Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
J Immunother Cancer ; 11(4)2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-37072346

RESUMEN

BACKGROUND: The wider application of T cells targeting viral tumor-antigens via their native receptors is hampered by the failure to expand potent tumor-specific T cells from patients. Here, we examine reasons for and solutions to this failure, taking as our model the preparation of Epstein-Barr virus (EBV)-specific T cells (EBVSTs) for the treatment of EBV-positive lymphoma. EBVSTs could not be manufactured from almost one-third of patients, either because they failed to expand, or they expanded, but lacked EBV specificity. We identified an underlying cause of this problem and established a clinically feasible approach to overcome it. METHODS: CD45RO+CD45RA- memory compartment residing antigen-specific T cells were enriched by depleting CD45RA positive (+) peripheral blood mononuclear cells (PBMCs) that include naïve T cells, among other subsets, prior to EBV antigen stimulation. We then compared the phenotype, specificity, function and T-cell receptor (TCR) Vß repertoire of EBVSTs expanded from unfractionated whole (W)-PBMCs and CD45RA-depleted (RAD)-PBMCs on day 16. To identify the CD45RA component that inhibited EBVST outgrowth, isolated CD45RA+ subsets were added back to RAD-PBMCs followed by expansion and characterization. The in vivo potency of W-EBVSTs and RAD-EBVSTs was compared in a murine xenograft model of autologous EBV+ lymphoma. RESULTS: Depletion of CD45RA+ PBMCs before antigen stimulation increased EBVST expansion, antigen-specificity and potency in vitro and in vivo. TCR sequencing revealed a selective outgrowth in RAD-EBVSTs of clonotypes that expanded poorly in W-EBVSTs. Inhibition of antigen-stimulated T cells by CD45RA+ PBMCs could be reproduced only by the naïve T-cell fraction, while CD45RA+ regulatory T cells, natural killer cells, stem cell memory and effector memory subsets lacked inhibitory activity. Crucially, CD45RA depletion of PBMCs from patients with lymphoma enabled the outgrowth of EBVSTs that failed to expand from W-PBMCs. This enhanced specificity extended to T cells specific for other viruses. CONCLUSION: Our findings suggest that naïve T cells inhibit the outgrowth of antigen-stimulated memory T cells, highlighting the profound effects of intra-T-cell subset interactions. Having overcome our inability to generate EBVSTs from many patients with lymphoma, we have introduced CD45RA depletion into three clinical trials: NCT01555892 and NCT04288726 using autologous and allogeneic EBVSTs to treat lymphoma and NCT04013802 using multivirus-specific T cells to treat viral infections after hematopoietic stem cell transplantation.


Asunto(s)
Herpesvirus Humano 4 , Células de Memoria Inmunológica , Inmunoterapia , Linfoma , Linfocitos T , Linfocitos T/inmunología , Humanos , Linfoma/inmunología , Linfoma/terapia , Antígenos Comunes de Leucocito , Células de Memoria Inmunológica/inmunología , Leucocitos Mononucleares/inmunología , Células Asesinas Naturales/inmunología , Inmunoterapia/métodos , Inmunofenotipificación , Femenino , Animales , Ratones , Xenoinjertos , Trasplante de Neoplasias
2.
Front Immunol ; 14: 1043631, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-36865556

RESUMEN

Effective secondary response to antigen is a hallmark of immunological memory. However, the extent of memory CD8 T cell response to secondary boost varies at different times after a primary response. Considering the central role of memory CD8 T cells in long-lived protection against viral infections and tumors, a better understanding of the molecular mechanisms underlying the changing responsiveness of these cells to antigenic challenge would be beneficial. We examined here primed CD8 T cell response to boost in a BALB/c mouse model of intramuscular vaccination by priming with HIV-1 gag-encoding Chimpanzee adenovector, and boosting with HIV-1 gag-encoding Modified Vaccinia virus Ankara. We found that boost was more effective at day(d)100 than at d30 post-prime, as evaluated at d45 post-boost by multi-lymphoid organ assessment of gag-specific CD8 T cell frequency, CD62L-expression (as a guide to memory status) and in vivo killing. RNA-sequencing of splenic gag-primed CD8 T cells at d100 revealed a quiescent, but highly responsive signature, that trended toward a central memory (CD62L+) phenotype. Interestingly, gag-specific CD8 T cell frequency selectively diminished in the blood at d100, relative to the spleen, lymph nodes and bone marrow. These results open the possibility to modify prime/boost intervals to achieve an improved memory CD8 T cell secondary response.


Asunto(s)
Linfocitos T CD8-positivos , Inmunización Secundaria , Células de Memoria Inmunológica , Vacunas , Animales , Ratones , Linfocitos T CD8-positivos/inmunología , División Celular , Ratones Endogámicos BALB C , Vacunación , Células de Memoria Inmunológica/inmunología
3.
Mol Immunol ; 156: 111-126, 2023 04.
Artículo en Inglés | MEDLINE | ID: mdl-36921486

RESUMEN

COVID-19 is a severe respiratory illness that has emerged as a devasting health problem worldwide. The disease outcome is heterogeneous, which is most likely dependent on the immunity of an individual. Asymptomatic and mildly/moderate symptomatic (non-severe) patients likely develop an effective early immune response and clear the virus. However, severe symptoms dominate due to a failure in the generation of an effective and specific early immune response against SARS-CoV-2. Moreover, a late surge in pathogenic inflammation involves dysregulated innate and adaptive immune responses leading to local and systemic tissue damage and the emergence of severe disease symptoms. In this review, we describe the potential mechanisms of protective and pathogenic immune responses in "mild/moderate" and "severe" symptomatic SARS-CoV-2 infected people, respectively, and discuss the immune components that are currently targeted for therapeutic intervention.


Asunto(s)
COVID-19 , SARS-CoV-2 , SARS-CoV-2/inmunología , COVID-19/genética , COVID-19/inmunología , Humanos , Inmunidad Innata , Inmunidad Adaptativa , Predisposición Genética a la Enfermedad , Células de Memoria Inmunológica , Tratamiento Farmacológico de COVID-19 , Vacuna BCG/inmunología
4.
Scand J Immunol ; 97(5): e13262, 2023 May.
Artículo en Inglés | MEDLINE | ID: mdl-36853017

RESUMEN

Reservoirs of HIV remain a major obstacle to the complete eradication of virus despite regular anti-retroviral therapy (ART). Memory stem cells (Tscm), one of the major reservoirs, are relatively less studied owing to their presence in lower numbers and inaccessible anatomical locations. We have evaluated the molecular characteristics of Tscms in patients with ART interruption (n = 15) versus patients on uninterrupted ART (n = 12) using flow cytometry. RNA sequencing was done in the sorted Tscms to study the differential gene expression. Patients with ART interruption had significantly lower baseline CD4+T-cell counts and high viral loads as compared to patients on ART. The former group had significantly higher frequency of CD4+ and CD8+Tscms with a higher expression of PD-1 on CD8+Tscms. The transcriptome profile of Tscm was significantly different among the patient groups. The main pathways were cellular and metabolic pathways, cellular development pathways, cell differentiation and negative regulation of cellular migratory pathways. An increased yet dysfunctional CD8+ memory stem cells describe HIV-1-infected patients with break-in ART and a distinct transcriptional signature of CD4+ Tscm as compared to those of patients on ART. A more detailed understanding of the biology and dynamics of Tscm in future studies is warranted. Strategies to improve the functionality of the CD8+ Tscm will help these patients to tackle the outburst of viral replication that occurs after the cessation of therapy.


Asunto(s)
Antirretrovirales , Infecciones por VIH , Células de Memoria Inmunológica , Células Madre , Interrupción del Tratamiento , Adulto , Femenino , Humanos , Masculino , Antirretrovirales/uso terapéutico , Linfocitos T CD4-Positivos/patología , Linfocitos T CD4-Positivos/virología , Infecciones por VIH/dietoterapia , Infecciones por VIH/virología , Células de Memoria Inmunológica/virología , Células Madre/virología , Análisis de Secuencia de ARN
5.
Front Immunol ; 13: 958616, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35983034

RESUMEN

Upon antigen recognition by the T cell receptor (TCR), a complex signaling network orchestrated by protein-tyrosine kinases (PTKs) and protein-tyrosine phosphatases (PTPs) regulates the transmission of the extracellular signal to the nucleus. The role of the PTPs Src-homology 2 (SH2) domain-containing phosphatase 1 (SHP1, Ptpn6) and Src-homology 2 (SH2) domain-containing phosphatase 2 (SHP2, Ptpn11) have been studied in various cell types including T cells. Whereas SHP1 acts as an essential negative regulator of the proximal steps in T cell signalling, the role of SHP2 in T cell activation is still a matter of debate. Here, we analyzed the role of the constitutively active SHP2-D61Y-mutant in T cell activation using knock-in mice expressing the mutant form Ptpn11D61Y in T cells. We observed reduced numbers of CD8+ and increased numbers of CD4+ T cells in the bone marrow and spleen of young and aged SHP2-D61Y-mutant mice as well as in Influenza A Virus (IAV)-infected mice compared to controls. In addition, we found elevated frequencies of effector memory CD8+ T cells and an upregulation of the programmed cell death protein 1 (PD-1)-receptor on both CD4+ and CD8+ T cells. Functional analysis of SHP2-D61Y-mutated T cells revealed an induction of late apoptosis/necrosis, a reduced proliferation and altered signaling upon TCR stimulation. However, the ability of D61Y-mutant mice to clear viral infection was not affected. In conclusion, our data indicate an important regulatory role of SHP2 in T cell function, where the effect is determined by the kinetics of SHP2 phosphatase activity and differs in the presence of the permanently active and the temporally regulated phosphatase. Due to interaction of SHP2 with the PD-1-receptor targeting the protein-tyrosine phosphatase might be a valuable tool to enhance T cell activities in immunotherapy.


Asunto(s)
Linfocitos T CD4-Positivos , Linfocitos T CD8-positivos , Receptor de Muerte Celular Programada 1 , Proteína Tirosina Fosfatasa no Receptora Tipo 11 , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Células de Memoria Inmunológica , Activación de Linfocitos , Ratones , Proteína Tirosina Fosfatasa no Receptora Tipo 11/metabolismo , Receptores de Antígenos de Linfocitos T/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Dominios Homologos src
6.
Proc Natl Acad Sci U S A ; 119(25): e2122379119, 2022 06 21.
Artículo en Inglés | MEDLINE | ID: mdl-35696582

RESUMEN

Acute myeloid leukemia (AML) remains a therapeutic challenge, and a paucity of tumor-specific targets has significantly hampered the development of effective immune-based therapies. Recent paradigm-changing studies have shown that natural killer (NK) cells exhibit innate memory upon brief activation with IL-12 and IL-18, leading to cytokine-induced memory-like (CIML) NK cell differentiation. CIML NK cells have enhanced antitumor activity and have shown promising results in early phase clinical trials in patients with relapsed/refractory AML. Here, we show that arming CIML NK cells with a neoepitope-specific chimeric antigen receptor (CAR) significantly enhances their antitumor responses to nucleophosphmin-1 (NPM1)-mutated AML while avoiding off-target toxicity. CIML NK cells differentiated from peripheral blood NK cells were efficiently transduced to express a TCR-like CAR that specifically recognizes a neoepitope derived from the cytosolic oncogenic NPM1-mutated protein presented by HLA-A2. These CAR CIML NK cells displayed enhanced activity against NPM1-mutated AML cell lines and patient-derived leukemic blast cells. CAR CIML NK cells persisted in vivo and significantly improved AML outcomes in xenograft models. Single-cell RNA sequencing and mass cytometry analyses identified up-regulation of cell proliferation, protein folding, immune responses, and major metabolic pathways in CAR-transduced CIML NK cells, resulting in tumor-specific, CAR-dependent activation and function in response to AML target cells. Thus, efficient arming of CIML NK cells with an NPM1-mutation-specific TCR-like CAR substantially improves their innate antitumor responses against an otherwise intracellular mutant protein. These preclinical findings justify evaluating this approach in clinical trials in HLA-A2+ AML patients with NPM1c mutations.


Asunto(s)
Memoria Inmunológica , Células de Memoria Inmunológica , Inmunoterapia Adoptiva , Células Asesinas Naturales , Leucemia Mieloide Aguda , Nucleofosmina , Receptores Quiméricos de Antígenos , Antígeno HLA-A2/inmunología , Humanos , Células de Memoria Inmunológica/inmunología , Células de Memoria Inmunológica/trasplante , Inmunoterapia Adoptiva/métodos , Células Asesinas Naturales/inmunología , Células Asesinas Naturales/trasplante , Leucemia Mieloide Aguda/genética , Leucemia Mieloide Aguda/terapia , Mutación , Nucleofosmina/genética , Nucleofosmina/inmunología , Receptores Quiméricos de Antígenos/genética , Receptores Quiméricos de Antígenos/inmunología
7.
J Ovarian Res ; 15(1): 29, 2022 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-35227285

RESUMEN

BACKGROUND: Ovarian cancer (OC) is one of the most common and lethal malignant tumors worldwide and the prognosis of OC remains unsatisfactory. Transcription factors (TFs) are demonstrated to be associated with the clinical outcome of many types of cancers, yet their roles in the prognostic prediction and gene regulatory network in patients with OC need to be further investigated. METHODS: TFs from GEO datasets were collected and analyzed. Differential expression analysis, WGCNA and Cox-LASSO regression model were used to identify the hub-TFs and a prognostic signature based on these TFs was constructed and validated. Moreover, tumor-infiltrating immune cells were analyzed, and a nomogram containing age, histology, FIGO_stage and TFs-based signature were established. Potential biological functions, pathways and the gene regulatory network of TFs in signature was also explored. RESULTS: In this study, 6 TFs significantly associated with the prognosis of OC were identified. These TFs were used to build up a TFs-based signature for predicting the survival of patients with OC. Patients with OC in training and testing datasets were divided into high-risk and low-risk groups, according to the median value of risk scores determined by the signature. The two groups were further used to validate the performance of the signature, and the results showed the TFs-based signature had effective prediction ability. Immune infiltrating analysis was conducted and abundance of B cells naïve, T cells CD4 memory resting, Macrophages M2 and Mast cells activated were significantly higher in high-risk group. A nomogram based on the signature was established and illustrated good predictive efficiencies for 1, 2, and 3-year overall survival. Furthermore, the construction of the TFs-target gene regulatory network revealed the potential mechanisms of TFs in OC. CONCLUSIONS: To our best knowledge, it is for the first time to develop a prognostic signature based on TFs in OC. The TFs-based signature is proven to be effective in predicting the survival of patients with OC. Our study may facilitate the clinical decision-making for patients with OC and help to elucidate the underlying mechanism of TFs in OC.


Asunto(s)
Nomogramas , Neoplasias Ováricas , Factores de Transcripción , Femenino , Humanos , Factores de Edad , Linfocitos B , Factores de Transcripción Básicos con Cremalleras de Leucinas y Motivos Hélice-Asa-Hélice/genética , Linfocitos T CD4-Positivos , Bases de Datos Genéticas , Proteínas de Unión al ADN/genética , Factores de Transcripción del Choque Térmico/genética , Células de Memoria Inmunológica , Linfocitos Infiltrantes de Tumor , Macrófagos , Mastocitos , Estadificación de Neoplasias , Neoplasias Ováricas/genética , Neoplasias Ováricas/patología , Pronóstico , Curva ROC , Factores de Transcripción de la Familia Snail/genética , Tasa de Supervivencia , Factores de Transcripción/genética
8.
mBio ; 12(5): e0244721, 2021 10 26.
Artículo en Inglés | MEDLINE | ID: mdl-34544282

RESUMEN

Future HIV-1 curative therapies require a thorough understanding of the distribution of genetically-intact HIV-1 within T-cell subsets during antiretroviral therapy (ART) and the cellular mechanisms that maintain this reservoir. Therefore, we sequenced near-full-length HIV-1 genomes and identified genetically-intact and genetically-defective genomes from resting naive, stem-cell memory, central memory, transitional memory, effector memory, and terminally-differentiated CD4+ T-cells with known cellular half-lives from 11 participants on ART. We find that a higher infection frequency with any HIV-1 genome was significantly associated with a shorter cellular half-life, such as transitional and effector memory cells. A similar enrichment of genetically-intact provirus was observed in these cells with relatively shorter half-lives. We found that effector memory and terminally-differentiated cells also had significantly higher levels of expansions of genetically-identical sequences, while only transitional and effector memory cells contained genetically-intact proviruses that were part of a cluster of identical sequences. Expansions of identical sequences were used to infer cellular proliferation from clonal expansion. Altogether, this indicates that specific cellular mechanisms such as short half-life and proliferative potential contribute to the persistence of genetically-intact HIV-1. IMPORTANCE The design of future HIV-1 curative therapies requires a more thorough understanding of the distribution of genetically-intact HIV-1 within T-cell subsets as well as the cellular mechanisms that maintain this reservoir. These genetically-intact and presumably replication-competent proviruses make up the latent HIV-1 reservoir. Our investigations into the possible cellular mechanisms maintaining the HIV-1 reservoir in different T-cell subsets have revealed a link between the half-lives of T-cells and the level of proviruses they contain. Taken together, we believe our study shows that more differentiated and proliferative cells, such as transitional and effector memory T-cells, contain the highest levels of genetically-intact proviruses, and the rapid turnover rate of these cells contributes to the expansion of genetically-intact proviruses within them. Therefore, our study delivers an in-depth assessment of the cellular mechanisms, such as cellular proliferation and half-life, that contribute to and maintain the latent HIV-1 reservoir.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , Genoma Viral/inmunología , VIH-1/genética , VIH-1/inmunología , Células de Memoria Inmunológica , Latencia del Virus/inmunología , Adulto , Linfocitos T CD4-Positivos/fisiología , Reservorios de Enfermedades/virología , Infecciones por VIH/virología , Semivida , Humanos , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Provirus/genética
9.
Comput Math Methods Med ; 2021: 5802110, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-35003322

RESUMEN

BACKGROUND: The advance of new treatment strategies for more effective management of oral cancer requires identification of novel biological targets. Therefore, the purpose of this study is to identify novel biomarkers associated with oral tumorigenesis and prognostic signature by comparing gene expression profile of oral squamous cell carcinomas (OSCCs). METHODS: Four datasets including GSE25099, GSE30784, GSE37991, and GSE41613 were collected from Gene Expression Omnibus (GEO) database. Gene Ontology (GO) and the Kyoto Encyclopedia of Genes and Genomes (KEGG) analysis, Cox model analysis, identification of key genes, and Kaplan-Meier analysis were also performed. The xCell was utilized to analyze the infiltration levels of immune cells. RESULTS: A total of 235 differentially expressed genes (DEGs) were found to be dysregulated in OSCC. These genes were mainly enriched in ECM receptor interaction and focal adhesion. Cox regression analysis identified 10 genes considered as key genes. Kaplan-Meier analysis showed that low expression of SERPINE1 (also known as PAI-1), high expression of CD1C, and C-X3-C motif chemokine receptor 1 (CX3CR1) were associated with well prognostic status in OSCC patients. In addition, we constructed a 3-immune-cell signature (myeloid dendritic cell, T cell CD4+ central memory, and common myeloid progenitor) that may be used to predict the survival status of OSCC patients. CONCLUSION: Three key genes and 3-immune-cell signature were potential biomarkers for the prognosis of OSCC, and they may serve as potential targets for the treatment of OSCC patients.


Asunto(s)
Neoplasias de la Boca/genética , Neoplasias de la Boca/inmunología , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/inmunología , Biomarcadores de Tumor/genética , Biología Computacional , Bases de Datos Genéticas/estadística & datos numéricos , Regulación Neoplásica de la Expresión Génica , Ontología de Genes , Redes Reguladoras de Genes , Humanos , Células de Memoria Inmunológica/inmunología , Estimación de Kaplan-Meier , Pronóstico , Modelos de Riesgos Proporcionales , Transducción de Señal/genética , Transducción de Señal/inmunología , Transcriptoma , Microambiente Tumoral/genética , Microambiente Tumoral/inmunología
10.
J Infect Dis ; 223(7): 1183-1195, 2021 04 08.
Artículo en Inglés | MEDLINE | ID: mdl-32777077

RESUMEN

Direct acting antiviral therapies rapidly clear chronic hepatitis C virus (HCV) infection and restore natural killer (NK) cell function. We investigated NK-cell memory formation following HCV clearance by examining NK-cell phenotype and responses from control and chronic HCV patients before and after therapy following sustained virologic response at 12 weeks post therapy (SVR12). NK-cell phenotype at SVR12 differed significantly from paired pretreatment samples, with an increase in maturation markers CD16, CD57, and KLRG1. HCV patients possessed stronger cytotoxic responses against HCV-infected cells as compared to healthy controls; a response that further increased following SVR12. The antigen-specific response was mediated by KLRG1+ NK cells, as demonstrated by increased degranulation and proliferation in response to HCV antigen only. Our data suggest that KLRG1+ HCV-specific memory NK cells develop following viral infection, providing insight into their role in HCV clearance and relevance with regard to vaccine design.


Asunto(s)
Antivirales , Hepatitis C Crónica , Células de Memoria Inmunológica/efectos de los fármacos , Células Asesinas Naturales/efectos de los fármacos , Antivirales/uso terapéutico , Hepacivirus , Hepatitis C Crónica/tratamiento farmacológico , Humanos , Células de Memoria Inmunológica/virología , Interferones , Células Asesinas Naturales/virología , Lectinas Tipo C , Receptores Inmunológicos
11.
Ocul Immunol Inflamm ; 29(7-8): 1338-1344, 2021 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-32400241

RESUMEN

Purpose: Investigating the existence of inflammation in the distal outflow system posterior of the Schlemm's canal in primary open-angle glaucoma (POAG).Methods: Scleral biopsies (n = 62) from POAG-patients were taken during deep sclerectomy and fixed either in formalin or RNAlater®. Histologic (hematoxylin & eosin) and immunohistological staining for CD 3 and CD 45RO were performed.Results: Cellular infiltration of immunocompetent cells (CD 3 and CD 45RO positive cells) exists around collector channels (CC). This inflammation is limited to the area around the CCs. Ninety-two percent of the biopsies are positive for inflammation. Untreated, dysgenetic glaucoma eyes and 8% of POAG eyes were negative for inflammation. Neither the use of benzalkonium chloride nor the number and type of preoperative antiglaucomatous medication correlated to the immunohistological result.Conclusion: In POAG a diverse cellular infiltration exists around the CCs in the vast majority of biopsies. This could have major diagnostic and therapeutic consequences for the treatment of POAG.Abbreviations: POAG: primary open-angle glaucoma; CC: collector channel; AH: aqueous humor; TM: trabecular meshwork; SC: Schlemm's canal; HE: hematoxylin & eosin; APC: antigen-presenting cell.


Asunto(s)
Glaucoma de Ángulo Abierto/diagnóstico , Escleritis/diagnóstico , Malla Trabecular/patología , Anciano , Biopsia , Complejo CD3/metabolismo , Femenino , Humanos , Células de Memoria Inmunológica/metabolismo , Inflamación/diagnóstico , Antígenos Comunes de Leucocito/metabolismo , Masculino , Persona de Mediana Edad , Esclerostomía , Linfocitos T/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...