Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 51
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 24(11): 1908-1920, 2023 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-37828379

RESUMEN

Co-inhibitory and checkpoint molecules suppress T cell function in the tumor microenvironment, thereby rendering T cells dysfunctional. Although immune checkpoint blockade is a successful treatment option for multiple human cancers, severe autoimmune-like adverse effects can limit its application. Here, we show that the gene encoding peptidoglycan recognition protein 1 (PGLYRP1) is highly coexpressed with genes encoding co-inhibitory molecules, indicating that it might be a promising target for cancer immunotherapy. Genetic deletion of Pglyrp1 in mice led to decreased tumor growth and an increased activation/effector phenotype in CD8+ T cells, suggesting an inhibitory function of PGLYRP1 in CD8+ T cells. Surprisingly, genetic deletion of Pglyrp1 protected against the development of experimental autoimmune encephalomyelitis, a model of autoimmune disease in the central nervous system. PGLYRP1-deficient myeloid cells had a defect in antigen presentation and T cell activation, indicating that PGLYRP1 might function as a proinflammatory molecule in myeloid cells during autoimmunity. These results highlight PGLYRP1 as a promising target for immunotherapy that, when targeted, elicits a potent antitumor immune response while protecting against some forms of tissue inflammation and autoimmunity.


Asunto(s)
Encefalomielitis Autoinmune Experimental , Neoplasias , Animales , Humanos , Ratones , Linfocitos T CD8-positivos/metabolismo , Citocinas/metabolismo , Encefalomielitis Autoinmune Experimental/genética , Inmunoterapia , Inflamación , Enfermedades Neuroinflamatorias , Microambiente Tumoral
2.
Nat Immunol ; 23(4): 632-642, 2022 04.
Artículo en Inglés | MEDLINE | ID: mdl-35301508

RESUMEN

Although inhibition of T cell coinhibitory receptors has revolutionized cancer therapy, the mechanisms governing their expression on human T cells have not been elucidated. In the present study, we show that type 1 interferon (IFN-I) regulates coinhibitory receptor expression on human T cells, inducing PD-1/TIM-3/LAG-3 while inhibiting TIGIT expression. High-temporal-resolution mRNA profiling of IFN-I responses established the dynamic regulatory networks uncovering three temporal transcriptional waves. Perturbation of key transcription factors (TFs) and TF footprint analysis revealed two regulator modules with different temporal kinetics that control expression of coinhibitory receptors and IFN-I response genes, with SP140 highlighted as one of the key regulators that differentiates LAG-3 and TIGIT expression. Finally, we found that the dynamic IFN-I response in vitro closely mirrored T cell features in acute SARS-CoV-2 infection. The identification of unique TFs controlling coinhibitory receptor expression under IFN-I response may provide targets for enhancement of immunotherapy in cancer, infectious diseases and autoimmunity.


Asunto(s)
COVID-19 , Interferón Tipo I , Redes Reguladoras de Genes , Humanos , Interferón Tipo I/genética , Receptores de Antígenos de Linfocitos T/metabolismo , Receptores Inmunológicos/genética , SARS-CoV-2 , Linfocitos T
3.
Cell ; 163(6): 1413-27, 2015 Dec 03.
Artículo en Inglés | MEDLINE | ID: mdl-26607793

RESUMEN

Th17 cells play a critical role in host defense against extracellular pathogens and tissue homeostasis but can induce autoimmunity. The mechanisms implicated in balancing "pathogenic" and "non-pathogenic" Th17 cell states remain largely unknown. We used single-cell RNA-seq to identify CD5L/AIM as a regulator expressed in non-pathogenic, but not in pathogenic Th17 cells. Although CD5L does not affect Th17 differentiation, it is a functional switch that regulates the pathogenicity of Th17 cells. Loss of CD5L converts non-pathogenic Th17 cells into pathogenic cells that induce autoimmunity. CD5L mediates this effect by modulating the intracellular lipidome, altering fatty acid composition and restricting cholesterol biosynthesis and, thus, ligand availability for Rorγt, the master transcription factor of Th17 cells. Our study identifies CD5L as a critical regulator of the Th17 cell functional state and highlights the importance of lipid metabolism in balancing immune protection and disease induced by T cells.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/metabolismo , Encefalomielitis Autoinmune Experimental/patología , Metabolismo de los Lípidos , Receptores Inmunológicos/metabolismo , Células Th17/patología , Animales , Diferenciación Celular , Sistema Nervioso Central/patología , Colesterol/biosíntesis , Encefalomielitis Autoinmune Experimental/inmunología , Ácidos Grasos Insaturados/metabolismo , Humanos , Ganglios Linfáticos/patología , Ratones , Miembro 3 del Grupo F de la Subfamilia 1 de Receptores Nucleares/metabolismo , Receptores Huérfanos Similares al Receptor Tirosina Quinasa/metabolismo , Receptores Depuradores , Análisis de la Célula Individual , Células Th17/inmunología
4.
Nat Immunol ; 18(3): 344-353, 2017 03.
Artículo en Inglés | MEDLINE | ID: mdl-28114290

RESUMEN

Although master transcription factors (TFs) are key to the development of specific T cell subsets, whether additional transcriptional regulators are induced by the same stimuli that dominantly repress the development of other, non-specific T cell lineages has not been fully elucidated. Through the use of regulatory T cells (Treg cells) induced by transforming growth factor-ß (TGF-ß), we identified the TF musculin (MSC) as being critical for the development of induced Treg cells (iTreg cells) by repression of the T helper type 2 (TH2) transcriptional program. Loss of MSC reduced expression of the Treg cell master TF Foxp3 and induced TH2 differentiation even under iTreg-cell-differentiation conditions. MSC interrupted binding of the TF GATA-3 to the locus encoding TH2-cell-related cytokines and diminished intrachromosomal interactions within that locus. MSC-deficient (Msc-/-) iTreg cells were unable to suppress TH2 responses, and Msc-/- mice spontaneously developed gut and lung inflammation with age. MSC therefore enforced Foxp3 expression and promoted the unidirectional induction of iTreg cells by repressing the TH2 developmental program.


Asunto(s)
Diferenciación Celular , Inflamación , Mucosa Intestinal/inmunología , Neumonía/inmunología , Linfocitos T Reguladores/fisiología , Células Th2/fisiología , Factores de Transcripción/metabolismo , Animales , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico , Células Cultivadas , Factores de Transcripción Forkhead/metabolismo , Factor de Transcripción GATA3/metabolismo , Regulación de la Expresión Génica , Inflamación/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factores de Transcripción/genética , Transcripción Genética , Factor de Crecimiento Transformador beta/metabolismo
5.
Nat Immunol ; 18(4): 412-421, 2017 04.
Artículo en Inglés | MEDLINE | ID: mdl-28166218

RESUMEN

Type 1 regulatory T cells (Tr1 cells) are induced by interleukin-27 (IL-27) and have critical roles in the control of autoimmunity and resolution of inflammation. We found that the transcription factors IRF1 and BATF were induced early on after treatment with IL-27 and were required for the differentiation and function of Tr1 cells in vitro and in vivo. Epigenetic and transcriptional analyses revealed that both transcription factors influenced chromatin accessibility and expression of the genes required for Tr1 cell function. IRF1 and BATF deficiencies uniquely altered the chromatin landscape, suggesting that these factors serve a pioneering function during Tr1 cell differentiation.


Asunto(s)
Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/metabolismo , Diferenciación Celular/inmunología , Cromatina/metabolismo , Factor 1 Regulador del Interferón/metabolismo , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/metabolismo , Animales , Enfermedades Autoinmunes/genética , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/metabolismo , Autoinmunidad , Factores de Transcripción con Cremalleras de Leucina de Carácter Básico/genética , Diferenciación Celular/genética , Cromatina/genética , Análisis por Conglomerados , Citocinas/metabolismo , Citocinas/farmacología , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Factor 1 Regulador del Interferón/genética , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , Subgrupos de Linfocitos T/efectos de los fármacos , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo , Linfocitos T Reguladores/citología , Linfocitos T Reguladores/efectos de los fármacos , Factores de Transcripción/metabolismo , Transcriptoma
6.
Immunity ; 53(3): 658-671.e6, 2020 09 15.
Artículo en Inglés | MEDLINE | ID: mdl-32937153

RESUMEN

Identifying signals in the tumor microenvironment (TME) that shape CD8+ T cell phenotype can inform novel therapeutic approaches for cancer. Here, we identified a gradient of increasing glucocorticoid receptor (GR) expression and signaling from naïve to dysfunctional CD8+ tumor-infiltrating lymphocytes (TILs). Conditional deletion of the GR in CD8+ TILs improved effector differentiation, reduced expression of the transcription factor TCF-1, and inhibited the dysfunctional phenotype, culminating in tumor growth inhibition. GR signaling transactivated the expression of multiple checkpoint receptors and promoted the induction of dysfunction-associated genes upon T cell activation. In the TME, monocyte-macrophage lineage cells produced glucocorticoids and genetic ablation of steroidogenesis in these cells as well as localized pharmacologic inhibition of glucocorticoid biosynthesis improved tumor growth control. Active glucocorticoid signaling associated with failure to respond to checkpoint blockade in both preclinical models and melanoma patients. Thus, endogenous steroid hormone signaling in CD8+ TILs promotes dysfunction, with important implications for cancer immunotherapy.


Asunto(s)
Linfocitos T CD8-positivos/inmunología , Glucocorticoides/metabolismo , Macrófagos/metabolismo , Melanoma Experimental/patología , Microambiente Tumoral/inmunología , Animales , Linfocitos T CD8-positivos/citología , Línea Celular Tumoral , Hematopoyesis/inmunología , Factor Nuclear 1-alfa del Hepatocito/biosíntesis , Inhibidores de Puntos de Control Inmunológico , Activación de Linfocitos/inmunología , Macrófagos/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Receptores de Glucocorticoides/genética , Receptores de Glucocorticoides/metabolismo , Transducción de Señal/inmunología
7.
Immunity ; 50(1): 181-194.e6, 2019 01 15.
Artículo en Inglés | MEDLINE | ID: mdl-30635236

RESUMEN

An improved understanding of the anti-tumor CD8+ T cell response after checkpoint blockade would enable more informed and effective therapeutic strategies. Here we examined the dynamics of the effector response of CD8+ tumor-infiltrating lymphocytes (TILs) after checkpoint blockade therapy. Bulk and single-cell RNA profiles of CD8+ TILs after combined Tim-3+PD-1 blockade in preclinical models revealed significant changes in the transcriptional profile of PD-1- TILs. These cells could be divided into subsets bearing characterstics of naive-, effector-, and memory-precursor-like cells. Effector- and memory-precursor-like TILs contained tumor-antigen-specific cells, exhibited proliferative and effector capacity, and expanded in response to different checkpoint blockade therapies across different tumor models. The memory-precursor-like subset shared features with CD8+ T cells associated with response to checkpoint blockade in patients and was compromised in the absence of Tcf7. Expression of Tcf7/Tcf1 was requisite for the efficacy of diverse immunotherapies, highlighting the importance of this transcriptional regulator in the development of effective CD8+ T cell responses upon immunotherapy.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Linfocitos T CD8-positivos/inmunología , Linfocitos Infiltrantes de Tumor/inmunología , Neoplasias Experimentales/terapia , Receptor de Muerte Celular Programada 1/antagonistas & inhibidores , Subgrupos de Linfocitos T/inmunología , Animales , Antígenos de Neoplasias/inmunología , Linfocitos T CD8-positivos/efectos de los fármacos , Proliferación Celular , Receptor 2 Celular del Virus de la Hepatitis A/antagonistas & inhibidores , Factor Nuclear 1-alfa del Hepatocito/genética , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Humanos , Memoria Inmunológica/genética , Inmunoterapia , Linfocitos Infiltrantes de Tumor/efectos de los fármacos , Melanoma Experimental , Ratones , Ratones Endogámicos C57BL , Neoplasias Experimentales/inmunología , Transcriptoma
8.
Immunity ; 47(3): 566-581.e9, 2017 09 19.
Artículo en Inglés | MEDLINE | ID: mdl-28930663

RESUMEN

Microglia play a pivotal role in the maintenance of brain homeostasis but lose homeostatic function during neurodegenerative disorders. We identified a specific apolipoprotein E (APOE)-dependent molecular signature in microglia from models of amyotrophic lateral sclerosis (ALS), multiple sclerosis (MS), and Alzheimer's disease (AD) and in microglia surrounding neuritic ß-amyloid (Aß)-plaques in the brains of people with AD. The APOE pathway mediated a switch from a homeostatic to a neurodegenerative microglia phenotype after phagocytosis of apoptotic neurons. TREM2 (triggering receptor expressed on myeloid cells 2) induced APOE signaling, and targeting the TREM2-APOE pathway restored the homeostatic signature of microglia in ALS and AD mouse models and prevented neuronal loss in an acute model of neurodegeneration. APOE-mediated neurodegenerative microglia had lost their tolerogenic function. Our work identifies the TREM2-APOE pathway as a major regulator of microglial functional phenotype in neurodegenerative diseases and serves as a novel target that could aid in the restoration of homeostatic microglia.


Asunto(s)
Apolipoproteínas E/metabolismo , Glicoproteínas de Membrana/metabolismo , Microglía/metabolismo , Enfermedades Neurodegenerativas/genética , Enfermedades Neurodegenerativas/metabolismo , Receptores Inmunológicos/metabolismo , Transducción de Señal , Transcriptoma , Enfermedad de Alzheimer/genética , Enfermedad de Alzheimer/metabolismo , Enfermedad de Alzheimer/patología , Péptidos beta-Amiloides/metabolismo , Precursor de Proteína beta-Amiloide/metabolismo , Animales , Apolipoproteínas E/deficiencia , Apolipoproteínas E/genética , Apoptosis/genética , Apoptosis/inmunología , Corteza Cerebral/metabolismo , Corteza Cerebral/patología , Análisis por Conglomerados , Modelos Animales de Enfermedad , Encefalomielitis Autoinmune Experimental , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Marcación de Gen , Humanos , Tolerancia Inmunológica , Ratones , Ratones Noqueados , Ratones Transgénicos , Microglía/inmunología , Monocitos/inmunología , Monocitos/metabolismo , Enfermedades Neurodegenerativas/inmunología , Neuronas/metabolismo , Fagocitosis/genética , Fagocitosis/inmunología , Fenotipo , Placa Amiloide/metabolismo , Placa Amiloide/patología , Superóxido Dismutasa-1/genética , Superóxido Dismutasa-1/metabolismo , Factor de Crecimiento Transformador beta/metabolismo
9.
Nature ; 558(7710): 454-459, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29899446

RESUMEN

The expression of co-inhibitory receptors, such as CTLA-4 and PD-1, on effector T cells is a key mechanism for ensuring immune homeostasis. Dysregulated expression of co-inhibitory receptors on CD4+ T cells promotes autoimmunity, whereas sustained overexpression on CD8+ T cells promotes T cell dysfunction or exhaustion, leading to impaired ability to clear chronic viral infections and diseases such as cancer1,2. Here, using RNA and protein expression profiling at single-cell resolution in mouse cells, we identify a module of co-inhibitory receptors that includes not only several known co-inhibitory receptors (PD-1, TIM-3, LAG-3 and TIGIT) but also many new surface receptors. We functionally validated two new co-inhibitory receptors, activated protein C receptor (PROCR) and podoplanin (PDPN). The module of co-inhibitory receptors is co-expressed in both CD4+ and CD8+ T cells and is part of a larger co-inhibitory gene program that is shared by non-responsive T cells in several physiological contexts and is driven by the immunoregulatory cytokine IL-27. Computational analysis identified the transcription factors PRDM1 and c-MAF as cooperative regulators of the co-inhibitory module, and this was validated experimentally. This molecular circuit underlies the co-expression of co-inhibitory receptors in T cells and identifies regulators of T cell function with the potential to control autoimmunity and tumour immunity.


Asunto(s)
Linfocitos T CD4-Positivos/citología , Linfocitos T CD4-Positivos/metabolismo , Linfocitos T CD8-positivos/citología , Linfocitos T CD8-positivos/metabolismo , Redes Reguladoras de Genes/genética , Melanoma/inmunología , Transcripción Genética , Animales , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD8-positivos/inmunología , Femenino , Tolerancia Inmunológica/genética , Tolerancia Inmunológica/inmunología , Interleucina-27/inmunología , Linfocitos Infiltrantes de Tumor/citología , Linfocitos Infiltrantes de Tumor/inmunología , Linfocitos Infiltrantes de Tumor/metabolismo , Masculino , Melanoma/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 1 de Unión al Dominio 1 de Regulación Positiva/metabolismo , Proteínas Proto-Oncogénicas c-maf/metabolismo , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Reproducibilidad de los Resultados
10.
Brain ; 145(9): 3288-3307, 2022 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-35899587

RESUMEN

Malignant brain tumours are the cause of a disproportionate level of morbidity and mortality among cancer patients, an unfortunate statistic that has remained constant for decades. Despite considerable advances in the molecular characterization of these tumours, targeting the cancer cells has yet to produce significant advances in treatment. An alternative strategy is to target cells in the glioblastoma microenvironment, such as tumour-associated astrocytes. Astrocytes control multiple processes in health and disease, ranging from maintaining the brain's metabolic homeostasis, to modulating neuroinflammation. However, their role in glioblastoma pathogenicity is not well understood. Here we report that depletion of reactive astrocytes regresses glioblastoma and prolongs mouse survival. Analysis of the tumour-associated astrocyte translatome revealed astrocytes initiate transcriptional programmes that shape the immune and metabolic compartments in the glioma microenvironment. Specifically, their expression of CCL2 and CSF1 governs the recruitment of tumour-associated macrophages and promotes a pro-tumourigenic macrophage phenotype. Concomitantly, we demonstrate that astrocyte-derived cholesterol is key to glioma cell survival, and that targeting astrocytic cholesterol efflux, via ABCA1, halts tumour progression. In summary, astrocytes control glioblastoma pathogenicity by reprogramming the immunological properties of the tumour microenvironment and supporting the non-oncogenic metabolic dependency of glioblastoma on cholesterol. These findings suggest that targeting astrocyte immunometabolic signalling may be useful in treating this uniformly lethal brain tumour.


Asunto(s)
Neoplasias Encefálicas , Glioblastoma , Glioma , Animales , Astrocitos/metabolismo , Neoplasias Encefálicas/metabolismo , Glioblastoma/metabolismo , Glioma/genética , Ratones , Microambiente Tumoral , Virulencia
11.
Eur J Immunol ; 47(11): 1970-1981, 2017 11.
Artículo en Inglés | MEDLINE | ID: mdl-28691750

RESUMEN

An association between T-cell lymphopenia and autoimmunity has long been proposed, but it remains to be elucidated whether T-cell lymphopenia affects B-cell responses to autoantigens. Human neonatal thymectomy (Tx) results in a decrease in T-cell numbers and we used this model to study the development of autoreactivity. Two cohorts of neonatally thymectomized individuals were examined, a cohort of young (1-5 years post-Tx, n = 10-27) and older children (>10 years, n = 26), and compared to healthy age-matched controls. T-cell and B-cell subsets were assessed and autoantibody profiling performed. Early post-Tx, a decrease in T-cell numbers (2.75 × 109 /L vs. 0.71 × 109 /L) and an increased proportion of memory T cells (19.72 vs. 57.43%) were observed. The presence of autoantibodies was correlated with an increased proportion of memory T cells in thymectomized children. No differences were seen in percentages of different B-cell subsets between the groups. The autoantigen microarray showed a skewed autoantibody response after Tx. In the cohort of older individuals, autoantibodies were present in 62% of the thymectomized children, while they were found in only 33% of the healthy controls. Overall, our data suggest that neonatal Tx skews the autoantibody profile. Preferential expansion and preservation of Treg (regulatory T) cell stability and function, may contribute to preventing autoimmune disease development after Tx.


Asunto(s)
Autoanticuerpos/inmunología , Autoinmunidad/inmunología , Linfocitos B/inmunología , Linfocitos T/inmunología , Timectomía/efectos adversos , Autoantígenos/inmunología , Niño , Preescolar , Femenino , Humanos , Memoria Inmunológica/inmunología , Lactante , Recién Nacido , Masculino
12.
PLoS Pathog ; 12(3): e1005490, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26967901

RESUMEN

While T cell immunity initially limits Mycobacterium tuberculosis infection, why T cell immunity fails to sterilize the infection and allows recrudescence is not clear. One hypothesis is that T cell exhaustion impairs immunity and is detrimental to the outcome of M. tuberculosis infection. Here we provide functional evidence for the development T cell exhaustion during chronic TB. Second, we evaluate the role of the inhibitory receptor T cell immunoglobulin and mucin domain-containing-3 (TIM3) during chronic M. tuberculosis infection. We find that TIM3 expressing T cells accumulate during chronic infection, co-express other inhibitory receptors including PD1, produce less IL-2 and TNF but more IL-10, and are functionally exhausted. Finally, we show that TIM3 blockade restores T cell function and improves bacterial control, particularly in chronically infected susceptible mice. These data show that T cell immunity is suboptimal during chronic M. tuberculosis infection due to T cell exhaustion. Moreover, in chronically infected mice, treatment with anti-TIM3 mAb is an effective therapeutic strategy against tuberculosis.


Asunto(s)
Mycobacterium tuberculosis/inmunología , Receptores Virales/metabolismo , Linfocitos T/inmunología , Tuberculosis/inmunología , Animales , Diferenciación Celular , Receptor 2 Celular del Virus de la Hepatitis A , Humanos , Inmunidad , Interleucina-10/metabolismo , Interleucina-2/metabolismo , Ratones , Receptores Virales/genética
13.
Genome Res ; 24(10): 1603-12, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25024161

RESUMEN

The T-cell receptor (TCR) repertoire is formed by random recombinations of genomic precursor elements; the resulting combinatorial diversity renders unlikely extensive TCR sharing between individuals. Here, we studied CDR3ß amino acid sequence sharing in a repertoire-wide manner, using high-throughput TCR-seq in 28 healthy mice. We uncovered hundreds of public sequences shared by most mice. Public CDR3 sequences, relative to private sequences, are two orders of magnitude more abundant on average, express restricted V/J segments, and feature high convergent nucleic acid recombination. Functionally, public sequences are enriched for MHC-diverse CDR3 sequences that were previously associated with autoimmune, allograft, and tumor-related reactions, but not with anti-pathogen-related reactions. Public CDR3 sequences are shared between mice of different MHC haplotypes, but are associated with different, MHC-dependent, V genes. Thus, despite their random generation process, TCR repertoires express a degree of uniformity in their post-genomic organization. These results, together with numerical simulations of TCR genomic rearrangements, suggest that biases and convergence in TCR recombination combine with ongoing selection to generate a restricted subset of self-associated, public CDR3 TCR sequences, and invite reexamination of the basic mechanisms of T-cell repertoire formation.


Asunto(s)
Linfocitos T CD4-Positivos/metabolismo , Motivos de Nucleótidos , Receptores de Antígenos de Linfocitos T alfa-beta/genética , Análisis de Secuencia de ARN/métodos , Animales , Linfocitos T CD4-Positivos/inmunología , Femenino , Ratones , Modelos Genéticos , Datos de Secuencia Molecular , Receptores de Antígenos de Linfocitos T alfa-beta/inmunología , Recombinación V(D)J
14.
J Immunol ; 194(11): 5272-81, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25917091

RESUMEN

In this work, we studied autoantibody repertoires and Ig isotypes in 71 mothers and their 104 healthy newborns (including twins and triplets delivered term or premature). Newborns receive maternal IgG Abs via the placenta before birth, but developing infants must produce their own IgM and IgA Abs. We used an Ag microarray analysis to detect binding to a selection of 295 self-Ags, compared with 27 standard foreign Ags. The magnitude of binding to specific self-Ags was found to be not less than that to the foreign Ags. As expected, each newborn shared with its mother a similar IgG repertoire-manifest as early as the 24th week of gestation. IgM and IgA autoantibody repertoires in cord sera were highly correlated among the newborns and differed from their mothers' repertoires; the latter differed in sera and milk. The autoantibodies bound to self-Ags known to be associated with tumors and to autoimmune diseases. Thus, autoantibody repertoires in healthy humans--the immunological homunculus--arise congenitally, differ in maternal milk and sera, and mark the potential of the immune system to attack tumors, beneficially, or healthy tissues, harmfully; regulation of the tissue site, the dynamics, and the response phenotype of homuncular autoimmunity very likely affects health.


Asunto(s)
Anticuerpos Antineoplásicos/sangre , Autoanticuerpos/sangre , Calostro/inmunología , Sangre Fetal/inmunología , Isotipos de Inmunoglobulinas/sangre , Anticuerpos Antineoplásicos/inmunología , Autoanticuerpos/inmunología , Autoantígenos/inmunología , Enfermedades Autoinmunes/inmunología , Humanos , Inmunoglobulina A/sangre , Inmunoglobulina A/inmunología , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Isotipos de Inmunoglobulinas/inmunología , Inmunoglobulina M/sangre , Inmunoglobulina M/inmunología , Recién Nacido , Leche Humana/inmunología , Neoplasias/inmunología
15.
Brain ; 139(Pt 7): 1939-57, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27246324

RESUMEN

SEE WINGER AND ZAMVIL DOI101093/BRAIN/AWW121 FOR A SCIENTIFIC COMMENTARY ON THIS ARTICLE: The innate immune system plays a central role in the chronic central nervous system inflammation that drives neurological disability in progressive forms of multiple sclerosis, for which there are no effective treatments. The mucosal immune system is a unique tolerogenic organ that provides a physiological approach for the induction of regulatory T cells. Here we report that nasal administration of CD3-specific antibody ameliorates disease in a progressive animal model of multiple sclerosis. This effect is IL-10-dependent and is mediated by the induction of regulatory T cells that share a similar transcriptional profile to Tr1 regulatory cells and that suppress the astrocyte inflammatory transcriptional program. Treatment results in an attenuated inflammatory milieu in the central nervous system, decreased microglia activation, reduced recruitment of peripheral monocytes, stabilization of the blood-brain barrier and less neurodegeneration. These findings suggest a new therapeutic approach for the treatment of progressive forms of multiple sclerosis and potentially other types of chronic central nervous system inflammation.


Asunto(s)
Astrocitos/inmunología , Complejo CD3/inmunología , Encefalomielitis Autoinmune Experimental/inmunología , Factores Inmunológicos/farmacología , Interleucina-10/inmunología , Muromonab-CD3/farmacología , Linfocitos T Reguladores/inmunología , Administración Intranasal , Animales , Encefalomielitis Autoinmune Experimental/tratamiento farmacológico , Femenino , Factores Inmunológicos/administración & dosificación , Ratones , Ratones Endogámicos C57BL , Ratones Endogámicos NOD , Muromonab-CD3/administración & dosificación , Neumonía Neumocócica/inmunología
16.
Bioinformatics ; 30(22): 3181-8, 2014 Nov 15.
Artículo en Inglés | MEDLINE | ID: mdl-25095879

RESUMEN

MOTIVATION: The clonal theory of adaptive immunity proposes that immunological responses are encoded by increases in the frequency of lymphocytes carrying antigen-specific receptors. In this study, we measure the frequency of different T-cell receptors (TcR) in CD4 + T cell populations of mice immunized with a complex antigen, killed Mycobacterium tuberculosis, using high throughput parallel sequencing of the TcRß chain. Our initial hypothesis that immunization would induce repertoire convergence proved to be incorrect, and therefore an alternative approach was developed that allows accurate stratification of TcR repertoires and provides novel insights into the nature of CD4 + T-cell receptor recognition. RESULTS: To track the changes induced by immunization within this heterogeneous repertoire, the sequence data were classified by counting the frequency of different clusters of short (3 or 4) continuous stretches of amino acids within the antigen binding complementarity determining region 3 (CDR3) repertoire of different mice. Both unsupervised (hierarchical clustering) and supervised (support vector machine) analyses of these different distributions of sequence clusters differentiated between immunized and unimmunized mice with 100% efficiency. The CD4 + TcR repertoires of mice 5 and 14 days postimmunization were clearly different from that of unimmunized mice but were not distinguishable from each other. However, the repertoires of mice 60 days postimmunization were distinct both from naive mice and the day 5/14 animals. Our results reinforce the remarkable diversity of the TcR repertoire, resulting in many diverse private TcRs contributing to the T-cell response even in genetically identical mice responding to the same antigen. However, specific motifs defined by short stretches of amino acids within the CDR3 region may determine TcR specificity and define a new approach to TcR sequence classification. AVAILABILITY AND IMPLEMENTATION: The analysis was implemented in R and Python, and source code can be found in Supplementary Data. CONTACT: b.chain@ucl.ac.uk SUPPLEMENTARY INFORMATION: Supplementary data are available at Bioinformatics online.


Asunto(s)
Linfocitos T CD4-Positivos/inmunología , Regiones Determinantes de Complementariedad/química , Receptores de Antígenos de Linfocitos T/química , Secuencia de Aminoácidos , Animales , Análisis por Conglomerados , Inmunización , Ratones , Mycobacterium tuberculosis/inmunología , Receptores de Antígenos de Linfocitos T/inmunología , Receptores de Antígenos de Linfocitos T alfa-beta/química , Análisis de Secuencia de Proteína , Máquina de Vectores de Soporte
17.
J Leukoc Biol ; 2024 Mar 13.
Artículo en Inglés | MEDLINE | ID: mdl-38478700

RESUMEN

Eosinophils have been mainly studied in allergic diseases and parasitic infections. Nonetheless, eosinophils accumulate in a variety of solid tumors, including colorectal cancer, where their presence is associated with improved prognosis. Eosinophils can promote anti-tumor immunity through various mechanisms, including direct cytotoxicity towards tumor cells and promoting T cell activation. However, the mechanisms by which tumor cells regulate eosinophil activities are largely unknown. Herein, we characterized the potential interactions between eosinophils and colorectal cancer cells using an unbiased transcriptomic and proteomic analyses approach. Human eosinophils were stimulated with colorectal cancer cell-conditioned media, containing tumor cell-secreted factors from multiple cancer cell lines. RNA sequencing analysis identified a "core" signature consisting of 101 genes that characterize a baseline transcriptional program for the response of human eosinophils to colorectal cancer cells. Among these, the increased expression of IL-3Rα and its ßc chain was identified and validated at the protein level. Secreted factors from tumor cells potentiated IL-3-induced expression of the adhesion molecule CD11a in eosinophils. Combining proteomics analysis of tumor cell-secreted factors with RNA sequencing revealed potential ligand-receptor pairs between tumor cells and eosinophils and the potential involvement of the adhesion molecule CD18 and F2RL3/PAR4. Subsequent functional analyses demonstrated that F2RL3/PAR4 suppresses eosinophil migration in response tumor cell-secreted factors. These findings add to the growing body of evidence that eosinophils are conditioned by their local microenvironment. Identifying mechanisms by which eosinophils interact with tumor cells could lead to the development of new immunotherapies for colorectal cancer and other solid tumors.

18.
NPJ Syst Biol Appl ; 10(1): 66, 2024 Jun 10.
Artículo en Inglés | MEDLINE | ID: mdl-38858414

RESUMEN

Cell-cell crosstalk involves simultaneous interactions of multiple receptors and ligands, followed by downstream signaling cascades working through receptors converging at dominant transcription factors, which then integrate and propagate multiple signals into a cellular response. Single-cell RNAseq of multiple cell subsets isolated from a defined microenvironment provides us with a unique opportunity to learn about such interactions reflected in their gene expression levels. We developed the interFLOW framework to map the potential ligand-receptor interactions between different cell subsets based on a maximum flow computation in a network of protein-protein interactions (PPIs). The maximum flow approach further allows characterization of the intracellular downstream signal transduction from differentially expressed receptors towards dominant transcription factors, therefore, enabling the association between a set of receptors and their downstream activated pathways. Importantly, we were able to identify key transcription factors toward which the convergence of multiple receptor signaling pathways occurs. These identified factors have a unique role in the integration and propagation of signaling following specific cell-cell interactions.


Asunto(s)
Transducción de Señal , Transducción de Señal/fisiología , Humanos , Factores de Transcripción/metabolismo , Factores de Transcripción/genética , Análisis de la Célula Individual/métodos , Comunicación Celular/fisiología , Biología Computacional/métodos , Ligandos , Mapas de Interacción de Proteínas/genética , Modelos Biológicos
19.
Cell Rep Med ; 5(7): 101640, 2024 Jul 16.
Artículo en Inglés | MEDLINE | ID: mdl-38959885

RESUMEN

CD8+ T cells must persist and function in diverse tumor microenvironments to exert their effects. Thus, understanding common underlying expression programs could better inform the next generation of immunotherapies. We apply a generalizable matrix factorization algorithm that recovers both shared and context-specific expression programs from diverse datasets to a single-cell RNA sequencing (scRNA-seq) compendium of 33,161 CD8+ T cells from 132 patients with seven human cancers. Our meta-single-cell analyses uncover a pan-cancer T cell dysfunction program that predicts clinical non-response to checkpoint blockade in melanoma and highlights CXCR6 as a pan-cancer marker of chronically activated T cells. Cxcr6 is trans-activated by AP-1 and repressed by TCF1. Using mouse models, we show that Cxcr6 deletion in CD8+ T cells increases apoptosis of PD1+TIM3+ cells, dampens CD28 signaling, and compromises tumor growth control. Our study uncovers a TCF1:CXCR6 axis that counterbalances PD1-mediated suppression of CD8+ cell responses and is essential for effective anti-tumor immunity.


Asunto(s)
Antígenos CD28 , Linfocitos T CD8-positivos , Factor Nuclear 1-alfa del Hepatocito , Receptores CXCR6 , Linfocitos T CD8-positivos/inmunología , Linfocitos T CD8-positivos/metabolismo , Animales , Humanos , Antígenos CD28/metabolismo , Antígenos CD28/genética , Antígenos CD28/inmunología , Factor Nuclear 1-alfa del Hepatocito/metabolismo , Factor Nuclear 1-alfa del Hepatocito/genética , Ratones , Receptores CXCR6/metabolismo , Receptores CXCR6/genética , Neoplasias/inmunología , Neoplasias/genética , Neoplasias/patología , Análisis de la Célula Individual/métodos , Transducción de Señal , Microambiente Tumoral/inmunología , Ratones Endogámicos C57BL
20.
Phys Biol ; 10(2): 025003, 2013 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-23492831

RESUMEN

Networks can be found everywhere-in technology, in nature and in our bodies. In this paper we present how antigen networks can be used as a model to study network interaction and architecture. Utilizing antigen microarray data of the reactivity of hundreds of antibodies of sera of ten mothers and their newborns, we reconstruct networks, either isotype specific (IgM or IgG) or person specific-mothers or newborns-and investigate the network properties. Such an approach makes it possible to decipher fundamental information regarding the personal immune network state and its unique characteristics. In the current paper we demonstrate how we are successful in studying the interaction between two dependent networks, the maternal IgG repertoire and the one of the offspring, using the concept of meta-network provides essential information regarding the biological phenomenon of cross placental transfer. Such an approach is useful in the study of coupled networks in variety of scientific fields.


Asunto(s)
Antígenos/inmunología , Inmunoglobulina G/inmunología , Inmunoglobulina M/inmunología , Mapas de Interacción de Proteínas , Femenino , Humanos , Recién Nacido
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA