Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 140
Filtrar
1.
Front Immunol ; 13: 806906, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35154123

RESUMEN

Memory CD8+ T cells accumulate with aging, while the naïve T cell compartment decreases, leading to an increased susceptibility to infections and a decreased vaccine efficiency. To get deeper insights into the underlying mechanisms, this study aims to determine the age-dependent expression profile of total versus memory CD8+ T cells from young and old donors. Total CD8+ and CD8+CD45RA- memory T cells isolated from young (<30 years) and old (>60 years) donors were stimulated with anti-CD3 and anti-CD28 antibodies for 48h before analyzing the cytokine secretion and activation markers by flow cytometry and changes in the expression profiles using RNA sequencing. Gene ontology (GO) term enrichment analyses were performed for up-regulated and uniquely expressed transcripts identified in the T cell populations of both age groups. Total and memory CD8+ T cells from old donors expressed significantly higher CD25 levels and have an increased cytokine secretion. While approximately 1,500 up-regulated transcripts were identified in all groups, CD8+CD45RA- memory T cells of old donors had approximately 500 more uniquely expressed transcripts. Four GO terms related to the JAK-STAT pathway were identified for up-regulated transcripts in the total CD8+ T cells of old donors, whereas CD8+CD45RA- memory T cells GO terms related to adjacent pathways, like JNK and MAPK/ERK, were found. Additionally, the unique transcripts of CD8+CD45RA- memory T cells of old donors were related to the JNK, MAPK and IL-12 pathways. For both T cell populations of the old donors, cytokine and JAK-STAT pathway transcripts were up-regulated. Thus, an age-dependent effect was observed on the transcriptomes of total and memory CD8+ T cells. The CD8+ CD45RA- memory T cells from old donors maintained the increased cytokine secretion of the total CD8+ T cell population and the increased JAK-STAT pathway transcripts, which have an impact on inflammation and senescence.


Asunto(s)
Envejecimiento , Donantes de Sangre , Linfocitos T CD8-positivos/inmunología , Perfilación de la Expresión Génica/métodos , Memoria Inmunológica/inmunología , Antígenos Comunes de Leucocito/genética , Células T de Memoria/inmunología , Adulto , Factores de Edad , Anciano , Envejecimiento/inmunología , Donantes de Sangre/estadística & datos numéricos , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/clasificación , Citocinas , Femenino , Citometría de Flujo , Voluntarios Sanos/estadística & datos numéricos , Humanos , Recuento de Linfocitos , Masculino , Persona de Mediana Edad , Adulto Joven
3.
Front Immunol ; 12: 708874, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34484208

RESUMEN

Regulatory T cells (Treg) are essential to maintain immune homeostasis and prevent autoimmune disorders. While the function and molecular regulation of Foxp3+CD4+ Tregs are well established, much of CD8+ Treg biology remains to be revealed. Here, we will review the heterogenous subsets of CD8+ T cells have been named "CD8+ Treg" and mainly focus on CD122hiLy49+CD8+ Tregs present in naïve mice. CD122hiLy49+CD8+ Tregs, which depends on transcription factor Helios and homeostatic cytokine IL-15, have been established as a non-redundant regulator of germinal center (GC) reaction. Recently, we have demonstrated that TGF-ß (Transforming growth factor-ß) and transcription factor Eomes (Eomesodermin) are essential for the function and homeostasis of CD8+ Tregs. In addition, we will discuss several open questions regarding the differentiation, function and true identity of CD8+ Tregs as well as a brief comparison between two regulatory T cell subsets critical to control GC reaction, namely CD4+ TFR (follicular regulatory T cells) and CD8+ Tregs.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Linfocitos T Reguladores/inmunología , Animales , Linfocitos T CD8-positivos/clasificación , Centro Germinal/inmunología , Humanos , Subunidad beta del Receptor de Interleucina-2/análisis , Ratones , Subfamilia A de Receptores Similares a Lectina de Células NK/análisis , Linfocitos T Reguladores/clasificación , Factor de Crecimiento Transformador beta/fisiología
4.
Nature ; 597(7875): 279-284, 2021 09.
Artículo en Inglés | MEDLINE | ID: mdl-34471285

RESUMEN

T cells are important in tumour immunity but a better understanding is needed of the differentiation of antigen-specific T cells in human cancer1,2. Here we studied CD8 T cells in patients with human papillomavirus (HPV)-positive head and neck cancer and identified several epitopes derived from HPV E2, E5 and E6 proteins that allowed us to analyse virus-specific CD8 T cells using major histocompatibility complex (MHC) class I tetramers. HPV-specific CD8 T cells expressed PD-1 and were detectable in the tumour at levels that ranged from 0.1% to 10% of tumour-infiltrating CD8 T lymphocytes (TILs) for a given epitope. Single-cell RNA-sequencing analyses of tetramer-sorted HPV-specific PD-1+ CD8 TILs revealed three transcriptionally distinct subsets. One subset expressed TCF7 and other genes associated with PD-1+ stem-like CD8 T cells that are critical for maintaining T cell responses in conditions of antigen persistence. The second subset expressed more effector molecules, representing a transitory cell population, and the third subset was characterized by a terminally differentiated gene signature. T cell receptor clonotypes were shared between the three subsets and pseudotime analysis suggested a hypothetical differentiation trajectory from stem-like to transitory to terminally differentiated cells. More notably, HPV-specific PD-1+TCF-1+ stem-like TILs proliferated and differentiated into more effector-like cells after in vitro stimulation with the cognate HPV peptide, whereas the more terminally differentiated cells did not proliferate. The presence of functional HPV-specific PD-1+TCF-1+CD45RO+ stem-like CD8 T cells with proliferative capacity shows that the cellular machinery to respond to PD-1 blockade exists in HPV-positive head and neck cancer, supporting the further investigation of PD-1 targeted therapies in this malignancy. Furthermore, HPV therapeutic vaccination efforts have focused on E6 and E7 proteins; our results suggest that E2 and E5 should also be considered for inclusion as vaccine antigens to elicit tumour-reactive CD8 T cell responses of maximal breadth.


Asunto(s)
Alphapapillomavirus/inmunología , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Neoplasias de Cabeza y Cuello/inmunología , Neoplasias de Cabeza y Cuello/virología , Linfocitos Infiltrantes de Tumor/inmunología , Receptor de Muerte Celular Programada 1/metabolismo , Células Madre/citología , Alphapapillomavirus/aislamiento & purificación , Linfocitos T CD8-positivos/clasificación , Linfocitos T CD8-positivos/metabolismo , Vacunas contra el Cáncer/inmunología , Diferenciación Celular , Proliferación Celular , Proteínas de Unión al ADN/inmunología , Humanos , Linfocitos Infiltrantes de Tumor/clasificación , Linfocitos Infiltrantes de Tumor/citología , Linfocitos Infiltrantes de Tumor/metabolismo , Proteínas Oncogénicas Virales/inmunología , Infecciones por Papillomavirus/inmunología , Infecciones por Papillomavirus/virología , Vacunas contra Papillomavirus/inmunología , RNA-Seq , Receptores de Antígenos de Linfocitos T/inmunología , Análisis de la Célula Individual , Células Madre/inmunología , Factor 1 de Transcripción de Linfocitos T/metabolismo , Linfocitos T/inmunología , Transcripción Genética
5.
Sci Rep ; 11(1): 16134, 2021 08 09.
Artículo en Inglés | MEDLINE | ID: mdl-34373557

RESUMEN

Head and neck squamous carcinoma (HNSCC) is highly infiltrated by immune cells, including tumor-infiltrating lymphocytes and myeloid lineage cells. In the tumor microenvironment, tumor cells orchestrate a highly immunosuppressive microenvironment by secreting immunosuppressive mediators, expressing immune checkpoint ligands, and downregulating human leukocyte antigen expression. In the present study, we aimed to comprehensively profile the immune microenvironment of HNSCC using gene expression data obtained from public database. We calculated enrichment scores of 33 immune cell types based on gene expression data of HNSCC tissues and adjacent non-cancer tissues. Based on these scores, we performed non-supervised clustering and identified three immune signatures-cold, lymphocyte, and myeloid/dendritic cell (DC)-based on the clustering results. We then compared the clinical and biological features of the three signatures. Among HNSCC and non-cancer tissues, human papillomavirus (HPV)-positive HNSCCs exhibited the highest scores in various immune cell types, including CD4+ T cells, CD8+ T cells, B cells, plasma cells, basophils, and their subpopulations. Among the three immune signatures, the proportions of HPV-positive tumors, oropharyngeal cancers, early T tumors, and N factor positive cases were significantly higher in the lymphocyte signature than in other signatures. Among the three signatures, the lymphocyte signature showed the longest overall survival (OS), especially in HPV-positive patients, whereas the myeloid/DC signature demonstrated the shortest OS in these patients. Gene set enrichment analysis revealed the upregulation of several pathways related to inflammatory and proinflammatory responses in the lymphocyte signature. The expression of PRF1, IFNG, GZMB, CXCL9, CXCL10, PDCD1, LAG3, CTLA4, HAVCR2, and TIGIT was the highest in the lymphocyte signature. Meanwhile, the expression of PD-1 ligand genes CD274 and PDCD1LG2 was highest in the myeloid/DC signature. Herein, our findings revealed the transcriptomic landscape of the immune microenvironment that closely reflects the clinical and biological significance of HNSCC, indicating that molecular profiling of the immune microenvironment can be employed to develop novel biomarkers and precision immunotherapies for HNSCC.


Asunto(s)
Neoplasias de Cabeza y Cuello/inmunología , Carcinoma de Células Escamosas de Cabeza y Cuello/inmunología , Microambiente Tumoral/inmunología , Linfocitos T CD8-positivos/clasificación , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/patología , Neoplasias de Cabeza y Cuello/genética , Neoplasias de Cabeza y Cuello/patología , Humanos , Linfocitos Infiltrantes de Tumor/clasificación , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/patología , Células Mieloides/clasificación , Células Mieloides/inmunología , Células Mieloides/patología , Papillomaviridae/aislamiento & purificación , Pronóstico , Carcinoma de Células Escamosas de Cabeza y Cuello/genética , Carcinoma de Células Escamosas de Cabeza y Cuello/patología , Transcriptoma , Microambiente Tumoral/genética
6.
Mol Cells ; 44(6): 401-407, 2021 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-34120892

RESUMEN

Infection with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes coronavirus disease 2019 (COVID-19), which is an ongoing pandemic disease. SARS-CoV-2-specific CD4+ and CD8+ T-cell responses have been detected and characterized not only in COVID-19 patients and convalescents, but also unexposed individuals. Here, we review the phenotypes and functions of SARS-CoV-2-specific T cells in COVID-19 patients and the relationships between SARS-CoV-2-specific T-cell responses and COVID-19 severity. In addition, we describe the phenotypes and functions of SARS-CoV-2-specific memory T cells after recovery from COVID-19 and discuss the presence of SARS-CoV-2-reactive T cells in unexposed individuals and SARS-CoV-2-specific T-cell responses elicited by COVID-19 vaccines. A better understanding of T-cell responses is important for effective control of the current COVID-19 pandemic.


Asunto(s)
Anticuerpos Neutralizantes/biosíntesis , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , COVID-19/inmunología , Inmunidad Celular , SARS-CoV-2/patogenicidad , Anticuerpos Antivirales/biosíntesis , Linfocitos T CD4-Positivos/clasificación , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/clasificación , Linfocitos T CD8-positivos/virología , COVID-19/patología , COVID-19/prevención & control , COVID-19/virología , Vacunas contra la COVID-19/administración & dosificación , Convalecencia , Citocinas/biosíntesis , Citotoxicidad Inmunológica , Interacciones Huésped-Patógeno/inmunología , Humanos , Inmunidad Humoral , Memoria Inmunológica , Inmunofenotipificación , SARS-CoV-2/inmunología , Índice de Severidad de la Enfermedad
7.
Mol Cell ; 81(11): 2477-2493.e10, 2021 06 03.
Artículo en Inglés | MEDLINE | ID: mdl-33891860

RESUMEN

CD8 T cells play an essential role in defense against viral and bacterial infections and in tumor immunity. Deciphering T cell loss of functionality is complicated by the conspicuous heterogeneity of CD8 T cell states described across experimental and clinical settings. By carrying out a unified analysis of over 300 assay for transposase-accessible chromatin sequencing (ATAC-seq) and RNA sequencing (RNA-seq) experiments from 12 studies of CD8 T cells in cancer and infection, we defined a shared differentiation trajectory toward dysfunction and its underlying transcriptional drivers and revealed a universal early bifurcation of functional and dysfunctional T cell states across models. Experimental dissection of acute and chronic viral infection using single-cell ATAC (scATAC)-seq and allele-specific single-cell RNA (scRNA)-seq identified state-specific drivers and captured the emergence of similar TCF1+ progenitor-like populations at an early branch point, at which functional and dysfunctional T cells diverge. Our atlas of CD8 T cell states will facilitate mechanistic studies of T cell immunity and translational efforts.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Epigénesis Genética/inmunología , Inmunidad Celular , Coriomeningitis Linfocítica/genética , Neoplasias/genética , Factores de Transcripción/genética , Enfermedad Aguda , Atlas como Asunto , Linfocitos T CD8-positivos/clasificación , Linfocitos T CD8-positivos/patología , Cromatina/química , Cromatina/inmunología , Enfermedad Crónica , Perfilación de la Expresión Génica , Redes Reguladoras de Genes , Secuenciación de Nucleótidos de Alto Rendimiento/métodos , Humanos , Activación de Linfocitos , Coriomeningitis Linfocítica/inmunología , Coriomeningitis Linfocítica/patología , Virus de la Coriomeningitis Linfocítica/inmunología , Virus de la Coriomeningitis Linfocítica/patogenicidad , Neoplasias/inmunología , Neoplasias/patología , Análisis de Componente Principal , Análisis de la Célula Individual , Factores de Transcripción/clasificación , Factores de Transcripción/inmunología , Transcripción Genética , Transposasas/genética , Transposasas/metabolismo
8.
J Infect Dis ; 223(6): 1029-1039, 2021 03 29.
Artículo en Inglés | MEDLINE | ID: mdl-32725203

RESUMEN

BACKGROUND: Inflammation persists among persons with human immunodeficiency virus (PWH) despite effective antiretroviral therapy and may contribute to T-cell dysfunction. Alcohol use is prevalent among PWH and promotes intestinal leak, dysbiosis, and a proinflammatory milieu. Whether alcohol use is associated with T-cell late differentiation remains to be investigated. METHODS: Data and samples from PWH (N = 359 of 365) enrolled in the New Orleans Alcohol Use in HIV Study were used. Alcohol use was assessed by self-report (Alcohol Use Disorders Identification Test; lifetime alcohol exposure; 30-day Alcohol Timeline Followback) and phosphatidylethanol (PEth) quantitation. In a subset of participants, fecal bacterial content was assessed by ribosomal 16S marker gene deep sequencing and quantitative polymerase chain reaction. Intestinal leak was assessed by fecal-to-plasma α-1-antitrypsin (A1AT) enzyme-linked immunosorbent assay ratio. Peripheral T-cell populations were quantified by flow cytometry. RESULTS: Alcohol Use Disorder Identification Test scores were positively associated with activated-senescent, exhausted, and terminal effector memory CD45RA+CD8+ but not CD4+ T cells (cells/µL) after confounder adjustment (P < .050). Phosphatidylethanol was positively associated with A1AT (P < .050). The PEth and activated-senescent CD8+ were associated with bacterial ß-diversity (P < .050) and positively associated with the relative abundance of coabundant Prevotellaceae members (q < .100). CONCLUSIONS: Alcohol use among PWH is associated with CD8+ T-cell late differentiation, intestinal leak, and dysbiosis. Alcohol-associated dysbiosis is implicated in CD8+ T-cell senescence.


Asunto(s)
Alcoholismo , Linfocitos T CD8-positivos/clasificación , Disbiosis , Infecciones por VIH , Alcoholismo/complicaciones , Disbiosis/complicaciones , Infecciones por VIH/complicaciones , Humanos , Fenotipo
9.
Eur J Immunol ; 51(1): 151-166, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32762051

RESUMEN

Tissue-resident memory CD8+ T cells (TRM ) localize to barrier tissues and mediate local protection against reinvading pathogens. Circulating central memory (TCM ) and effector memory CD8+ T cells (TEM ) also contribute to tissue recall responses, but their potential to form mucosal TRM remains unclear. Here, we employed adoptive transfer and lymphocytic choriomeningitis virus reinfection models to specifically assess secondary responses of TCM and TEM at mucosal sites. Donor TCM and TEM exhibited robust systemic recall responses, but only limited accumulation in the small intestine, consistent with reduced expression of tissue-homing and -retention molecules. Murine and human circulating memory T cells also exhibited limited CD103 upregulation following TGF-ß stimulation. Upon pathogen clearance, TCM and TEM readily gave rise to secondary TEM . TCM also formed secondary central memory in lymphoid tissues and TRM in internal tissues, for example, the liver. Both TCM and TEM failed to substantially contribute to resident mucosal memory in the small intestine, while activated intestinal TRM , but not liver TRM , efficiently reformed CD103+ TRM . Our findings demonstrate that circulating TCM and TEM are limited in generating mucosal TRM upon reinfection. This may pose important implications on cell therapy and vaccination strategies employing memory CD8+ T cells for protection at mucosal sites.


Asunto(s)
Antígenos CD/inmunología , Linfocitos T CD8-positivos/inmunología , Inmunidad Mucosa , Memoria Inmunológica , Cadenas alfa de Integrinas/inmunología , Inmunidad Adaptativa , Traslado Adoptivo , Animales , Antígenos CD/metabolismo , Linfocitos T CD8-positivos/clasificación , Linfocitos T CD8-positivos/citología , Diferenciación Celular/inmunología , Femenino , Humanos , Cadenas alfa de Integrinas/metabolismo , Mucosa Intestinal/citología , Mucosa Intestinal/inmunología , Intestino Delgado/citología , Intestino Delgado/inmunología , Activación de Linfocitos , Virus de la Coriomeningitis Linfocítica/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Membrana Mucosa/citología , Membrana Mucosa/inmunología , Factor de Crecimiento Transformador beta/inmunología
10.
Eur J Immunol ; 51(1): 247-249, 2021 01.
Artículo en Inglés | MEDLINE | ID: mdl-32671842

RESUMEN

Increased incidence of narcolepsy type 1 (NT1) was observed following Pandemrix®-vaccination, initiated as a preventive measure against the 2009 Influenza pandemic. Here, single cell analysis was conducted to suggest a lower number of CD8+ CD27+ T cells among these patients. These findings provide understanding into the autoimmune pathogenesis of NT1.


Asunto(s)
Vacunas contra la Influenza/efectos adversos , Narcolepsia/etiología , Narcolepsia/inmunología , Algoritmos , Basófilos/inmunología , Linfocitos T CD8-positivos/clasificación , Linfocitos T CD8-positivos/inmunología , Estudios de Casos y Controles , Citometría de Flujo , Cadenas beta de HLA-DQ/inmunología , Humanos , Vacunas contra la Influenza/inmunología , Hermanos , Análisis de la Célula Individual
11.
JCI Insight ; 6(3)2021 02 08.
Artículo en Inglés | MEDLINE | ID: mdl-33351781

RESUMEN

Clinical trials of biologic therapies in type 1 diabetes (T1D) aim to mitigate autoimmune destruction of pancreatic ß cells through immune perturbation and serve as resources to elucidate immunological mechanisms in health and disease. In the T1DAL trial of alefacept (LFA3-Ig) in recent-onset T1D, endogenous insulin production was preserved in 30% of subjects for 2 years after therapy. Given our previous findings linking exhausted-like CD8+ T cells to beneficial response in T1D trials, we applied unbiased analyses to sorted CD8+ T cells to evaluate their potential role in T1DAL. Using RNA sequencing, we found that greater insulin C-peptide preservation was associated with a module of activation- and exhaustion-associated genes. This signature was dissected into 2 CD8 memory phenotypes through correlation with cytometry data. These cells were hypoproliferative, shared expanded rearranged TCR junctions, and expressed exhaustion-associated markers including TIGIT and KLRG1. The 2 phenotypes could be distinguished by reciprocal expression of CD8+ T and NK cell markers (GZMB, CD57, and inhibitory killer cell immunoglobulin-like receptor [iKIR] genes), versus T cell activation and differentiation markers (PD-1 and CD28). These findings support previous evidence linking exhausted-like CD8+ T cells to successful immune interventions for T1D, while suggesting that multiple inhibitory mechanisms can promote this beneficial cell state.


Asunto(s)
Alefacept/uso terapéutico , Péptido C/biosíntesis , Linfocitos T CD8-positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Diabetes Mellitus Tipo 1/terapia , Adolescente , Adulto , Péptido C/genética , Antígenos CD57/metabolismo , Linfocitos T CD8-positivos/clasificación , Linfocitos T CD8-positivos/metabolismo , Niño , Diabetes Mellitus Tipo 1/metabolismo , Método Doble Ciego , Femenino , Humanos , Factores Inmunológicos/uso terapéutico , Memoria Inmunológica/genética , Inmunofenotipificación , Células Asesinas Naturales/inmunología , Lectinas Tipo C/metabolismo , Activación de Linfocitos , Masculino , Receptor de Muerte Celular Programada 1/metabolismo , RNA-Seq , Receptores Inmunológicos/metabolismo , Adulto Joven
12.
Nat Med ; 26(9): 1468-1479, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32778827

RESUMEN

PD-1 blockade is highly effective in classical Hodgkin lymphomas (cHLs), which exhibit frequent copy-number gains of CD274 (PD-L1) and PDC1LG2 (PD-L2) on chromosome 9p24.1. However, in this largely MHC-class-I-negative tumor, the mechanism of action of anti-PD-1 therapy remains undefined. We utilized the complementary approaches of T cell receptor (TCR) sequencing and cytometry by time-of-flight analysis to obtain a peripheral immune signature of responsiveness to PD-1 blockade in 56 patients treated in the CheckMate 205 phase II clinical trial (NCT02181738). Anti-PD-1 therapy was most effective in patients with a diverse baseline TCR repertoire and an associated expansion of singleton clones during treatment. CD4+, but not CD8+, TCR diversity significantly increased during therapy, most strikingly in patients who had achieved complete responses. Additionally, patients who responded to therapy had an increased abundance of activated natural killer cells and a newly identified CD3-CD68+CD4+GrB+ subset. These studies highlight the roles of recently expanded, clonally diverse CD4+ T cells and innate effectors in the efficacy of PD-1 blockade in cHL.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Enfermedad de Hodgkin/tratamiento farmacológico , Células Asesinas Naturales/inmunología , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Antineoplásicos Inmunológicos/uso terapéutico , Linfocitos T CD4-Positivos/clasificación , Linfocitos T CD8-positivos/clasificación , Humanos , Activación de Linfocitos/inmunología , Nivolumab/uso terapéutico , Receptores de Antígenos de Linfocitos T/genética , Microambiente Tumoral/inmunología
13.
Artículo en Inglés | MEDLINE | ID: mdl-32839203

RESUMEN

Immunological memory, defined as the ability to respond in an enhanced manner upon secondary encounter with the same pathogen, can provide substantial protection against infectious disease. The improved protection is mediated in part by different populations of memory CD8 T cells that are retained after primary infection. Memory cells persist in the absence of pathogen-derived antigens and enable secondary CD8 T-cell responses with accelerated kinetics and of larger magnitude after reencounter with the same pathogen. At least three subsets of memory T cells have been defined that are referred to as central memory CD8 T cells (Tcm), effector memory CD8 T cells (Tem), and tissue-resident memory CD8 T cells (Trm). Tcm and Tem are circulating memory T cells that mediate bodywide immune surveillance in search of invading pathogens. In contrast, Trm permanently reside in peripheral barrier tissues, where they form a stationary defensive line of sentinels that alert the immune system upon pathogen reencounter. The characterization of these different subsets has been instrumental in our understanding of the strategies that memory T cells employ to counter invading pathogens. It is clear that memory T cells not only have a numerical advantage over naive T cells resulting in improved protection in secondary responses, but also acquire distinct sets of competencies that assist in pathogen clearance. Nevertheless, inherent challenges are associated with the allocation of memory T cells to a limited number of subsets. The classification of memory T cells into Tcm, Tem, and Trm may not take into account the full extent of the heterogeneity that is observed in the memory population. Therefore, in this review, we will revisit the current classification of memory subsets, elaborate on functional and migratory properties attributed to Tcm, Tem, and Trm, and discuss how potential heterogeneity within these populations arises and persists.


Asunto(s)
Linfocitos T CD8-positivos/fisiología , Memoria Inmunológica/fisiología , Subgrupos de Linfocitos T/fisiología , Animales , Linfocitos T CD8-positivos/clasificación , Diferenciación Celular , Humanos , Activación de Linfocitos , Subgrupos de Linfocitos T/clasificación
14.
PLoS One ; 15(6): e0234323, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32530943

RESUMEN

We investigated the phenotype and molecular signatures of CD8+ T cell subsets in kidney-transplant recipients (KTRs) with biopsy-proven T cell-mediated rejection (TCMR). We included 121 KTRs and divided them into three groups according to the pathologic or clinical diagnosis: Normal biopsy control (NC)(n = 32), TCMR (n = 50), and long-term graft survival (LTGS)(n = 39). We used flowcytometry and microarray to analyze the phenotype and molecular signatures of CD8+ T cell subsets using peripheral blood from those patients and analyzed significant gene expressions according to CD8+ T cell subsets. We investigated whether the analysis of CD8+ T cell subsets is useful for predicting the development of TCMR. CCR7+CD8+ T cells significantly decreased, but CD28nullCD57+CD8+ T cells and CCR7-CD45RA+CD8+ T cells showed an increase in the TCMR group compared to other groups (p<0.05 for each); hence CCR7+CD8+ T cells showed significant negative correlations to both effector CD8+ T cells. We identified genes significantly associated with the change of CCR7+CD8+ T, CCR7-CD45RA+CD8+ T, and CD28nullCD57+CD8+ T cells in an ex vivo study and found that most of them were included in the significant genes on in vitro CCR7+CD8+ T cells. Finally, the decrease of CCR7+CD8+ T cells relative to CD28nullCD57+ T or CCR7-CD45RA+CD8+ T cells can predict TCMR significantly in the whole clinical cohort. In conclusion, phenotype and molecular signature of CD8+ T subsets showed a significant relationship to the development of TCMR; hence monitoring of CD8+ T cell subsets may be a useful for predicting TCMR in KTRs.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Rechazo de Injerto/inmunología , Trasplante de Riñón/efectos adversos , Subgrupos de Linfocitos T/inmunología , Adulto , Antígenos CD28/genética , Antígenos CD28/inmunología , Antígenos CD57/genética , Antígenos CD57/inmunología , Linfocitos T CD8-positivos/clasificación , Estudios Transversales , Femenino , Perfilación de la Expresión Génica , Rechazo de Injerto/etiología , Rechazo de Injerto/genética , Voluntarios Sanos , Humanos , Inmunofenotipificación , Técnicas In Vitro , Masculino , Persona de Mediana Edad , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Receptores CCR7/genética , Receptores CCR7/inmunología , Subgrupos de Linfocitos T/clasificación
15.
Front Immunol ; 11: 896, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32477365

RESUMEN

Intestinal tissue-resident memory CD8 T cells (Trm) are non-recirculating effector cells ideally positioned to detect and react to microbial infections in the gut mucosa. There is an emerging understanding of Trm cell differentiation and functions, but their implication in inflammatory bowel diseases, such as Crohn's disease (CD), is still unknown. Here, we describe CD8 cells in the human intestine expressing KLRG1 or CD103, two receptors of E-cadherin. While CD103 CD8 T cells are present in high numbers in the mucosa of CD patients and controls, KLRG1 CD8 T cells are increased in inflammatory conditions. Mucosal CD103 CD8 T cells are more responsive to TCR restimulation, but KLRG1 CD8 T cells show increased cytotoxic and proliferative potential. CD103 CD8 T cells originate mostly from KLRG1 negative cells after TCR triggering and TGFß stimulation. Interestingly, mucosal CD103 CD8 T cells from CD patients display major changes in their transcriptomic landscape compared to controls. They express Th17 related genes including CCL20, IL22, and IL26, which could contribute to the pathogenesis of CD. Overall, these findings suggest that CD103 CD8 T cells in CD induce a tissue-wide alert increasing innate immune responses and recruitment of effector cells such as KLRG1 CD8 T cells.


Asunto(s)
Antígenos CD/metabolismo , Linfocitos T CD8-positivos/inmunología , Enfermedad de Crohn/inmunología , Memoria Inmunológica , Cadenas alfa de Integrinas/metabolismo , Mucosa Intestinal/inmunología , Lectinas Tipo C/metabolismo , Receptores Inmunológicos/metabolismo , Linfocitos T CD8-positivos/clasificación , Ensayos Clínicos como Asunto , Enfermedad de Crohn/fisiopatología , Perfilación de la Expresión Génica , Humanos , Inmunidad Innata , Inflamación , Interleucinas/inmunología , Subgrupos de Linfocitos T/inmunología
16.
J Leukoc Biol ; 107(6): 953-970, 2020 06.
Artículo en Inglés | MEDLINE | ID: mdl-32125017

RESUMEN

Tissue resident memory T cells (Trm) are critical for local protection against reinfection. The accumulation of T cells in the tissues requires a post-priming signal from TNFR superfamily members, referred to as signal 4. Glucocorticoid-induced TNFR-related protein (GITR; TNFRSF18) signaling is important for this post-priming signal and for Trm formation during respiratory infection with influenza virus. As GITR signaling impacts both effector T cell accumulation and Trm formation, we asked if GITR differentially affects subsets of effector cells with different memory potential. Effector CD4+ T cells can be subdivided into 2 populations based on expression of lymphocyte antigen 6C (Ly6C), whereas effector CD8+ cells can be divided into 3 populations based on Ly6C and CX3CR1. The Ly6Chi and CX3CR1hi T cell populations represent the most differentiated effector T cells. Upon transfer, the Ly6Clo CD4+ effector T cells preferentially enter the lung parenchyma, compared to the Ly6Chi CD4+ T cells. We show that GITR had a similar effect on the accumulation of both the Ly6Chi and Ly6Clo CD4+ T cell subsets. In contrast, whereas GITR increased the accumulation of all three CD8+ T cell subsets defined by CX3CR1 and Ly6C expression, it had a more substantial effect on the least differentiated Ly6Clo CX3CR1lo subset. Moreover, GITR selectively up-regulated CXCR6 on the less differentiated CX3CR1lo CD8+ T cell subsets and induced a small but significant increase in CD127 selectively on the Ly6Clo CD4+ T cell subset. Thus, GITR contributes to accumulation of both differentiated effector cells as well as memory precursors, but with some differences between subsets.


Asunto(s)
Antígenos Ly/genética , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Proteína Relacionada con TNFR Inducida por Glucocorticoide/genética , Virus de la Influenza A/inmunología , Infecciones por Orthomyxoviridae/inmunología , Animales , Antígenos Ly/inmunología , Linfocitos T CD4-Positivos/clasificación , Linfocitos T CD4-Positivos/patología , Linfocitos T CD4-Positivos/virología , Linfocitos T CD8-positivos/clasificación , Linfocitos T CD8-positivos/patología , Linfocitos T CD8-positivos/virología , Receptor 1 de Quimiocinas CX3C/genética , Receptor 1 de Quimiocinas CX3C/inmunología , Diferenciación Celular , Movimiento Celular , Femenino , Regulación de la Expresión Génica , Proteína Relacionada con TNFR Inducida por Glucocorticoide/deficiencia , Proteína Relacionada con TNFR Inducida por Glucocorticoide/inmunología , Memoria Inmunológica , Inmunofenotipificación , Virus de la Influenza A/crecimiento & desarrollo , Subunidad alfa del Receptor de Interleucina-7/genética , Subunidad alfa del Receptor de Interleucina-7/inmunología , Pulmón/inmunología , Pulmón/patología , Pulmón/virología , Recuento de Linfocitos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/patología , Infecciones por Orthomyxoviridae/virología , Transducción de Señal , Bazo/inmunología , Bazo/patología , Bazo/virología
17.
J Leukoc Biol ; 108(3): 851-857, 2020 09.
Artículo en Inglés | MEDLINE | ID: mdl-32052478

RESUMEN

The cellular origin of CD4- CD8- (double negative, DNT) TCR-α/ß+ T cells remains unknown. Available evidence indicates that they may derive from CD8+ T cells, but most published data have been obtained using cells that bear an invariant transgenic T cell receptor that recognizes an Ag that is not present in normal mice. Here, we have used complementary fate mapping and adoptive transfer experiments to identify the cellular lineage of origin of DNT cells in wild-type mice with a polyclonal T cell repertoire. We show that TCR-α/ß+ DNT cells can be traced back to CD8+ and CD4+ CD8+ double positive cells in the thymus. We also demonstrate that polyclonal DNT cells generated in secondary lymphoid organs proliferate upon adoptive transfer and can regain CD8 expression in lymphopenic environment. These results demonstrate the cellular origin of DNT cells and provide a conceptual framework to understand their presence in pathological circumstances.


Asunto(s)
Antígenos CD4/análisis , Antígenos CD8/análisis , Linfocitos T CD8-positivos/citología , Linfopoyesis , Receptores de Antígenos de Linfocitos T alfa-beta/análisis , Subgrupos de Linfocitos T/citología , Traslado Adoptivo , Animales , Antígenos CD8/biosíntesis , Antígenos CD8/genética , Linfocitos T CD8-positivos/clasificación , Linaje de la Célula , Tejido Linfoide/citología , Tejido Linfoide/inmunología , Ratones , Organismos Libres de Patógenos Específicos , Regulación hacia Arriba
18.
Biochem Biophys Res Commun ; 523(2): 328-335, 2020 03 05.
Artículo en Inglés | MEDLINE | ID: mdl-31864702

RESUMEN

Intraepithelial lymphocytes (IELs) are very unique in the intestinal immune system. They include γδT cells and CD4-CD8-TCRαß+T cells (double negative: DNT), both of which are specific for the intestine, in addition to CD4+ and CD8+ T cells. IELs exist within the monolayer of the intestinal epithelial cells and dynamically move between lamina propria (LP) and intraepithelial (IE) region. The localization and movement patterns of IEL subsets and the regulatory factors have been unknown. Here, we developed a novel in vitro live imaging system and quantified the motility and morphological changes among subsets of IELs. We identified CD8αα as the key regulatory factor. IELs, especially γδ and DNT cells, showed amoeboid shape and frequent morphological change, while most T cells in MLN or SP showed round shape in vitro. TCR signal, IL-15, gut microbes, CCL25, and integrin αEß7 expression were non-essential for IEL movement in vitro. CD8αα+ cells showed higher motility and larger morphological changes than CD8αα- cells. Adoptive transferred CD8αα+CD4-IELs localized to IE region of recipient NSG mice, while CD8αα-CD4-IELs localized to the LP. Our results showed that the CD8αα/TL signal is essential for the localization of IELs to IE region in vivo. CD8αα/TL may be an effective target to increase the number of IELs, which protects against intestinal infection, allergy, tumorigenesis or inflammation.


Asunto(s)
Antígenos CD8/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/inmunología , Linfocitos Intraepiteliales/citología , Linfocitos Intraepiteliales/inmunología , Traslado Adoptivo , Animales , Linfocitos T CD8-positivos/clasificación , Movimiento Celular/inmunología , Forma de la Célula , Quimiocinas CC/metabolismo , Femenino , Inmunidad Mucosa , Interleucina-15/metabolismo , Intestino Delgado/citología , Intestino Delgado/inmunología , Linfocitos Intraepiteliales/clasificación , Microscopía Intravital , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Ratones Transgénicos
19.
PLoS One ; 14(2): e0211446, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30735510

RESUMEN

It is common practice for researchers to use antibodies to remove a specific cell type to infer its function. However, it is difficult to completely eliminate a cell type and there is often limited or no information as to how the cells which survive depletion are affected. This is particularly important for CD8+ T cells for two reasons. First, they are more resistant to mAb-mediated depletion than other lymphocytes. Second, targeting either the CD8α or CD8ß chain could induce differential effects. We show here that two commonly used mAbs, against either the CD8α or CD8ß subunit, can differentially affect cellular metabolism. Further, in vivo treatment leaves behind a population of CD8+ T cells with different phenotypic and functional attributes relative to each other or control CD8+ T cells. The impact of anti-CD8 antibodies on CD8+ T cell phenotype and function indicates the need to carefully consider the use of these, and possibly other "depleting" antibodies, as they could significantly complicate the interpretation of results or change the outcome of an experiment. These observations could impact how immunotherapy and modulation of CD8+ T cell activation is pursued.


Asunto(s)
Antígenos CD8/inmunología , Linfocitos T CD8-positivos/inmunología , Depleción Linfocítica/métodos , Traslado Adoptivo , Animales , Anticuerpos Monoclonales , Antígenos CD8/metabolismo , Linfocitos T CD8-positivos/clasificación , Linfocitos T CD8-positivos/citología , Diferenciación Celular/inmunología , Citotoxicidad Inmunológica , Femenino , Memoria Inmunológica , Inmunofenotipificación , Activación de Linfocitos , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos
20.
PLoS One ; 14(1): e0210839, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30650147

RESUMEN

Type 1 diabetes is an autoimmune disease where autoreactive T lymphocytes destroy pancreatic beta cells. We previously reported a defect in CD4+ Tregs cell proliferation and reduced CD4+ Tregs PD-1 expression in patients. Another 'memory-like' regulatory subset, CD8+ Tregs, evaluated as CD8+CD25+FOXP3+, has recently raised interest for their effective suppressive activity. Different CD8+ T cell populations, their proliferation capacity and expression of PD-1 molecule were evaluated by flow-cytometer analysis in newly diagnosed, long-term Type 1 diabetes patients compared to healthy normal donors. Under basal conditions, CD8+ Tregs and CD8+ Teffs were seemingly represented among study groups while there was evidence of diminished expression of PD-1 in Teff subsets of long-term patients. After 3 days of PMA/ionomycin stimulation, patients CD8+ Tregs showed decreased percentage in respect to control group. CD8+ Teffs were instead increased in long-term diabetics versus controls. PD-1+CD8+ Tregs were represented at a much lower percentage in long-term diabetic patients, in respect to controls. Importantly, patients CD8+ Tregs and CD8+ Teffs presented a significant proliferation defect in respect to the control group. In conclusion, our study indicates that a defect of CD8+ Tregs is observed in diabetics. This subset could thus represent a novel target of immunotherapy in patients.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Diabetes Mellitus Tipo 1/inmunología , Linfocitos T Reguladores/inmunología , Adolescente , Adulto , Biomarcadores/sangre , Linfocitos T CD8-positivos/clasificación , Linfocitos T CD8-positivos/efectos de los fármacos , Estudios de Casos y Controles , Proliferación Celular/efectos de los fármacos , Niño , Preescolar , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/diagnóstico , Femenino , Hemoglobina Glucada/metabolismo , Humanos , Inmunoterapia , Técnicas In Vitro , Ionomicina/farmacología , Masculino , Proyectos Piloto , Receptor de Muerte Celular Programada 1/metabolismo , Linfocitos T Reguladores/clasificación , Linfocitos T Reguladores/efectos de los fármacos , Acetato de Tetradecanoilforbol/farmacología , Adulto Joven
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...