Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 58
Filtrar
Más filtros

Bases de datos
Tipo del documento
Intervalo de año de publicación
1.
Annu Rev Immunol ; 34: 317-34, 2016 05 20.
Artículo en Inglés | MEDLINE | ID: mdl-27168241

RESUMEN

CD4(+) T helper (Th) cells play a central role in the adaptive immune response by providing help to B cells and cytotoxic T cells and by releasing different types of cytokines in tissues to mediate protection against a wide range of pathogenic microorganisms. These functions are performed by different types of Th cells endowed with distinct migratory capacities and effector functions. Here we discuss how studies of the human T cell response to microbes have advanced our understanding of Th cell functional heterogeneity, in particular with the discovery of a distinct Th1 subset involved in the response to Mycobacteria and the characterization of two types of Th17 cells specific for extracellular bacteria or fungi. We also review new approaches to dissect at the clonal level the human CD4(+) T cell response induced by pathogens or vaccines that have revealed an unexpected degree of intraclonal diversification and propose a progressive and selective model of CD4(+) T cell differentiation.


Asunto(s)
Inmunidad Adaptativa , Infecciones Bacterianas/inmunología , Biodiversidad , Micosis/inmunología , Células TH1/inmunología , Células Th17/inmunología , Vacunas/inmunología , Animales , Antígenos CD4/metabolismo , Diferenciación Celular , Selección Clonal Mediada por Antígenos , Células Clonales , Citotoxicidad Inmunológica , Humanos , Inmunidad Humoral , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo
2.
Cell ; 186(19): 4216-4234.e33, 2023 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-37714135

RESUMEN

Chronic stimulation can cause T cell dysfunction and limit the efficacy of cellular immunotherapies. Improved methods are required to compare large numbers of synthetic knockin (KI) sequences to reprogram cell functions. Here, we developed modular pooled KI screening (ModPoKI), an adaptable platform for modular construction of DNA KI libraries using barcoded multicistronic adaptors. We built two ModPoKI libraries of 100 transcription factors (TFs) and 129 natural and synthetic surface receptors (SRs). Over 30 ModPoKI screens across human TCR- and CAR-T cells in diverse conditions identified a transcription factor AP4 (TFAP4) construct that enhanced fitness of chronically stimulated CAR-T cells and anti-cancer function in vitro and in vivo. ModPoKI's modularity allowed us to generate an ∼10,000-member library of TF combinations. Non-viral KI of a combined BATF-TFAP4 polycistronic construct enhanced fitness. Overexpressed BATF and TFAP4 co-occupy and regulate key gene targets to reprogram T cell function. ModPoKI facilitates the discovery of complex gene constructs to program cellular functions.


Asunto(s)
Tratamiento Basado en Trasplante de Células y Tejidos , Ejercicio Físico , Humanos , Biblioteca de Genes , Inmunoterapia , Investigación
3.
Cell ; 175(7): 1958-1971.e15, 2018 12 13.
Artículo en Inglés | MEDLINE | ID: mdl-30449619

RESUMEN

Human T cells are central effectors of immunity and cancer immunotherapy. CRISPR-based functional studies in T cells could prioritize novel targets for drug development and improve the design of genetically reprogrammed cell-based therapies. However, large-scale CRISPR screens have been challenging in primary human cells. We developed a new method, single guide RNA (sgRNA) lentiviral infection with Cas9 protein electroporation (SLICE), to identify regulators of stimulation responses in primary human T cells. Genome-wide loss-of-function screens identified essential T cell receptor signaling components and genes that negatively tune proliferation following stimulation. Targeted ablation of individual candidate genes characterized hits and identified perturbations that enhanced cancer cell killing. SLICE coupled with single-cell RNA sequencing (RNA-seq) revealed signature stimulation-response gene programs altered by key genetic perturbations. SLICE genome-wide screening was also adaptable to identify mediators of immunosuppression, revealing genes controlling responses to adenosine signaling. The SLICE platform enables unbiased discovery and characterization of functional gene targets in primary cells.


Asunto(s)
Sistemas CRISPR-Cas , Genoma Humano , Linfocitos T/inmunología , Proteína 9 Asociada a CRISPR/genética , Proteína 9 Asociada a CRISPR/inmunología , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Técnicas de Inactivación de Genes , Estudio de Asociación del Genoma Completo , Humanos , Linfocitos T/citología
4.
Molecules ; 28(8)2023 Apr 08.
Artículo en Inglés | MEDLINE | ID: mdl-37110545

RESUMEN

Cytotoxic T-lymphocyte-associated protein 4 (CTLA-4) is a check point protein expressed on the surface of T cells and plays a central role in regulating the immune response. In recent years, CTLA-4 has become a popular target for cancer immunotherapy in which blocking CTLA-4 can restore T-cell function and enhance the immune response against cancer. Currently, there are many CTLA-4 inhibitors in a variety of modalities, including cell therapies, which are being developed in both preclinical and clinical stages to further harness the potential of the target for the treatment of certain types of cancer. In drug discovery research, measuring the level of CTLA-4 in T cells is important for drug discovery and development because it provides key information for quantitative assessment of the pharmacodynamics, efficacy, and safety of the CTLA-4-based therapies. However, to our best knowledge, there is still no report of a sensitive, specific, accurate, and reliable assay for CTLA-4 measurement. In this work, an LC/MS-based method was developed to measure CTLA-4 in human T cells. The assay demonstrated high specificity with an LLOQ of 5 copies of CTLA-4 per cell when using 2.5 million T cells for analysis. As shown in the work, the assay was successfully used to measure CTLA-4 levels in subtype T-cell samples from individual healthy subjects. The assay could be applied in supporting the studies of CTLA-4-based cancer therapies.


Asunto(s)
Neoplasias , Humanos , Antígeno CTLA-4/metabolismo , Neoplasias/tratamiento farmacológico , Inmunoterapia/métodos , Linfocitos T/metabolismo
5.
Eur J Immunol ; 51(6): 1365-1376, 2021 06.
Artículo en Inglés | MEDLINE | ID: mdl-33682083

RESUMEN

Studies in murine models show that subthreshold TCR interactions with self-peptide are required for thymic development and peripheral survival of naïve T cells. Recently, differences in the strength of tonic TCR interactions with self-peptide, as read-out by cell surface levels of CD5, were associated with distinct effector potentials among sorted populations of T cells in mice. However, whether CD5 can also be used to parse functional heterogeneity among human T cells is less clear. Our study demonstrates that CD5 levels correlate with TCR signal strength in human naïve CD4+ T cells. Further, we describe a relationship between CD5 levels on naïve human CD4+ T cells and binding affinity to foreign peptide, in addition to a predominance of CD5hi T cells in the memory compartment. Differences in gene expression and biases in cytokine production potential between CD5lo and CD5hi naïve human CD4+ T cells are consistent with observations in mice. Together, these data validate the use of CD5 surface levels as a marker of heterogeneity among human naïve CD4+ T cells with important implications for the identification of functionally biased T- cell populations that can be exploited to improve the efficacy of adoptive cell therapies.


Asunto(s)
Biomarcadores/metabolismo , Linfocitos T CD4-Positivos/inmunología , Antígenos CD5/metabolismo , Inmunoterapia Adoptiva/métodos , Receptores de Antígenos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/inmunología , Animales , Autoantígenos/metabolismo , Células Cultivadas , Selección Clonal Mediada por Antígenos , Humanos , Memoria Inmunológica , Sinapsis Inmunológicas , Ratones , Ratones Endogámicos C57BL , Unión Proteica , Transducción de Señal
6.
Cryobiology ; 103: 92-100, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34508713

RESUMEN

Cryopreservation of human T lymphocytes has become an essential tool for some cell-based immunotherapy. However, the cryopreservation procedure of the cells has not been systematically studied. In particular, the key factors of ice seeding and cryoprotective agents (CPA) driving the success of cryopreservation remain unclear. We systematically investigated the key factors, including cooling rate, ice-seeding temperature, CPA concentration, and types of CPA, during cryopreservation of human T lymphocytes with controlled ice nucleation. We found that ice seeding at below -10 °C could enable human T lymphocytes to be cooled at 90 °C min-1 with high relative viability and recovery after rewarming, 94.9% and 90.2%, respectively, which are significantly higher than those without ice seeding (P < 0.001). After optimization, the concentration of dimethyl sulphoxide was as low as 2% (v/v) with relative viability and recovery of 95.4% and 100.8%, respectively, at the cooling rate of 90 °C min-1 after ice seeding at -16 °C. The cryopreservation procedure developed in this study could facilitate the understanding of the mechanism for ice seeding and cell injury and offer a promising cryopreservation method with a high cooling rate and extremely low toxicity for extensive clinical application of immunotherapy.


