Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 6 de 6
Filter
Add more filters










Database
Language
Publication year range
1.
Immunity ; 56(4): 813-828.e10, 2023 04 11.
Article in English | MEDLINE | ID: mdl-36809763

ABSTRACT

T cell factor 1 (Tcf-1) expressing CD8+ T cells exhibit stem-like self-renewing capacity, rendering them key for immune defense against chronic viral infection and cancer. Yet, the signals that promote the formation and maintenance of these stem-like CD8+ T cells (CD8+SL) remain poorly defined. Studying CD8+ T cell differentiation in mice with chronic viral infection, we identified the alarmin interleukin-33 (IL-33) as pivotal for the expansion and stem-like functioning of CD8+SL as well as for virus control. IL-33 receptor (ST2)-deficient CD8+ T cells exhibited biased end differentiation and premature loss of Tcf-1. ST2-deficient CD8+SL responses were restored by blockade of type I interferon signaling, suggesting that IL-33 balances IFN-I effects to control CD8+SL formation in chronic infection. IL-33 signals broadly augmented chromatin accessibility in CD8+SL and determined these cells' re-expansion potential. Our study identifies the IL-33-ST2 axis as an important CD8+SL-promoting pathway in the context of chronic viral infection.


Subject(s)
CD8-Positive T-Lymphocytes , Interleukin-33 , Lymphocytic Choriomeningitis , Animals , Mice , Alarmins/metabolism , Interleukin-1 Receptor-Like 1 Protein/metabolism , Interleukin-33/metabolism , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus , Mice, Inbred C57BL , Persistent Infection , T Cell Transcription Factor 1/metabolism
2.
Cell Rep Med ; 2(3): 100209, 2021 03 16.
Article in English | MEDLINE | ID: mdl-33763654

ABSTRACT

Therapeutic vaccination regimens inducing clinically effective tumor-specific CD8+ T lymphocyte (CTL) responses are an unmet medical need. We engineer two distantly related arenaviruses, Pichinde virus and lymphocytic choriomeningitis virus, for therapeutic cancer vaccination. In mice, life-replicating vector formats of these two viruses delivering a self-antigen in a heterologous prime-boost regimen induce tumor-specific CTL responses up to 50% of the circulating CD8 T cell pool. This CTL attack eliminates established solid tumors in a significant proportion of animals, accompanied by protection against tumor rechallenge. The magnitude of CTL responses is alarmin driven and requires combining two genealogically distantly related arenaviruses. Vector-neutralizing antibodies do not inhibit booster immunizations by the same vector or by closely related vectors. Rather, CTL immunodominance hierarchies favor vector backbone-targeted responses at the expense of self-reactive CTLs. These findings establish an arenavirus-based immunotherapy regimen that allows reshuffling of immunodominance hierarchies and breaking self-directed tolerance for efficient tumor control.


Subject(s)
Cancer Vaccines/administration & dosage , Immunotherapy/methods , Lymphocytic choriomeningitis virus/immunology , Mastocytoma/therapy , Pichinde virus/immunology , T-Lymphocytes, Cytotoxic/immunology , Alarmins/genetics , Alarmins/immunology , Animals , Antibodies, Neutralizing/pharmacology , Cancer Vaccines/genetics , Cancer Vaccines/immunology , Female , Gene Expression , Genetic Engineering/methods , Genetic Vectors/classification , Genetic Vectors/immunology , Guinea Pigs , Immunization, Secondary , Lymphocytic choriomeningitis virus/classification , Lymphocytic choriomeningitis virus/genetics , Mastocytoma/genetics , Mastocytoma/immunology , Mastocytoma/mortality , Mice , Mice, Inbred C57BL , Phylogeny , Pichinde virus/classification , Pichinde virus/genetics , Self Tolerance , Survival Analysis , Vaccination/methods
3.
Eur J Immunol ; 51(1): 76-90, 2021 01.
Article in English | MEDLINE | ID: mdl-32700362

ABSTRACT

Upon viral infection, stressed or damaged cells can release alarmins like IL-33 that act as endogenous danger signals alerting innate and adaptive immune cells. IL-33 coming from nonhematopoietic cells has been identified as important factor triggering the expansion of antiviral CD8+ T cells. In LN the critical cellular source of IL-33 is unknown, as is its potential cell-intrinsic function as a chromatin-associated factor. Using IL-33-GFP reporter mice, we identify fibroblastic reticular cells (FRC) and lymphatic endothelial cells (LEC) as the main IL-33 source. In homeostasis, IL-33 is dispensable as a transcriptional regulator in FRC, indicating it functions mainly as released cytokine. Early during infection with lymphocytic choriomeningitis virus (LCMV) clone 13, both FRC and LEC lose IL-33 protein expression suggesting cytokine release, correlating timewise with IL-33 receptor expression by reactive CD8+ T cells and their greatly augmented expansion in WT versus ll33-/- mice. Using mice lacking IL-33 selectively in FRC versus LEC, we identify FRC as key IL-33 source driving acute and chronic antiviral T-cell responses. Collectively, these findings show that LN T-zone FRC not only regulate the homeostasis of naïve T cells but also their expansion and differentiation several days into an antiviral response.


Subject(s)
Interleukin-33/metabolism , Lymphocytic Choriomeningitis/immunology , Acute Disease , Adaptive Immunity , Animals , CD8-Positive T-Lymphocytes/immunology , Chronic Disease , Endothelial Cells/immunology , Fibroblasts/immunology , Homeostasis , Humans , Immunity, Innate , Interleukin-33/deficiency , Interleukin-33/genetics , Lymph Nodes/immunology , Lymphocytic choriomeningitis virus/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Models, Immunological
4.
J Immunother Cancer ; 7(1): 67, 2019 03 12.
Article in English | MEDLINE | ID: mdl-30871609

ABSTRACT

BACKGROUND: Cancer cells are known to develop mechanisms to circumvent effective anti-tumor immunity. The two ectonucleotidases CD39 and CD73 are promising drug targets, as they act in concert to convert extracellular immune-stimulating ATP to adenosine. CD39 is expressed by different immune cell populations as well as cancer cells of different tumor types and supports the tumor in escaping immune recognition and destruction. Thus, increasing extracellular ATP and simultaneously reducing adenosine concentrations in the tumor can lead to effective anti-tumor immunity. METHODS: We designed locked nucleic acid (LNA)-modified antisense oligonucleotides (ASOs) with specificity for human or mouse CD39 that do not need a transfection reagent or delivery system for efficient target knockdown. Knockdown efficacy of ASOs on mRNA and protein level was investigated in cancer cell lines and in primary human T cells. The effect of CD39 knockdown on ATP-degrading activity was evaluated by measuring levels of ATP in tumor cell supernatants and analysis of T cell proliferation in the presence of extracellular ATP. The in vivo effects of CD39-specific ASOs on target expression, anti-tumor immune responses and on tumor growth were analyzed in syngeneic mouse tumor models using multi-color flow cytometry. RESULTS: CD39-specific ASOs suppressed expression of CD39 mRNA and protein in different murine and human cancer cell lines and in primary human T cells. Degradation of extracellular ATP was strongly reduced by CD39-specific ASOs. Strikingly, CD39 knockdown by ASOs was associated with improved CD8+ T cell proliferation. Treatment of tumor-bearing mice with CD39-specific ASOs led to dose-dependent reduction of CD39-protein expression in regulatory T cells (Tregs) and tumor-associated macrophages. Moreover, frequency of intratumoral Tregs was substantially reduced in CD39 ASO-treated mice. As a consequence, the ratio of CD8+ T cells to Tregs in tumors was improved, while PD-1 expression was induced in CD39 ASO-treated intratumoral CD8+ T cells. Consequently, CD39 ASO treatment demonstrated potent reduction in tumor growth in combination with anti-PD-1 treatment. CONCLUSION: Targeting of CD39 by ASOs represents a promising state-of-the art therapeutic approach to improve immune responses against tumors.


Subject(s)
Apyrase/genetics , Gene Silencing , Immunity/genetics , Neoplasms/genetics , Neoplasms/immunology , Oligonucleotides, Antisense/genetics , Adenosine/metabolism , Adenosine Triphosphate/metabolism , Animals , Antineoplastic Agents, Immunological/pharmacology , Biomarkers, Tumor , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cells, Cultured , Disease Models, Animal , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Mice , Neoplasms/pathology , Oligonucleotides, Antisense/administration & dosage , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , Xenograft Model Antitumor Assays
5.
Immunity ; 47(5): 928-942.e7, 2017 11 21.
Article in English | MEDLINE | ID: mdl-29166590

ABSTRACT

Pancreatic-islet inflammation contributes to the failure of ß cell insulin secretion during obesity and type 2 diabetes. However, little is known about the nature and function of resident immune cells in this context or in homeostasis. Here we show that interleukin (IL)-33 was produced by islet mesenchymal cells and enhanced by a diabetes milieu (glucose, IL-1ß, and palmitate). IL-33 promoted ß cell function through islet-resident group 2 innate lymphoid cells (ILC2s) that elicited retinoic acid (RA)-producing capacities in macrophages and dendritic cells via the secretion of IL-13 and colony-stimulating factor 2. In turn, local RA signaled to the ß cells to increase insulin secretion. This IL-33-ILC2 axis was activated after acute ß cell stress but was defective during chronic obesity. Accordingly, IL-33 injections rescued islet function in obese mice. Our findings provide evidence that an immunometabolic crosstalk between islet-derived IL-33, ILC2s, and myeloid cells fosters insulin secretion.


Subject(s)
Insulin/metabolism , Interleukin-33/pharmacology , Islets of Langerhans/drug effects , Lymphocytes/drug effects , Myeloid Cells/metabolism , Tretinoin/metabolism , Animals , Humans , Inflammation/immunology , Insulin Secretion , Interleukin-33/biosynthesis , Islets of Langerhans/immunology , Islets of Langerhans/pathology , Lymphocytes/physiology , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Vitamin A/physiology
6.
Nat Commun ; 8: 15327, 2017 05 26.
Article in English | MEDLINE | ID: mdl-28548102

ABSTRACT

Viral infections lead to alarmin release and elicit potent cytotoxic effector T lymphocyte (CTLeff) responses. Conversely, the induction of protective tumour-specific CTLeff and their recruitment into the tumour remain challenging tasks. Here we show that lymphocytic choriomeningitis virus (LCMV) can be engineered to serve as a replication competent, stably-attenuated immunotherapy vector (artLCMV). artLCMV delivers tumour-associated antigens to dendritic cells for efficient CTL priming. Unlike replication-deficient vectors, artLCMV targets also lymphoid tissue stroma cells expressing the alarmin interleukin-33. By triggering interleukin-33 signals, artLCMV elicits CTLeff responses of higher magnitude and functionality than those induced by replication-deficient vectors. Superior anti-tumour efficacy of artLCMV immunotherapy depends on interleukin-33 signalling, and a massive CTLeff influx triggers an inflammatory conversion of the tumour microenvironment. Our observations suggest that replicating viral delivery systems can release alarmins for improved anti-tumour efficacy. These mechanistic insights may outweigh safety concerns around replicating viral vectors in cancer immunotherapy.


Subject(s)
Alarmins/immunology , Cancer Vaccines/immunology , Immunotherapy/methods , Lymphocytic choriomeningitis virus/genetics , Neoplasms/therapy , T-Lymphocytes, Cytotoxic/immunology , Animals , Antigens, Neoplasm/immunology , Cancer Vaccines/therapeutic use , Cell Line, Tumor , Dendritic Cells/immunology , Gene Expression Profiling , Genetic Engineering , Genetic Vectors/genetics , Genetic Vectors/immunology , Genetic Vectors/therapeutic use , HEK293 Cells , Humans , Interleukin-33/genetics , Interleukin-33/immunology , Lymphocyte Activation/immunology , Mesocricetus , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred DBA , Mice, Knockout , Neoplasms/immunology , Tumor Microenvironment/immunology , Vaccines, Live, Unattenuated/immunology , Virus Replication/genetics , Virus Replication/immunology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL
...