Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Int J Mol Sci ; 24(18)2023 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-37762169

RESUMEN

Regeneration of functional naïve T lymphocytes following the onset of human immunodeficiency virus (HIV) infection remains a crucial issue for people living with HIV (PLWH), even when adhering to antiretroviral therapy (ART). Thus far, reports on the impact of HIV-1 infection on the entry of thymic precursors and the egress of functional naïve T lymphocytes to and from the thymus are limited. We examined the impact of HIV-1 on Sphingosine-1-phosphate (S1P) signaling, which governs the egress of functional naïve thymocytes from the thymus to the periphery. Using in vitro experiments with primary human thymocytes and in vivo and ex vivo studies with humanized mice, we show that HIV-1 infection results in upregulation of the expression of S1P receptor 1 (S1PR1) in the human thymus. Intriguingly, this upregulation occurs during intrathymic infection (direct infection of the human thymic implant) as well as systemic infection in humanized mice. Moreover, considering the dysregulation of pro- and anti-inflammatory cytokines in infected thymi, the increased expression of S1PR1 in response to in vitro exposure to Interferon-Beta (IFN-ß) and Tumor Necrosis Factor-Alpha (TNF-α) indicates that cytokine dysregulation following HIV infection may contribute to upregulation of S1PR1. Finally, an increased presence of CD3hiCD69- (fully mature) as well as CD3hiCD69+ (less mature) T cells in the spleen during HIV infection in humanized mice, combined with earlier expression of S1PR1 during thymocyte development, suggests that upregulation of S1PR1 may translate to increased or accelerated egress from the thymus. The egress of thymocytes that are not functionally mature from the thymus to peripheral blood and lymphoid organs may have implications for the immune function of PLWH.


Asunto(s)
Infecciones por VIH , Receptores de Esfingosina-1-Fosfato , Timo , Animales , Humanos , Ratones , Infecciones por VIH/metabolismo , VIH-1 , Lisofosfolípidos/metabolismo , Receptores de Esfingosina-1-Fosfato/metabolismo , Timocitos/metabolismo
2.
J Neurovirol ; 24(3): 350-361, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29582356

RESUMEN

Events leading to and propagating neurocognitive impairment (NCI) in HIV-1-infected (HIV+) persons are largely mediated by peripheral blood monocytes. We previously identified expression levels of individual genes and gene networks in peripheral blood monocytes that correlated with neurocognitive functioning in HIV+ adults. Here, we expand upon those findings by examining if gene expression data at baseline is predictive of change in neurocognitive functioning 2 years later. We also attempt to validate the original findings in a new sample of HIV+ patients and determine if the findings are HIV specific by including HIV-uninfected (HIV-) participants as a comparison group. At two time points, messenger RNA (mRNA) was isolated from the monocytes of 123 HIV+ and 60 HIV- adults enrolled in the Multicenter AIDS Cohort Study and analyzed with the Illumina HT-12 v4 Expression BeadChip. All participants received baseline and follow-up neurocognitive testing 2 years after mRNA analysis. Data were analyzed using standard gene expression analysis and weighted gene co-expression network analysis with correction for multiple testing. Gene sets were analyzed for GO term enrichment. Only weak reproducibility of associations of single genes with neurocognitive functioning was observed, indicating that such measures are unreliable as biomarkers for HIV-related NCI; however, gene networks were generally preserved between time points and largely reproducible, suggesting that these may be more reliable. Several gene networks associated with variables related to HIV infection were found (e.g., MHC I antigen processing, TNF signaling, interferon gamma signaling, and antiviral defense); however, no significant associations were found for neurocognitive function. Furthermore, neither individual gene probes nor gene networks predicted later neurocognitive change. This study did not validate our previous findings and does not support the use of monocyte gene expression profiles as a biomarker for current or future HIV-associated neurocognitive impairment.


Asunto(s)
Disfunción Cognitiva/genética , Redes Reguladoras de Genes , Infecciones por VIH/genética , Monocitos/metabolismo , Transcriptoma , Adulto , Biomarcadores/sangre , Estudios de Casos y Controles , Disfunción Cognitiva/complicaciones , Disfunción Cognitiva/diagnóstico , Disfunción Cognitiva/inmunología , Femenino , Regulación de la Expresión Génica , Ontología de Genes , Infecciones por VIH/complicaciones , Infecciones por VIH/diagnóstico , Infecciones por VIH/inmunología , Antígenos de Histocompatibilidad Clase I/sangre , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Interferón gamma/sangre , Interferón gamma/genética , Interferón gamma/inmunología , Masculino , Persona de Mediana Edad , Anotación de Secuencia Molecular , Monocitos/inmunología , Factores de Necrosis Tumoral/sangre , Factores de Necrosis Tumoral/genética , Factores de Necrosis Tumoral/inmunología
3.
Retrovirology ; 13(1): 49, 2016 07 26.
Artículo en Inglés | MEDLINE | ID: mdl-27459960

RESUMEN

BACKGROUND: Eradication of HIV cannot be achieved with combination antiretroviral therapy (cART) because of the persistence of long-lived latently infected resting memory CD4(+) T cells. We previously reported that HIV latency could be established in resting CD4(+) T cells in the presence of the chemokine CCL19. To define how CCL19 facilitated the establishment of latent HIV infection, the role of chemokine receptor signalling was explored. RESULTS: In resting CD4(+) T cells, CCL19 induced phosphorylation of RAC-alpha serine/threonine-protein kinase (Akt), nuclear factor kappa B (NF-κB), extracellular-signal-regulated kinase (ERK) and p38. Inhibition of the phosphoinositol-3-kinase (PI3K) and Ras/Raf/Mitogen-activated protein kinase/ERK kinase (MEK)/ERK signalling pathways inhibited HIV integration, without significant reduction in HIV nuclear entry (measured by Alu-LTR and 2-LTR circle qPCR respectively). Inhibiting activation of MEK1/ERK1/2, c-Jun N-terminal kinase (JNK), activating protein-1 (AP-1) and NF-κB, but not p38, also inhibited HIV integration. We also show that HIV integrases interact with Pin1 in CCL19-treated CD4(+) T cells and inhibition of JNK markedly reduced this interaction, suggesting that CCL19 treatment provided sufficient signals to protect HIV integrase from degradation via the proteasome pathway. Infection of CCL19-treated resting CD4(+) T cells with mutant strains of HIV, lacking NF-κB binding sites in the HIV long terminal repeat (LTR) compared to infection with wild type virus, led to a significant reduction in integration by up to 40-fold (range 1-115.4, p = 0.03). This was in contrast to only a modest reduction of 5-fold (range 1.7-11, p > 0.05) in fully activated CD4(+) T cells infected with the same mutants. Finally, we demonstrated significant differences in integration sites following HIV infection of unactivated, CCL19-treated, and fully activated CD4(+) T cells. CONCLUSIONS: HIV integration in CCL19-treated resting CD4(+) T cells depends on NF-κB signalling and increases the stability of HIV integrase, which allow subsequent integration and establishment of latency. These findings have implications for strategies needed to prevent the establishment, and potentially reverse, latent infection.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Quimiocina CCL19/farmacología , FN-kappa B/metabolismo , Receptores CCR/genética , Integración Viral , Latencia del Virus , Linfocitos T CD4-Positivos/efectos de los fármacos , Linfocitos T CD4-Positivos/fisiología , Regulación Viral de la Expresión Génica/efectos de los fármacos , Integrasa de VIH/genética , VIH-1/enzimología , VIH-1/fisiología , Humanos , FN-kappa B/genética , Receptores CCR/metabolismo , Transducción de Señal/efectos de los fármacos , Integración Viral/efectos de los fármacos , Latencia del Virus/efectos de los fármacos , Replicación Viral/efectos de los fármacos
5.
Sci Rep ; 5: 10010, 2015 Jun 18.
Artículo en Inglés | MEDLINE | ID: mdl-26084721

RESUMEN

Cocaine abuse has been shown to have broad-ranging effects on human immunity. With regards to HIV infection, in vitro studies have shown that cocaine enhances infection of stimulated lymphocytes. Moreover, cohort studies in the pre- and post-HAART era have linked stimulant abuse with increased HIV pathogenesis. The latter data, however, have been undermined by a series of confounding factors underscoring the importance of controlled in vivo models to fully assess the impact of cocaine use and abuse on HIV infection and pathogenesis. Here, we have infected humanized mice with HIV-1 following acute cocaine exposure to assess the impact on infection. Stimulant exposure resulted in increased inflammatory cytokine expression, accelerated HIV infection, while blunting effector function of cytotoxic T lymphocytes. These data demonstrate cocaine's multifactorial impact on HIV infection that extends beyond high-risk behavior.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Cocaína/efectos adversos , Citocinas/inmunología , Regulación de la Expresión Génica/efectos de los fármacos , Infecciones por VIH/inmunología , VIH-1/inmunología , Animales , Linfocitos T CD8-positivos/patología , Cocaína/farmacología , Modelos Animales de Enfermedad , Regulación de la Expresión Génica/inmunología , Infecciones por VIH/patología , Humanos , Ratones
6.
Mol Ther Nucleic Acids ; 4: e236, 2015 Apr 14.
Artículo en Inglés | MEDLINE | ID: mdl-25872029

RESUMEN

We described earlier a dual-combination anti-HIV type 1 (HIV-1) lentiviral vector (LVsh5/C46) that downregulates CCR5 expression of transduced cells via RNAi and inhibits HIV-1 fusion via cell surface expression of cell membrane-anchored C46 antiviral peptide. This combinatorial approach has two points of inhibition for R5-tropic HIV-1 and is also active against X4-tropic HIV-1. Here, we utilize the humanized bone marrow, liver, thymus (BLT) mouse model to characterize the in vivo efficacy of LVsh5/C46 (Cal-1) vector to engineer cellular resistance to HIV-1 pathogenesis. Human CD34+ hematopoietic stem/progenitor cells (HSPC) either nonmodified or transduced with LVsh5/C46 vector were transplanted to generate control and treatment groups, respectively. Control and experimental groups displayed similar engraftment and multilineage hematopoietic differentiation that included robust CD4+ T-cell development. Splenocytes isolated from the treatment group were resistant to both R5- and X4-tropic HIV-1 during ex vivo challenge experiments. Treatment group animals challenged with R5-tropic HIV-1 displayed significant protection of CD4+ T-cells and reduced viral load within peripheral blood and lymphoid tissues up to 14 weeks postinfection. Gene-marking and transgene expression were confirmed stable at 26 weeks post-transplantation. These data strongly support the use of LVsh5/C46 lentiviral vector in gene and cell therapeutic applications for inhibition of HIV-1 infection.

7.
Sci Rep ; 5: 8670, 2015 Mar 02.
Artículo en Inglés | MEDLINE | ID: mdl-25728014

RESUMEN

Prenatal exposure to cocaine is a significant source of fetal and neonatal developmental defects. While cocaine associated neurological and cardiac pathologies are well-documented, it is apparent that cocaine use has far more diverse physiological effects. It is known that in some cell types, the sigma-1 receptor mediates many of cocaine's cellular effects. Here we present a novel and concise investigation into the mechanism that underlies cocaine associated hematopoietic pathology. Indeed, this is the first examination of the effects of cocaine on hematopoiesis. We show that cocaine impairs multilineage hematopoiesis from human progenitors from multiple donors and tissue types. We go on to present the first demonstration of the expression of the sigma-1 receptor in human CD34 + human hematopoietic stem/progenitor cells. Furthermore, we demonstrate that these cocaine-induced hematopoietic defects can be reversed through sigma-1 receptor blockade.


Asunto(s)
Linaje de la Célula/efectos de los fármacos , Cocaína/farmacología , Hematopoyesis/efectos de los fármacos , Células Madre Hematopoyéticas/citología , Células Madre Hematopoyéticas/metabolismo , Receptores sigma/metabolismo , Adulto , Antígenos CD34/metabolismo , Médula Ósea/efectos de los fármacos , Médula Ósea/metabolismo , Separación Celular , Sangre Fetal/efectos de los fármacos , Sangre Fetal/metabolismo , Células Madre Hematopoyéticas/efectos de los fármacos , Humanos , Hígado/efectos de los fármacos , Hígado/embriología , Hígado/metabolismo , Receptor Sigma-1
8.
J Clin Invest ; 125(5): 1815-26, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25822024

RESUMEN

Adoptive transfer of tumor-reactive T cells can successfully reduce tumor burden; however, in rare cases, lethal on-target/off-tumor effects have been reported. A noninvasive method to track engineered cells with high sensitivity and resolution would allow observation of correct cell homing and/or identification of dangerous off-target locations in preclinical and clinical applications. Human deoxycytidine kinase triple mutant (hdCK3mut) is a nonimmunogenic PET reporter that was previously shown to be an effective tool to monitor whole-body hematopoiesis. Here, we engineered a construct in which hdCK3mut is coexpressed with the anti-melanoma T cell receptor F5, introduced this construct into human CD34 cells or PBMCs, and evaluated this approach in multiple immunotherapy models. Expression of hdCK3mut allowed engrafted cells to be visualized within recipient bone marrow, while accumulation of [18F]-L-FMAU in hdCK3mut-expressing T cells permitted detection of intratumoral homing. Animals that received T cells coexpressing hdCK3mut and the anti-melanoma T cell receptor had demonstrably higher signals in HLA-matched tumors compared with those in animals that received cells solely expressing hdCK3mut. Engineered T cells caused cytotoxicity in HLA/antigen-matched tumors and induced IFN-γ production and activation. Moreover, hdCK3mut permitted simultaneous monitoring of engraftment and tumor infiltration, without affecting T cell function. Our findings suggest that hdCK3mut reporter imaging can be applied in clinical immunotherapies for whole-body detection of engineered cell locations.


Asunto(s)
Desoxicitidina Quinasa/análisis , Genes Reporteros , Inmunoterapia/métodos , Linfocitos Infiltrantes de Tumor/inmunología , Melanoma Experimental/terapia , Tomografía de Emisión de Positrones , Linfocitos T/inmunología , Animales , Médula Ósea/diagnóstico por imagen , Quimiotaxis de Leucocito , Pruebas Inmunológicas de Citotoxicidad , Desoxicitidina Quinasa/genética , Genes Sintéticos , Vectores Genéticos/genética , Supervivencia de Injerto , Antígeno HLA-A2/inmunología , Trasplante de Células Madre Hematopoyéticas , Células Madre Hematopoyéticas/química , Células Madre Hematopoyéticas/fisiología , Humanos , Inmunoterapia Adoptiva , Ensayos de Liberación de Interferón gamma , Lentivirus/genética , Leucocitos Mononucleares/química , Leucocitos Mononucleares/fisiología , Leucocitos Mononucleares/trasplante , Antígeno MART-1/inmunología , Melanoma Experimental/inmunología , Ratones , Mutación , Receptores de Antígenos de Linfocitos T/análisis , Receptores de Antígenos de Linfocitos T/genética , Proteínas Recombinantes de Fusión/análisis , Proteínas Recombinantes de Fusión/genética , Retroviridae/genética , Linfocitos T/trasplante , Timo/trasplante
9.
Blood ; 122(13): 2195-204, 2013 Sep 26.
Artículo en Inglés | MEDLINE | ID: mdl-23886835

RESUMEN

HIV infection has been associated with defective hematopoiesis since the earliest days of the HIV/AIDS epidemic. Generation of all hematopoietic lineages suffers in the face of infection. The mechanisms by which HIV impairs normal blood cell development remain unclear, and direct infection of intermediate hematopoietic progenitors has not been established as a source of HIV-associated hematopoietic pathology. Here, we demonstrate infection of multiple subsets of highly purified intermediate hematopoietic progenitors by wild-type HIV both in vitro and in vivo. Although direct infection is clearly cytotoxic, we find that some infected progenitors can survive and harbor proviral DNA. We report intermediate hematopoietic progenitors to be a novel target of infection and their permissivity to infection increases with development. Further, the nonobese diabetic severe combined immunodeficiency common γ chain knockout-bone marrow-liver-thymus humanized mouse provides a unique model for studying the impact of HIV infection on bone marrow-based human hematopoiesis.


