Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 14 de 14
Filtrer
Plus de filtres










Gamme d'année
1.
Cancer Immunol Immunother ; 73(9): 174, 2024 Jul 02.
Article de Anglais | MEDLINE | ID: mdl-38953978

RÉSUMÉ

Γδ T cell infiltration into tumours usually correlates with improved patient outcome, but both tumour-promoting and tumoricidal effects of γδ T cells have been documented. Human γδ T cells can be divided into functionally distinct subsets based on T cell receptor (TCR) Vδ usage. Still, the contribution of these different subsets to tumour immunity remains elusive. Here, we provide a detailed γδ T cell profiling in colon tumours, using mass and flow cytometry, mRNA quantification, and TCR sequencing. δ chain usage in both the macroscopically unaffected colon mucosa and tumours varied considerably between patients, with substantial fractions of Vδ1, Vδ2, and non-Vδ1 Vδ2 cells. Sequencing of the Vδ complementarity-determining region 3 showed that almost all non-Vδ1 Vδ2 cells used Vδ3 and that tumour-infiltrating γδ clonotypes were unique for every patient. Non-Vδ1Vδ2 cells from colon tumours expressed several activation markers but few NK cell receptors and exhaustion markers. In addition, mRNA analyses showed that non-Vδ1 Vδ2 cells expressed several genes for proteins with tumour-promoting functions, such as neutrophil-recruiting chemokines, Galectin 3, and transforming growth factor-beta induced. In summary, our results show a large variation in γδ T cell subsets between individual tumours, and that Vδ3 cells make up a substantial proportion of γδ T cells in colon tumours. We suggest that individual γδ T cell composition in colon tumours may contribute to the balance between favourable and adverse immune responses, and thereby also patient outcome.


Sujet(s)
Adénocarcinome , Tumeurs du côlon , Récepteur lymphocytaire T antigène, gamma-delta , Humains , Récepteur lymphocytaire T antigène, gamma-delta/métabolisme , Récepteur lymphocytaire T antigène, gamma-delta/immunologie , Récepteur lymphocytaire T antigène, gamma-delta/génétique , Tumeurs du côlon/immunologie , Tumeurs du côlon/anatomopathologie , Tumeurs du côlon/génétique , Adénocarcinome/immunologie , Adénocarcinome/anatomopathologie , Adénocarcinome/génétique , Phénotype , Femelle , Mâle , Sous-populations de lymphocytes T/immunologie , Sous-populations de lymphocytes T/métabolisme , Sujet âgé , Adulte d'âge moyen , Lymphocytes TIL/immunologie , Lymphocytes TIL/métabolisme
2.
Cancers (Basel) ; 15(5)2023 Feb 24.
Article de Anglais | MEDLINE | ID: mdl-36900249

RÉSUMÉ

The incidence of left-sided colon and rectal cancer in young people are increasing worldwide, but its causes are poorly understood. It is not clear if the tumor microenvironment is dependent on age of onset, and little is known about the composition of tumor-infiltrating T cells in early-onset colorectal cancer (EOCRC). To address this, we investigated T-cell subsets and performed gene expression immune profiling in sporadic EOCRC tumors and matched average-onset colorectal cancer (AOCRC) tumors. Left-sided colon and rectal tumors from 40 cases were analyzed; 20 EOCRC (<45 years) patients were matched 1:1 to AOCRC (70-75 years) patients by gender, tumor location, and stage. Cases with germline pathogenic variants, inflammatory bowel disease or neoadjuvant-treated tumors were excluded. For T cells in tumors and stroma, a multiplex immunofluorescence assay combined with digital image analysis and machine learning algorithms was used. Immunological mediators in the tumor microenvironment were assessed by NanoString gene expression profiling of mRNA. Immunofluorescence revealed no significant difference between EOCRC and AOCRC with regard to infiltration of total T cells, conventional CD4+ and CD8+ T cells, regulatory T cells, or γδ T cells. Most T cells were located in the stroma in both EOCRC and AOCRC. Immune profiling by gene expression revealed higher expression in AOCRC of the immunoregulatory cytokine IL-10, the inhibitory NK cell receptors KIR3DL3 and KLRB1 (CD161), and IFN-a7 (IFNA7). In contrast, the interferon-induced gene IFIT2 was more highly expressed in EOCRC. However, in a global analysis of 770 tumor immunity genes, no significant differences could be detected. T-cell infiltration and expression of inflammatory mediators are similar in EOCRC and AOCRC. This may indicate that the immune response to cancer in left colon and rectum is not related to age of onset and that EOCRC is likely not driven by immune response deficiency.

3.
Clin. transl. oncol. (Print) ; 24(9): 1818–1827, septiembre 2022. ilus, tab
Article de Anglais | IBECS | ID: ibc-206267

RÉSUMÉ

BackgroundTumor-associated macrophages (TAM) are known to facilitate colorectal cancer (CRC) growth. High macrophage infiltration in thymidine phosphorylase (TYMP) expressing CRC may correspond to poor prognosis. The prognostic impact of the expression CD163, a receptor associated with TAM, and TYMP in stroma, respectively, tumor tissue is not yet established. The aim of this study was to identify the potential associations between TYMP and CD163 expression levels and relapse-free survival (RFS) of patients with stage II CRC, and if microdissection is of importance.MethodsStage II CRC patients, radically resected with relapse (n = 104), were matched to patients with a 5-year relapse-free follow-up (n = 206). Gene expression of TYMP and CD163 was analyzed in snap-frozen tumor tissues and in microdissected formalin-fixed tumor tissues separated into tumor epithelium and stroma.ResultsTYMP expression was high in poorly differentiated tumors, right-sided CRC, and tumors with high microsatellite instability CD163-expressing macrophages near tumor epithelial cells had high expression in poorly differentiated and T4 tumors. High TYMP expression in tumor epithelial cells was in the multivariate analyses associated with shorter relapse-free survival (hazard ratio 1.66; 95% confidence interval: 1.09–2.56; p < 0.05).ConclusionsTYMP expression in tumor epithelial cells was associated with RFS and emphasizes the need for tissue microdissection. Additional studies are needed to establish whether TYMP and CD163 could add clinically relevant information to identify high-risk stage II patients that could benefit from adjuvant chemotherapy. (AU)


Sujet(s)
Humains , Antigènes CD/génétique , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Tumeurs colorectales/anatomopathologie , Récepteurs de surface cellulaire , Pronostic
4.
Clin Transl Oncol ; 24(9): 1818-1827, 2022 Sep.
Article de Anglais | MEDLINE | ID: mdl-35567733

RÉSUMÉ

BACKGROUND: Tumor-associated macrophages (TAM) are known to facilitate colorectal cancer (CRC) growth. High macrophage infiltration in thymidine phosphorylase (TYMP) expressing CRC may correspond to poor prognosis. The prognostic impact of the expression CD163, a receptor associated with TAM, and TYMP in stroma, respectively, tumor tissue is not yet established. The aim of this study was to identify the potential associations between TYMP and CD163 expression levels and relapse-free survival (RFS) of patients with stage II CRC, and if microdissection is of importance. METHODS: Stage II CRC patients, radically resected with relapse (n = 104), were matched to patients with a 5-year relapse-free follow-up (n = 206). Gene expression of TYMP and CD163 was analyzed in snap-frozen tumor tissues and in microdissected formalin-fixed tumor tissues separated into tumor epithelium and stroma. RESULTS: TYMP expression was high in poorly differentiated tumors, right-sided CRC, and tumors with high microsatellite instability CD163-expressing macrophages near tumor epithelial cells had high expression in poorly differentiated and T4 tumors. High TYMP expression in tumor epithelial cells was in the multivariate analyses associated with shorter relapse-free survival (hazard ratio 1.66; 95% confidence interval: 1.09-2.56; p < 0.05). CONCLUSIONS: TYMP expression in tumor epithelial cells was associated with RFS and emphasizes the need for tissue microdissection. Additional studies are needed to establish whether TYMP and CD163 could add clinically relevant information to identify high-risk stage II patients that could benefit from adjuvant chemotherapy.


Sujet(s)
Antigènes CD , Antigènes de différenciation des myélomonocytes , Tumeurs colorectales , Thymidine phosphorylase , Antigènes CD/génétique , Antigènes de différenciation des myélomonocytes/génétique , Marqueurs biologiques tumoraux/génétique , Marqueurs biologiques tumoraux/métabolisme , Tumeurs colorectales/anatomopathologie , Humains , Récidive tumorale locale/anatomopathologie , Pronostic , Récepteurs de surface cellulaire , Thymidine phosphorylase/génétique
5.
Front Immunol ; 13: 813203, 2022.
Article de Anglais | MEDLINE | ID: mdl-35355990

RÉSUMÉ

B cells interact with T follicular helper (Tfh) cells in germinal centers (GCs) to generate high-affinity antibodies. Much less is known about how cognate T-B-cell interactions influence Th cells that enter circulation and peripheral tissues. Therefore, we generated mice lacking MHC-II expressing B cells and, by thoracic duct cannulation, analyzed Th cells in the efferent lymph at defined intervals post-immunization. Focusing on gut-draining mesenteric lymph nodes (MLNs), we show that antigen-specific α4ß7+ gut-homing effector Th cells enter the circulation prior to CXCR5+PD-1+ Tfh-like cells. B cells appear to have no or limited impact on the early generation and egress of gut-homing Th cells but are critical for the subsequent appearance of Tfh-like cells that peak in the lymph before GCs have developed. At this stage, antigen-presenting B cells also reduce the proportion of α4ß7+ Th cells in the MLN and efferent lymph. Furthermore, cognate B-cell interaction drives a broad transcriptional program in Th cells, including IL-4 that is confined to the Tfh cell lineage. The IL-4-producing Tfh-like cells originate from Bcl6+ precursors in the LNs and have gut-homing capacity. Hence, B cells program the efferent lymph Th cell response within a limited window of time after antigenic challenge.


Sujet(s)
Interleukine-4 , Lymphocytes T auxiliaires , Animaux , Lymphocytes B , Centre germinatif , Souris , Récepteurs CXCR5/génétique
6.
Cancers (Basel) ; 13(20)2021 Oct 19.
Article de Anglais | MEDLINE | ID: mdl-34680397

RÉSUMÉ

Although mouse models of CRC treatments have demonstrated robust immune activation, it remains unclear to what extent CRC patients' APCs and TILs interact to fuel or quench treatment-induced immune responses. Our ex vivo characterization of tumor and adjacent colon cell suspensions suggest that contrasting environments in these tissues promoted inversed expression of T cell co-stimulatory CD80, and co-inhibitory programmed death (PD)-ligand1 (PD-L1) on intratumoral vs. colonic APCs. While putative tumor-specific CD103+CD39+CD8+ TILs expressed lower CD69 (early activation marker) and higher PD-1 (extended activation/exhaustion marker) than colonic counterparts, the latter had instead higher CD69 and lower PD-1 levels. Functional comparisons showed that intratumoral APCs were inferior to colonic APCs regarding protein uptake and upregulation of CD80 and PD-L1 after protein degradation. Our attempt to model CRC treatment-induced T cell activation in vitro showed less interferon (IFN)-γ production by TILs than colonic T cells. In this model, we also measured APCs' CD80 and PD-L1 expression in response to activated co-residing T cells. These markers were comparable in the two tissues, despite higher IFN- γ exposure for colonic APCs. Thus, APCs within distinct intratumoral and colonic milieus showed different activation and functional status, but were similarly responsive to signals from induced T cell activation.

7.
BMC Immunol ; 22(1): 58, 2021 08 19.
Article de Anglais | MEDLINE | ID: mdl-34407765

RÉSUMÉ

BACKGROUND: Intratumoral regulatory T cells (Treg) in colon cancer are a heterogeneous cell population, with potential impact on patient outcome. Generally, a high Treg infiltration has been correlated to a worse patient outcome, but it is still unclear how the composition of different Treg subsets affects patient relapse and survival. In this study, we used mass and flow cytometry to characterize Treg in colon tumors and corresponding unaffected tissue, followed by a correlation to clinical parameters and patient outcome. RESULTS: Using mass cytometry, we defined 13 clusters of intestinal Treg, three of which were enriched in the tumors. The two most enriched clusters were defined by their expression of the proliferation marker Ki67 and CD56, respectively. The Treg accumulating in the tumors expressed inducible T-cell co-stimulator (ICOS), OX-40, and CD39, indicating that they were effector Treg (eTreg). Intratumoral CD39+ Treg also had a higher expression of Foxp3, suggesting a higher suppressive activity, and we subsequently used CD39 as a marker for eTreg. Our further studies showed that colon tumors can be divided into two tumor groups, based on the proportion of CD39+ putative eTreg in the tumors. This property was independent of both tumor microsatellite status and tumor stage, which are important factors in predicting cancer disease progression. In a prospective study of forty-four colon cancer patients, we also showed that patients with a high CD39 expression on tumor-infiltrating Treg have a tendency towards a less favorable patient outcome in terms of cumulative cancer-specific survival. CONCLUSIONS: This study uncovers novel subsets of tumor-infiltrating Treg in colon cancer, and suggests that CD39 may be a potential therapeutic target in patients with microsatellite stable colon tumors, which are usually refractory to checkpoint blockade therapy.


Sujet(s)
Tumeurs du côlon/immunologie , Lymphocytes TIL/immunologie , Sous-populations de lymphocytes T/immunologie , Lymphocytes T régulateurs/immunologie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Antigènes CD/métabolisme , Apyrase/métabolisme , Tumeurs du côlon/mortalité , Femelle , Facteurs de transcription Forkhead/métabolisme , Humains , Activation des lymphocytes , Mâle , Adulte d'âge moyen , Stadification tumorale , Pronostic , Analyse de survie
8.
Eur J Immunol ; 51(9): 2317-2329, 2021 09.
Article de Anglais | MEDLINE | ID: mdl-34272885

RÉSUMÉ

Endothelial cells are key regulators of transendothelial migration and their secretion of chemokines and expression of adhesion molecules facilitates lymphocyte entry into tissues. Previously, we demonstrated that Tregs can reduce transendothelial migration of T cells into tumors by decreasing endothelial CXCL10 secretion, but the mechanism by which this occurs is still not known. In this study, we aimed to define how Tregs decrease transendothelial migration into tumors. mRNA sequencing of intestinal tumor endothelial cells from Treg depleted mice identified neutral sphingomyelinase 2 (nSMase2) as a gene downregulated in the presence of Tregs. nSMase2 is expressed in human umbilical vein endothelial cells (HUVECs) and was decreased after coculture with Tregs. Furthermore, blocking of nSMase2 activity in vitro decreased VCAM1, CX3CL1, and CXCL10 expression in HUVECs, mirroring the same decrease found in Treg cocultures. In the APCmin/+ mouse model of intestinal cancer, nSMase2 is lower in tumor endothelial cells than in unaffected small intestine and chronic treatment with a nSMase2 inhibitor suppressed the increased migration that is otherwise seen in the absence of Tregs. We conclude that nSMase2 is an important mediator in endothelial cells supporting transendothelial migration, which may be targeted by Tregs to reduce T-cell migration into tumors.


Sujet(s)
Chimiokine CXCL10/métabolisme , Tumeurs du côlon/anatomopathologie , Lymphocytes TIL/immunologie , Sphingomyeline phosphodiesterase/métabolisme , Lymphocytes T régulateurs/immunologie , Migration transendothéliale et transépithéliale/physiologie , Animaux , Molécules d'adhérence cellulaire/biosynthèse , Lignée cellulaire , Chimiokine CX3CL1/biosynthèse , Chimiokine CXCL10/biosynthèse , Tumeurs du côlon/immunologie , Régulation négative , Femelle , Cellules endothéliales de la veine ombilicale humaine , Humains , Mâle , Souris , Souris transgéniques , Sous-populations de lymphocytes T/immunologie , Versicanes/biosynthèse
9.
Cancer Immunol Immunother ; 70(12): 3461-3475, 2021 Dec.
Article de Anglais | MEDLINE | ID: mdl-33885944

RÉSUMÉ

Mucosal-associated invariant T (MAIT) cells are unconventional T cells recognizing microbial metabolites, presented by the invariant MR1 protein. Upon activation, MAIT cells rapidly secrete cytokines and exert cytotoxic functions, and may thus be highly relevant also in tumor immunity. MAIT cells accumulate in colon tumors, but in contrast to other cytotoxic T cell subsets, their presence in tumors has been associated with worse patient outcome. Here we investigated if exhaustion may contribute to reduced anti-tumor immunity by MAIT cells. Freshly isolated lymphocytes from colon tumors, unaffected tissue and blood from the same patients were analyzed by flow cytometry to detect MAIT cells with effector functions that are relevant for tumor immunity, and their expression of inhibitory receptors and other exhaustion markers. Our studies show that MAIT cells with a PD-1highTim-3+CD39+ terminally exhausted phenotype and an increased proliferation accumulate in colon tumors. The exhausted MAIT cells have reduced polyfunctionality with regard to production of important anti-tumor effector molecules, and blocking antibodies to PD-1 partly improved activation of tumor-infiltrating MAIT cells in vitro. We conclude that the tumor microenvironment leads to exhaustion not only of conventional T cells, but also MAIT cells, and that checkpoint blockade therapy may be useful also to reinvigorate tumor-infiltrating MAIT cells.


Sujet(s)
Tumeurs du côlon/immunologie , Cellules T invariantes associées aux muqueuses/immunologie , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Antigènes CD/immunologie , Apyrase/immunologie , Marqueurs biologiques/métabolisme , Prolifération cellulaire/physiologie , Cytokines/immunologie , Femelle , Récepteur cellulaire-2 du virus de l'hépatite A/immunologie , Humains , Activation des lymphocytes/immunologie , Mâle , Adulte d'âge moyen , Phénotype , Récepteur-1 de mort cellulaire programmée/immunologie , Microenvironnement tumoral/immunologie
10.
J Crohns Colitis ; 15(8): 1346-1361, 2021 Aug 02.
Article de Anglais | MEDLINE | ID: mdl-33537747

RÉSUMÉ

BACKGROUND AND AIMS: Uncontrolled activation of intestinal mononuclear phagocytes [MNPs] drives chronic inflammation in inflammatory bowel disease [IBD]. Triggering receptor expressed on myeloid cells 1 [TREM-1] has been implicated in the pathogenesis of IBD. However, the role of TREM-1+ cell subsets in driving IBD pathology and the link with clinical parameters are not understood. We investigated TREM-1 expression in human intestinal MNP subsets and examined blocking TREM-1 as a potential IBD therapy. METHODS: TREM-1 gene expression was analysed in intestinal mucosa, enriched epithelial and lamina propria [LP] layers, and purified cells from controls and IBD patients. TREM-1 protein on immune cells was assessed by flow cytometry and immunofluorescence microscopy. Blood monocyte activation was examined by large-scale gene expression using a TREM-1 agonist or LP conditioned media [LP-CM] from patients in the presence or absence of TREM-1 and tumour necrosis factor [TNF] antagonist antibodies. RESULTS: TREM-1 gene expression increases in intestinal mucosa from IBD patients and correlates with disease score. TREM-1+ cells, which are mainly immature macrophages and CD11b+ granulocytes, increase among LP cells from Crohn's disease patients and their frequency correlates with inflammatory molecules in LP-CM. LP-CM from Crohn's disease patients induces an inflammatory transcriptome in blood monocytes, including increased IL-6 expression, which is reduced by simultaneous blocking of TREM-1 and TNF. CONCLUSIONS: High intestinal TREM-1 expression, reflecting a high frequency of TREM-1+ immature macrophages and TREM-1+CD11b+ granulocytes, is linked to the deleterious inflammatory microenvironment in IBD patients. Therefore, blocking the TREM-1 pathway, especially simultaneously with anti-TNF therapy, has potential as a new IBD therapy.


Sujet(s)
Maladie de Crohn/anatomopathologie , Muqueuse intestinale/métabolisme , Macrophages/métabolisme , Récepteur de déclenchement de type-1 exprimé sur les cellules myéloïdes/métabolisme , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Antigènes CD11b/métabolisme , Études cas-témoins , Femelle , Expression des gènes , Granulocytes/métabolisme , Humains , Interleukine-6/métabolisme , Muqueuse intestinale/anatomopathologie , Mâle , Adulte d'âge moyen , Monocytes/métabolisme , Jeune adulte
11.
Cancer Immunol Immunother ; 69(7): 1279-1292, 2020 Jul.
Article de Anglais | MEDLINE | ID: mdl-32185408

RÉSUMÉ

The presence of activated T cells in colorectal cancer tissues is a strong predictor of patient survival. Our previous studies have shown that regulatory T cells (Treg) are able to reduce T cell transendothelial migration in vitro and accumulation of effector T cells in intestinal tumors in vivo in the murine APCMin/+ model for microsatellite stable intestinal tumors. In this study, we investigated the effect of Treg depletion on the density and effector functions of different TCRαß+ and TCRγδ+ T cell populations in intestinal tumors. We used the APCMin/+\DEREG mouse model, which harbor a diphtheria toxin receptor under the control of the FOXP3 promoter, to deplete Treg in tumor bearing mice. We found that the density of conventional TCRαß+CD8αß+ T cells was significantly increased in Treg-depleted tumors in comparison with Treg-proficient tumors. Furthermore, TCRαß+CD8αß+ T cells showed increased proliferation and activation as well as increased Granzyme B and IFN-γ production in Treg-depleted tumors. In sharp contrast, the densities and effector functions of TCRαß+CD8αα+ T cells and TCRγδ+ T cells remained unchanged by Treg depletion. We also documented a distinct population of IL-17A+TNF+ TCRγδ+CD8- T cells in tumors, which were not affected by Treg depletion. We conclude that Treg depletion affects only conventional TCRαß+CD8αß+ T cells in intestinal tumors, while unconventional T cells and T cells in unaffected tissue are not altered. Immunotherapies aimed at depleting Treg from tumors may thus be a viable option for reinvigoration of conventional cytotoxic T cells with a Th1 cytokine profile.


Sujet(s)
Protéine de la polypose adénomateuse colique/physiologie , Antigènes CD8/immunologie , Lymphocytes T CD8+/immunologie , Tumeurs de l'intestin/immunologie , Lymphocytes TIL/immunologie , Récepteur lymphocytaire T antigène, alpha-bêta/immunologie , Lymphocytes T régulateurs/immunologie , Animaux , Antigènes CD8/métabolisme , Lymphocytes T CD8+/métabolisme , Femelle , Humains , Tumeurs de l'intestin/métabolisme , Tumeurs de l'intestin/anatomopathologie , Activation des lymphocytes , Déplétion lymphocytaire , Mâle , Souris , Souris de lignée C57BL , Souris transgéniques , Récepteur lymphocytaire T antigène, alpha-bêta/métabolisme
12.
Oncotarget ; 10(29): 2810-2823, 2019 Apr 19.
Article de Anglais | MEDLINE | ID: mdl-31073372

RÉSUMÉ

Mucosal-associated invariant T (MAIT) cells all express a semi-invariable T cell receptor recognizing microbial metabolites presented on the MHC class I-like molecule MR1. Upon activation, they rapidly secrete cytokines and increase their cytotoxic potential. We showed recently that MAIT cells with Th1 phenotype accumulate in human colon adenocarcinomas. Here, we investigated the cytotoxic potential of tumor-infiltrating MAIT cells in colon adenocarcinomas, and to what extent it may be affected by the tumor microenvironment. Activation of MAIT cells from tumors induced increased Granzyme B, and to a lesser extent, perforin expression. Degranulation was assessed by surface expression of CD107a, and was also seen in response to cognate antigen recognition. The cytotoxic potential of tumor-associated MAIT cells was very similar to that of MAIT cells from unaffected colon. MAIT cells were also identified by immunofluorescence in direct contact with tumor cells in sections from colon cancer specimens. To summarize, tumor-associated MAIT cells from colon tumors have strong cytotoxic potential and are not compromised in this regard compared to MAIT cells from the unaffected colon. We conclude that MAIT cells may contribute significantly to the protective immune response to tumors, both by secretion of Th1-associated cytokines and by direct killing of tumor cells.

13.
Cancer Immunol Immunother ; 67(7): 1067-1077, 2018 07.
Article de Anglais | MEDLINE | ID: mdl-29671006

RÉSUMÉ

Tumor-infiltrating lymphocytes are crucial for anti-tumor immunity. We have previously shown that regulatory T cells (Treg) are able to reduce T-cell transendothelial migration in vitro and accumulation of effector T cells in intestinal tumors in vivo. Treg depletion also resulted in increased levels of the chemokines CXCL9 and CXCL10 specifically in the tumors. In this study, we investigated the mechanisms for Treg mediated suppression of T-cell migration into intestinal tumors in the APCmin/+ mouse model. By breeding APCmin/+ mice with DEREG mice, which harbour a high affinity diphtheria toxin receptor under the control of the FOXP3 promoter, we were able to deplete Treg in tumor-bearing mice. Using adoptive transfer experiments, we could document a markedly increased migration of T cells specifically into Treg depleted tumors, and that Treg depletion results in increased production of the CXCR3 ligand CXCL10 from endothelial cells in the tumors. Furthermore, we were able to demonstrate that T cells use CXCR3 to migrate into intestinal tumors. In addition, human colon adenocarcinomas express high levels of mRNA CXCR3 ligands and tumor endothelial cells produce CXCL9 and CXCL10 ex vivo. In conclusion, this study demonstrates that Treg reduce endothelial CXCL10 production, inhibit T-cell migration into tumors and that CXCR3 mediated signalling is crucial for lymphocyte accumulation in intestinal tumors. Thus, immunotherapy aimed at Treg depletion may be effective by increasing not only T effector cell activity, but also their accumulation in tumors.


Sujet(s)
Mouvement cellulaire , Chimiokine CXCL9/métabolisme , Endothélium vasculaire/métabolisme , Tumeurs de l'intestin/immunologie , Lymphocytes TIL/immunologie , Récepteurs CXCR3/métabolisme , Lymphocytes T régulateurs/immunologie , Adénocarcinome/immunologie , Adénocarcinome/métabolisme , Adénocarcinome/anatomopathologie , Animaux , Tumeurs du côlon/immunologie , Tumeurs du côlon/métabolisme , Tumeurs du côlon/anatomopathologie , Modèles animaux de maladie humaine , Endothélium vasculaire/immunologie , Femelle , Humains , Tumeurs de l'intestin/métabolisme , Tumeurs de l'intestin/anatomopathologie , Déplétion lymphocytaire , Lymphocytes TIL/métabolisme , Lymphocytes TIL/anatomopathologie , Mâle , Souris , Souris de lignée C57BL , Lymphocytes T régulateurs/métabolisme , Lymphocytes T régulateurs/anatomopathologie , Cellules cancéreuses en culture
14.
Sci Immunol ; 2(18)2017 12 01.
Article de Anglais | MEDLINE | ID: mdl-29196450

RÉSUMÉ

T follicular helper (Tfh) cells are a subset of CD4+ T cells that promote antibody production during vaccination. Conventional dendritic cells (cDCs) efficiently prime Tfh cells; however, conclusions regarding which cDC instructs Tfh cell differentiation have differed between recent studies. We found that these discrepancies might exist because of the unusual sites used for immunization in murine models, which differentially bias which DC subsets access antigen. We used intranasal immunization as a physiologically relevant route of exposure that delivers antigen to all tissue DC subsets. Using a combination of mice in which the function of individual DC subsets is impaired and different antigen formulations, we determined that CD11b+ migratory type 2 cDCs (cDC2s) are necessary and sufficient for Tfh induction. DC-specific deletion of the guanine nucleotide exchange factor DOCK8 resulted in an isolated loss of CD11b+ cDC2, but not CD103+ cDC1, migration to lung-draining lymph nodes. Impaired cDC2 migration or development in DC-specific Dock8 or Irf4 knockout mice, respectively, led to reduced Tfh cell priming, whereas loss of CD103+ cDC1s in Batf3-/- mice did not. Loss of cDC2-dependent Tfh cell priming impaired antibody-mediated protection from live influenza virus challenge. We show that migratory cDC2s uniquely carry antigen into the subanatomic regions of the lymph node where Tfh cell priming occurs-the T-B border. This work identifies the DC subset responsible for Tfh cell-dependent antibody responses, particularly when antigen dose is limiting or is encountered at a mucosal site, which could ultimately inform the formulation and delivery of vaccines.


Sujet(s)
Anticorps/immunologie , Antigènes CD11b/immunologie , Cellules dendritiques/immunologie , Lymphocytes T auxiliaires/immunologie , Animaux , Facteurs de transcription à motif basique et à glissière à leucines/déficit , Facteurs de transcription à motif basique et à glissière à leucines/immunologie , Prolifération cellulaire , Cellules dendritiques/métabolisme , Femelle , Mâle , Souris , Souris de lignée C57BL , Souris knockout , Souris transgéniques , Protéines de répression/déficit , Protéines de répression/immunologie , Lymphocytes T auxiliaires/cytologie
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...