Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 41
Filter
1.
BMC Immunol ; 23(1): 61, 2022 12 10.
Article in English | MEDLINE | ID: mdl-36496363

ABSTRACT

BACKGROUND: Multiple antigenic stimulations are crucial to immune system training during early post-natal life. These stimulations can be either due to commensals, which accounts for the acquisition and maintenance of tolerance, or to pathogens, which triggers immunity. In pig, only few works previously explored the influence of natural exposition to pathogens upon immune competence. We propose herein the results of a multicentric, field study, conducted on 265 piglets exposed to contrasted pathogen levels in their living environment. Piglets were housed in 15 different commercial farms, sorted in two groups, low (HSLOW)- and high (HSHIGH)-health status farms, depending on their recurrent exposition to five common swine pathogens. RESULTS: Using animal-based measures, we compared the immune competence and growth performances of HSLOW and HSHIGH pigs around weaning. As expected, we observed a rise in the number of circulating leucocytes with age, which affected different cell populations. Monocyte, antigen-experienced and cytotoxic lymphocyte subpopulation counts were higher in piglets reared in HSLOW farms as compared to their HSHIGH homologs. Also, the age-dependent evolution in γδ T cell and neutrophil counts was significantly affected by the health status. With age, circulating IFNα level decreased and IgM level increased while being greater in HSLOW piglets at any time. After weaning, LPS-stimulated blood cells derived from HSLOW piglets were more prone to secrete IL-8 than those derived from HSHIGH pigs did. Monocytes and granulocytes issued from HSLOW pigs also exhibited comparable phagocytosis capacity. Altogether our data emphasize the more robust immunophenotype of HSLOW piglets. Finally, piglets raised under higher pathogen pressure grew less than HSHIGH piglets did and exhibited a different metabolic profile. The higher cost of the immune responses associated with the low farm health status may account for lower HSLOW piglet performances. CONCLUSIONS: Altogether, our data, obtained in field conditions, provide evidence that early exposure to pathogens shapes the immune competence of piglets. They also document the negative impact of an overstimulation of the immune system on piglets' growth.


Subject(s)
Neutrophils , Phagocytosis , Swine , Animals , Weaning , Leukocyte Count , Leukocytes
2.
Sci Rep ; 12(1): 4681, 2022 03 18.
Article in English | MEDLINE | ID: mdl-35304495

ABSTRACT

A bioartificial pancreas (BAP) encapsulating high pancreatic islets concentration is a promising alternative for type 1 diabetes therapy. However, the main limitation of this approach is O2 supply, especially until graft neovascularization. Here, we described a methodology to design an optimal O2-balanced BAP using statistical design of experiment (DoE). A full factorial DoE was first performed to screen two O2-technologies on their ability to preserve pseudo-islet viability and function under hypoxia and normoxia. Then, response surface methodology was used to define the optimal O2-carrier and islet seeding concentrations to maximize the number of viable pseudo-islets in the BAP containing an O2-generator under hypoxia. Monitoring of viability, function and maturation of neonatal pig islets for 15 days in vitro demonstrated the efficiency of the optimal O2-balanced BAP. The findings should allow the design of a more realistic BAP for humans with high islets concentration by maintaining the O2 balance in the device.


Subject(s)
Diabetes Mellitus, Type 1 , Islets of Langerhans Transplantation , Islets of Langerhans , Pancreas, Artificial , Diabetes Mellitus, Type 1/therapy , Humans , Hypoxia , Islets of Langerhans/physiology , Islets of Langerhans Transplantation/methods , Pancreas/physiology
3.
Med Sci (Paris) ; 37(12): 1139-1145, 2021 Dec.
Article in French | MEDLINE | ID: mdl-34928218

ABSTRACT

Regulation of immune responses was among the first functions of extracellular vesicles to be identified, more than twenty years ago. What exactly defines the outcome of an immune response remains a challenging issue. Owing to their reduced size, extracellular vesicles easily diffuse in interstitial and lymphatic fluids, where they can interact with the multiple effectors of the immune system. By accelerating and amplifying immune interactions, these ultra-mobile units may contribute to local and systemic coordination for efficient adaption to external and internal changes. Here we introduce the related ground-breaking studies of extracellular vesicle-mediated immune effects and present ongoing considerations on their potential roles in health and the development of immune disorders.


TITLE: Les vésicules extracellulaires - Un maillon essentiel du système immunitaire. ABSTRACT: L'implication des vésicules extracellulaires dans les échanges immunitaires a été parmi les premières fonctions mises en évidence pour ces vésicules, il y a plus de vingt ans. Du fait de leur petite taille, elles diffusent en effet aisément via les fluides interstitiels et la lymphe où elles interagissent avec les multiples effecteurs du système immunitaire. En accélérant et en amplifiant les échanges, il est concevable que ces unités ultra-mobiles favorisent la concertation entre cellules à l'échelle locale et globale, en réponse aux changements que subit l'organisme, que ceux-ci soient internes ou externes. Ici, vous sont présentées les découvertes clés sur les rôles des vésicules extracellulaires dans l'immunité, dont l'impact sur la santé commence tout juste maintenant à être mesuré.


Subject(s)
Extracellular Vesicles , Humans
5.
Front Immunol ; 11: 1814, 2020.
Article in English | MEDLINE | ID: mdl-33101266

ABSTRACT

Beta cell failure and apoptosis following islet inflammation have been associated with autoimmune type 1 diabetes pathogenesis. As conveyors of biological active material, extracellular vesicles (EV) act as mediators in communication with immune effectors fostering the idea that EV from inflamed beta cells may contribute to autoimmunity. Evidence accumulates that beta exosomes promote diabetogenic responses, but relative contributions of larger vesicles as well as variations in the composition of the beta cell's vesiculome due to environmental changes have not been explored yet. Here, we made side-by-side comparisons of the phenotype and function of apoptotic bodies (AB), microvesicles (MV) and small EV (sEV) isolated from an equal amount of MIN6 beta cells exposed to inflammatory, hypoxic or genotoxic stressors. Under normal conditions, large vesicles represent 93% of the volume, but only 2% of the number of the vesicles. Our data reveal a consistently higher release of AB and sEV and to a lesser extent of MV, exclusively under inflammatory conditions commensurate with a 4-fold increase in the total volume of the vesiculome and enhanced export of immune-stimulatory material including the autoantigen insulin, microRNA, and cytokines. Whilst inflammation does not change the concentration of insulin inside the EV, specific Toll-like receptor-binding microRNA sequences preferentially partition into sEV. Exposure to inflammatory stress engenders drastic increases in the expression of monocyte chemoattractant protein 1 in all EV and of interleukin-27 solely in AB suggesting selective sorting toward EV subspecies. Functional in vitro assays in mouse dendritic cells and macrophages reveal further differences in the aptitude of EV to modulate expression of cytokines and maturation markers. These findings highlight the different quantitative and qualitative imprints of environmental changes in subpopulations of beta EV that may contribute to the spread of inflammation and sustained immune cell recruitment at the inception of the (auto-) immune response.


Subject(s)
Cytokines/metabolism , Extracellular Vesicles/metabolism , Inflammation/metabolism , Insulin-Secreting Cells/metabolism , Insulin/metabolism , Animals , Apoptosis , Cell Hypoxia , Cell Line, Tumor , DNA Damage , Dendritic Cells/immunology , Dendritic Cells/metabolism , Extracellular Vesicles/immunology , Extracellular Vesicles/ultrastructure , Female , Inflammation/immunology , Inflammation/pathology , Insulin-Secreting Cells/immunology , Insulin-Secreting Cells/ultrastructure , Macrophage Activation , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Inbred NOD , MicroRNAs/metabolism , Phenotype , RAW 264.7 Cells , Secretory Pathway , Signal Transduction
6.
Immunol Res ; 65(6): 1156-1163, 2017 12.
Article in English | MEDLINE | ID: mdl-29134568

ABSTRACT

Adrenergic receptor agonists and antagonists are extensively used as drugs in medicine for a broad spectrum of indications. We examined the consequences of ß2-adrenergic stimulation of murine dendritic cells (DCs) on CD4+ T cell activation. We demonstrated in vitro that treatment of LPS-matured DCs with the ß2-agonist salbutamol reduced their ability to trigger OT-II T cell proliferation specific for ovalbumin antigen. Salbutamol also induced a decrease in MHC class II molecule expression by DC through Gi protein activation. Co-culture of CD4+ T cells with salbutamol-conditioned mature DC impaired TNFα and IL-6 secretion while preserving IL-10 production by T cells. Using a vaccination protocol in mice, we showed that salbutamol favored IL-10-producing CD4+ T cells. None of these effects was observed when working with ß2-adrenoreceptor deficient mice. Finally, we suggest that ß2-adrenergic stimulation of DC could be an interesting way to shape CD4+ T cell responses for the purposes of immunotherapy.


Subject(s)
Adrenergic beta-2 Receptor Agonists/pharmacology , Albuterol/pharmacology , CD4-Positive T-Lymphocytes/immunology , Dendritic Cells/immunology , Immunotherapy, Adoptive/methods , Receptors, Adrenergic, beta-2/metabolism , Animals , CD4-Positive T-Lymphocytes/transplantation , Cell Differentiation , Cell Proliferation , Cells, Cultured , Coculture Techniques , Dendritic Cells/drug effects , Humans , Interleukin-10/metabolism , Lipopolysaccharides/immunology , Lymphocyte Activation , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Adrenergic, beta-2/genetics
7.
Dev Comp Immunol ; 76: 169-176, 2017 11.
Article in English | MEDLINE | ID: mdl-28633932

ABSTRACT

The cross-talk between sympatho-adreno-medullar axis and innate immunity players was mainly studied in rodents. In intensive husbandry, pigs are exposed to multiple stressors inducing repeated releases of catecholamines that bind to adrenoreceptors (AR) on target cells. Among adrenoreceptors, the ß2-AR is largely expressed by immune cells including macrophages. We report herein on the effects of catecholamines, through ß2-AR stimulation, on pig macrophage functions activated by LPS. ß2-AR stimulation of porcine macrophages prevented the LPS-induced increase in TNFα and IL-8 secretion while increasing IL-10 secretion. In contrast, treatment with a ß2-agonist had no effect on anti-microbial functions. Lastly, ß2-AR stimulation of macrophages reduced the expression of genes up-regulated by LPS. Altogether, we demonstrated that ß2-AR stimulation of porcine macrophages prevented polarization towards a pro-inflammatory phenotype. Since porcine macrophages are a suitable model for human macrophages, our results might be relevant to appreciate catecholamine effects on human macrophages.


Subject(s)
Lipopolysaccharides/pharmacology , Macrophages/drug effects , Macrophages/metabolism , Receptors, Adrenergic, beta-2/metabolism , Adrenergic beta-2 Receptor Agonists/pharmacology , Animals , Anti-Infective Agents/pharmacology , Catecholamines/pharmacology , Immunity, Innate/drug effects , Interleukin-10/metabolism , Interleukin-8/metabolism , Swine , Tumor Necrosis Factor-alpha/metabolism , Up-Regulation/drug effects
8.
Physiol Behav ; 169: 1-8, 2017 02 01.
Article in English | MEDLINE | ID: mdl-27867043

ABSTRACT

Pig husbandry is known as an intensive breeding system, piglets being submitted to multiple stressful events such as early weaning, successive mixing, crowding and shipping. These stressors are thought to impair immune defences and might contribute, at least partly, to the prophylactic use of antibiotics. Robustness was recently defined as the ability of an individual to express a high-production potential in a wide variety of environmental conditions. Increasing robustness thus appears as a valuable option to improve resilience to stressors and could be obtained by selecting piglets upon their adrenocortical activity. In this study, we aimed at depicting the consequences of an acute social stress on the immune capacity of piglets genetically selected upon divergent hypothalamic-pituitary-adrenocortical (HPA) axis activity. For this purpose, we monitored neuroendocrine and immune parameters, in high- (HPAhi) and low- (HPAlo) responders to ACTH, just before and immediately after a one-hour mixing with unfamiliar conspecifics. As expected, stressed piglets displayed higher levels of circulating cortisol and norepinephrine. Blood cell count analysis combined to flow cytometry revealed a stress-induced leukocyte mobilization in the bloodstream with a specific recruitment of CD8α+ lymphocytes. Besides, one-hour mixing decreased LPS-induced IL-8 and TNFα secretions in whole-blood assays (WBA) and reduced mononuclear cell phagocytosis. Altogether, our data demonstrate that acute social stress alters immune competence of piglets from both groups, and bring new insights in favour of good farming practices. While for most parameters high- and low-responders to ACTH behaved similarly, HPAhi piglets displayed higher number of CD4+ CD8α- T cells, as well as increased cytokine production in WBA (LPS-induced TNFα and PIL-induced IL-8), which could confer them increased resistance to pathogens. Finally, a principal component analysis including all parameters highlighted that overall stress effects were less pronounced on piglets with a strong HPA axis. Thus, selection upon adrenocortical axis activity seems to reduce the magnitude of response to stress and appears as a good tool to increase piglet robustness.


Subject(s)
Adrenocorticotropic Hormone/pharmacology , Cytokines/metabolism , Hydrocortisone/blood , Norepinephrine/blood , Stress, Psychological/immunology , Stress, Psychological/physiopathology , Animals , Antigens, CD/metabolism , Chi-Square Distribution , Dose-Response Relationship, Drug , Flow Cytometry , Leukocyte Count , Leukocytes/physiology , Lipopolysaccharides/pharmacology , Male , Peptide Fragments/pharmacology , Phagocytosis/drug effects , Principal Component Analysis , Swine
9.
PLoS One ; 9(8): e105907, 2014.
Article in English | MEDLINE | ID: mdl-25170840

ABSTRACT

BACKGROUND: The importance of immune responses in the control of melanoma growth is well known. However, the implication of these antitumor immune responses in the efficacy of dacarbazine, a cytotoxic drug classically used in the treatment of melanoma, remains poorly understood in humans. METHODS: In this prospective observational study, we performed an immunomonitoring of eleven metastatic or locally advanced patients treated with dacarbazine as a first line of treatment. We assessed by flow cytometry lymphoid populations and their activation state; we also isolated NK cells to perform in vitro cytotoxicity tests. RESULTS: We found that chemotherapy induces lymphopenia and that a significantly higher numbers of naïve CD4+ T cells and lower proportion of Treg before chemotherapy are associated with disease control after dacarbazine treatment. Interestingly, NK cell cytotoxicity against dacarbazine-pretreated melanoma cells is only observed in NK cells from patients who achieved disease control. CONCLUSION: Together, our data pinpoint that some immune factors could help to predict the response of melanoma patients to dacarbazine. Future larger scale studies are warranted to test their validity as prediction markers.


Subject(s)
Dacarbazine/therapeutic use , Melanoma/drug therapy , Skin Neoplasms/drug therapy , Adult , Aged , Aged, 80 and over , Antineoplastic Agents, Alkylating/therapeutic use , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/genetics , Female , Flow Cytometry , Gene Expression/drug effects , Humans , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Leukocyte Count , Male , Melanoma/blood , Melanoma/pathology , Middle Aged , Neoplasm Metastasis , Prognosis , Prospective Studies , Reverse Transcriptase Polymerase Chain Reaction , Skin Neoplasms/blood , Skin Neoplasms/pathology , T-Lymphocytes, Regulatory/drug effects , T-Lymphocytes, Regulatory/metabolism , Transcription Factors/genetics , Treatment Outcome
10.
Nat Immunol ; 15(8): 758-66, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24973819

ABSTRACT

The TH9 subset of helper T cells was initially shown to contribute to the induction of autoimmune and allergic diseases, but subsequent evidence has suggested that these cells also exert antitumor activities. However, the molecular events that account for their effector properties are elusive. Here we found that the transcription factor IRF1 enhanced the effector function of TH9 cells and dictated their anticancer properties. Under TH9-skewing conditions, interleukin 1ß (IL-1ß) induced phosphorylation of the transcription factor STAT1 and subsequent expression of IRF1, which bound to the promoters of Il9 and Il21 and enhanced secretion of the cytokines IL-9 and IL-21 from TH9 cells. Furthermore, IL-1ß-induced TH9 cells exerted potent anticancer functions in an IRF1- and IL-21-dependent manner. Our findings thus identify IRF1 as a target for controlling the function of TH9 cells.


Subject(s)
Interferon Regulatory Factor-1/immunology , Interleukins/immunology , Melanoma, Experimental/immunology , T-Lymphocytes, Helper-Inducer/immunology , 3T3 Cells , Animals , Base Sequence , Cell Line , Female , Green Fluorescent Proteins/biosynthesis , Green Fluorescent Proteins/genetics , Interferon Regulatory Factor-1/biosynthesis , Interferon-gamma/genetics , Interferon-gamma/immunology , Interleukin-10/antagonists & inhibitors , Interleukin-10/immunology , Interleukin-9/genetics , Interleukin-9/immunology , Interleukin-9/metabolism , Interleukins/genetics , Interleukins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Ovalbumin/immunology , Phosphorylation/immunology , Proto-Oncogene Proteins c-fyn/antagonists & inhibitors , Proto-Oncogene Proteins c-fyn/genetics , RNA Interference , RNA, Small Interfering , STAT1 Transcription Factor/immunology , Sequence Analysis, RNA , T-Lymphocytes, Helper-Inducer/metabolism
11.
Oncoimmunology ; 2(9): e25737, 2013 Sep 01.
Article in English | MEDLINE | ID: mdl-24327935

ABSTRACT

In addition to cytotoxic effects, anticancer agents can exert multiple immunomodulatory functions. We have recently described the molecular mechanisms whereby bleomycin can 1) promote endoplasmic reticulum stress, causing the immunogenic death of cancer cells and hence strengthening antitumor CD8+ T cell responses; and 2) induce the secretion of transforming growth factor ß (TGFß), which stimulates regulatory T cells. This suggests that bleomycin may be favorably combined with TGFß-targeting strategies.

12.
Science ; 342(6161): 971-6, 2013 Nov 22.
Article in English | MEDLINE | ID: mdl-24264990

ABSTRACT

Cyclophosphamide is one of several clinically important cancer drugs whose therapeutic efficacy is due in part to their ability to stimulate antitumor immune responses. Studying mouse models, we demonstrate that cyclophosphamide alters the composition of microbiota in the small intestine and induces the translocation of selected species of Gram-positive bacteria into secondary lymphoid organs. There, these bacteria stimulate the generation of a specific subset of "pathogenic" T helper 17 (pT(H)17) cells and memory T(H)1 immune responses. Tumor-bearing mice that were germ-free or that had been treated with antibiotics to kill Gram-positive bacteria showed a reduction in pT(H)17 responses, and their tumors were resistant to cyclophosphamide. Adoptive transfer of pT(H)17 cells partially restored the antitumor efficacy of cyclophosphamide. These results suggest that the gut microbiota help shape the anticancer immune response.


Subject(s)
Antineoplastic Agents/therapeutic use , Bacterial Translocation/drug effects , Cyclophosphamide/therapeutic use , Immunosuppressive Agents/therapeutic use , Intestine, Small/microbiology , Microbiota/physiology , Neoplasms/drug therapy , Neoplasms/immunology , Adoptive Transfer , Animals , Anti-Bacterial Agents/administration & dosage , Germ-Free Life , Gram-Positive Bacteria/drug effects , Gram-Positive Bacteria/physiology , Immunologic Memory , Lymphoid Tissue/immunology , Lymphoid Tissue/microbiology , Mice , Microbiota/drug effects , Th17 Cells/immunology , Th17 Cells/transplantation
13.
Oncoimmunology ; 2(4): e23714, 2013 Apr 01.
Article in English | MEDLINE | ID: mdl-23734324

ABSTRACT

Melanoma is a highly chemoresistant and metastatic tumor that, in the absence of BRAF mutations, is generally treated with the alkylating agent dacarbazine (DTIC). We discovered that DTIC upregulates the expression of NKG2D ligands on tumor cells, leading to the activation of natural killer (NK) and CD8+ T cells. These observations underscore the immunogenic properties of DTIC and provide a rationale to combine DTIC with immunotherapeutic agents.

14.
PLoS One ; 8(6): e65181, 2013.
Article in English | MEDLINE | ID: mdl-23762310

ABSTRACT

Bleomycin (BLM) is an anticancer drug currently used for the treatment of testis cancer and Hodgkin lymphoma. This drug triggers cancer cell death via its capacity to generate radical oxygen species (ROS). However, the putative contribution of anticancer immune responses to the efficacy of BLM has not been evaluated. We make here the observation that BLM induces immunogenic cell death. In particular, BLM is able to induce ROS-mediated reticulum stress and autophagy, which result in the surface exposure of chaperones, including calreticulin and ERp57, and liberation of HMBG1 and ATP. BLM induces anti-tumor immunity which relies on calreticulin, CD8(+) T cells and interferon-γ. We also find that, in addition to its capacity to trigger immunogenic cell death, BLM induces expansion of Foxp3+ regulatory T (Treg) cells via its capacity to induce transforming growth factor beta (TGFß) secretion by tumor cells. Accordingly, Treg cells or TGFß depletion dramatically potentiates the antitumor effect of BLM. We conclude that BLM induces both anti-tumor CD8(+) T cell response and a counteracting Treg proliferation. In the future, TGFß or Treg inhibition during BLM treatment could greatly enhance BLM anti-tumor efficacy.


Subject(s)
Antineoplastic Agents/pharmacology , Bleomycin/pharmacology , Immunity/drug effects , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/pathology , Animals , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Death/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Female , Humans , Immunity, Innate/drug effects , Mice , T-Lymphocytes, Regulatory/drug effects , Transforming Growth Factor beta/metabolism
15.
Cancer Res ; 73(12): 3578-90, 2013 Jun 15.
Article in English | MEDLINE | ID: mdl-23619236

ABSTRACT

Activation of the transcription factor PPARγ by the n-3 fatty acid docosahexaenoic acid (DHA) is implicated in controlling proinflammatory cytokine secretion, but the intracellular signaling pathways engaged by PPARγ are incompletely characterized. Here, we identify the adapter-encoding gene SOCS3 as a critical transcriptional target of PPARγ. SOCS3 promoter binding and gene transactivation by PPARγ was associated with a repression in differentiation of proinflammatory T-helper (TH)17 cells. Accordingly, TH17 cells induced in vitro displayed increased SOCS3 expression and diminished capacity to produce interleukin (IL)-17 following activation of PPARγ by DHA. Furthermore, naïve CD4 T cells derived from mice fed a DHA-enriched diet displayed less capability to differentiate into TH17 cells. In two different mouse models of cancer, DHA prevented tumor outgrowth and angiogenesis in an IL-17-dependent manner. Altogether, our results uncover a novel molecular pathway by which PPARγ-induced SOCS3 expression prevents IL-17-mediated cancer growth.


Subject(s)
Mammary Neoplasms, Experimental/genetics , PPAR gamma/genetics , Suppressor of Cytokine Signaling Proteins/genetics , Transcriptional Activation , Animals , Blotting, Western , CD4-Positive T-Lymphocytes/drug effects , CD4-Positive T-Lymphocytes/metabolism , Cell Differentiation/drug effects , Cell Line, Tumor , Diet , Docosahexaenoic Acids/administration & dosage , Docosahexaenoic Acids/pharmacology , Female , Interleukin-17/metabolism , Mammary Neoplasms, Experimental/pathology , Mammary Neoplasms, Experimental/prevention & control , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Mice, Nude , PPAR gamma/agonists , PPAR gamma/metabolism , Promoter Regions, Genetic/genetics , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Suppressor of Cytokine Signaling 3 Protein , Suppressor of Cytokine Signaling Proteins/metabolism , Th17 Cells/drug effects , Th17 Cells/metabolism , Tumor Burden/drug effects , Tumor Burden/genetics
16.
J Invest Dermatol ; 133(2): 499-508, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22951720

ABSTRACT

Dacarbazine (DTIC) is a cytotoxic drug widely used for melanoma treatment. However, the putative contribution of anticancer immune responses in the efficacy of DTIC has not been evaluated. By testing how DTIC affects host immune responses to cancer in a mouse model of melanoma, we unexpectedly found that both natural killer (NK) and CD8(+) T cells were indispensable for DTIC therapeutic effect. Although DTIC did not directly affect immune cells, it triggered the upregulation of NKG2D ligands on tumor cells, leading to NK cell activation and IFNγ secretion in mice and humans. NK cell-derived IFNγ subsequently favored upregulation of major histocompatibility complex class I molecules on tumor cells, rendering them sensitive to cytotoxic CD8(+) T cells. Accordingly, DTIC markedly enhanced cytotoxic T lymphocyte antigen 4 inhibition efficacy in vivo in an NK-dependent manner. These results underscore the immunogenic properties of DTIC and provide a rationale to combine DTIC with immunotherapeutic agents that relieve immunosuppression in vivo.


Subject(s)
CD8-Positive T-Lymphocytes/drug effects , Dacarbazine/pharmacology , Killer Cells, Natural/drug effects , Melanoma, Experimental/drug therapy , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Skin Neoplasms/drug therapy , Animals , Antineoplastic Agents, Alkylating/pharmacology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/metabolism , Cell Line, Tumor , Disease Models, Animal , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Ligands , Melanoma, Experimental/immunology , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mice, Nude , NK Cell Lectin-Like Receptor Subfamily K/immunology , Skin Neoplasms/immunology , Skin Neoplasms/pathology , Up-Regulation/drug effects , Up-Regulation/immunology
17.
Nat Med ; 19(1): 57-64, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23202296

ABSTRACT

Chemotherapeutic agents are widely used for cancer treatment. In addition to their direct cytotoxic effects, these agents harness the host's immune system, which contributes to their antitumor activity. Here we show that two clinically used chemotherapeutic agents, gemcitabine (Gem) and 5-fluorouracil (5FU), activate the NOD-like receptor family, pyrin domain containing-3 protein (Nlrp3)-dependent caspase-1 activation complex (termed the inflammasome) in myeloid-derived suppressor cells (MDSCs), leading to production of interleukin-1ß (IL-1ß), which curtails anticancer immunity. Chemotherapy-triggered IL-1ß secretion relied on lysosomal permeabilization and the release of cathepsin B, which bound to Nlrp3 and drove caspase-1 activation. MDSC-derived IL-1ß induced secretion of IL-17 by CD4(+) T cells, which blunted the anticancer efficacy of the chemotherapy. Accordingly, Gem and 5FU exerted higher antitumor effects when tumors were established in Nlrp3(-/-) or Casp1(-/-) mice or wild-type mice treated with interleukin-1 receptor antagonist (IL-1Ra). Altogether, these results identify how activation of the Nlrp3 inflammasome in MDSCs by 5FU and Gem limits the antitumor efficacy of these chemotherapeutic agents.


Subject(s)
Carrier Proteins/metabolism , Caspase 1/metabolism , Deoxycytidine/analogs & derivatives , Fluorouracil/pharmacology , Inflammasomes/metabolism , Neoplasms/metabolism , Animals , Antineoplastic Agents/pharmacology , CD4-Positive T-Lymphocytes/metabolism , Carrier Proteins/genetics , Caspase 1/genetics , Cathepsin B/genetics , Cathepsin B/metabolism , Cell Line, Tumor , Cell Proliferation , Dendritic Cells/metabolism , Deoxycytidine/pharmacology , Female , Humans , Interleukin-17/metabolism , Interleukin-1beta/biosynthesis , Macrophages/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , NLR Family, Pyrin Domain-Containing 3 Protein , Neoplasms/genetics , RNA Interference , RNA, Small Interfering , Receptors, Interleukin-1/antagonists & inhibitors , Signal Transduction/immunology , bcl-2-Associated X Protein/genetics , Gemcitabine
18.
Sci Transl Med ; 4(143): 143ra99, 2012 Jul 18.
Article in English | MEDLINE | ID: mdl-22814852

ABSTRACT

Some successful chemotherapeutics, notably anthracyclines and oxaliplatin, induce a type of cell stress and death that is immunogenic, hence converting the patient's dying cancer cells into a vaccine that stimulates antitumor immune responses. By means of a fluorescence microscopy platform that allows for the automated detection of the biochemical hallmarks of such a peculiar cell death modality, we identified cardiac glycosides (CGs) as exceptionally efficient inducers of immunogenic cell death, an effect that was associated with the inhibition of the plasma membrane Na(+)- and K(+)-dependent adenosine triphosphatase (Na(+)/K(+)-ATPase). CGs exacerbated the antineoplastic effects of DNA-damaging agents in immunocompetent but not immunodeficient mice. Moreover, cancer cells succumbing to a combination of chemotherapy plus CGs could vaccinate syngeneic mice against a subsequent challenge with living cells of the same type. Finally, retrospective clinical analyses revealed that the administration of the CG digoxin during chemotherapy had a positive impact on overall survival in cohorts of breast, colorectal, head and neck, and hepatocellular carcinoma patients, especially when they were treated with agents other than anthracyclines and oxaliplatin.


Subject(s)
Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Cardiac Glycosides/pharmacology , Cardiac Glycosides/therapeutic use , Neoplasms/drug therapy , Animals , Anthracyclines/pharmacology , Anthracyclines/therapeutic use , Biosensing Techniques , Cell Line, Tumor , Digoxin/pharmacology , Digoxin/therapeutic use , Humans , Mice , Neoplasms/metabolism , Organoplatinum Compounds/pharmacology , Organoplatinum Compounds/therapeutic use , Oxaliplatin
19.
Immunity ; 36(3): 362-73, 2012 Mar 23.
Article in English | MEDLINE | ID: mdl-22406269

ABSTRACT

Although Th17 cells are known to promote tissue inflammation and autoimmunity, their role during cancer progression remains elusive. Here, we showed that in vitro Th17 cells generated with the cytokines IL-6 and TGF-ß expressed CD39 and CD73 ectonucleotidases, leading to adenosine release and the subsequent suppression of CD4(+) and CD8(+) T cell effector functions. The IL-6-mediated activation of the transcription factor Stat3 and the TGF-ß-driven downregulation of Gfi-1 transcription factor were both essential for the expression of ectonucleotidases during Th17 cell differentiation. Stat3 supported whereas Gfi-1 repressed CD39 and CD73 expression by binding to their promoters. Accordingly, Th17 cells differentiated with IL-1ß, IL-6, and IL-23 but without TGF-ß did not express ectonucleotidases and were not immunosuppressive. Finally, adoptive transfer of Th17 cells induced by TGF-ß and IL-6 promoted tumor growth in a CD39-dependent manner. Thus, ectonucleotidase expression supports the immunosuppressive fate of Th17 cells in cancer.


Subject(s)
5'-Nucleotidase/genetics , Antigens, CD/genetics , Apyrase/genetics , DNA-Binding Proteins/immunology , STAT3 Transcription Factor/immunology , Th17 Cells/immunology , Th17 Cells/metabolism , Transcription Factors/immunology , Animals , Binding Sites/genetics , DNA-Binding Proteins/metabolism , Gene Expression Regulation , Interleukin-6/pharmacology , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Promoter Regions, Genetic , RNA, Small Interfering/genetics , STAT3 Transcription Factor/antagonists & inhibitors , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Th17 Cells/drug effects , Transcription Factors/metabolism , Transforming Growth Factor beta/pharmacology
20.
Science ; 334(6062): 1573-7, 2011 Dec 16.
Article in English | MEDLINE | ID: mdl-22174255

ABSTRACT

Antineoplastic chemotherapies are particularly efficient when they elicit immunogenic cell death, thus provoking an anticancer immune response. Here we demonstrate that autophagy, which is often disabled in cancer, is dispensable for chemotherapy-induced cell death but required for its immunogenicity. In response to chemotherapy, autophagy-competent, but not autophagy-deficient, cancers attracted dendritic cells and T lymphocytes into the tumor bed. Suppression of autophagy inhibited the release of adenosine triphosphate (ATP) from dying tumor cells. Conversely, inhibition of extracellular ATP-degrading enzymes increased pericellular ATP in autophagy-deficient tumors, reestablished the recruitment of immune cells, and restored chemotherapeutic responses but only in immunocompetent hosts. Thus, autophagy is essential for the immunogenic release of ATP from dying cells, and increased extracellular ATP concentrations improve the efficacy of antineoplastic chemotherapies when autophagy is disabled.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/physiology , Neoplasms/immunology , Adenosine Triphosphate/metabolism , Animals , Antineoplastic Agents/therapeutic use , Autophagy/drug effects , Calreticulin/pharmacology , Cell Death/immunology , Cell Line, Tumor , Dendritic Cells/immunology , Humans , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mitoxantrone/pharmacology , Neoplasms/drug therapy
SELECTION OF CITATIONS
SEARCH DETAIL
...