Asunto(s)
Infecciones por VIH/patología , VIH-1/inmunología , Células Madre Hematopoyéticas/virología , Animales , ADN Viral , Modelos Animales de Enfermedad , Citometría de Flujo , Infecciones por VIH/inmunología , Hematopoyesis/inmunología , Humanos , Ratones , Reacción en Cadena en Tiempo Real de la Polimerasa
10.
J Leukoc Biol ; 94(4): 835-43, 2013 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-23817564

RESUMEN

In vivo and in vitro exposure to stimulants has been associated with increased levels of HIV infection in PBMCs. Among these lymphocyte subsets, quiescent CD4(+) T cells make up the majority of circulating T cells in the blood. Others and we have demonstrated that HIV infects this population of cells inefficiently. However, minor changes in their cell state can render them permissive to infection, significantly impacting the viral reservoir. We have hypothesized that stimulants, such as cocaine, may perturb the activation state of quiescent cells enhancing permissiveness to infection. Quiescent T cells isolated from healthy human donors were exposed to cocaine and infected with HIV. Samples were harvested at different time-points to assess the impact of cocaine on their susceptibility to infection at various stages of the HIV life cycle. Our data show that a 3-day exposure to cocaine enhanced infection of quiescent cells, an effect that appears to be mediated by σ1R and D4R. Overall, our results indicate that cocaine-mediated effects on quiescent T cells may increase the pool of infection-susceptible T cells. The latter underscores the impact that stimulants have on HIV-seropositive individuals and the challenges posed for treatment.


Asunto(s)
Cocaína/farmacología , Infecciones por VIH/patología , VIH/fisiología , Linfocitos T/virología , Internalización del Virus/efectos de los fármacos , VIH/efectos de los fármacos , Humanos , Cinética , Fenotipo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores de Dopamina D4/metabolismo , Receptores sigma/metabolismo , Transcripción Reversa/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Proteínas Virales/genética , Proteínas Virales/metabolismo , Receptor Sigma-1
11.
Retrovirology ; 10: 37, 2013 Apr 04.
Artículo en Inglés | MEDLINE | ID: mdl-23557201

RESUMEN

The restriction of the Human Immunodeficiency Virus (HIV) infection in quiescent CD4⁺ T cells has been an area of active investigation. Early studies have suggested that this T cell subset is refractory to infection by the virus. Subsequently it was demonstrated that quiescent cells could be infected at low levels; nevertheless these observations supported the earlier assertions of debilitating defects in the viral life cycle. This phenomenon raised hopes that identification of the block in quiescent cells could lead to the development of new therapies against HIV. As limiting levels of raw cellular factors such as nucleotides did not account for the block to infection, a number of groups pursued the identification of cellular proteins whose presence or absence may impact the permissiveness of quiescent T cells to HIV infection. A series of studies in the past few years have identified a number of host factors implicated in the block to infection. In this review, we will present the progress made, other avenues of investigation and the potential impact these studies have in the development of more effective therapies against HIV.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH/inmunología , Replicación Viral , Humanos
12.
Mol Ther ; 21(5): 1055-63, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23481324

RESUMEN

Current tumor immunotherapy approaches include the genetic modification of peripheral T cells to express tumor antigen-specific T-cell receptors (TCRs). The approach, tested in melanoma, has led to some limited success of tumor regression in patients. Yet, the introduction of exogenous TCRs into mature T cells entails an underlying risk; the generation of autoreactive clones due to potential TCR mispairing, and the lack of effective negative selection, as these peripheral cells do not undergo thymic selection following introduction of the exogenous TCR. We have successfully generated MART-1-specific CD8 T cells from genetically modified human hematopoietic stem cells (hHSC) in a humanized mouse model. The advantages of this approach include a long-term source of antigen specific T cells and proper T-cell selection due to thymopoiesis following expression of the TCR. In this report, we examine the molecular processes occurring on endogenous TCR expression and demonstrate that this approach results in exclusive cell surface expression of the newly introduced TCR, and the exclusion of endogenous TCR cell surface expression. This suggests that this stem cell based approach can provide a potentially safer approach for anticancer immunotherapy due to the involvement of thymic selection.


Asunto(s)
Regulación de la Expresión Génica , Células Madre Hematopoyéticas/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Animales , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Diferenciación Celular/inmunología , Selección Clonal Mediada por Antígenos/inmunología , Expresión Génica , Reordenamiento Génico de Linfocito T , Células Madre Hematopoyéticas/citología , Humanos , Implantes Experimentales , Ratones , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Linfocitos T/citología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Transgenes
13.
J Vis Exp ; (70): e4181, 2012 Dec 18.
Artículo en Inglés | MEDLINE | ID: mdl-23271478

RESUMEN

Small animal models such as mice have been extensively used to study human disease and to develop new therapeutic interventions. Despite the wealth of information gained from these studies, the unique characteristics of mouse immunity as well as the species specificity of viral diseases such as human immunodeficiency virus (HIV) infection led to the development of humanized mouse models. The earlier models involved the use of C. B 17 scid/scid mice and the transplantation of human fetal thymus and fetal liver termed thy/liv (SCID-hu) (1, 2) or the adoptive transfer of human peripheral blood leukocytes (SCID-huPBL) (3). Both models were mainly utilized for the study of HIV infection. One of the main limitations of both of these models was the lack of stable reconstitution of human immune cells in the periphery to make them a more physiologically relevant model to study HIV disease. To this end, the BLT humanized mouse model was developed. BLT stands for bone marrow/liver/thymus. In this model, 6 to 8 week old NOD.Cg-Prkdcscid Il2rgtm1Wjl/SzJ (NSG) immunocompromised mice receive the thy/liv implant as in the SCID-hu mouse model only to be followed by a second human hematopoietic stem cell transplant (4). The advantage of this system is the full reconstitution of the human immune system in the periphery. This model has been used to study HIV infection and latency (5-8). We have generated a modified version of this model in which we use genetically modified human hematopoietic stem cells (hHSC) to construct the thy/liv implant followed by injection of transduced autologous hHSC (7, 9). This approach results in the generation of genetically modified lineages. More importantly, we adapted this system to examine the potential of generating functional cytotoxic T cells (CTL) expressing a melanoma specific T cell receptor. Using this model we were able to assess the functionality of our transgenic CTL utilizing live positron emission tomography (PET) imaging to determine tumor regression (9). The goal of this protocol is to describe the process of generating these transgenic mice and assessing in vivo efficacy using live PET imaging. As a note, since we use human tissues and lentiviral vectors, our facilities conform to CDC NIH guidelines for Biosafety Level 2 (BSL2) with special precautions (BSL2+). In addition, the NSG mice are severely immunocompromised thus, their housing and maintenance must conform to the highest health standards (http://jaxmice.jax.org/research/immunology/005557-housing.html).


Asunto(s)
Trasplante de Médula Ósea/métodos , Modelos Animales de Enfermedad , Terapia Genética/métodos , Células Madre Hematopoyéticas/fisiología , Trasplante de Hígado/métodos , Neoplasias Experimentales/terapia , Timo/trasplante , Animales , Antígenos CD34/biosíntesis , Antígenos CD34/inmunología , Femenino , Células Madre Hematopoyéticas/inmunología , Humanos , Masculino , Ratones , Ratones SCID , Ratones Transgénicos , Neoplasias Experimentales/genética , Trasplante Heterólogo
14.
Oncoimmunology ; 1(4): 539-540, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22754779

RESUMEN

Using a humanized mouse model, we developed melanoma specific CD8 T cells from genetically modified human hematopoietic stem cells (hHSC). The transgenic T cells were functional both in vivo and ex vivo, effectively limiting and clearing tumor growth. Finally, the transduced hHSC stably populated the bone marrow.

15.
PLoS One ; 6(11): e26894, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-22073215

RESUMEN

The HIV-1 Trans-Activator of Transcription (Tat) protein binds to multiple host cellular factors and greatly enhances the level of transcription of the HIV genome. While Tat's control of viral transcription is well-studied, much less is known about the interaction of Tat with the human genome. Here, we report the genome-wide binding map of Tat to the human genome in Jurkat T cells using chromatin immunoprecipitation combined with next-generation sequencing. Surprisingly, we found that ~53% of the Tat target regions are within DNA repeat elements, greater than half of which are Alu sequences. The remaining target regions are located in introns and distal intergenic regions; only ~7% of Tat-bound regions are near transcription start sites (TSS) at gene promoters. Interestingly, Tat binds to promoters of genes that, in Jurkat cells, are bound by the ETS1 transcription factor, the CBP histone acetyltransferase and/or are enriched for histone H3 lysine 4 tri-methylation (H3K4me3) and H3K27me3. Tat binding is associated with genes enriched with functions in T cell biology and immune response. Our data reveal that Tat's interaction with the host genome is more extensive than previously thought, with potentially important implications for the viral life cycle.


Asunto(s)
Productos del Gen tat/metabolismo , Genoma Humano , VIH-1/metabolismo , Inmunoprecipitación de Cromatina , Humanos , Células Jurkat , Unión Proteica
16.
Proc Natl Acad Sci U S A ; 108(51): E1408-16, 2011 Dec 20.
Artículo en Inglés | MEDLINE | ID: mdl-22123951

RESUMEN

The goal of cancer immunotherapy is the generation of an effective, stable, and self-renewing antitumor T-cell population. One such approach involves the use of high-affinity cancer-specific T-cell receptors in gene-therapy protocols. Here, we present the generation of functional tumor-specific human T cells in vivo from genetically modified human hematopoietic stem cells (hHSC) using a human/mouse chimera model. Transduced hHSC expressing an HLA-A*0201-restricted melanoma-specific T-cell receptor were introduced into humanized mice, resulting in the generation of a sizeable melanoma-specific naïve CD8(+) T-cell population. Following tumor challenge, these transgenic CD8(+) T cells, in the absence of additional manipulation, limited and cleared human melanoma tumors in vivo. Furthermore, the genetically enhanced T cells underwent proper thymic selection, because we did not observe any responses against non-HLA-matched tumors, and no killing of any kind occurred in the absence of a human thymus. Finally, the transduced hHSC established long-term bone marrow engraftment. These studies present a potential therapeutic approach and an important tool to understand better and to optimize the human immune response to melanoma and, potentially, to other types of cancer.


Asunto(s)
Antineoplásicos/farmacología , Linfocitos T CD8-positivos/citología , Células Madre Hematopoyéticas/citología , Animales , Antígenos CD34/biosíntesis , Linfocitos T CD8-positivos/inmunología , Línea Celular Tumoral , Citometría de Flujo/métodos , Ingeniería Genética/métodos , Humanos , Inmunoterapia/métodos , Linfocitos Infiltrantes de Tumor/citología , Ratones , Ratones SCID , Modelos Genéticos , Trasplante de Neoplasias , Células Madre/citología , Timo/metabolismo , Transgenes
17.
Clin Vaccine Immunol ; 18(10): 1776-80, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21832097

RESUMEN

We examined the effect of two leader sequences, one from a transmembrane molecule (H2-L(d)) and another from a secreted molecule (rat KC chemokine), on the immunogenicity of DNA epitope vaccines. The chemokine leader enhanced vaccine immunogenicity, thus underscoring the importance of the leader sequence in DNA epitope vaccine design.


Asunto(s)
Epítopos/genética , Epítopos/inmunología , Señales de Clasificación de Proteína/genética , Vacunas de ADN/inmunología , Animales , Pruebas Inmunológicas de Citotoxicidad , Inmunización Secundaria/métodos , Interferón gamma/metabolismo , Interleucina-2/metabolismo , Ratones , Ratones Endogámicos BALB C , Linfocitos T/inmunología , Vacunación/métodos , Vacunas de ADN/administración & dosificación , Vacunas de ADN/genética
18.
Immunol Res ; 48(1-3): 110-21, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20725862

RESUMEN

The ability of HIV to infect quiescent CD4+ T cells has been a topic of intense debate. While early studies suggested that the virus could not infect this particular T cell subset, subsequent studies using more sensitive protocols demonstrated that these cells could inefficiently support HIV infection. Additional studies showed that the kinetics of infection in quiescent cells was delayed and multiple stages of the viral life cycle were marred by inefficiencies. Despite that, proviral DNA has been found in these cells presenting them as a potential viral reservoir. Therefore, a better understanding of the relationship between HIV and quiescent T cells may lead to further advances in the field of HIV.


Asunto(s)
Linfocitos T CD4-Positivos/fisiología , Reservorios de Enfermedades/virología , Infecciones por VIH/inmunología , VIH/fisiología , Subgrupos de Linfocitos T/fisiología , Linfocitos T CD4-Positivos/virología , Ciclo Celular/genética , Ciclo Celular/inmunología , Regulación Viral de la Expresión Génica , VIH/patogenicidad , Infecciones por VIH/virología , Interacciones Huésped-Patógeno , Humanos , Subgrupos de Linfocitos T/virología , Virulencia/genética , Virulencia/inmunología , Latencia del Virus/genética , Latencia del Virus/inmunología , Replicación Viral/inmunología
19.
J Virol ; 83(12): 6222-33, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19369341

RESUMEN

Until very recently, quiescent CD4(+) T cells were thought to be resistant to human immunodeficiency virus (HIV) infection. Subsequent studies, attempting to fully elucidate the mechanisms of resistance, showed that quiescent cells could become infected by HIV at low efficiency and form a latently infected population. In this study, we set out to identify the sites of viral integration and to assess the efficiency of the overall integration process in quiescent cells. Based on our results, HIV integration in quiescent CD4(+) T cells occurs in sites similar to those of their prestimulated counterparts. While site selections are similar, the integration process in quiescent cells is plagued by the formation of high levels of incorrectly processed viral ends and abortive two-long-terminal-repeat circles.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Infecciones por VIH/virología , VIH/fisiología , Integración Viral , Linfocitos T CD4-Positivos/citología , Línea Celular , Mapeo Cromosómico , ADN Viral , VIH/genética , Duplicado del Terminal Largo de VIH , Humanos , Activación de Linfocitos , Análisis de Secuencia de ADN
20.
J Virol ; 83(7): 3374-8, 2009 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-19129455

RESUMEN

The activation state of CD4(+) T cells plays a crucial role in the establishment of a productive human immunodeficiency virus infection. Here, we show that T cells stimulated for 1 day demonstrated delayed kinetics of viral reverse transcription and integration compared to cells stimulated for 2 days prior to infection. As a result, the efficiency of reverse transcription and integration inhibitors differs in these differentially stimulated cells. These studies increase our understanding of how T cells support viral replication and provide insight regarding the efficiency of antiretroviral therapy in lymphoid compartments.


Asunto(s)
Fármacos Anti-VIH/farmacología , Linfocitos T CD4-Positivos/virología , Infecciones por VIH/inmunología , VIH/fisiología , Activación de Linfocitos , Transcripción Reversa , Linfocitos T CD4-Positivos/inmunología , Células Cultivadas , VIH/efectos de los fármacos , VIH/inmunología , Humanos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA