Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Eur J Immunol ; 51(9): 2317-2329, 2021 09.
Article in English | MEDLINE | ID: mdl-34272885

ABSTRACT

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.


Subject(s)
Chemokine CXCL10/metabolism , Colonic Neoplasms/pathology , Lymphocytes, Tumor-Infiltrating/immunology , Sphingomyelin Phosphodiesterase/metabolism , T-Lymphocytes, Regulatory/immunology , Transendothelial and Transepithelial Migration/physiology , Animals , Cell Adhesion Molecules/biosynthesis , Cell Line , Chemokine CX3CL1/biosynthesis , Chemokine CXCL10/biosynthesis , Colonic Neoplasms/immunology , Down-Regulation , Female , Human Umbilical Vein Endothelial Cells , Humans , Male , Mice , Mice, Transgenic , T-Lymphocyte Subsets/immunology , Versicans/biosynthesis
2.
Cancer Immunol Immunother ; 69(7): 1279-1292, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32185408

ABSTRACT

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.


Subject(s)
Adenomatous Polyposis Coli Protein/physiology , CD8 Antigens/immunology , CD8-Positive T-Lymphocytes/immunology , Intestinal Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Receptors, Antigen, T-Cell, alpha-beta/immunology , T-Lymphocytes, Regulatory/immunology , Animals , CD8 Antigens/metabolism , CD8-Positive T-Lymphocytes/metabolism , Female , Humans , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Lymphocyte Activation , Lymphocyte Depletion , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Receptors, Antigen, T-Cell, alpha-beta/metabolism
3.
PLoS One ; 13(9): e0204181, 2018.
Article in English | MEDLINE | ID: mdl-30235302

ABSTRACT

The gut homing receptor integrin α4ß7 is essential for the migration of pro-inflammatory T cells into the gut mucosa. Since intestinal neoplasia has been associated with chronic inflammation, we investigated whether interfering with gut-homing affects intestinal tumorigenesis. Using chemically induced and spontaneous intestinal tumor models we showed that lack of ß7 integrin significantly impairs tumor growth without affecting tumor frequencies, with a mild translatable effect on overall survival. This correlates with human data showing lower MAdCAM-1 expression and disease-free survival in colorectal cancer patients. Thus, paradoxically in contrast to extra-intestinal tumors, blocking migration of immune cells into the gut might have a positive therapeutic effect on intestinal neoplasia.


Subject(s)
Integrin beta Chains/metabolism , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Animals , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Proliferation , Inflammation/pathology , Kaplan-Meier Estimate , Mice , Mice, Inbred C57BL , Survival Analysis , T-Lymphocytes/immunology , Tumor Burden
4.
Cancer Immunol Immunother ; 67(7): 1067-1077, 2018 07.
Article in English | MEDLINE | ID: mdl-29671006

ABSTRACT

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.


Subject(s)
Cell Movement , Chemokine CXCL9/metabolism , Endothelium, Vascular/metabolism , Intestinal Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Receptors, CXCR3/metabolism , T-Lymphocytes, Regulatory/immunology , Adenocarcinoma/immunology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , Disease Models, Animal , Endothelium, Vascular/immunology , Female , Humans , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Lymphocyte Depletion , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Male , Mice , Mice, Inbred C57BL , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , Tumor Cells, Cultured
5.
Oncotarget ; 9(97): 36993-37007, 2018 Dec 11.
Article in English | MEDLINE | ID: mdl-30651930

ABSTRACT

Increasing knowledge of the function and regulation of tumor-infiltrating lymphocytes has led to new insights in cancer immunotherapy. Regulatory T cells (Treg) accumulate in colon tumors, and we recently showed that CD39+ Treg from cancer patients inhibit transendothelial migration of conventional T cells. CD39 mediates the hydrolysis of ATP to immunosuppressive adenosine and adds to the immunosuppressive effects of Treg. Here, we further investigated the regulatory features of intratumoral CD39+ Treg in colon cancer. Using flow cytometry analyses of cells from 46 colon cancer patients, we confirm the accumulation of CD39+ Treg in the tumor tissue compared to unaffected colon tissue, and also show that tumor-infiltrating Treg express more CD39 and Foxp3 on a per cell basis. Furthermore, CD39+ Treg in tumors express markers indicating increased turnover and suppressive ability. In particular, tumor-infiltrating CD39+ Treg have high expression of surface molecules related to immunosuppression, such as ICOS, PD-L1 and CTLA-4. Functional suppression assays also indicate potent suppressive capacity of CD39+ Treg on proliferation and IFN-γ secretion by conventional T cells. In conclusion, our results identify tumor-infiltrating CD39+ Treg as a numerous and potentially important immunosuppressive subset, and suggest that immunotherapy aimed at reducing the activity of CD39+ Treg may be particularly useful in the setting of colon cancer.

6.
Immunology ; 152(1): 74-88, 2017 09.
Article in English | MEDLINE | ID: mdl-28437001

ABSTRACT

CD4+ Foxp3+ regulatory T (Treg) cells include differentiated populations of effector Treg cells characterized by the expression of specific transcription factors. Tumours, including intestinal malignancies, often present with local accumulation of Treg cells that can prevent tumour clearance, but how tumour progression leads to Treg cell accumulation is incompletely understood. Here using genetically modified mouse models we show that ablation of E-cadherin, a process associated with epithelial to mesenchymal transition and tumour progression, promotes the accumulation of intestinal Treg cells by the specific accumulation of the KLRG1+ GATA3+ Treg subset. Epithelial E-cadherin ablation activates the ß-catenin pathway, and we find that increasing ß-catenin signals in intestinal epithelial cells also boosts Treg cell frequencies through local accumulation of KLRG1+ GATA3+ Treg cells. Both E-cadherin ablation and increased ß-catenin signals resulted in epithelial cells with higher levels of interleukin-33, a cytokine that preferentially expands KLRG1+ GATA3+ Treg cells. Tumours often present reduced E-cadherin expression and increased ß-catenin signalling and interleukin-33 production. Accordingly, Treg cell accumulation in intestinal tumours from APCmin/+ mice was exclusively due to the increase in KLRG1+ GATA3+ Treg cells. Our data identify a novel axis through which epithelial cells control local Treg cell subsets, which may be activated during intestinal tumorigenesis.


Subject(s)
Epithelial Cells/immunology , GATA3 Transcription Factor/immunology , Intestinal Mucosa/immunology , Intestinal Neoplasms/immunology , Lymphocytes, Tumor-Infiltrating/immunology , Receptors, Immunologic/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Cadherins/immunology , Cadherins/metabolism , Cdh1 Proteins/genetics , Cdh1 Proteins/immunology , Cdh1 Proteins/metabolism , Cells, Cultured , Chemotaxis, Leukocyte , Epithelial Cells/metabolism , Epithelial Cells/pathology , Forkhead Transcription Factors/immunology , Forkhead Transcription Factors/metabolism , GATA3 Transcription Factor/metabolism , Genes, APC , Genetic Predisposition to Disease , Interleukin-33/immunology , Interleukin-33/metabolism , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestinal Neoplasms/genetics , Intestinal Neoplasms/metabolism , Intestinal Neoplasms/pathology , Lectins, C-Type , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Mice, Inbred C57BL , Mice, Transgenic , Phenotype , Receptors, Immunologic/metabolism , Signal Transduction , T-Lymphocytes, Regulatory/metabolism , T-Lymphocytes, Regulatory/pathology , beta Catenin/genetics , beta Catenin/immunology , beta Catenin/metabolism
7.
Immun Inflamm Dis ; 4(2): 191-200, 2016 06.
Article in English | MEDLINE | ID: mdl-27957327

ABSTRACT

Several Butyrophilin (BTN) and Btn-like (BTNL) molecules control T lymphocyte responses, and are genetically associated with inflammatory disorders and cancer. In this study, we present a comprehensive expression analysis of human and murine BTN and BTNL genes in conditions associated with intestinal inflammation and cancer. Using real-time PCR, expression of human BTN and BTNL genes was analyzed in samples from patients with ulcerative colitis, irritable bowel syndrome, and colon tumors. Expression of murine Btn and Btnl genes was examined in mouse models of spontaneous colitis (Muc2-/-) and intestinal tumorigenesis (ApcMin/+). Our analysis indicates a strong association of several of the human genes with ulcerative colitis and colon cancer; while especially BTN1A1, BTN2A2, BTN3A3, and BTNL8 were significantly altered in inflammation, colonic tumors exhibited significantly decreased levels of BTNL2, BTNL3, BTNL8, and BTNL9 as compared to unaffected tissue. Colonic inflammation in Muc2-/- mice significantly down-regulated the expression of particularly Btnl1, Btnl4, and Btnl6 mRNA, and intestinal polyps derived from ApcMin/+ mice displayed altered levels of Btn1a1, Btn2a2, and Btnl1 transcripts. Thus, our data present an association of BTN and BTNL genes with intestinal inflammation and cancer and represent a valuable resource for further studies of this gene family.


Subject(s)
Butyrophilins/metabolism , Colonic Neoplasms/metabolism , Inflammation/metabolism , Animals , Colitis, Ulcerative/metabolism , Colon , Humans , Intestinal Mucosa , Mice , Mice, Inbred C57BL , T-Lymphocytes
8.
J Immunol ; 195(7): 3472-81, 2015 Oct 01.
Article in English | MEDLINE | ID: mdl-26297765

ABSTRACT

Mucosa-associated invariant T (MAIT) cells are innate-like T cells with a conserved TCR α-chain recognizing bacterial metabolites presented on the invariant MHC-related 1 molecule. MAIT cells are present in intestinal tissues and liver, and they rapidly secrete IFN-γ and IL-17 in response to bacterial insult. In colon cancer, IL-17-driven inflammation promotes tumor progression, whereas IFN-γ production is essential for antitumor immunity. Thus, tumor-associated MAIT cells may affect antitumor immune responses by their secreted cytokines. However, the knowledge of MAIT cell presence and function in tumors is virtually absent. In this study, we determined the frequency, phenotype, and functional capacity of MAIT cells in colon adenocarcinomas and unaffected colon lamina propria. Flow cytometric analyses showed significant accumulation of MAIT cells in tumor tissue, irrespective of tumor stage or localization. Colonic MAIT cells displayed an activated memory phenotype and expression of chemokine receptors CCR6 and CCR9. Most MAIT cells in unaffected colon tissues produced IFN-γ, whereas only few produced IL-17. Colonic MAIT cells also produced TNF-α, IL-2, and granzyme B. In the tumors, significantly lower frequencies of IFN-γ-producing MAIT cells were seen, whereas there were no differences in the other cytokines analyzed, and in vitro studies showed that secreted factors from tumor tissue reduced IFN-γ production from MAIT cells. In conclusion, MAIT cells infiltrate colon tumors but their ability to produce IFN-γ is substantially reduced. We suggest that MAIT cells have the capacity to promote local immune responses to tumors, but factors in the tumor microenvironment act to reduce MAIT cell IFN-γ production.


Subject(s)
Adenocarcinoma/pathology , Colonic Neoplasms/pathology , Interferon-gamma/biosynthesis , Intestinal Mucosa/immunology , T-Lymphocytes/immunology , Adult , Aged , Aged, 80 and over , Female , Granzymes/biosynthesis , Humans , Inflammation/immunology , Interferon-gamma/immunology , Interleukin-17/biosynthesis , Interleukin-17/immunology , Interleukin-2 , Intestinal Mucosa/cytology , Liver/cytology , Liver/immunology , Lymphocyte Activation/immunology , Male , Middle Aged , Receptors, CCR/biosynthesis , Receptors, CCR6/biosynthesis , T-Lymphocyte Subsets/immunology , Tumor Necrosis Factor-alpha/biosynthesis
9.
Eur J Immunol ; 45(6): 1654-66, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25754875

ABSTRACT

Colorectal cancer (CRC) is one of the most prevalent tumor types worldwide and tumor-infiltrating T cells are crucial for anti-tumor immunity. We previously demonstrated that Treg cells from CRC patients inhibit transendothelial migration of conventional T cells. However, it remains unclear if local Treg cells affect lymphocyte migration into colonic tumors. By breeding APC(Min/+) mice with depletion of regulatory T cells mice, expressing the diphtheria toxin receptor under the control of the FoxP3 promoter, we were able to selectively deplete Treg cells in tumor-bearing mice, and investigate the impact of these cells on the infiltration of conventional T cells into intestinal tumors. Short-term Treg-cell depletion led to a substantial increase in the frequencies of T cells in the tumors, attributed by both increased infiltration and proliferation of T cells in the Treg-cell-depleted tumors. We also demonstrate a selective increase of the chemokines CXCL9 and CXCL10 in Treg-cell-depleted tumors, which were accompanied by accumulation of CXCR3(+) T cells, and increased IFN-γ mRNA expression. In conclusion, Treg-cell depletion increases the accumulation of conventional T cells in intestinal tumors, and targeting Treg cells could be a possible anti-tumor immunotherapy, which not only affects T-cell effector functions, but also their recruitment to tumors.


Subject(s)
Chemokines/biosynthesis , Colorectal Neoplasms/immunology , Colorectal Neoplasms/metabolism , Receptors, CXCR3/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/immunology , Adenomatous Polyposis Coli Protein/metabolism , Animals , Cell Adhesion Molecules/metabolism , Chemotaxis/immunology , Cytokines/biosynthesis , Disease Models, Animal , Female , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Lymphocyte Activation/immunology , Lymphocyte Depletion , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Mucoproteins , Phenotype , Receptors, Chemokine/metabolism , Th1 Cells/immunology , Th1 Cells/metabolism , Vascular Cell Adhesion Molecule-1/metabolism
10.
Cancer Immunol Immunother ; 63(8): 807-19, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24777614

ABSTRACT

Tumor progression in the colon moves from aberrant crypt foci to adenomatous polyps to invasive carcinomas. The composition of the tumor-infiltrating leukocyte population affects the ability of the immune system to fight the tumor. T cell infiltration into colorectal adenocarcinomas, particularly T helper 1 (Th1) type T cells as well as increased regulatory T cell (Treg) frequencies, is correlated with improved prognosis. However, whether Th1 cells and Tregs are already present at the adenoma stage is not known. In this study, the APC(Min/+) mouse model of intestinal adenomatous polyposis was used to investigate tumor-associated lymphocyte subsets and the mechanisms of their accumulation into gastrointestinal adenomas. Compared to unaffected tissue, adenomas accumulated CD4(+)FoxP3(+) putative Treg in parallel with lower frequencies of conventional T cells and B cells. The accumulation of Treg was also observed in human adenomatous polyps. Despite high Treg numbers, the function of conventional T cells present in the APC(Min/+) adenomas was not different from those in the unaffected tissue. Adenomas displayed an altered chemokine balance, with higher CCL17 and lower CXCL11 and CCL25 expression than in the unaffected tissue. In parallel, CXCR3(+) Tregs were largely absent from adenomas. The data indicate that already in early stages of tumor development, the balance of lymphocyte-recruiting chemokines is altered possibly contributing to the observed shift toward higher frequencies of Treg.


Subject(s)
Adenocarcinoma/immunology , Adenoma/immunology , Chemokines/biosynthesis , Colorectal Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Aged , Animals , Chemokines/immunology , Female , Humans , Male , Mice , Mice, Inbred C57BL , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...