Asunto(s)
Criopreservación , Hielo , Criopreservación/métodos , Crioprotectores/farmacología , Congelación , Humanos , Linfocitos T
7.
Int J Mol Sci ; 20(9)2019 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-31083348

RESUMEN

Detrimental health consequences from exposure to space radiation are a major concern for long-duration human exploration missions to the Moon or Mars. Cellular responses to radiation are expected to be heterogeneous for space radiation exposure, where only high-energy protons and other particles traverse a fraction of the cells. Therefore, assessing DNA damage and DNA damage response in individual cells is crucial in understanding the mechanisms by which cells respond to different particle types and energies in space. In this project, we identified a cell-specific signature for radiation response by using single-cell transcriptomics of human lymphocyte subpopulations. We investigated gene expression in individual human T lymphocytes 3 h after ex vivo exposure to 2-Gy gamma rays while using the single-cell sequencing technique (10X Genomics). In the process, RNA was isolated from ~700 irradiated and ~700 non-irradiated control cells, and then sequenced with ~50 k reads/cell. RNA in each of the cells was distinctively barcoded prior to extraction to allow for quantification for individual cells. Principal component and clustering analysis of the unique molecular identifier (UMI) counts classified the cells into three groups or sub-types, which correspond to CD4+, naïve, and CD8+/NK cells. Gene expression changes after radiation exposure were evaluated using negative binomial regression. On average, BBC3, PCNA, and other TP53 related genes that are known to respond to radiation in human T cells showed increased activation. While most of the TP53 responsive genes were upregulated in all groups of cells, the expressions of IRF1, STAT1, and BATF were only upregulated in the CD4+ and naïve groups, but were unchanged in the CD8+/NK group, which suggests that the interferon-gamma pathway does not respond to radiation in CD8+/NK cells. Thus, single-cell RNA sequencing technique was useful for simultaneously identifying the expression of a set of genes in individual cells and T lymphocyte subpopulation after gamma radiation exposure. The degree of dependence of UMI counts between pairs of upregulated genes was also evaluated to construct a similarity matrix for cluster analysis. The cluster analysis identified a group of TP53-responsive genes and a group of genes that are involved in the interferon gamma pathway, which demonstrate the potential of this method for identifying previously unknown groups of genes with similar expression patterns.


Asunto(s)
Exposición a la Radiación , Factor de Transcripción STAT1/metabolismo , Análisis de Secuencia de ARN , Transducción de Señal , Análisis de la Célula Individual , Subgrupos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/efectos de la radiación , Proteína p53 Supresora de Tumor/metabolismo , Análisis por Conglomerados , Rayos gamma , Humanos , Inmunofenotipificación , Reproducibilidad de los Resultados , Transducción de Señal/genética , Transducción de Señal/efectos de la radiación , Regulación hacia Arriba/genética , Regulación hacia Arriba/efectos de la radiación
8.
Breast Cancer Res ; 20(1): 111, 2018 09 12.
Artículo en Inglés | MEDLINE | ID: mdl-30208917

RESUMEN

BACKGROUND: Antimicrobial T cells play key roles in the disease progression of cancers arising in mucosal epithelial tissues, such as the colon. However, little is known about microbe-reactive T cells within human breast ducts and whether these impact breast carcinogenesis. METHODS: Epithelial ducts were isolated from primary human breast tissue samples, and the associated T lymphocytes were characterized using flow cytometric analysis. Functional assays were performed to determine T-cell cytokine secretion in response to bacterially treated human breast carcinoma cells. RESULTS: We show that human breast epithelial ducts contain mucosal associated invariant T (MAIT) cells, an innate T-cell population that recognizes specific bacterial metabolites presented by nonclassical MR1 antigen-presenting molecules. The MAIT cell population from breast ducts resembled that of peripheral blood in its innate lymphocyte phenotype (i.e., CD161, PLZF, and interleukin [IL]-18 receptor coexpression), but the breast duct MAIT cell population had a distinct T-cell receptor Vß use profile and was markedly enriched for IL-17-producing cells compared with blood MAIT cells. Breast carcinoma cells that had been exposed to Escherichia coli activated MAIT cells in an MR1-dependent manner. However, whereas phorbol 12-myristate 13-acetate/ionomycin stimulation induced the production of both interferon-γ and IL-17 by breast duct MAIT cells, bacterially exposed breast carcinoma cells elicited a strongly IL-17-biased response. Breast carcinoma cells also showed upregulated expression of natural killer group 2 member D (NKG2D) ligands compared with primary breast epithelial cells, and the NKG2D receptor contributed to MAIT cell activation by the carcinoma cells. CONCLUSIONS: These results demonstrate that MAIT cells from human breast ducts mediate a selective T-helper 17 cell response to human breast carcinoma cells that were exposed to E. coli. Thus, cues from the breast microbiome and the expression of stress-associated ligands by neoplastic breast duct epithelial cells may shape MAIT cell responses during breast carcinogenesis.


Asunto(s)
Neoplasias de la Mama/inmunología , Células Epiteliales/inmunología , Interleucina-17/inmunología , Células T Invariantes Asociadas a Mucosa/inmunología , Mama/citología , Neoplasias de la Mama/metabolismo , Neoplasias de la Mama/microbiología , Línea Celular Tumoral , Células Cultivadas , Citocinas/inmunología , Citocinas/metabolismo , Células Epiteliales/metabolismo , Escherichia coli/inmunología , Escherichia coli/fisiología , Femenino , Antígenos de Histocompatibilidad Clase I/genética , Antígenos de Histocompatibilidad Clase I/inmunología , Humanos , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-17/metabolismo , Activación de Linfocitos/inmunología , Antígenos de Histocompatibilidad Menor/genética , Antígenos de Histocompatibilidad Menor/inmunología , Células T Invariantes Asociadas a Mucosa/metabolismo
9.
Malar J ; 17(1): 114, 2018 Mar 14.
Artículo en Inglés | MEDLINE | ID: mdl-29540197

RESUMEN

BACKGROUND: Human-immune-system humanized mouse models can bridge the gap between humans and conventional mice for testing human vaccines. The HLA-expressing humanized DRAGA (HLA-A2.HLA-DR4.Rag1KO.IL2RγcKO.NOD) mice reconstitute a functional human-immune-system and sustain the complete life cycle of Plasmodium falciparum. Herein, the DRAGA mice were investigated for immune responses following immunization with live P. falciparum sporozoites under chloroquine chemoprophylaxis (CPS-CQ), an immunization approach that showed in human trials to confer pre-erythrocytic immunity. RESULTS: The CPS-CQ immunized DRAGA mice (i) elicited human CD4 and CD8 T cell responses to antigens expressed by P. falciparum sporozoites (Pfspz) and by the infected-red blood cells (iRBC). The Pfspz-specific human T cell responses were found to be systemic (spleen and liver), whereas the iRBCs-specific human T cell responses were more localized to the liver, (ii) elicited stronger antibody responses to the Pfspz than to the iRBCs, and (iii) they were protected against challenge with infectious Pfspz but not against challenge with iRBCs. CONCLUSIONS: The DRAGA mice represent a new pre-clinical model to investigate the immunogenicity and protective efficacy of P. falciparum malaria vaccine candidates.


Asunto(s)
Anticuerpos Antiprotozoarios/sangre , Cloroquina/uso terapéutico , Vacunas contra la Malaria/inmunología , Malaria Falciparum/prevención & control , Plasmodium falciparum/inmunología , Esporozoítos/inmunología , Animales , Formación de Anticuerpos , Antimaláricos/uso terapéutico , Humanos , Ratones , Ratones Transgénicos
10.
Proc Natl Acad Sci U S A ; 112(33): 10437-42, 2015 Aug 18.
Artículo en Inglés | MEDLINE | ID: mdl-26216948

RESUMEN

T-cell genome engineering holds great promise for cell-based therapies for cancer, HIV, primary immune deficiencies, and autoimmune diseases, but genetic manipulation of human T cells has been challenging. Improved tools are needed to efficiently "knock out" genes and "knock in" targeted genome modifications to modulate T-cell function and correct disease-associated mutations. CRISPR/Cas9 technology is facilitating genome engineering in many cell types, but in human T cells its efficiency has been limited and it has not yet proven useful for targeted nucleotide replacements. Here we report efficient genome engineering in human CD4(+) T cells using Cas9:single-guide RNA ribonucleoproteins (Cas9 RNPs). Cas9 RNPs allowed ablation of CXCR4, a coreceptor for HIV entry. Cas9 RNP electroporation caused up to ∼40% of cells to lose high-level cell-surface expression of CXCR4, and edited cells could be enriched by sorting based on low CXCR4 expression. Importantly, Cas9 RNPs paired with homology-directed repair template oligonucleotides generated a high frequency of targeted genome modifications in primary T cells. Targeted nucleotide replacement was achieved in CXCR4 and PD-1 (PDCD1), a regulator of T-cell exhaustion that is a validated target for tumor immunotherapy. Deep sequencing of a target site confirmed that Cas9 RNPs generated knock-in genome modifications with up to ∼20% efficiency, which accounted for up to approximately one-third of total editing events. These results establish Cas9 RNP technology for diverse experimental and therapeutic genome engineering applications in primary human T cells.


Asunto(s)
Proteínas Bacterianas/genética , Endonucleasas/genética , Ribonucleoproteínas/genética , Linfocitos T/citología , Proteínas Bacterianas/química , Linfocitos T CD4-Positivos/citología , Línea Celular , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas , Electroporación , Endonucleasas/química , Técnicas de Sustitución del Gen , Ingeniería Genética/métodos , Genoma , Secuenciación de Nucleótidos de Alto Rendimiento , Humanos , Leucocitos Mononucleares/citología , Receptores CXCR4/metabolismo , Ribonucleoproteínas/química
11.
Eur J Immunol ; 46(3): 560-9, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26635029

RESUMEN

In the thymus, a T-cell repertoire able to confer protection against infectious and noninfectious agents in a peptide-dependent, self-MHC-restricted manner is selected. Direct detection of Ag-specific thymocytes, and analysis of the impact of the expression of the MHC-restricting allele on their frequency or function has never been studied in humans because of the extremely low precursor frequency. Here, we used a tetramer-based enrichment protocol to analyze the ex vivo frequency and activation-phenotype of human thymocytes specific for self, viral and tumor-antigens presented by HLA-A*0201 (A2) in individuals expressing or not this allele. Ag-specific thymocytes were quantified within both CD4CD8 double or single-positive compartments in every donor. Our data indicate that the maturation efficiency of Ag-specific thymocytes is poorly affected by HLA-A2 expression, in terms of frequencies. Nevertheless, A2-restricted T-cell lines from A2(+) donors reacted to A2(+) cell lines in a highly peptide-specific fashion, whereas their alloreactive counterparts showed off-target activity. This first ex vivo analysis of human antigen-specific thymocytes at different stages of human T-cell development should open new perspectives in the understanding of the human thymic selection process.


Asunto(s)
Antígenos de Neoplasias/inmunología , Antígenos Virales/inmunología , Autoantígenos/inmunología , Epítopos , Antígeno HLA-A2/inmunología , Linfocitos T/inmunología , Timocitos/fisiología , Presentación de Antígeno , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Línea Celular , Antígeno HLA-A2/genética , Humanos , Péptidos/inmunología , Timocitos/inmunología , Timo/citología , Timo/inmunología
12.
Transpl Int ; 30(10): 1061-1074, 2017 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-28543637

RESUMEN

The PI3K/mTOR signaling cascade is fundamental in T-cell activation and fate decisions. We showed the distinct regulation of PI3K/mTOR in regulatory and effector T-cells and proposed the potential therapeutic benefit of targeting this pathway to control the balance between effector and regulatory T-cell activities. Substantial adverse effects in long-term clinical usage of rapamycin suggest the use of alternative treatments in restraining effector T-cell function in transplant patients. We hypothesize that dual PI3K/mTOR inhibitors may represent an immunosuppressant alternative. Here we show that dual PI3K/mTOR PI-103 and PKI-587 inhibitors interfered IL-2-dependent responses in T-cells. However, in contrast to the inhibitory effects in non-Treg T-cell proliferation and effector functions, dual inhibitors increased the differentiation, preferential expansion, and suppressor activity of iTregs. Rapamycin, PI-103, and PKI-587 targeted different signaling events and induced different metabolic patterns in primary T-cells. Similar to rapamycin, in vivo administration of PI-103 and PKI-587 controlled effectively the immunological response against allogeneic skin graft. These results characterize specific regulatory mechanisms of dual PI3K/mTOR inhibitors in T-cells and support their potential as a novel therapeutic option in transplantation.


Asunto(s)
Furanos/farmacología , Morfolinas/farmacología , Piridinas/farmacología , Pirimidinas/farmacología , Linfocitos T/efectos de los fármacos , Inmunología del Trasplante , Triazinas/farmacología , Animales , Evaluación Preclínica de Medicamentos , Humanos , Interleucina-2/metabolismo , Ratones , Inhibidores de las Quinasa Fosfoinosítidos-3 , Sirolimus , Serina-Treonina Quinasas TOR/antagonistas & inhibidores
13.
Proc Natl Acad Sci U S A ; 111(47): 16814-9, 2014 Nov 25.
Artículo en Inglés | MEDLINE | ID: mdl-25385601

RESUMEN

Rho-associated kinase 2 (ROCK2) regulates the secretion of proinflammatory cytokines and the development of autoimmunity in mice. Data from a phase 1 clinical trial demonstrate that oral administration of KD025, a selective ROCK2 inhibitor, to healthy human subjects down-regulates the ability of T cells to secrete IL-21 and IL-17 by 90% and 60%, respectively, but not IFN-γ in response to T-cell receptor stimulation in vitro. Pharmacological inhibition with KD025 or siRNA-mediated inhibition of ROCK2, but not ROCK1, significantly diminished STAT3 phosphorylation and binding to IL-17 and IL-21 promoters and reduced IFN regulatory factor 4 and nuclear hormone RAR-related orphan receptor γt protein levels in T cells derived from healthy subjects or rheumatoid arthritis patients. Simultaneously, treatment with KD025 also promotes the suppressive function of regulatory T cells through up-regulation of STAT5 phosphorylation and positive regulation of forkhead box p3 expression. The administration of KD025 in vivo down-regulates the progression of collagen-induced arthritis in mice via targeting of the Th17-mediated pathway. Thus, ROCK2 signaling appears to be instrumental in regulating the balance between proinflammatory and regulatory T-cell subsets. Targeting of ROCK2 in man may therefore restore disrupted immune homeostasis and have a role in the treatment of autoimmunity.


Asunto(s)
Linfocitos T CD4-Positivos/efectos de los fármacos , Interleucina-17/metabolismo , Interleucinas/metabolismo , Inhibidores de Proteínas Quinasas/farmacología , Factor de Transcripción STAT3/fisiología , Quinasas Asociadas a rho/antagonistas & inhibidores , Administración Oral , Linfocitos T CD4-Positivos/metabolismo , Células Cultivadas , Humanos , Interleucina-17/genética , Interleucinas/genética , Fosforilación , Regiones Promotoras Genéticas , Inhibidores de Proteínas Quinasas/administración & dosificación , Factor de Transcripción STAT3/metabolismo , Transcripción Genética
14.
Int J Mol Sci ; 18(9)2017 Sep 08.
Artículo en Inglés | MEDLINE | ID: mdl-28885562

RESUMEN

Breast cancer is a common malignancy among women. The innate and adaptive immune responses failed to be activated owing to immune modulation in the tumour microenvironment. Decades of scientific study links the overexpression of human epidermal growth factor receptor 2 (ERBB2) antigen with aggressive tumours. The Chimeric Antigen Receptor (CAR) coding for specific tumour-associated antigens could initiate intrinsic T-cell signalling, inducing T-cell activation, and cytotoxic activity without the need for major histocompatibility complex recognition. This renders CAR as a potentially universal immunotherapeutic option. Herein, we aimed to establish CAR in CD3+ T-cells, isolated from human peripheral blood mononucleated cells that could subsequently target and induce apoptosis in the ERBB2 overexpressing human breast cancer cell line, SKBR3. Constructed CAR was inserted into a lentiviral plasmid containing a green fluorescent protein tag and produced as lentiviral particles that were used to transduce activated T-cells. Transduced CAR-T cells were then primed with SKBR3 cells to evaluate their functionality. Results showed increased apoptosis in SKBR3 cells co-cultured with CAR-T cells compared to the control (non-transduced T-cells). This study demonstrates that CAR introduction helps overcome the innate limitations of native T-cells leading to cancer cell apoptosis. We recommend future studies should focus on in vivo cytotoxicity of CAR-T cells against ERBB2 expressing tumours.


Asunto(s)
Neoplasias de la Mama/inmunología , Neoplasias de la Mama/metabolismo , Inmunoterapia Adoptiva , Receptor ErbB-2/metabolismo , Receptores de Antígenos de Linfocitos T/metabolismo , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Apoptosis/genética , Apoptosis/inmunología , Biomarcadores , Neoplasias de la Mama/patología , Neoplasias de la Mama/terapia , Línea Celular Tumoral , Técnicas de Cocultivo , Citocinas/biosíntesis , Femenino , Expresión Génica , Vectores Genéticos/genética , Humanos , Inmunofenotipificación , Inmunoterapia Adoptiva/métodos , Interferón gamma/metabolismo , Lentivirus/genética , Activación de Linfocitos/genética , Activación de Linfocitos/inmunología , Receptor ErbB-2/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos T/genética , Proteínas Recombinantes de Fusión , Anticuerpos de Cadena Única/genética , Anticuerpos de Cadena Única/inmunología , Anticuerpos de Cadena Única/metabolismo , Transducción Genética
15.
Eur J Immunol ; 45(9): 2480-3, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26256443

RESUMEN

Cellular metabolism is emerging as a key determinant of T-lymphocyte differentiation and function. While this new paradigm has been primarily characterized in murine systems, research is now characterizing a role for different aspects of cellular metabolism in controlling human T-lymphocyte biology. In this issue of the European Journal of Immunology, Renner et al. [Eur. J. Immunol. 2015. 45: 2504-2516] analyze the glycolytic and mitochondrial activity of activated human CD4(+) and CD8(+) T cells, and correlate it to T-cell function. The authors show that although neither glucose deprivation nor mitochondrial restriction affects cytokine production, the glycolytic inhibitor 2-deoxyglucose severely affects T-cell function.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/metabolismo , Desoxiglucosa/farmacología , Glucosa/deficiencia , Mitocondrias/metabolismo , Subgrupos de Linfocitos T/metabolismo , Humanos
16.
J Infect Dis ; 212(5): 830-8, 2015 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-25737563

RESUMEN

INTRODUCTION: Though Staphylococcus aureus is a major pathogen, vaccine trials have failed. In contrast, class-switched antibodies specific to S. aureus are common, implying immune memory formation and suggesting a large pool of S. aureus-reactive helper T-cells. OBJECTIVE: To elucidate the cellular arm of S. aureus-specific immune memory, the T-cell response in humans was characterized. METHODS: The proliferative response of human peripheral blood mononuclear cells (PBMCs) to S. aureus antigens and the frequency of S. aureus-specific T-cells were quantified by (3)H-thymidine incorporation; cytokine release was measured by flow cytometry. RESULTS: Staphylococcus aureus particles and extracellular proteins elicited pronounced proliferation in PBMCs of healthy adults. This reflected a memory response with high frequencies of T-cells being activated by single S. aureus antigens. The whole S. aureus-specific T-cell pool was estimated to comprise 3.6% of T-cells with 35-fold differences between individuals (range, 0.2%-5.7%). When exposed to S. aureus antigens, the T-cells released predominantly but not solely T helper (Th)1/Th17 cytokines. CONCLUSIONS: The large number of S. aureus antigen-reactive memory T-lymphocytes is likely to influence the course of S. aureus infection. To enable rational vaccine design, the naturally acquired human T-cell memory needs to be explored at high priority.


Asunto(s)
Memoria Inmunológica , Staphylococcus aureus/inmunología , Linfocitos T/inmunología , Adulto , Antígenos Bacterianos/inmunología , Proliferación Celular , Citocinas/metabolismo , Citometría de Flujo , Humanos , Marcaje Isotópico , Leucocitos Mononucleares/inmunología
17.
Clin Exp Immunol ; 182(2): 162-72, 2015 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-26212126

RESUMEN

Multiple sclerosis (MS) is an inflammatory, demyelinating and neurodegenerative disease triggered by infiltration of activated T cells into the central nervous system. Interferon (IFN)-ß is an established, safe and effective treatment for patients with relapsing-remitting MS (RRMS). The cytokine can inhibit leucocyte infiltration into the central nervous system; however, little is known about the precise molecular mechanisms. Previously, in vitro application of IFN-ß1b was shown to reduce CXCL12/CXCR4-mediated monocyte migration. Here, we analysed the effects of IFN-ß1b on CXCR4-dependent T cell function. In vitro exposure to IFN-ß1b (1000 U/ml) for 20 h reduced CXCR4-dependent chemotaxis of primary human T cells from healthy individuals and patients with RRMS. Investigating the IFN-ß1b/CXCR4 signalling pathways, we found no difference in phosphorylation of ZAP70, ERK1/2 and AKT despite an early induction of the negative regulator of G-protein signalling, RGS1 by IFN-ß1b. However, CXCR4 surface expression was reduced. Quantitative real time-PCR revealed a similar reduction in CXCR4-mRNA, and the requirement of several hours' exposure to IFN-ß1b supports a transcriptional regulation. Interestingly, T cells from MS patients showed a lower CXCR4 expression than T cells from healthy controls, which was not reduced further in patients under IFN-ß1b therapy. Furthermore, we observed no change in CXCL12-dependent chemotaxis in RRMS patients. Our results demonstrate clearly that IFN-ß1b can impair the functional response to CXCR4 by down-regulating its expression, but also points to the complex in vivo effects of IFN-ß1b therapy.


Asunto(s)
Quimiotaxis/efectos de los fármacos , Interferon beta-1b/farmacología , Receptores CXCR4/metabolismo , Linfocitos T/efectos de los fármacos , Adulto , Western Blotting , Movimiento Celular/efectos de los fármacos , Células Cultivadas , Femenino , Expresión Génica/efectos de los fármacos , Expresión Génica/inmunología , Humanos , Masculino , Persona de Mediana Edad , Esclerosis Múltiple Recurrente-Remitente/genética , Esclerosis Múltiple Recurrente-Remitente/inmunología , Receptores CXCR4/genética , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Linfocitos T/inmunología , Linfocitos T/metabolismo , Factores de Tiempo , Adulto Joven
18.
Cytokine ; 72(2): 224-5, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25649044

RESUMEN

Balanced regulation of cytokine secretion in T cells is critical for maintenance of immune homeostasis and prevention of autoimmunity. The Rho-associated kinase (ROCK) 2 signaling pathway was previously shown to be involved in controlling of cellular movement and shape. However, recent work from our group and others has demonstrated a new and important role of ROCK2 in regulating cytokine secretion in T cells. We found that ROCK2 promotes pro-inflammatory cytokines such as IL-17 and IL-21, whereas IL-2 and IL-10 secretion are negatively regulated by ROCK2 under Th17-skewing activation. Also, in disease, but not in steady state conditions, ROCK2 contributes to regulation of IFN-γ secretion in T cells from rheumatoid arthritis patients. Thus, ROCK2 signaling is a key pathway in modulation of T-cell mediated immune responses underscoring the therapeutic potential of targeted inhibition of ROCK2 in autoimmunity.


Asunto(s)
Citocinas/metabolismo , Linfocitos T/inmunología , Quinasas Asociadas a rho/metabolismo , Enfermedades Autoinmunes/inmunología , Autoinmunidad , Citocinas/genética , Humanos , Interferón gamma/inmunología , Interferón gamma/metabolismo , Interleucina-17/genética , Interleucina-17/inmunología , Interleucinas/genética , Interleucinas/inmunología , Interleucinas/metabolismo , Transducción de Señal , Linfocitos T/metabolismo , Balance Th1 - Th2 , Quinasas Asociadas a rho/genética
19.
J Appl Toxicol ; 35(5): 459-65, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25056757

RESUMEN

Perfluorinated compounds, such as perfluorooctane sulfonate (PFOS) and perfluorooctanoic acid (PFOA), have been shown to alter various immune functions suggesting they are immunotoxic. This study assessed the effects of PFOS and PFOA on interleukin (IL)-2 production in the human Jurkat T-cell line and PFOS in healthy human primary T cells. Jurkat cells were stimulated with phytohemagglutinin (PHA)/phorbol myristate acetate (PMA), anti CD-3/anti CD-28, or anti CD-3, and dosed with 0, 0.05, 0.1, 0.5, 1, 5, 10, 50, 75, or 100 µg ml(-1) PFOS or 0, 0.005, 0.01, 0.05, 0.1, 0.5, 1, 5, or 10 µg ml(-1) PFOA. Jurkat cells stimulated with PHA/PMA or anti CD-3 exhibited decreased IL-2 production beginning at 50 µg PFOS ml(-1) and 5 µg PFOS ml(-1) respectively, but stimulation with anti-CD3/anti-CD28 resulted in no changes compared with the control. Addition of the PPAR-alpha antagonist GW6471 to PFOS-dosed cells stimulated with PHA/PMA resulted in decreases in IL-2 production starting at 50 µg PFOS ml(-1), which suggests PFOS affected T-cell IL-2 production via PPAR-alpha-independent mechanisms. Exposure to PFOA, PFOA + GW6471, or PFOS + PFOA in Jurkat cells resulted in no significant differences in IL-2 production. In vitro dosing studies using healthy primary human CD4+ T cells were consistent with the Jurkat results. These data demonstrated that PFOA did not impact IL-2 production, but PFOS suppressed IL-2 production in both a human cell line and human primary cells at dose levels within the high end of the human exposure range. A decrease in IL-2 production is characteristic of autoimmune diseases such as systemic lupus erythematosus and should be further investigated.


Asunto(s)
Ácidos Alcanesulfónicos/toxicidad , Linfocitos T CD4-Positivos/efectos de los fármacos , Caprilatos/toxicidad , Fluorocarburos/toxicidad , Interleucina-2/metabolismo , Linfocitos T CD4-Positivos/metabolismo , Supervivencia Celular/efectos de los fármacos , Femenino , Humanos , Células Jurkat , Masculino , PPAR alfa/metabolismo , Fitohemaglutininas , Acetato de Tetradecanoilforbol
20.
Immunology ; 2014 Sep 22.
Artículo en Inglés | MEDLINE | ID: mdl-25251258

RESUMEN

The protein CD46 protects cells from complement attack by regulating cleavage of C3b and C3d. CD46 also regulates the adaptive immune response by controlling T cell activation and differentiation. Co-engagement of the T cell receptor and CD46 notably drives T cell differentiation by switching production of IFNγ to secretion of anti-inflammatory IL-10. This regulatory pathway is altered in several chronic inflammatory diseases highlighting its key role for immune homeostasis. The manipulation of the CD46 pathway may therefore provide a powerful means to regulate immune responses. Herein, we investigated the effect of recombinant proteins derived from the fiber knob of the adenovirus serotype 35 (Ad35) that uses CD46 as its entry receptor, on human T cell activation. We compared the effects of Ad35K++, engineered to exhibit enhanced affinity to CD46, and of Ad35K-, mutated in the binding site for CD46. Ad35K++ profoundly affects T cell activation by decreasing the levels of CD46 at the surface of primary T cells, and impairing T cell co-activation, shown by decreased CD25 expression, reduced proliferation and lower secretion of IL-10 and IFNγ. In contrast, Ad35K- acts a potent coactivator of T cells, enhancing T cell proliferation and cytokine production. These data show that recombinant Ad35 proteins are potent modulators of human T cell activation, and support their further development as potential drugs targeting T cell responses. This article is protected by copyright. All rights reserved.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA