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










Base de datos
Intervalo de año de publicación
1.
Biomedicines ; 9(6)2021 Jun 09.
Artículo en Inglés | MEDLINE | ID: mdl-34207853

RESUMEN

NK cells are the first line of defense against viruses and malignant cells, and their natural functionality makes these cells a promising candidate for cancer cell therapy. The genetic modifications of NK cells, allowing them to overcome some of their inherent limitations, such as low proliferative potential, can enable their use as a therapeutic product. We demonstrate that hTERT-engineered NK cell cultures maintain a high percentage of cells in the S/G2 phase for an extended time after transduction, while the life span of NK cells is measurably extended. Bulk and clonal NK cell cultures pre-activated in vitro with IL-2 and K562-mbIL21 feeder cells can be transduced with hTERT more efficiently compared with the cells activated with IL-2 alone. Overexpressed hTERT was functionally active in transduced NK cells, which displayed upregulated expression of the activation marker HLA-DR, and decreased expression of the maturation marker CD57 and activating receptor NKp46. Larger numbers of KIR2DL2/3+ cells in hTERT-engineered populations may indicate that NK cells with this phenotype are more susceptible to transduction. The hTERT-modified NK cells demonstrated a high natural cytotoxic response towards K562 cells and stably expressed Ki67, a proliferation marker. Overall, our data show that ectopic hTERT expression in NK cells enhances their activation and proliferation, extends in vitro life span, and can be a useful tool in developing NK-based cancer cell therapies.

2.
J Virol ; 93(21)2019 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-31434738

RESUMEN

Adoptive cell transfer (ACT) is a powerful experimental approach to directly study T-cell-mediated immunity in vivo In the rhesus macaque AIDS virus model, infusing simian immunodeficiency virus (SIV)-infected animals with CD8 T cells engineered to express anti-SIV T-cell receptor specificities enables direct experimentation to better understand antiviral T-cell immunity in vivo Limiting factors in ACT experiments include suboptimal trafficking to, and poor persistence in, the secondary lymphoid tissues targeted by AIDS viruses. Previously, we redirected CD8 T cells to B-cell follicles by ectopic expression of the CXCR5 homing protein. Here, we modify peripheral blood mononuclear cell (PBMC)-derived CD8 T cells to express the CCR9 chemokine receptor, which induces preferential homing of the engineered cells to the small intestine, a site of intense early AIDS virus replication and pathology in rhesus macaques. Additionally, we increase in vivo persistence and overall systemic distribution of infused CD8 T cells, especially in secondary lymphoid tissues, by minimizing ex vivo culture/manipulation, thereby avoiding the loss of CD28+/CD95+ central memory T cells by differentiation in culture. These proof-of-principle results establish the feasibility of preferentially localizing PBMC-derived CD8 T cells to the small intestine and enables the direct experimental ACT-based assessment of the potential role of the quality and timing of effective antiviral CD8 T-cell responses to inhibit viral infection and subsequent replication in small intestine CD4 T cells. More broadly, these results support the engineered expression of homing proteins to direct CD8 T cells to target tissues as a means for both experimental and potential therapeutic advances in T-cell immunotherapies, including cancer.IMPORTANCEAdoptive cell transfer (ACT) of T cells engineered with antigen-specific effector properties can deliver targeted immune responses against malignancies and infectious diseases. Current T-cell-based therapeutic ACT relies on circulatory distribution to deliver engineered T cells to their targets, an approach which has proven effective for some leukemias but provided only limited efficacy against solid tumors. Here, engineered expression of the CCR9 homing receptor redirected CD8 T cells to the small intestine in rhesus macaque ACT experiments. Targeted homing of engineered T-cell immunotherapies holds promise to increase the effectiveness of adoptively transferred cells in both experimental and clinical settings.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Quimiotaxis de Leucocito/inmunología , Intestino Delgado/inmunología , Receptores CCR/metabolismo , Traslado Adoptivo , Animales , Antígenos CD28/metabolismo , Linfocitos T CD8-positivos/metabolismo , Quimiocinas CC/metabolismo , Memoria Inmunológica , Intestino Delgado/virología , Leucocitos Mononucleares/inmunología , Ganglios Linfáticos/inmunología , Macaca mulatta , Transducción de Señal , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Virus de la Inmunodeficiencia de los Simios/inmunología
3.
PLoS One ; 13(3): e0195246, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29590210

RESUMEN

[This corrects the article DOI: 10.1371/journal.pone.0023703.].

4.
J Immunol Methods ; 450: 90-94, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28802832

RESUMEN

Natural killer (NK) cells are capable of rapidly recognizing and efficiently killing tumor cells. This makes them a potentially promising agent for cancer immunotherapy. Additional genetic modifications of NK cells may further improve their anti-tumor efficacy. Numerous technical challenges associated with gene delivery into NK cells have significantly tempered this approach. We achieved efficient retroviral vector transduction of primary human NK cells that were stimulated by a combination of IL-2 and engineered K562 cells expressing membrane-bound IL-21. The activated NK cells were in less differentiated state and expressed NK cell activation receptors NKG2D, NKp30, CD16, and were highly HLA-DR-positive. This NK cell population was highly susceptible to the transduction by both GFP- and NGFR-expressing retroviral vectors, with transduction efficiency exceeding 50%. More mature CD57+ NK cell population was generally resistant to retroviral vector transduction because of poor response to the stimulation. Our findings may facilitate retroviral vector-mediated genetic engineering of human primary NK cells for future immunotherapies.


Asunto(s)
Vectores Genéticos , Inmunoterapia/métodos , Interleucina-2/metabolismo , Interleucinas/metabolismo , Células Asesinas Naturales/metabolismo , Leucemia Eritroblástica Aguda/terapia , Activación de Linfocitos , Retroviridae/genética , Transducción Genética , Transfección/métodos , Antígenos CD57/inmunología , Antígenos CD57/metabolismo , Diferenciación Celular , Técnicas de Cocultivo , Células Nutrientes , Proteínas Ligadas a GPI/inmunología , Proteínas Ligadas a GPI/metabolismo , Proteínas Fluorescentes Verdes/genética , Proteínas Fluorescentes Verdes/metabolismo , Antígenos HLA-DR/inmunología , Antígenos HLA-DR/metabolismo , Humanos , Interleucina-2/inmunología , Interleucinas/inmunología , Células Jurkat , Células K562 , Células Asesinas Naturales/inmunología , Leucemia Eritroblástica Aguda/genética , Leucemia Eritroblástica Aguda/inmunología , Leucemia Eritroblástica Aguda/metabolismo , Subfamilia K de Receptores Similares a Lectina de Células NK/inmunología , Subfamilia K de Receptores Similares a Lectina de Células NK/metabolismo , Receptor 3 Gatillante de la Citotoxidad Natural/inmunología , Receptor 3 Gatillante de la Citotoxidad Natural/metabolismo , Proteínas del Tejido Nervioso/genética , Proteínas del Tejido Nervioso/metabolismo , Receptores de IgG/inmunología , Receptores de IgG/metabolismo , Receptores de Factor de Crecimiento Nervioso/genética , Receptores de Factor de Crecimiento Nervioso/metabolismo
5.
J Virol ; 90(21): 9942-9952, 2016 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-27558423

RESUMEN

AIDS virus infections are rarely controlled by cell-mediated immunity, in part due to viral immune evasion and immunodeficiency resulting from CD4+ T-cell infection. One likely aspect of this failure is that antiviral cellular immune responses are either absent or present at low levels during the initial establishment of infection. To test whether an extensive, timely, and effective response could reduce the establishment of infection from a high-dose inoculum, we adoptively transferred large numbers of T cells that were molecularly engineered with anti-simian immunodeficiency virus (anti-SIV) activity into rhesus macaques 3 days following an intrarectal SIV inoculation. To measure in vivo antiviral activity, we assessed the number of viruses transmitted using SIVmac239X, a molecularly tagged viral stock containing 10 genotypic variants, at a dose calculated to transmit 12 founder viruses. Single-genome sequencing of plasma virus revealed that the two animals receiving T cells expressing SIV-specific T-cell receptors (TCRs) had significantly fewer viral genotypes than the two control animals receiving non-SIV-specific T cells (means of 4.0 versus 7.5 transmitted viral genotypes; P = 0.044). Accounting for the likelihood of transmission of multiple viruses of a particular genotype, the calculated means of the total number of founder viruses transmitted were 4.5 and 14.5 in the experimental and control groups, respectively (P = 0.021). Thus, a large antiviral T-cell response timed with virus exposure can limit viral transmission. The presence of strong, preexisting T-cell responses, including those induced by vaccines, might help prevent the establishment of infection at the lower-exposure doses in humans that typically transmit only a single virus. IMPORTANCE: The establishment of AIDS virus infection in an individual is essentially a race between the spreading virus and host immune defenses. Cell-mediated immune responses induced by infection or vaccination are important contributors in limiting viral replication. However, in human immunodeficiency virus (HIV)/SIV infection, the virus usually wins the race, irreversibly crippling the immune system before an effective cellular immune response is developed and active. We found that providing an accelerated response by adoptively transferring large numbers of antiviral T cells shortly after a high-dose mucosal inoculation, while not preventing infection altogether, limited the number of individual viruses transmitted. Thus, the presence of strong, preexisting T-cell responses, including those induced by vaccines, might prevent infection in humans, where the virus exposure is considerably lower.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/virología , Macaca mulatta/inmunología , Macaca mulatta/virología , Virus de la Inmunodeficiencia de los Simios/inmunología , Traslado Adoptivo/métodos , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Células Cultivadas , Humanos , Inmunidad Celular/inmunología , Vacunas contra el SIDAS/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Síndrome de Inmunodeficiencia Adquirida del Simio/virología , Vacunación/métodos , Replicación Viral/genética
6.
J Virol ; 87(22): 12237-48, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24006441

RESUMEN

Kaposi's sarcoma-associated herpesvirus (KSHV) encodes 12 pre-microRNAs that can produce 25 KSHV mature microRNAs. We previously reported single-nucleotide polymorphisms (SNPs) in KSHV-encoded pre-microRNA and mature microRNA sequences from clinical samples (V. Marshall et al., J. Infect. Dis., 195:645-659, 2007). To determine whether microRNA SNPs affect pre-microRNA processing and, ultimately, mature microRNA expression levels, we performed a detailed comparative analysis of (i) mature microRNA expression levels, (ii) in vitro Drosha/Dicer processing, and (iii) RNA-induced silencing complex-dependent targeting of wild-type (wt) and variant microRNA genes. Expression of pairs of wt and variant pre-microRNAs from retroviral vectors and measurement of KSHV mature microRNA expression by real-time reverse transcription-PCR (RT-PCR) revealed differential expression levels that correlated with the presence of specific sequence polymorphisms. Measurement of KSHV mature microRNA expression in a panel of primary effusion lymphoma cell lines by real-time RT-PCR recapitulated some observed expression differences but suggested a more complex relationship between sequence differences and expression of mature microRNA. Furthermore, in vitro maturation assays demonstrated significant SNP-associated changes in Drosha/DGCR8 and/or Dicer processing. These data demonstrate that SNPs within KSHV-encoded pre-microRNAs are associated with differential microRNA expression levels. Given the multiple reports on the involvement of microRNAs in cancer, the biological significance of these phenotypic and genotypic variants merits further studies in patients with KSHV-associated malignancies.


Asunto(s)
Regulación Viral de la Expresión Génica , Infecciones por Herpesviridae/genética , Herpesvirus Humano 8/patogenicidad , Linfoma de Efusión Primaria/virología , MicroARNs/genética , Polimorfismo de Nucleótido Simple/genética , Procesamiento Postranscripcional del ARN/genética , ARN Viral/genética , Células Cultivadas , Células HEK293 , Infecciones por Herpesviridae/virología , Herpesvirus Humano 8/genética , Humanos , Luciferasas/metabolismo , Linfoma de Efusión Primaria/genética , MicroARNs/fisiología , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
7.
Curr Protoc Immunol ; Chapter 7: Unit 7.21B, 2011 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22048804

RESUMEN

Human and rhesus macaque primary antigen-specific T cells derived from infected or immunized individuals or animals are a valuable material with which to study cellular immune responses against pathogens and tumors. Antigen-specific T cells can be expanded in vitro but have a finite proliferative life span. After a limited period in culture, primary T cells undergo replicative senescence and stop dividing. This restricts their applicability to short-term experiments and complicates their use in adoptive immunotherapy. The proliferative life span of primary human and rhesus macaque T cells can be considerably extended by ectopically expressed human telomerase reverse transcriptase (TERT). Antigen-specific T cells transduced with TERT-expressing retroviral vectors can proliferate and expand in culture for long periods of time while maintaining their primary T cell characteristics, including antigen-specific responses. Thus, TERT-immortalized T cells are an important and valuable resource for studying T cell-mediated immune responses and, potentially, for adoptive immunotherapy.


Asunto(s)
Complejo CD3/inmunología , Macaca mulatta/inmunología , Linfocitos T/inmunología , Telomerasa/inmunología , Animales , Vectores Genéticos , Humanos , Inmunoterapia Adoptiva , Virus de la Leucemia Murina/genética , Telomerasa/genética
8.
PLoS One ; 6(8): e23703, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21886812

RESUMEN

BACKGROUND: The SIV/rhesus macaque model for HIV/AIDS is a powerful system for examining the contribution of T cells in the control of AIDS viruses. To better our understanding of CD8(+) T-cell control of SIV replication in CD4(+) T cells, we asked whether TCRs isolated from rhesus macaque CD8(+) T-cell clones that exhibited varying abilities to suppress SIV replication could convey their suppressive properties to CD8(+) T cells obtained from an uninfected/unvaccinated animal. PRINCIPAL FINDINGS: We transferred SIV-specific TCR genes isolated from rhesus macaque CD8(+) T-cell clones with varying abilities to suppress SIV replication in vitro into CD8(+) T cells obtained from an uninfected animal by retroviral transduction. After sorting and expansion, transduced CD8(+) T-cell lines were obtained that specifically bound their cognate SIV tetramer. These cell lines displayed appropriate effector function and specificity, expressing intracellular IFNγ upon peptide stimulation. Importantly, the SIV suppression properties of the transduced cell lines mirrored those of the original TCR donor clones: cell lines expressing TCRs transferred from highly suppressive clones effectively reduced wild-type SIV replication, while expression of a non-suppressing TCR failed to reduce the spread of virus. However, all TCRs were able to suppress the replication of an SIV mutant that did not downregulate MHC-I, recapitulating the properties of their donor clones. CONCLUSIONS: Our results show that antigen-specific SIV suppression can be transferred between allogenic T cells simply by TCR gene transfer. This advance provides a platform for examining the contributions of TCRs versus the intrinsic effector characteristics of T-cell clones in virus suppression. Additionally, this approach can be applied to develop non-human primate models to evaluate adoptive T-cell transfer therapy for AIDS and other diseases.


Asunto(s)
Linfocitos T CD8-positivos/metabolismo , Genes Codificadores de los Receptores de Linfocitos T/genética , Macaca mulatta/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Especificidad del Receptor de Antígeno de Linfocitos T/inmunología , Transducción Genética , Replicación Viral/inmunología , Animales , Antígenos Virales , Técnicas de Cultivo de Célula , Células Clonales/inmunología , Humanos , Macaca mulatta/genética
9.
Nanomedicine (Lond) ; 6(7): 1175-88, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21675859

RESUMEN

AIMS: While numerous studies have reported on nanoparticle uptake by phagocytic cells, the mechanisms of this uptake are poorly understood. A metastudy of research focusing on biological particulate matter has postulated that nanoparticles cannot be phagocytosed and therefore must enter cells via pinocytosis. The purpose of this study was to identify the route(s) of uptake of gold nanoparticles in vitro and to determine if these route(s) depend on particle size. MATERIALS & METHODS: The parent RAW264.7 cell line and its derivatives, transduced with a virus carrying siRNA to macrophage scavenger receptor A, were used as model phagocytes. Citrate-stabilized gold colloids were used as model nanoparticles. We used chemical inhibitors known to interfere with specific routes of particulate uptake. We developed multifocal light microscopy methods including multifocal stack analysis with NIH ImageJ software to analyze cell uptake. RESULTS: Irrespective of size, gold nanoparticles are internalized by macrophages via multiple routes, including both phagocytosis and pinocytosis. If either route was blocked, the particles entered cells via the other route. CONCLUSION: Gold nanoparticles with hydrodynamic sizes below 100 nm can be phagocytosed. Phagocytosis of anionic gold colloids by RAW264.7 cells is mediated by macrophage scavenger receptor A.


Asunto(s)
Oro Coloide/metabolismo , Macrófagos/citología , Receptores Depuradores/metabolismo , Receptores Depuradores de Clase A/metabolismo , Animales , Línea Celular , Macrófagos/metabolismo , Ratones , Fagocitosis , Pinocitosis
10.
Immunotherapy ; 3(3): 407-21, 2011 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-21395382

RESUMEN

Telomeres are specialized repeats, present at the end of chromosomes, whose loss during cell division is followed by growth arrest, a central mechanism of replicative senescence in human cells. Telomere length in stem cells is maintained by telomerase, a specialized reverse transcriptase, whose function is to restore shortening telomeres. Unlike most somatic cell types, human T lymphocytes are capable of briefly reactivating telomerase expression at the time of stimulation. Telomerase expression in T lymphocytes is modulated by a variety of external stimuli and by viral infections. However, telomerase reactivation in stimulated, proliferating human T lymphocytes is limited and cannot prevent the ultimate onset of senescence. Ectopic telomerase expression can rescue human and macaque antigen-specific T cells from senescence. Primary T cells have been engineered with telomerase to have substantially extended replicative lifespans without the loss of primary cell functions or malignant transformation. 'Immortal' antigen-specific T-cell lines and clones overexpressing telomerase are an invaluable source of well-characterized quasi-primary T cells for research of T-cell biology and are potentially useful for immunotherapy of cancer and AIDS.


Asunto(s)
Supervivencia Celular/genética , Inmunoterapia Adoptiva/métodos , Linfocitos T Citotóxicos , Telomerasa , Animales , Técnicas de Cultivo de Célula , División Celular , Células Cultivadas , Senescencia Celular/genética , Cromosomas/metabolismo , Humanos , Macaca , Secuencias Repetitivas de Ácidos Nucleicos , Transducción de Señal , Linfocitos T/metabolismo , Linfocitos T Citotóxicos/enzimología , Linfocitos T Citotóxicos/metabolismo , Telomerasa/biosíntesis , Telomerasa/genética , Telomerasa/fisiología , Telómero/metabolismo , Transformación Genética
11.
Virology ; 409(1): 132-40, 2011 Jan 05.
Artículo en Inglés | MEDLINE | ID: mdl-21035160

RESUMEN

Studies using transformed human cell lines suggest that most SIV strains use CCR5 as co-receptor. Our analysis of primary rhesus macaque CD4(+) T-cell clones revealed marked differences in susceptibility to SIV(mac)239 infection. We investigated whether different levels of CCR5 expression account for clonal differences in SIV(mac)239 susceptibility. Macaque CD4(+) T-cells showed significant CCR5 downregulation 1-2days following CD3 mAb stimulation, which gradually recovered at resting state, 7-10days after activation. Exposure of clones to SIV(mac)239 during their CCR5(low) or CCR5(high) expression states revealed differences in SIV susceptibility independent of surface CCR5 levels. Furthermore, a CCR5 antagonist similarly reduced SIV(mac)239 infection of clones during their CCR5(low) or CCR5(high) expression states. Our data suggest a model where i) very low levels of CCR5 are sufficient for efficient SIV infection, ii) CCR5 levels above this threshold do not enhance infection, and iii) low level infection can occur in the absence of CCR5.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Regulación hacia Abajo , Receptores de Antígenos de Linfocitos T/inmunología , Receptores CCR5/metabolismo , Síndrome de Inmunodeficiencia Adquirida del Simio/inmunología , Virus de la Inmunodeficiencia de los Simios/patogenicidad , Animales , Anticuerpos Monoclonales/inmunología , Anticuerpos Monoclonales/metabolismo , Antagonistas de los Receptores CCR5 , Complejo CD3/inmunología , Complejo CD3/metabolismo , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Línea Celular , ADN Viral/análisis , Femenino , Productos del Gen gag/genética , Productos del Gen gag/inmunología , Productos del Gen gag/metabolismo , Humanos , Macaca mulatta , Masculino , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores CCR5/genética , Síndrome de Inmunodeficiencia Adquirida del Simio/virología
12.
PLoS One ; 5(11): e13872, 2010 Nov 08.
Artículo en Inglés | MEDLINE | ID: mdl-21079786

RESUMEN

BACKGROUND: BORIS/CTCFL is a paralogue of CTCF, the major epigenetic regulator of vertebrate genomes. BORIS is normally expressed only in germ cells but is aberrantly activated in numerous cancers. While recent studies demonstrated that BORIS is a transcriptional activator of testis-specific genes, little is generally known about its biological and molecular functions. METHODOLOGY/PRINCIPAL FINDINGS: Here we show that BORIS is expressed as 23 isoforms in germline and cancer cells. The isoforms are comprised of alternative N- and C-termini combined with varying numbers of zinc fingers (ZF) in the DNA binding domain. The patterns of BORIS isoform expression are distinct in germ and cancer cells. Isoform expression is activated by downregulation of CTCF, upregulated by reduction in CpG methylation caused by inactivation of DNMT1 or DNMT3b, and repressed by activation of p53. Studies of ectopically expressed isoforms showed that all are translated and localized to the nucleus. Using the testis-specific cerebroside sulfotransferase (CST) promoter and the IGF2/H19 imprinting control region (ICR), it was shown that binding of BORIS isoforms to DNA targets in vitro is methylation-sensitive and depends on the number and specific composition of ZF. The ability to bind target DNA and the presence of a specific long amino terminus (N258) in different isoforms are necessary and sufficient to activate CST transcription. Comparative sequence analyses revealed an evolutionary burst in mammals with strong conservation of BORIS isoproteins among primates. CONCLUSIONS: The extensive repertoire of spliced BORIS variants in humans that confer distinct DNA binding and transcriptional activation properties, and their differential patterns of expression among germ cells and neoplastic cells suggest that the gene is involved in a range of functionally important aspects of both normal gametogenesis and cancer development. In addition, a burst in isoform diversification may be evolutionarily tied to unique aspects of primate speciation.


Asunto(s)
Empalme Alternativo , Proteínas de Unión al ADN/genética , Gametogénesis/genética , Regiones Promotoras Genéticas/genética , Secuencia de Aminoácidos , Animales , Secuencia de Bases , Factor de Unión a CCCTC , Metilación de ADN , Proteínas de Unión al ADN/metabolismo , Femenino , Regulación de la Expresión Génica , Células HCT116 , Células HEK293 , Humanos , Células K562 , Proteínas Luminiscentes/genética , Proteínas Luminiscentes/metabolismo , Masculino , Microscopía Fluorescente , Datos de Secuencia Molecular , Neoplasias/genética , Neoplasias/patología , Isoformas de Proteínas/genética , Proteínas Represoras/genética , Proteínas Represoras/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Homología de Secuencia de Aminoácido , Testículo/citología , Testículo/metabolismo , Proteína p53 Supresora de Tumor/genética , Proteína p53 Supresora de Tumor/metabolismo , Dedos de Zinc
13.
Virology ; 391(1): 130-9, 2009 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-19555986

RESUMEN

CD8(+) T lymphocytes (CTL) play a role in controlling HIV/SIV infection. CTL antiviral activity is dependent on recognition of antigenic peptides associated with MHC class I molecules on infected target cells, and CTL activation can be impaired by Nef-mediated down-regulation of MHC class I molecules. We tested the ability of a series of rhesus macaque CD8(+) T-cell clones specific for the SIV Gag CM9 peptide to suppress SIV infection of autologous CD4(+) T cells. We used a set of SIV(mac)239 viruses with either wild-type Nef or Nef mutations that impair MHC class I down-regulation. All CTL clones efficiently suppressed virus replication in cells infected with mutant viruses with altered Nef function, phenotypically MHC class I(high) or MHC class I(intermediate). However, the ability of the clones to suppress virus replication was variably reduced in the presence of wild-type Nef (MHC class I(low)) despite the observations that all CTL clones showed similar IFN-gamma responses to titrated amounts of cognate peptide as well as to SIV-infected cells. In addition, the CTL clones showed variable CD107a (CTL degranulation marker) responses that did not correlate with their capacity to suppress virus replication. Thus, the clonal differences are not attributable to TCR avidity or typical effector responses, and point to a potential as yet unknown mechanism for CTL-mediated suppression of viral replication. These data emphasize that current assays for evaluating CTL responses in infected or vaccinated individuals do not fully capture the complex requirements for effective CTL-mediated control of virus replication.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Productos del Gen nef/metabolismo , Antígenos de Histocompatibilidad Clase I/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/virología , Células Cultivadas , Regulación hacia Abajo , Regulación de la Expresión Génica , Interferón gamma/inmunología , Proteína 1 de la Membrana Asociada a los Lisosomas/inmunología , Macaca mulatta/inmunología , Macaca mulatta/virología , ARN Viral/metabolismo , Virus de la Inmunodeficiencia de los Simios/fisiología , Replicación Viral
14.
Methods Mol Biol ; 511: 143-58, 2009.
Artículo en Inglés | MEDLINE | ID: mdl-19347296

RESUMEN

Antigen-specific T cells play a key role in cellular immune response against cancer. The ability to isolate, maintain, and characterize tumor-specific T cells is a prerequisite to studying anticancer immune response and developing novel strategies for cancer immunotherapy. However, the life span of human T cells in vitro is usually short and is limited by the onset of cellular senescence. To establish long-term, antigen-specific T-cell lines and clones, we selectively immortalized antigen-responsive T cells from human peripheral blood mononuclear cells (PBMCs). PBMCs were stimulated with antigens, and then infected with a murine leukemia virus-based retroviral vector carrying an immortalizing gene, the human telomerase-reverse transcriptase gene. Since such vectors can only integrate in dividing cells, only antigen-activated T cells are efficiently transduced. Using this approach, we generated immortalized T-cell lines that maintained strictly IL-2-dependent growth and MHC-restricted, antigen-specific responsiveness, some of which have been in continuous culture for longer than 1 year, far in excess of the survival of parallel control nonimmortalized cultures. These lines showed antigen-specific proliferation with induced cytokine and chemokine production, and, in the case of CD8+ T-cell lines, antigen-specific cytolytic activity. When applied to the tumor antigen-specific T cells, the approach provides a convenient, reproducible means for generating a stable, continuously renewable source of antigen-specific T lymphocytes for a variety of studies on anticancer immunity.


Asunto(s)
Técnicas de Cultivo de Célula , Línea Celular Tumoral , Neoplasias , Linfocitos T/citología , Separación Celular/métodos , Transformación Celular Neoplásica , Humanos , Neoplasias/inmunología , Neoplasias/patología , Retroviridae/genética , Retroviridae/metabolismo , Linfocitos T/inmunología , Telomerasa/genética , Telomerasa/metabolismo
15.
Virology ; 375(1): 307-14, 2008 May 25.
Artículo en Inglés | MEDLINE | ID: mdl-18328525

RESUMEN

CD8(+) cytotoxic T lymphocytes (CTL) play an important role in controlling virus replication in HIV- and SIV-infected humans and monkeys, respectively. Three well-studied SIV CTL determinants are the two Mamu A()01-restricted epitopes Gag CM9 and Tat SL8, and the Mamu B()17-restricted epitope Nef IW9. Point mutations leading to amino acid replacements in these epitopes have been reported to mediate SIV escape from CTL control. We found that synthetic peptides containing mutations in SIV Gag CM9 and Tat SL8 were no longer recognized by the respective CTL. On the other hand, the described I-to-T replacement at the N-terminal amino acid residue of the SIV Nef IW9 epitope only moderately affected CTL recognition of the variant peptide, TW9. In an attempt to dissect the mechanism of escape of the Nef TW9 mutation, we investigated the effect of this mutation on CTL recognition of CD4(+)T cells infected with an engineered SIV(mac)239 that contained the TW9 mutation in Nef. Although, the wild type and mutant virus both infected and efficiently replicated in rhesus macaque CD4(+)T cells, the TW9 mutant virus failed to induce IFN-gamma expression in an SIV Nef IW9-specific CTL clone. Thus, unlike escape from Gag CM9- or Tat SL8-specfic CTL control presumably by loss of epitope binding, these results point to a defect at the level of processing and/or presentation of the variant TW9 epitope with resultant loss of triggering of the cognate TCR on CTL generated against the wild type peptide. Our data highlight the value of functional assays using virus-infected target cells as opposed to peptide-pulsed APC when assessing relevant escape mutations in CTL epitopes.


Asunto(s)
Sustitución de Aminoácidos , Epítopos de Linfocito T/genética , Epítopos de Linfocito T/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Proteínas Reguladoras y Accesorias Virales/genética , Proteínas Reguladoras y Accesorias Virales/inmunología , Replicación Viral , Animales , Linfocitos T CD4-Positivos/virología , Células Cultivadas , Interferón gamma/biosíntesis , Macaca mulatta , Mutación Missense , Virus de la Inmunodeficiencia de los Simios/fisiología , Linfocitos T Citotóxicos/inmunología , Replicación Viral/fisiología
16.
Virology ; 372(2): 430-41, 2008 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-18068748

RESUMEN

CD8(+) cytotoxic T lymphocyte (CTL) responses play an important role in controlling the replication of primate lentiviruses. Induction of these responses is a key objective for most current AIDS vaccine approaches. Despite a variety of approaches for measuring properties and activities of CTL, the functions responsible for controlling viral replication in vivo have not been clearly identified. Assays measuring CTL-mediated suppression of viral replication in vitro are beginning to be used as possible correlates of in vivo virus suppressive activity, but the utility and interpretive value of these assays are typically limited by properties of the cells that have been used. We investigated the capacity of SIV-specific CTL clones (effectors), immortalized by transduction with human telomerase reverse transcriptase (hTERT), to suppress SIV replication in autologous hTERT immortalized CD4(+) T-cell clones (targets). Immortalized and non-immortalized SIV-specific effector cells showed IFN-gamma production and degranulation in response to viral antigen specific stimulation and significantly inhibited SIV(mac)239 replication (2 to 4 log decrease in viral RNA or cell-associated proviral DNA) (p<0.0005). Our in vitro assays of inhibition of viral replication, using T-cell clones as effectors and targets, provide a well-defined approach for evaluating possible mechanisms of CTL-mediated control of viral production which may involve direct killing of infected target cells and/or release of proinflammatory cytokines such as IFN-gamma and TNF-alpha. The use of hTERT immortalized effector and target cells for such assays preserves relevant functional properties while providing a convenient, reproducible means of conducting studies over time.


Asunto(s)
Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/inmunología , Virus de la Inmunodeficiencia de los Simios/inmunología , Replicación Viral/inmunología , Animales , Línea Celular , Regulación de la Expresión Génica , Proteína 1 de la Membrana Asociada a los Lisosomas/metabolismo , Macaca mulatta , Virus de la Inmunodeficiencia de los Simios/fisiología
17.
AIDS Res Hum Retroviruses ; 23(3): 456-65, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17411379

RESUMEN

T cell lines and clones play a key role in basic studies of cellular immunology, and are also finding applications in adoptive immunotherapy. However, with proliferative expansion, T cells ultimately undergo cellular senescence and death, so that long-term culture of T cell clones is difficult to achieve. Expression of telomerase reverse transcriptase (TERT) in differentiated cells can maintain telomere length over many cell divisions, preventing senescence. We used a retroviral vector that expresses the human TERT (hTERT) gene to transduce a rhesus macaque-derived CD8(+) T cell clone specific for the MamuA*01-restricted immunodominant SIV gag epitope CM9. Extensive in vitro characterization revealed that the untransduced parental cells and the hTERT-transduced cells displayed comparable proliferation capacity, effector memory surface marker profiles, cytolytic activities, and cytokine profiles following antigen stimulation. The hTERT-transduced cells showed improved survival compared to parallel nontransduced cultures during in vitro propagation in long-term culture. Such immortalized T cells may be useful as a source of consistent controls for in vitro assays of cellular immune function, and as a potentially important reagent for autologous adoptive cellular immunotherapy studies in macaques.


Asunto(s)
Linfocitos T CD8-positivos , Macaca mulatta/virología , Virus de la Inmunodeficiencia de los Simios/inmunología , Linfocitos T Citotóxicos/metabolismo , Telomerasa/metabolismo , Transducción Genética/métodos , Animales , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Linfocitos T CD8-positivos/virología , Línea Celular , Células Clonales/inmunología , Células Clonales/metabolismo , Femenino , Humanos , Linfocitos T Citotóxicos/virología
18.
J Virol ; 80(18): 9039-52, 2006 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-16940516

RESUMEN

Human immunodeficiency virus type 1 (HIV-1) infects CD4(+) T lymphocytes and monocytes/macrophages, incorporating host proteins in the process of assembly and budding. Analysis of the host cell proteins incorporated into virions can provide insights into viral biology. We characterized proteins in highly purified HIV-1 virions produced from human monocyte-derived macrophages (MDM), within which virus buds predominantly into intracytoplasmic vesicles, in contrast to the plasmalemmal budding of HIV-1 typically seen with infected T cells. Liquid chromatography-linked tandem mass spectrometry of highly purified virions identified many cellular proteins, including 33 previously described proteins in HIV-1 preparations from other cell types. Proteins involved in many different cellular structures and functions were present, including those from the cytoskeleton, adhesion, signaling, intracellular trafficking, chaperone, metabolic, ubiquitin/proteasomal, and immune response systems. We also identified annexins, annexin-binding proteins, Rab proteins, and other proteins involved in membrane organization, vesicular trafficking, and late endosomal function, as well as apolipoprotein E, which participates in cholesterol transport, immunoregulation, and modulation of cell growth and differentiation. Several tetraspanins, markers of the late endosomal compartment, were also identified. MDM-derived HIV contained 26 of 37 proteins previously found in exosomes, consistent with the idea that HIV uses the late endosome/multivesicular body pathway during virion budding from macrophages.


Asunto(s)
VIH-1/aislamiento & purificación , VIH-1/fisiología , Macrófagos/citología , Monocitos/citología , Proteómica/métodos , Linfocitos T/virología , Línea Celular , Proteína gp120 de Envoltorio del VIH/química , VIH-1/ultraestructura , Humanos , Antígenos Comunes de Leucocito/biosíntesis , Macrófagos/virología , Monocitos/virología , Proteína Tirosina Fosfatasa no Receptora Tipo 1 , Espectrometría de Masa por Ionización de Electrospray , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Tripsina/farmacología , Proteínas Virales/química , Replicación Viral
19.
Immunol Lett ; 105(1): 26-37, 2006 May 15.
Artículo en Inglés | MEDLINE | ID: mdl-16442639

RESUMEN

To establish long-term, antigen-specific T-cell lines and clones, we selectively immortalized antigen-responsive T cells from human peripheral blood mononuclear cells (PBMCs). PBMCs were stimulated with either alloantigen or soluble antigen, then infected with a murine leukemia virus-based retroviral vector carrying an immortalizing gene, either the Tax gene from human T-cell leukemia virus type 1, or the human telomerase-reverse transcriptase gene. Since such vectors can only integrate in dividing cells, only antigen-activated T cells are efficiently transduced. This approach generated immortalized antigen-specific CD4+ and CD8+ T-cell lines that maintained strictly IL-2-dependent growth and HLA-restricted, antigen-specific responsiveness, some of which have been in continuous culture for longer than 1 year, far in excess of the survival of parallel control non-immortalized cultures. Clones derived from these lines showed antigen-specific proliferation with induced cytokine and chemokine production, and, in the case of a CD8+ T-cell clone, antigen-specific cytolytic activity. This approach provides a convenient, reproducible means for generating a stable, continuously renewable source of antigen-specific T lymphocytes for a variety of studies of T cell biology.


Asunto(s)
Técnicas de Cultivo de Célula/métodos , Linfocitos T/inmunología , Secuencia de Aminoácidos , Antígenos/administración & dosificación , Antígenos/genética , Línea Celular , Línea Celular Transformada , Proliferación Celular , Proteínas de Unión al ADN/genética , Toxoide Diftérico/administración & dosificación , Genes pX , Vectores Genéticos , Antígenos de Superficie de la Hepatitis B/administración & dosificación , Antígenos de Superficie de la Hepatitis B/genética , Humanos , Isoantígenos , Fragmentos de Péptidos/administración & dosificación , Fragmentos de Péptidos/genética , Fragmentos de Péptidos/inmunología , Linfocitos T/citología , Telomerasa/genética , Transducción Genética
20.
J Virol ; 79(16): 10258-67, 2005 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-16051819

RESUMEN

Previous attempts to extend the host range of the avian sarcoma/leukosis virus (ASLV)-based RCASBP vectors produced two viral vectors, RCASBP M2C (4070A) and RCASBP M2C (797-8), which replicate using the amphotropic murine leukemia virus 4070A Env protein (2). Both viruses were adapted to replicate efficiently in the avian cell line DF-1, but RCASBP M2C (4070A) caused extensive cytopathic effects (CPE) in DF-1 cells whereas RCASBP M2C (797-8) induced low levels of CPE. The two viruses differed only at amino acid 242 of the polyproline-rich region in the surface (SU) subunit of the Env protein. In RCASBP M2C (4070A), an isoleucine replaced the wild-type proline residue, whereas a threonine residue was found in RCASBP M2C (797-8). In the present study, we show that other amino acid substitutions at position 242 strongly influence the CPE and replication rate of the chimeric viruses. There was a correlation between the amount of unintegrated linear retroviral DNA present in infected DF-1 cells and the level of CPE. This suggests that there may be a role for superinfection in the CPE. The treatment of RCASBP M2C (4070A)-infected cells with dantrolene, which inhibits the release of calcium from the endoplasmic reticulum (ER), reduced the amount of CPE seen during infection with the highly cytotoxic virus. Dantrolene treatment did not appear to affect virus production, suggesting that Ca2+ release from the ER had a role in the CPE caused by these viruses.


Asunto(s)
Efecto Citopatogénico Viral , Productos del Gen env/química , Orthoreovirus Aviar/fisiología , Replicación Viral , Animales , Calcio/metabolismo , Línea Celular , Embrión de Pollo , ADN Viral/análisis , Dantroleno/farmacología , Retículo Endoplásmico/metabolismo , Productos del Gen env/fisiología , Vectores Genéticos , Orthoreovirus Aviar/patogenicidad , Péptidos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...