 infection remain to be fully understood. This review describes the functional properties of p13, an 87-amino acid protein coded by HTLV-1 open reading frame II (orf-II). p13 is mainly localized in the inner membrane of the mitochondria, where it induces potassium (K+) influx and reactive oxygen species (ROS) production, which can trigger either proliferation or apoptosis, depending on the ROS setpoint of the cell. Recent evidence indicates that p13 may influence the cell's innate immune response to viral infection and the infected cell phenotype. Association of the HTLV-1 transcriptional activator, Tax, with p13 increases p13's stability, leads to its partial co-localization with Tax in nuclear speckles, and reduces the ability of Tax to interact with the transcription cofactor CBP/p300. Comparison of p13 sequences isolated from HTLV-1-infected individuals revealed a small number of amino acid variations in the domains controlling the subcellular localization of the protein. Disruptive mutations of p13 were found in samples obtained from asymptomatic patients with low proviral load. p13 sequences of HTLV-1 subtype C isolates from indigenous Australian patients showed a high degree of identity among each other, with all samples containing a pattern of 5 amino acids that distinguished them from other subtypes. Further characterization of p13's functional properties and sequence variants may lead to a deeper understanding of the impact of p13 as a contributor to the clinical manifestations of HTLV-1 infection.


Asunto(s)
Variación Genética , Virus Linfotrópico T Tipo 1 Humano/genética , Proteínas de los Retroviridae/genética , Animales , Humanos , Sistemas de Lectura Abierta
7.
Retrovirology ; 16(1): 42, 2019 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-31852501

RESUMEN

The extraordinarily high prevalence of HTLV-1 subtype C (HTLV-1C) in some isolated indigenous communities in Oceania and the severity of the health conditions associated with the virus impress the great need for basic and translational research to prevent and treat HTLV-1 infection. The genome of the virus's most common subtype, HTLV-1A, encodes structural, enzymatic, and regulatory proteins that contribute to viral persistence and pathogenesis. Among these is the p30 protein encoded by the doubly spliced Tax-orf II mRNA, a nuclear/nucleolar protein with both transcriptional and post-transcriptional activity. The p30 protein inhibits the productive replication cycle via nuclear retention of the mRNA that encodes for both the viral transcriptional trans-activator Tax, and the Rex proteins that regulate the transport of incompletely spliced viral mRNA to the cytoplasm. In myeloid cells, p30 inhibits the PU-1 transcription factor that regulates interferon expression and is a critical mediator of innate and adaptive immunity. Furthermore, p30 alters gene expression, cell cycle progression, and DNA damage responses in T-cells, raising the hypothesis that p30 may directly contribute to T cell transformation. By fine-tuning viral expression while also inhibiting host innate responses, p30 is likely essential for viral infection and persistence. This concept is supported by the finding that macaques, a natural host for the closely genetically related simian T-cell leukemia virus 1 (STLV-1), exposed to an HTLV-1 knockout for p30 expression by a single point mutation do not became infected unless reversion and selection of the wild type HTLV-1 genotype occurs. All together, these data suggest that inhibition of p30 may help to curb and eventually eradicate viral infection by exposing infected cells to an effective host immune response.


Asunto(s)
Regulación Viral de la Expresión Génica , Virus Linfotrópico T Tipo 1 Humano/fisiología , Proteínas de los Retroviridae/genética , Latencia del Virus/genética , Animales , Línea Celular , Expresión Génica , Genotipo , Infecciones por HTLV-I/inmunología , Infecciones por HTLV-I/virología , Virus Linfotrópico T Tipo 1 Humano/genética , Virus Linfotrópico T Tipo 1 Humano/inmunología , Humanos , Macaca/virología , ARN Viral/genética , Proteínas de los Retroviridae/inmunología
8.
Retrovirology ; 16(1): 43, 2019 12 18.
Artículo en Inglés | MEDLINE | ID: mdl-31852543

RESUMEN

The human T cell leukemia virus type 1 (HTVL-1), first reported in 1980 by Robert Gallo's group, is the etiologic agent of both cancer and inflammatory diseases. Despite approximately 40 years of investigation, the prognosis for afflicted patients remains poor with no effective treatments. The virus persists in the infected host by evading the host immune response and inducing proliferation of infected CD4+ T-cells. Here, we will review the role that viral orf-I protein products play in altering intracellular signaling, protein expression and cell-cell communication in order to escape immune recognition and promote T-cell proliferation. We will also review studies of orf-I mutations found in infected patients and their potential impact on viral load, transmission and persistence. Finally, we will compare the orf-I gene in HTLV-1 subtypes as well as related STLV-1.


Asunto(s)
Infecciones por HTLV-I/transmisión , Infecciones por HTLV-I/virología , Virus Linfotrópico T Tipo 1 Humano/genética , Proteínas Reguladoras y Accesorias Virales/genética , Linfocitos T CD4-Positivos/virología , Proliferación Celular , Infecciones por HTLV-I/inmunología , Virus Linfotrópico T Tipo 1 Humano/inmunología , Virus Linfotrópico T Tipo 1 Humano/patogenicidad , Humanos , Evasión Inmune , Paraparesia Espástica Tropical/inmunología , Virus Linfotrópico T Tipo 1 de los Simios/genética , Carga Viral , Proteínas Reguladoras y Accesorias Virales/inmunología
9.
AIDS ; 33(2): 199-209, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30562171

RESUMEN

OBJECTIVE: To determine whether latency can be established and reversed in both proliferating and nonproliferating CD4+ T cells in the same model in vitro. METHODS: Activated CD4+ T cells were infected with either a nonreplication competent, luciferase reporter virus or wild-type full-length enhanced green fluorescent protein (EGFP) reporter virus and cultured for 12 days. The cells were then sorted by flow cytometry to obtain two distinct T-cell populations that did not express the T-cell activation markers, CD69, CD25 and human leukocyte antigen (HLA)-DR: CD69CD25HLA-DR small cells (nonblasts) that had not proliferated in vitro following mitogen stimulation and CD69CD25HLA-DR large cells (which we here call transitional blasts) that had proliferated. The cells were then reactivated with latency-reversing agents and either luciferase or EGFP quantified. RESULTS: Inducible luciferase expression, consistent with latent infection, was observed in nonblasts and transitional blasts following stimulation with either phorbol-myristate-acetate/phytohemagglutinin (3.8 ±â€Š1 and 2.9 ±â€Š0.5 fold above dimethyl sulfoxide, respectively) or romidepsin (2.1 ±â€Š0.6 and 1.8 ±â€Š0.2 fold above dimethyl sulfoxide, respectively). Constitutive expression of luciferase was higher in transitional blasts compared with nonblasts. Using wild-type full-length EGFP reporter virus, inducible virus was observed in nonblasts but not in transitional blasts. No significant difference was observed in the response to latency-reversing agents in either nonblasts or transitional blasts. CONCLUSION: HIV latency can be established in vitro in resting T cells that have not proliferated (nonblasts) and blasts that have proliferated (transitional blasts). This model could potentially be used to assess new strategies to eliminate latency.


Asunto(s)
Linfocitos T CD4-Positivos/fisiología , Linfocitos T CD4-Positivos/virología , Proliferación Celular , VIH/fisiología , Latencia del Virus , Antígenos CD/análisis , Antígenos de Diferenciación de Linfocitos T/análisis , Linfocitos T CD4-Positivos/química , Linfocitos T CD4-Positivos/clasificación , Células Cultivadas , Citometría de Flujo , Antígenos HLA-DR/análisis , Humanos , Subunidad alfa del Receptor de Interleucina-2/análisis , Lectinas Tipo C/análisis , Coloración y Etiquetado
10.
Proteomics ; 18(12): e1700253, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29437277

RESUMEN

The recognition of pathogen-derived peptides by T lymphocytes is the cornerstone of adaptive immunity, whereby intracellular antigens are degraded in the cytosol and short peptides assemble with class I human leukocyte antigen (HLA) molecules in the ER. These peptide-HLA complexes egress to the cell surface and are scrutinized by cytotoxic CD8+ T-cells leading to the eradication of the infected cell. Here, naturally presented HLA-B*57:01 bound peptides derived from the envelope protein of the human immunodeficiency virus (HIVenv) are identified. HIVenv peptides are present at a very small percentage of the overall HLA-B*57:01 peptidome (<0.1%) and both native and posttranslationally modified forms of two distinct HIV peptides are identified. Notably, a peptide bearing a natively encoded C-terminal tryptophan residue is also present in a modified form containing a kynurenine residue. Kynurenine is a major product of tryptophan catabolism and is abundant during inflammation and infection. Binding of these peptides at a molecular level and their immunogenicity in preliminary functional studies are examined. Modest immune responses are observed to the modified HIVenv peptide, highlighting a potential role for kynurenine-modified peptides in the immune response to HIV and other viral infections.


Asunto(s)
Linfocitos B/inmunología , Epítopos/inmunología , Productos del Gen env/inmunología , Antígenos VIH/inmunología , VIH-1/inmunología , Antígenos HLA-B/inmunología , Procesamiento Proteico-Postraduccional , Linfocitos B/virología , Células Cultivadas , Epítopos/metabolismo , Productos del Gen env/metabolismo , Antígenos VIH/metabolismo , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Antígenos HLA-B/química , Antígenos HLA-B/metabolismo , Humanos
11.
AIDS ; 31(8): 1137-1141, 2017 05 15.
Artículo en Inglés | MEDLINE | ID: mdl-28301423

RESUMEN

OBJECTIVE: To determine the long-term effects of vorinostat on safety and virological parameters in HIV-infected individuals on suppressive antiretroviral therapy (ART). DESIGN: Prospective longitudinal observational extended follow-up of 20 HIV-infected individuals on ART previously enrolled in a clinical trial of daily vorinostat 400 mg for 14 days. Extended follow-up included visits at 6, 12, 18 and 24 months postenrolment in the initial clinical trial. METHODS: Cell-associated unspliced HIV RNA, total HIV DNA and plasma HIV RNA were quantified by PCR, and CD4 and CD8 T cells quantified by flow cytometry. Changes over time in each parameter were assessed using the Wilcoxon matched pair signed-rank test and generalized estimating equations for trend modelling. RESULTS: We recorded a total of 31 adverse events (26 grade 1 and five grade 2) in all study participants (n = 20). There were no significant changes in the number of CD4 or CD8 T cells or plasma HIV RNA over time. In 12 participants for whom baseline samples were available, there were no significant changes in total HIV DNA, cell-associated unspliced HIV RNA, plasma RNA or CD4 and CD8 T cells at 6, 12, 18 or 24 months. CONCLUSION: Extended follow-up for 24 months did not reveal any long-term toxicity or changes in markers of HIV persistence or transcription in participants on ART who had received 14 days of vorinostat.


Asunto(s)
Antirretrovirales/uso terapéutico , Antineoplásicos/uso terapéutico , Interacciones Farmacológicas , Infecciones por VIH/tratamiento farmacológico , Ácidos Hidroxámicos/uso terapéutico , Neoplasias/tratamiento farmacológico , Carga Viral , Antirretrovirales/efectos adversos , Recuento de Linfocito CD4 , Relación CD4-CD8 , Efectos Colaterales y Reacciones Adversas Relacionados con Medicamentos/epidemiología , Infecciones por VIH/complicaciones , Inhibidores de Histona Desacetilasas , Humanos , Ácidos Hidroxámicos/efectos adversos , Estudios Longitudinales , Estudios Prospectivos , Resultado del Tratamiento , Vorinostat
12.
J Neurovirol ; 22(4): 455-63, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-26727904

RESUMEN

Despite the success of combination antiretroviral therapy (cART), HIV persists in long lived latently infected cells in the blood and tissue, and treatment is required lifelong. Recent clinical studies have trialed latency-reversing agents (LRA) as a method to eliminate latently infected cells; however, the effects of LRA on the central nervous system (CNS), a well-known site of virus persistence on cART, are unknown. In this study, we evaluated the toxicity and potency of a panel of commonly used and well-known LRA (panobinostat, romidepsin, vorinostat, chaetocin, disulfiram, hexamethylene bisacetamide [HMBA], and JQ-1) in primary fetal astrocytes (PFA) as well as monocyte-derived macrophages as a cellular model for brain perivascular macrophages. We show that most LRA are non-toxic in these cells at therapeutic concentrations. Additionally, romidepsin, JQ-1, and panobinostat were the most potent at inducing viral transcription, with greater magnitude observed in PFA. In contrast, vorinostat, chaetocin, disulfiram, and HMBA all demonstrated little or no induction of viral transcription. Together, these data suggest that some LRA could potentially activate transcription in latently infected cells in the CNS. We recommend that future trials of LRA also examine the effects of these agents on the CNS via examination of cerebrospinal fluid.


Asunto(s)
VIH-1/efectos de los fármacos , Inhibidores de Histona Desacetilasas/farmacología , Neuronas/efectos de los fármacos , Activación Viral/efectos de los fármacos , Latencia del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos , Acetamidas/farmacología , Astrocitos/efectos de los fármacos , Astrocitos/metabolismo , Astrocitos/virología , Azepinas/farmacología , Línea Celular , Supervivencia Celular/efectos de los fármacos , Depsipéptidos/farmacología , Disulfiram/farmacología , Feto , VIH-1/genética , VIH-1/metabolismo , Humanos , Ácidos Hidroxámicos/farmacología , Indoles/farmacología , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/virología , Neuronas/metabolismo , Neuronas/virología , Panobinostat , Piperazinas/farmacología , Cultivo Primario de Células , Transcripción Genética/efectos de los fármacos , Triazoles/farmacología , Activación Viral/genética , Latencia del Virus/genética , Replicación Viral/genética , Vorinostat
13.
Arch Virol ; 160(12): 2945-55, 2015 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-26347284

RESUMEN

During attempts to clone retroviral determinants associated with a mouse model of Langerhans cell histiocytosis (LCH), suppression subtractive hybridization (SSH) was used to identify unique viruses in the liver of severe combined immunodeficiency (SCID) mice transplanted with LCH tissues. A partial genomic sequence of a murine coronavirus was identified, and the whole genome (31428 bp) of the coronavirus was subsequently sequenced using PCR cloning techniques. Nucleotide sequence comparisons revealed that the genome sequence of the new virus was 91-93% identical to those of known murine hepatitis viruses (MHVs). The predicted open reading frame from the nucleotide sequence encoded all known proteins of MHVs. Analysis at the protein level showed that the virus was closely related to the highly virulent MHV-JHM strain. The virus strain was named MHV-MI. No type D retroviruses were found. Degenerate PCR targeting of type D retrovirus and 5'-RACE targeting of other types of retroviruses confirmed the absence of any retroviral association with the LCH xenografted SCID mice.


Asunto(s)
Infecciones por Coronavirus/virología , Histiocitosis de Células de Langerhans/virología , Virus de la Hepatitis Murina/genética , Virus de la Hepatitis Murina/aislamiento & purificación , Técnicas de Hibridación Sustractiva/métodos , Adolescente , Animales , Secuencia de Bases , Modelos Animales de Enfermedad , Femenino , Genoma Viral , Humanos , Ratones , Ratones SCID , Datos de Secuencia Molecular , Virus de la Hepatitis Murina/clasificación , Sistemas de Lectura Abierta , Reacción en Cadena de la Polimerasa , Ensayos Antitumor por Modelo de Xenoinjerto
14.
PLoS One ; 10(4): e0122919, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25853889

RESUMEN

MYB transcriptional elongation is regulated by an attenuator sequence within intron 1 that has been proposed to encode a RNA stem loop (SLR) followed by a polyU tract. We report that NFκBp50 can bind the SLR polyU RNA and promote MYB transcriptional elongation together with NFκBp65. We identified a conserved lysine-rich motif within the Rel homology domain (RHD) of NFκBp50, mutation of which abrogated the interaction of NFκBp50 with the SLR polyU and impaired NFκBp50 mediated MYB elongation. We observed that the TAR RNA-binding region of Tat is homologous to the NFκBp50 RHD lysine-rich motif, a finding consistent with HIV Tat acting as an effector of MYB transcriptional elongation in an SLR dependent manner. Furthermore, we identify the DNA binding activity of NFκBp50 as a key component required for the SLR polyU mediated regulation of MYB. Collectively these results suggest that the MYB SLR polyU provides a platform for proteins to regulate MYB and reveals novel nucleic acid binding properties of NFκBp50 required for MYB regulation.


Asunto(s)
Proteínas de Unión al ADN/genética , Genes myb/genética , VIH-1/genética , Subunidad p50 de NF-kappa B/genética , Proteínas de Unión al ADN/metabolismo , VIH-1/patogenicidad , Humanos , Intrones/genética , Secuencias Invertidas Repetidas/genética , Mutación , Subunidad p50 de NF-kappa B/metabolismo , ARN Viral/genética
15.
Virology ; 442(1): 51-8, 2013 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-23602007

RESUMEN

Human immunodeficiency virus type 1 (HIV-1) resistance to CCR5 antagonists, including maraviroc (MVC), results from alterations in the HIV-1 envelope glycoproteins (Env) enabling recognition of antagonist-bound CCR5. Here, we characterized tropism alterations for CD4+ T-cell subsets and macrophages by Envs from two subjects who developed MVC resistance in vivo, which displayed either relatively efficient or inefficient recognition of MVC-bound CCR5. We show that MVC-resistant Env with efficient recognition of drug-bound CCR5 displays a tropism shift for CD4+ T-cell subsets associated with increased infection of central memory T-cells and reduced infection of effector memory and transitional memory T-cells, and no change in macrophage infectivity. In contrast, MVC-resistant Env with inefficient recognition of drug-bound CCR5 displays no change in tropism for CD4+ T-cell subsets, but exhibits a significant reduction in macrophage infectivity. The pattern of HIV-1 tropism alterations for susceptible cells may therefore be variable in subjects with MVC resistance.


Asunto(s)
Antagonistas de los Receptores CCR5 , Linfocitos T CD4-Positivos/virología , Ciclohexanos/farmacología , Farmacorresistencia Viral , Inhibidores de Fusión de VIH/farmacología , VIH-1/efectos de los fármacos , VIH-1/patogenicidad , Macrófagos/virología , Triazoles/farmacología , Línea Celular , Proteína gp120 de Envoltorio del VIH/metabolismo , Humanos , Maraviroc , Subgrupos de Linfocitos T/virología
16.
Antimicrob Agents Chemother ; 56(8): 4310-9, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22664963

RESUMEN

Bovine colostrum (first milk) contains very high concentrations of IgG, and on average 1 kg (500 g/liter) of IgG can be harvested from each immunized cow immediately after calving. We used a modified vaccination strategy together with established production systems from the dairy food industry for the large-scale manufacture of broadly neutralizing HIV-1 IgG. This approach provides a low-cost mucosal HIV preventive agent potentially suitable for a topical microbicide. Four cows were vaccinated pre- and/or postconception with recombinant HIV-1 gp140 envelope (Env) oligomers of clade B or A, B, and C. Colostrum and purified colostrum IgG were assessed for cross-clade binding and neutralization against a panel of 27 Env-pseudotyped reporter viruses. Vaccination elicited high anti-gp140 IgG titers in serum and colostrum with reciprocal endpoint titers of up to 1 × 10(5). While nonimmune colostrum showed some intrinsic neutralizing activity, colostrum from 2 cows receiving a longer-duration vaccination regimen demonstrated broad HIV-1-neutralizing activity. Colostrum-purified polyclonal IgG retained gp140 reactivity and neutralization activity and blocked the binding of the b12 monoclonal antibody to gp140, showing specificity for the CD4 binding site. Colostrum-derived anti-HIV antibodies offer a cost-effective option for preparing the substantial quantities of broadly neutralizing antibodies that would be needed in a low-cost topical combination HIV-1 microbicide.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Calostro/inmunología , Anticuerpos Anti-VIH/inmunología , Infecciones por VIH , VIH-1/inmunología , Productos del Gen env del Virus de la Inmunodeficiencia Humana/inmunología , Vacunas contra el SIDA , Animales , Anticuerpos Monoclonales/inmunología , Bovinos , Productos del Gen env/inmunología , Anticuerpos Anti-VIH/sangre , Infecciones por VIH/inmunología , Infecciones por VIH/prevención & control , Infecciones por VIH/virología , Inmunoglobulina G/inmunología , Pruebas de Neutralización , Vacunación
17.
J Infect Dis ; 204(10): 1532-40, 2011 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-21930607

RESUMEN

BACKGROUND: Despite virally suppressive combination antiretroviral therapy (cART), some HIV-infected patients exhibit suboptimal CD4(+) T-cell recovery. This study aimed to determine the effect of intensification of cART with raltegravir or addition of hyperimmune bovine colostrum (HIBC) on CD4(+) T-cell count in such patients. METHODS: We randomized 75 patients to 4 treatment groups to receive raltegravir, HIBC, placebo, or both raltegravir and HIBC in a factorial, double-blind study. The primary endpoint was time-weighted mean change in CD4(+) T-cell count from baseline to week 24. T-cell activation (CD38(+) and HLA-DR(+)), plasma markers of microbial translocation (lipopolysaccharide, 16S rDNA), monocyte activation (soluble (s) CD14), and HIV-RNA (lowest level of detection 4 copies/mL) were monitored. Analysis was performed using linear regression methods. RESULTS: Compared with placebo, the addition of neither raltegravir nor HIBC to cART for 24 weeks resulted in a significant change in CD4(+) T-cell count (mean difference, 95% confidence interval [CI]: 3.09 cells/µL, -14.27; 20.45, P = .724 and 9.43 cells/µL, -7.81; 26.68, P = .279, respectively, intention to treat). There was no significant interaction between HIBC and raltegravir (P = .275). No correlation was found between CD4(+) T-cell count and plasma lipopolysaccharide, 16S rDNA, sCD14, or HIV-RNA. CONCLUSION: The determinants of poor CD4(+) T-cell recovery following cART require further investigation. CLINICAL TRIALS REGISTRATION: ClinicalTrials.gov identifier: NCT00772590, Australia New Zealand Clinical Trials Registry: ACTRN12609000575235.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Calostro , Infecciones por VIH/tratamiento farmacológico , Infecciones por VIH/inmunología , Inhibidores de Integrasa VIH/farmacología , Pirrolidinonas/farmacología , Adulto , Animales , Recuento de Linfocito CD4 , Linfocitos T CD4-Positivos/metabolismo , Bovinos , Calostro/inmunología , Método Doble Ciego , Quimioterapia Combinada , Femenino , VIH/inmunología , VIH/aislamiento & purificación , Infecciones por VIH/sangre , Inhibidores de Integrasa VIH/uso terapéutico , Humanos , Modelos Lineales , Activación de Linfocitos , Masculino , Persona de Mediana Edad , Embarazo , Pirrolidinonas/uso terapéutico , ARN Viral/sangre , Raltegravir Potásico
18.
RNA Biol ; 8(6): 1035-46, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21955498

RESUMEN

We have previously reported induction of transcriptional gene silencing (TGS) of HIV-1 by short hairpin RNA (shRNA) expressed in MOLT-4 cells. The shRNA (termed shPromA) targets the highly conserved tandem NF-kB binding sequences of the HIV-1 promoter. Recent articles have reported that TGS mediated by promoter-targeted siRNAs was exclusively the result of sequence non-specific off-target effects. Specifically, several mismatched siRNAs to the target promoter sequences were reported to also induce significant TGS, suggesting TGS was a consequence of off-target effects. Here, following extensive investigation, we report that shPromA induces sequence specific transcriptional silencing in HIV-1 infection in MOLT4 cells, while four shRNA variants, mismatched by 2-3 nucleotides, fail to suppress viral replication. We confirm similar levels of shRNA expression from the U6 promoter and the presence of processed/cleaved siRNAs for each construct in transduced MOLT-4 cells. HIV-1 sequence specific shPromA does not suppress HIV-2, which has an alternate NF-kB binding sequence. As a result of the unique sequence targeted, shPromA does not induce down-regulation of other NF-kB driven genes, either at the mRNA or protein level. Furthermore, we confirmed shPromA does not have sequence non-specific off-target effects through unaltered expression of CD4, CXCR4, and CCR5, which are used for viral entry. Additionally, shPromA does not alter PKR, IFN levels, and three downstream mediators of IFN-a response genes. Our data clearly shows that shPromA achieved highly specific TGS of HIV-1, demonstrating that effective TGS can be induced with minimal off-target effects.


Asunto(s)
Silenciador del Gen , VIH-1/genética , Regiones Promotoras Genéticas/genética , ARN Interferente Pequeño/genética , Secuencia de Bases , Sitios de Unión/genética , Antígenos CD4/inmunología , Antígenos CD4/metabolismo , Línea Celular Tumoral , Exonucleasas/genética , Exorribonucleasas , Citometría de Flujo , Regulación de la Expresión Génica , VIH-1/inmunología , VIH-1/fisiología , Interacciones Huésped-Patógeno/genética , Interacciones Huésped-Patógeno/inmunología , Humanos , Leucemia , Datos de Secuencia Molecular , Leucemia-Linfoma Linfoblástico de Células Precursoras/genética , Leucemia-Linfoma Linfoblástico de Células Precursoras/patología , Leucemia-Linfoma Linfoblástico de Células Precursoras/virología , ARN Interferente Pequeño/metabolismo , Receptores CCR5/inmunología , Receptores CCR5/metabolismo , Receptores CXCR4/inmunología , Receptores CXCR4/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Ácido Nucleico , eIF-2 Quinasa/metabolismo
19.
PLoS Pathog ; 5(4): e1000378, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19360124

RESUMEN

Escape mutant (EM) virus that evades CD8+ T cell recognition is frequently observed following infection with HIV-1 or SIV. This EM virus is often less replicatively "fit" compared to wild-type (WT) virus, as demonstrated by reversion to WT upon transmission of HIV to a naïve host and the association of EM virus with lower viral load in vivo in HIV-1 infection. The rate and timing of reversion is, however, highly variable. We quantified reversion to WT of a series of SIV and SHIV viruses containing minor amounts of WT virus in pigtail macaques using a sensitive PCR assay. Infection with mixes of EM and WT virus containing > or =10% WT virus results in immediate and rapid outgrowth of WT virus at SIV Gag CD8 T cell epitopes within 7 days of infection of pigtail macaques with SHIV or SIV. In contrast, infection with biologically passaged SHIV(mn229) viruses with much smaller proportions of WT sequence, or a molecular clone of pure EM SIV(mac239), demonstrated a delayed or slow pattern of reversion. WT virus was not detectable until > or =8 days after inoculation and took > or =8 weeks to become the dominant quasispecies. A delayed pattern of reversion was associated with significantly lower viral loads. The diversity of the infecting inoculum determines the timing of reversion to WT virus, which in turn predicts the outcome of infection. The delay in reversion of fitness-reducing CD8 T cell escape mutations in some scenarios suggests opportunities to reduce the pathogenicity of HIV during very early infection.


Asunto(s)
Linfocitos T CD8-positivos/virología , Productos del Gen gag/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/genética , Animales , Linfocitos T CD8-positivos/inmunología , Productos del Gen gag/inmunología , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Macaca nemestrina , Mutación , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Replicación Viral
20.
Retrovirology ; 5: 18, 2008 Feb 04.
Artículo en Inglés | MEDLINE | ID: mdl-18241354

RESUMEN

BACKGROUND: Macrophages are important targets and long-lived reservoirs of HIV-1, which are not cleared of infection by currently available treatments. In the primary monocyte-derived macrophage model of infection, replication is initially productive followed by a decline in virion output over ensuing weeks, coincident with a decrease in the levels of the essential viral transactivator protein Tat. We investigated two possible mechanisms in macrophages for regulation of viral replication, which appears to be primarily regulated at the level of tat mRNA: 1) differential mRNA stability, used by cells and some viruses for the rapid regulation of gene expression and 2) control of HIV-1 alternative splicing, which is essential for optimal viral replication. RESULTS: Following termination of transcription at increasing times after infection in macrophages, we found that tat mRNA did indeed decay more rapidly than rev or nef mRNA, but with similar kinetics throughout infection. In addition, tat mRNA decayed at least as rapidly in peripheral blood lymphocytes. Expression of cellular splicing factors in uninfected and infected macrophage cultures from the same donor showed an inverse pattern over time between enhancing factors (members of the SR family of RNA binding proteins) and inhibitory factors (members of the hnRNP family). While levels of the SR protein SC35 were greatly up-regulated in the first week or two after infection, hnRNPs of the A/B and H groups were down-regulated. Around the peak of virus production in each culture, SC35 expression declined to levels in uninfected cells or lower, while the hnRNPs increased to control levels or above. We also found evidence for increased cytoplasmic expression of SC35 following long-term infection. CONCLUSION: While no evidence of differential regulation of tat mRNA decay was found in macrophages following HIV-1 infection, changes in the balance of cellular splicing factors which regulate alternative viral pre-mRNA splicing were observed. These changes correlated with changes in Tat expression and virus production and could play an important role in viral persistence in macrophages. This mechanism could provide a novel target for control of infection in this critical cell type, which would be necessary for eventual eradication of the virus from infected individuals.


Asunto(s)
Empalme Alternativo , Regulación Viral de la Expresión Génica , Infecciones por VIH/virología , VIH-1/fisiología , Células Cultivadas , Genes tat/genética , Humanos , Macrófagos/virología , Fosfoproteínas/metabolismo , ARN Mensajero/metabolismo , ARN Viral/metabolismo , Ribonucleoproteínas/metabolismo , Replicación Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA