Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 11 de 11
Filter
Add more filters










Publication year range
1.
Cell ; 184(21): 5338-5356.e21, 2021 10 14.
Article in English | MEDLINE | ID: mdl-34624222

ABSTRACT

The tumor microenvironment (TME) influences cancer progression and therapy response. Therefore, understanding what regulates the TME immune compartment is vital. Here we show that microbiota signals program mononuclear phagocytes in the TME toward immunostimulatory monocytes and dendritic cells (DCs). Single-cell RNA sequencing revealed that absence of microbiota skews the TME toward pro-tumorigenic macrophages. Mechanistically, we show that microbiota-derived stimulator of interferon genes (STING) agonists induce type I interferon (IFN-I) production by intratumoral monocytes to regulate macrophage polarization and natural killer (NK) cell-DC crosstalk. Microbiota modulation with a high-fiber diet triggered the intratumoral IFN-I-NK cell-DC axis and improved the efficacy of immune checkpoint blockade (ICB). We validated our findings in individuals with melanoma treated with ICB and showed that the predicted intratumoral IFN-I and immune compositional differences between responder and non-responder individuals can be transferred by fecal microbiota transplantation. Our study uncovers a mechanistic link between the microbiota and the innate TME that can be harnessed to improve cancer therapies.


Subject(s)
Interferon Type I/metabolism , Membrane Proteins/metabolism , Microbiota , Monocytes/metabolism , Tumor Microenvironment , Akkermansia/drug effects , Akkermansia/physiology , Animals , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Dietary Fiber/pharmacology , Dinucleoside Phosphates/administration & dosage , Dinucleoside Phosphates/pharmacology , Humans , Immune Checkpoint Inhibitors/pharmacology , Immunomodulation/drug effects , Killer Cells, Natural/drug effects , Killer Cells, Natural/metabolism , Macrophages/drug effects , Macrophages/metabolism , Melanoma/immunology , Melanoma/pathology , Mice, Inbred BALB C , Mice, Inbred C57BL , Microbiota/drug effects , Monocytes/drug effects , Phagocytes/drug effects , Phagocytes/metabolism , Transcription, Genetic/drug effects , Tumor Microenvironment/drug effects
3.
Nat Med ; 26(5): 781-791, 2020 05.
Article in English | MEDLINE | ID: mdl-32284588

ABSTRACT

Although immunotherapy has revolutionized cancer treatment, only a subset of patients demonstrate durable clinical benefit. Definitive predictive biomarkers and targets to overcome resistance remain unidentified, underscoring the urgency to develop reliable immunocompetent models for mechanistic assessment. Here we characterize a panel of syngeneic mouse models, representing a variety of molecular and phenotypic subtypes of human melanomas and exhibiting their diverse range of responses to immune checkpoint blockade (ICB). Comparative analysis of genomic, transcriptomic and tumor-infiltrating immune cell profiles demonstrated alignment with clinical observations and validated the correlation of T cell dysfunction and exclusion programs with resistance. Notably, genome-wide expression analysis uncovered a melanocytic plasticity signature predictive of patient outcome in response to ICB, suggesting that the multipotency and differentiation status of melanoma can determine ICB benefit. Our comparative preclinical platform recapitulates melanoma clinical behavior and can be employed to identify mechanisms and treatment strategies to improve patient care.


Subject(s)
Drug Screening Assays, Antitumor , Immunotherapy , Melanoma/pathology , Melanoma/therapy , Animals , Antineoplastic Agents, Immunological/therapeutic use , CTLA-4 Antigen/immunology , Cells, Cultured , Disease Models, Animal , Drug Screening Assays, Antitumor/methods , Female , Gene Expression Regulation, Neoplastic/drug effects , Genetic Heterogeneity , Humans , Immunotherapy/adverse effects , Immunotherapy/methods , Ipilimumab/therapeutic use , Melanoma/diagnosis , Melanoma/genetics , Mice , Mice, Inbred C57BL , Mice, Transgenic , Prognosis , Programmed Cell Death 1 Receptor/immunology , RNA-Seq , Treatment Outcome , Whole Genome Sequencing
4.
Methods Enzymol ; 632: 309-337, 2020.
Article in English | MEDLINE | ID: mdl-32000903

ABSTRACT

The tumor microenvironment (TME) is a highly complex and dynamic ensemble of cells of which a variety of immune cells are a major component. The unparalleled results obtained with immunotherapeutic approaches have underscored the importance of examining the immune landscape of the TME. Recent technological advances have incorporated high-throughput techniques at the single cell level, such as single cell RNA sequencing, mass cytometry, and multi-parametric flow cytometry to the characterization of the TME. Among them, flow cytometry is the most broadly used both in research and clinical settings and multi-color analysis is now routinely performed. The high dimensionality of the data makes the traditional manual gating strategy in 2D scatter plots very difficult. New unbiased visualization techniques provide a solution to this problem. Here we describe the steps to characterize the immune cell compartment in the TME in mouse tumor models by high-parametric flow cytometry, from the experimental setup to the analysis methodology with special emphasis on the use of unsupervised algorithms.


Subject(s)
Flow Cytometry/methods , Immune System/cytology , Neoplasms/immunology , Tumor Microenvironment , Algorithms , Animals , Cell Culture Techniques/methods , Cell Line, Tumor , Cluster Analysis , Immune System/immunology , Mice
5.
J Immunol ; 200(3): 1008-1015, 2018 02 01.
Article in English | MEDLINE | ID: mdl-29282306

ABSTRACT

NK cells play important roles during immunosurveillance against tumors and viruses as they trigger cytotoxicity against susceptible cells and secrete proinflammatory cytokines such as IFN-γ. In addition, upon activation, macrophages can become proinflammatory (M1) or anti-inflammatory (M2) cells. Although the consequences of the cross-talk between M1 and NK cells are known, the outcome of the cross-talk between M2 and NK cells remains ill-defined. Therefore, in the current work, we investigated the outcome and the underlying mechanisms of the interaction between resting or stimulated human NK cells with M1 or M2. We observed a lower percentage of activated NK cells that produced less IFN-γ upon coculture with M2. Also, CD56dim NK cells cocultured with M2 displayed lower degranulation and cytotoxic activity than NK cells cocultured with M1. Soluble TGF-ß and M2-driven upregulation of CD85j (ILT-2) on NK cells accounted for the diminished IFN-γ production by CD56bright NK cells, whereas M2-driven upregulation of CD85j on NK cells accounted for the generation of hyporesponsive CD56dim NK cells with limited degranulation and cytotoxic capacity. Accordingly, M2 expressed higher amounts of HLA-G, the main ligand for CD85j, than M1. Hyporesponsiveness to degranulation in NK cells was not restored at least for several hours upon removal of M2. Therefore, alternatively activated macrophages restrain NK cell activation and effector functions through different mechanisms, leading to NK cells that display diminished IFN-γ production and at least a transiently impaired degranulation ability. These results unravel an inhibitory circuit of possible relevance in pathological situations.


Subject(s)
Cell Communication/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Macrophage Activation/immunology , Transforming Growth Factor beta/metabolism , Antigens, CD/immunology , CD56 Antigen/metabolism , Cells, Cultured , Coculture Techniques , HLA-G Antigens/metabolism , Humans , Interferon-gamma/immunology , Leukocyte Immunoglobulin-like Receptor B1/immunology , Macrophages/immunology , Transforming Growth Factor beta/immunology
6.
Cancer Cell ; 31(2): 161-163, 2017 02 13.
Article in English | MEDLINE | ID: mdl-28196588

ABSTRACT

Type I interferons have been shown to play a major role in anti-cancer immunity. In this issue of Cancer Cell, Katlinski et al. describe tumor-induced degradation of type I interferon receptor IFNAR1 chain as a new immune-evasion mechanism in colorectal cancers. Stabilizing IFNAR1 inhibits tumor growth and improves immunotherapy efficacy.


Subject(s)
Interferon Type I/metabolism , Tumor Escape , Humans
7.
Immunity ; 45(4): 714-716, 2016 10 18.
Article in English | MEDLINE | ID: mdl-27760335

ABSTRACT

It has recently become apparent that the gut microbiota modulates the response to cancer therapy. In this issue of Immunity, Daillère et al. (2016) identified two bacterial species potentiating the anti-tumor effect of cyclophosphamide that are kept in check by the sensor NOD2.


Subject(s)
Bacteria/metabolism , Gastrointestinal Microbiome/physiology , Neoplasms/microbiology , Nod2 Signaling Adaptor Protein/metabolism , Animals , Cyclophosphamide/pharmacology , Cyclophosphamide/therapeutic use , Humans , Neoplasms/drug therapy
8.
J Immunol ; 197(3): 953-61, 2016 08 01.
Article in English | MEDLINE | ID: mdl-27342842

ABSTRACT

Despite the classical function of NK cells in the elimination of tumor and of virus-infected cells, evidence for a regulatory role for NK cells has been emerging in different models of autoimmunity, transplantation, and viral infections. However, this role has not been fully explored in the context of a growing tumor. In this article, we show that NK cells can limit spontaneous cross-priming of tumor Ag-specific CD8(+) T cells, leading to reduced memory responses. After challenge with MC57 cells transduced to express the model Ag SIY (MC57.SIY), NK cell-depleted mice exhibited a significantly higher frequency of SIY-specific CD8(+) T cells, with enhanced IFN-γ production and cytotoxic capability. Depletion of NK cells resulted in a CD8(+) T cell population skewed toward an effector memory T phenotype that was associated with enhanced recall responses and delayed tumor growth after a secondary tumor challenge with B16.SIY cells. Dendritic cells (DCs) from NK cell-depleted tumor-bearing mice exhibited a more mature phenotype. Interestingly, tumor-infiltrating and tumor-draining lymph node NK cells displayed an upregulated expression of the inhibitory molecule programmed death ligand 1 that, through interaction with programmed death-1 expressed on DCs, limited DC activation, explaining their reduced ability to induce tumor-specific CD8(+) T cell priming. Our results suggest that NK cells can, in certain contexts, have an inhibitory effect on antitumor immunity, a finding with implications for immunotherapy in the clinic.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Cross-Priming/immunology , Dendritic Cells/immunology , Killer Cells, Natural/immunology , Lymphocyte Activation/immunology , Neoplasms, Experimental/immunology , Animals , B7-H1 Antigen/immunology , Cell Line, Tumor , Cell Separation , Flow Cytometry , Humans , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/immunology , Real-Time Polymerase Chain Reaction , Signal Transduction/immunology
9.
Am J Physiol Gastrointest Liver Physiol ; 311(1): G40-9, 2016 07 01.
Article in English | MEDLINE | ID: mdl-27151946

ABSTRACT

Celiac disease (CD) is an immune-mediated enteropathy triggered by gluten in genetically susceptible individuals. Innate immunity contributes to the pathogenesis of CD, but the mechanisms remain poorly understood. Although previous in vitro work suggests that gliadin peptide p31-43 acts as an innate immune trigger, the underlying pathways are unclear and have not been explored in vivo. Here we show that intraluminal delivery of p31-43 induces morphological changes in the small intestinal mucosa of normal mice consistent with those seen in CD, including increased cell death and expression of inflammatory mediators. The effects of p31-43 were dependent on MyD88 and type I IFNs, but not Toll-like receptor 4 (TLR4), and were enhanced by coadministration of the TLR3 agonist polyinosinic:polycytidylic acid. Together, these results indicate that gliadin peptide p31-43 activates the innate immune pathways in vivo, such as IFN-dependent inflammation, relevant to CD. Our findings also suggest a common mechanism for the potential interaction between dietary gluten and viral infections in the pathogenesis of CD.


Subject(s)
Celiac Disease/immunology , Gliadin/toxicity , Immunity, Innate/drug effects , Immunity, Mucosal/drug effects , Intestinal Mucosa/drug effects , Intestine, Small/drug effects , Peptide Fragments/toxicity , Animals , Apoptosis/drug effects , Apoptosis Regulatory Proteins/metabolism , Celiac Disease/metabolism , Celiac Disease/pathology , Gene Expression Regulation , Genotype , Gliadin/administration & dosage , Inflammation Mediators/metabolism , Interferon Type I/genetics , Interferon Type I/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestinal Mucosa/pathology , Intestine, Small/immunology , Intestine, Small/metabolism , Intestine, Small/pathology , Male , Mice, Inbred C57BL , Mice, Knockout , Myeloid Differentiation Factor 88/metabolism , Peptide Fragments/administration & dosage , Phenotype , Poly I-C/pharmacology , Receptor, Interferon alpha-beta/deficiency , Receptor, Interferon alpha-beta/genetics , Signal Transduction/drug effects , Toll-Like Receptor 3/agonists , Toll-Like Receptor 3/metabolism
10.
PLoS One ; 9(6): e99236, 2014.
Article in English | MEDLINE | ID: mdl-24915573

ABSTRACT

Systemic administration of polyinosinic:polycytidylic acid (poly I:C), mimics virally-induced activation of TLR3 signalling causing acute small intestine damage, but whether and how mucosal administration of poly I:C causes enteropathy is less clear. Our aim was to investigate the inflammatory pathways elicited after intraluminal administration of poly I:C and determine acute and delayed consequences of this locally induced immune activation. Intraluminal poly I:C induced rapid mucosal immune activation in C57BL/6 mice involving IFNß and the CXCL10/CXCR3 axis, that may drive inflammation towards a Th1 profile. Intraluminal poly I:C also caused enteropathy and gut dysfunction in gliadin-sensitive NOD-DQ8 mice, and this was prolonged by concomitant oral administration of gliadin. Our results indicate that small intestine pathology can be induced in mice by intraluminal administration of poly I:C and that this is exacerbated by subsequent oral delivery of a relevant dietary antigen.


Subject(s)
Disease Progression , Gliadin/administration & dosage , Gliadin/adverse effects , Intestinal Diseases/chemically induced , Intestinal Diseases/pathology , Poly I-C/administration & dosage , Poly I-C/adverse effects , Administration, Oral , Animals , Cytokines/blood , DEAD Box Protein 58 , DEAD-box RNA Helicases/genetics , DEAD-box RNA Helicases/metabolism , Female , Gene Expression Regulation/drug effects , Inflammation Mediators/metabolism , Interferon-Induced Helicase, IFIH1 , Intestinal Diseases/blood , Intestinal Mucosa/drug effects , Intestine, Small/drug effects , Intestine, Small/metabolism , Mice, Inbred C57BL , Mice, Inbred NOD , RNA, Messenger/genetics , RNA, Messenger/metabolism , Real-Time Polymerase Chain Reaction , Toll-Like Receptor 3/genetics , Toll-Like Receptor 3/metabolism
11.
PLoS One ; 9(2): e89068, 2014.
Article in English | MEDLINE | ID: mdl-24586509

ABSTRACT

Lymphocytic infiltration in the lamina propria (LP), which is primarily composed of CD4(+) Th1 cells and plasma cells, and increased numbers of intraepithelial lymphocytes (IELs), is a characteristic finding in active celiac disease (CD). Signals for this selective cell recruitment have not been fully established. CXCR3 and its ligands, particularly CXCL10, have been suggested to be one of the most relevant pathways in the attraction of cells into inflamed tissues. In addition, CXCR3 is characteristically expressed by Th1 cells. The aim of this work was to investigate the participation of the chemokine CXCL10/CXCR3 axis in CD pathogenesis. A higher concentration of CXCL10 was found in the serum of untreated CD patients. The mRNA levels of CXCL10 and CXCL11 but not CXCL9 were significantly higher in duodenal biopsies from untreated CD patients compared with non-CD controls or treated patients. The results demonstrate that CXCL10 is abundantly produced in untreated CD and reduced in treated patients, and the expression of CXCL10 was found to be correlated with the IFNγ levels in the tissue. Plasma cells and enterocytes were identified as CXCL10-producing cells. Moreover, the CXCL10 expression in intestinal tissues was upregulated by poly I:C and IL-15. IELs, LP T lymphocytes, and plasma cells, which infiltrate the intestinal mucosa in untreated CD, express CXCR3. The CXCR3/CXCL10 signalling axis is overactivated in the small intestinal mucosa in untreated patients, and this finding explains the specific recruitment of the major cell populations that infiltrate the epithelium and the LP in CD.


Subject(s)
Celiac Disease/immunology , Celiac Disease/metabolism , Chemokine CXCL10/metabolism , Intestine, Small/immunology , Plasma Cells/immunology , Receptors, CXCR3/metabolism , T-Lymphocytes/immunology , Adult , Celiac Disease/blood , Celiac Disease/pathology , Chemokine CXCL10/biosynthesis , Chemokine CXCL10/blood , Chemokine CXCL11/metabolism , Chemokine CXCL9/metabolism , Child , Gene Expression Regulation/immunology , Humans , Interferon-beta/metabolism , Interferon-gamma/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Intestine, Small/metabolism , Signal Transduction/immunology , Tumor Necrosis Factor-alpha/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...