Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 7.753
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 42(1): 347-373, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38941603

ABSTRACT

Plasmacytoid dendritic cells (pDCs) represent a unique cell type within the innate immune system. Their defining property is the recognition of pathogen-derived nucleic acids through endosomal Toll-like receptors and the ensuing production of type I interferon and other soluble mediators, which orchestrate innate and adaptive responses. We review several aspects of pDC biology that have recently come to the fore. We discuss emerging questions regarding the lineage affiliation and origin of pDCs and argue that these cells constitute an integral part of the dendritic cell lineage. We emphasize the specific function of pDCs as innate sentinels of virus infection, particularly their recognition of and distinct response to virus-infected cells. This essential evolutionary role of pDCs has been particularly important for the control of coronaviruses, as demonstrated by the recent COVID-19 pandemic. Finally, we highlight the key contribution of pDCs to systemic lupus erythematosus, in which therapeutic targeting of pDCs is currently underway.


Subject(s)
COVID-19 , Dendritic Cells , Immunity, Innate , Lupus Erythematosus, Systemic , SARS-CoV-2 , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , COVID-19/immunology , Animals , SARS-CoV-2/immunology , SARS-CoV-2/physiology , Lupus Erythematosus, Systemic/immunology , Toll-Like Receptors/metabolism , Cell Differentiation , Cell Lineage
2.
Annu Rev Immunol ; 40: 559-587, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35113732

ABSTRACT

The immune system employs recognition tools to communicate with its microbial evolutionary partner. Among all the methods of microbial perception, T cells enable the widest spectrum of microbial recognition resolution, ranging from the crudest detection of whole groups of microbes to the finest detection of specific antigens. The application of this recognition capability to the crucial task of combatting infections has been the focus of classical immunology. We now appreciate that the coevolution of the immune system and the microbiota has led to development of a lush immunological decision tree downstream of microbial recognition, of which an inflammatory response is but one branch. In this review we discuss known T cell-microbe interactions in the gut and place them in the context of an algorithmic framework of recognition, context-dependent interpretation, and response circuits across multiple levels of microbial recognition resolution. The malleability of T cells in response to the microbiota presents an opportunity to edit immune response cellularity, identity, and functionality by utilizing microbiota-controlled pathways to promote human health.


Subject(s)
Microbiota , T-Lymphocytes , Animals , Humans
3.
Annu Rev Immunol ; 39: 131-166, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33481643

ABSTRACT

Dendritic cells (DCs) possess the ability to integrate information about their environment and communicate it to other leukocytes, shaping adaptive and innate immunity. Over the years, a variety of cell types have been called DCs on the basis of phenotypic and functional attributes. Here, we refocus attention on conventional DCs (cDCs), a discrete cell lineage by ontogenetic and gene expression criteria that best corresponds to the cells originally described in the 1970s. We summarize current knowledge of mouse and human cDC subsets and describe their hematopoietic development and their phenotypic and functional attributes. We hope that our effort to review the basic features of cDC biology and distinguish cDCs from related cell types brings to the fore the remarkable properties of this cell type while shedding some light on the seemingly inordinate complexity of the DC field.


Subject(s)
Dendritic Cells , Immunity, Innate , Animals , Cell Lineage , Humans , Mice
4.
Annu Rev Immunol ; 39: 759-790, 2021 04 26.
Article in English | MEDLINE | ID: mdl-33710920

ABSTRACT

As the professional antigen-presenting cells of the immune system, dendritic cells (DCs) sense the microenvironment and shape the ensuing adaptive immune response. DCs can induce both immune activation and immune tolerance according to the peripheral cues. Recent work has established that DCs comprise several phenotypically and functionally heterogeneous subsets that differentially regulate T lymphocyte differentiation. This review summarizes both mouse and human DC subset phenotypes, development, diversification, and function. We focus on advances in our understanding of how different DC subsets regulate distinct CD4+ T helper (Th) cell differentiation outcomes, including Th1, Th2, Th17, T follicular helper, and T regulatory cells. We review DC subset intrinsic properties, local tissue microenvironments, and other immune cells that together determine Th cell differentiation during homeostasis and inflammation.


Subject(s)
Immune Tolerance , Lymphocyte Activation , Animals , Dendritic Cells , Humans , Mice , T-Lymphocytes, Regulatory , Th17 Cells
5.
Annu Rev Immunol ; 37: 269-293, 2019 04 26.
Article in English | MEDLINE | ID: mdl-30649988

ABSTRACT

Myeloid cells are a major cellular compartment of the immune system comprising monocytes, dendritic cells, tissue macrophages, and granulocytes. Models of cellular ontogeny, activation, differentiation, and tissue-specific functions of myeloid cells have been revisited during the last years with surprising results; for example, most tissue macrophages are yolk sac derived, monocytes and macrophages follow a multidimensional model of activation, and tissue signals have a significant impact on the functionality of all these cells. While these exciting results have brought these cells back to center stage, their enormous plasticity and heterogeneity, during both homeostasis and disease, are far from understood. At the same time, the ongoing revolution in single-cell genomics, with single-cell RNA sequencing (scRNA-seq) leading the way, promises to change this. Prevailing models of hematopoiesis with distinct intermediates are challenged by scRNA-seq data suggesting more continuous developmental trajectories in the myeloid cell compartment. Cell subset structures previously defined by protein marker expression need to be revised based on unbiased analyses of scRNA-seq data. Particularly in inflammatory conditions, myeloid cells exhibit substantially vaster heterogeneity than previously anticipated, and work performed within large international projects, such as the Human Cell Atlas, has already revealed novel tissue macrophage subsets. Based on these exciting developments, we propose the next steps to a full understanding of the myeloid cell compartment in health and diseases.


Subject(s)
Cell Differentiation , Cellular Microenvironment , Inflammation/immunology , Myeloid Cells/physiology , Animals , Biomarkers , Cell Plasticity , Homeostasis , Humans , Sequence Analysis, RNA
6.
Annu Rev Immunol ; 36: 435-459, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29400984

ABSTRACT

The initiation and maintenance of adaptive immunity require multifaceted modes of communication between different types of immune cells, including direct intercellular contact, secreted soluble signaling molecules, and extracellular vesicles (EVs). EVs can be formed as microvesicles directly pinched off from the plasma membrane or as exosomes secreted by multivesicular endosomes. Membrane receptors guide EVs to specific target cells, allowing directional transfer of specific and complex signaling cues. EVs are released by most, if not all, immune cells. Depending on the type and status of their originating cell, EVs may facilitate the initiation, expansion, maintenance, or silencing of adaptive immune responses. This review focusses on EVs from professional antigen-presenting cells, their demonstrated and speculated roles, and their potential for cancer immunotherapy.


Subject(s)
Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Extracellular Vesicles/metabolism , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Biological Transport , Cell-Derived Microparticles/metabolism , Dendritic Cells/immunology , Dendritic Cells/metabolism , Epithelial Cells/metabolism , Exosomes/metabolism , Histocompatibility Antigens/genetics , Histocompatibility Antigens/immunology , Humans , Immune Tolerance , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Macrophages/immunology , Macrophages/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
7.
Annu Rev Immunol ; 36: 717-753, 2018 04 26.
Article in English | MEDLINE | ID: mdl-29490164

ABSTRACT

Antigen cross-presentation is an adaptation of the cellular process of loading MHC-I molecules with endogenous peptides during their biosynthesis within the endoplasmic reticulum. Cross-presented peptides derive from internalized proteins, microbial pathogens, and transformed or dying cells. The physical separation of internalized cargo from the endoplasmic reticulum, where the machinery for assembling peptide-MHC-I complexes resides, poses a challenge. To solve this problem, deliberate rewiring of organelle communication within cells is necessary to prepare for cross-presentation, and different endocytic receptors and vesicular traffic patterns customize the emergent cross-presentation compartment to the nature of the peptide source. Three distinct pathways of vesicular traffic converge to form the ideal cross-presentation compartment, each regulated differently to supply a unique component that enables cross-presentation of a diverse repertoire of peptides. Delivery of centerpiece MHC-I molecules is the critical step regulated by microbe-sensitive Toll-like receptors. Defining the subcellular sources of MHC-I and identifying sites of peptide loading during cross-presentation remain key challenges.


Subject(s)
Antigen Presentation/immunology , Antigens/immunology , Cross-Priming/immunology , Immunomodulation , Animals , Biological Transport , Dendritic Cells/immunology , Dendritic Cells/metabolism , Endocytosis/immunology , Endoplasmic Reticulum/metabolism , Endosomes/metabolism , Epitopes/immunology , Epitopes/metabolism , Histocompatibility Antigens Class I/immunology , Histocompatibility Antigens Class I/metabolism , Humans , Intracellular Space/metabolism , Phagocytosis/immunology , Proteolysis , Receptors, Cell Surface/metabolism
8.
Annu Rev Immunol ; 35: 469-499, 2017 04 26.
Article in English | MEDLINE | ID: mdl-28226228

ABSTRACT

Professional antigen-presenting cells (APCs) in the skin include dendritic cells, monocytes, and macrophages. They are highly dynamic, with the capacity to enter skin from the peripheral circulation, patrol within tissue, and migrate through lymphatics to draining lymph nodes. Skin APCs are endowed with antigen-sensing, -processing, and -presenting machinery and play key roles in initiating, modulating, and resolving cutaneous inflammation. Skin APCs are a highly heterogeneous population with functionally specialized subsets that are developmentally imprinted and modulated by local tissue microenvironmental and inflammatory cues. This review explores recent advances that have allowed for a more accurate taxonomy of APC subsets found in both mouse and human skin. It also examines the functional specificity of individual APC subsets and their collaboration with other immune cell types that together promote adaptive T cell and regional cutaneous immune responses during homeostasis, inflammation, and disease.


Subject(s)
Antigen-Presenting Cells/immunology , Dendritic Cells/immunology , Langerhans Cells/immunology , Macrophages/immunology , Monocytes/immunology , Skin/immunology , T-Lymphocytes/immunology , Animals , Antigen Presentation , Cell Movement , Homeostasis , Humans , Inflammation , Lymphocyte Activation , Mice
9.
Cell ; 187(2): 375-389.e18, 2024 01 18.
Article in English | MEDLINE | ID: mdl-38242085

ABSTRACT

Immune checkpoint inhibition treatment using aPD-1 monoclonal antibodies is a promising cancer immunotherapy approach. However, its effect on tumor immunity is narrow, as most patients do not respond to the treatment or suffer from recurrence. We show that the crosstalk between conventional type I dendritic cells (cDC1) and T cells is essential for an effective aPD-1-mediated anti-tumor response. Accordingly, we developed a bispecific DC-T cell engager (BiCE), a reagent that facilitates physical interactions between PD-1+ T cells and cDC1. BiCE treatment promotes the formation of active dendritic/T cell crosstalk in the tumor and tumor-draining lymph nodes. In vivo, single-cell and physical interacting cell analysis demonstrates the distinct and superior immune reprogramming of the tumors and tumor-draining lymph nodes treated with BiCE as compared to conventional aPD-1 treatment. By bridging immune cells, BiCE potentiates cell circuits and communication pathways needed for effective anti-tumor immunity.


Subject(s)
Antibodies, Bispecific , Neoplasms , Humans , Antibodies, Bispecific/therapeutic use , Dendritic Cells/immunology , Immunotherapy , Neoplasms/immunology , Neoplasms/therapy , T-Lymphocytes/immunology
10.
Cell ; 187(11): 2817-2837.e31, 2024 May 23.
Article in English | MEDLINE | ID: mdl-38701783

ABSTRACT

FMS-related tyrosine kinase 3 ligand (FLT3L), encoded by FLT3LG, is a hematopoietic factor essential for the development of natural killer (NK) cells, B cells, and dendritic cells (DCs) in mice. We describe three humans homozygous for a loss-of-function FLT3LG variant with a history of various recurrent infections, including severe cutaneous warts. The patients' bone marrow (BM) was hypoplastic, with low levels of hematopoietic progenitors, particularly myeloid and B cell precursors. Counts of B cells, monocytes, and DCs were low in the patients' blood, whereas the other blood subsets, including NK cells, were affected only moderately, if at all. The patients had normal counts of Langerhans cells (LCs) and dermal macrophages in the skin but lacked dermal DCs. Thus, FLT3L is required for B cell and DC development in mice and humans. However, unlike its murine counterpart, human FLT3L is required for the development of monocytes but not NK cells.


Subject(s)
Killer Cells, Natural , Membrane Proteins , Animals , Female , Humans , Male , Mice , B-Lymphocytes/metabolism , B-Lymphocytes/cytology , Bone Marrow/metabolism , Cell Lineage , Dendritic Cells/metabolism , Hematopoiesis , Hematopoietic Stem Cells/metabolism , Hematopoietic Stem Cells/cytology , Killer Cells, Natural/metabolism , Killer Cells, Natural/immunology , Langerhans Cells/metabolism , Membrane Proteins/metabolism , Membrane Proteins/genetics , Monocytes/metabolism , Skin/metabolism , Mice, Inbred C57BL
11.
Cell ; 2024 Jun 12.
Article in English | MEDLINE | ID: mdl-38870946

ABSTRACT

Defective host defenses later in life are associated with changes in immune cell activities, suggesting that age-specific considerations are needed in immunotherapy approaches. In this study, we found that PD-1 and CTLA4-based cancer immunotherapies are unable to eradicate tumors in elderly mice. This defect in anti-tumor activity correlated with two known age-associated immune defects: diminished abundance of systemic naive CD8+ T cells and weak migratory activities of dendritic cells (DCs). We identified a vaccine adjuvant, referred to as a DC hyperactivator, which corrects DC migratory defects in the elderly. Vaccines containing tumor antigens and DC hyperactivators induced T helper type 1 (TH1) CD4+ T cells with cytolytic activity that drive anti-tumor immunity in elderly mice. When administered early in life, DC hyperactivators were the only adjuvant identified that elicited anti-tumor CD4+ T cells that persisted into old age. These results raise the possibility of correcting age-associated immune defects through DC manipulation.

12.
Cell ; 186(21): 4583-4596.e13, 2023 10 12.
Article in English | MEDLINE | ID: mdl-37725977

ABSTRACT

The CD1 system binds lipid antigens for display to T cells. Here, we solved lipidomes for the four human CD1 antigen-presenting molecules, providing a map of self-lipid display. Answering a basic question, the detection of >2,000 CD1-lipid complexes demonstrates broad presentation of self-sphingolipids and phospholipids. Whereas peptide antigens are chemically processed, many lipids are presented in an unaltered form. However, each type of CD1 protein differentially edits the self-lipidome to show distinct capture motifs based on lipid length and chemical composition, suggesting general antigen display mechanisms. For CD1a and CD1d, lipid size matches the CD1 cleft volume. CD1c cleft size is more variable, and CD1b is the outlier, where ligands and clefts show an extreme size mismatch that is explained by uniformly seating two small lipids in one cleft. Furthermore, the list of compounds that comprise the integrated CD1 lipidome supports the ongoing discovery of lipid blockers and antigens for T cells.


Subject(s)
Antigens, CD1 , Lipids , Humans , Antigen Presentation , Antigens, CD1/chemistry , Antigens, CD1/metabolism , Lipidomics , Lipids/chemistry , T-Lymphocytes , Amino Acid Motifs
13.
Cell ; 186(25): 5536-5553.e22, 2023 12 07.
Article in English | MEDLINE | ID: mdl-38029747

ABSTRACT

Mycobacterium tuberculosis (Mtb) causes 1.6 million deaths annually. Active tuberculosis correlates with a neutrophil-driven type I interferon (IFN) signature, but the cellular mechanisms underlying tuberculosis pathogenesis remain poorly understood. We found that interstitial macrophages (IMs) and plasmacytoid dendritic cells (pDCs) are dominant producers of type I IFN during Mtb infection in mice and non-human primates, and pDCs localize near human Mtb granulomas. Depletion of pDCs reduces Mtb burdens, implicating pDCs in tuberculosis pathogenesis. During IFN-driven disease, we observe abundant DNA-containing neutrophil extracellular traps (NETs) described to activate pDCs. Cell-type-specific disruption of the type I IFN receptor suggests that IFNs act on IMs to inhibit Mtb control. Single-cell RNA sequencing (scRNA-seq) indicates that type I IFN-responsive cells are defective in their response to IFNγ, a cytokine critical for Mtb control. We propose that pDC-derived type I IFNs act on IMs to permit bacterial replication, driving further neutrophil recruitment and active tuberculosis disease.


Subject(s)
Interferon Type I , Tuberculosis , Humans , Mice , Animals , Macrophages/microbiology , Cytokines , Neutrophils , Dendritic Cells
14.
Annu Rev Immunol ; 33: 227-56, 2015.
Article in English | MEDLINE | ID: mdl-25581310

ABSTRACT

The diverse microbial populations constituting the intestinal microbiota promote immune development and differentiation, but because of their complex metabolic requirements and the consequent difficulty culturing them, they remained, until recently, largely uncharacterized and mysterious. In the last decade, deep nucleic acid sequencing platforms, new computational and bioinformatics tools, and full-genome characterization of several hundred commensal bacterial species facilitated studies of the microbiota and revealed that differences in microbiota composition can be associated with inflammatory, metabolic, and infectious diseases, that each human is colonized by a distinct bacterial flora, and that the microbiota can be manipulated to reduce and even cure some diseases. Different bacterial species induce distinct immune cell populations that can play pro- and anti-inflammatory roles, and thus the composition of the microbiota determines, in part, the level of resistance to infection and susceptibility to inflammatory diseases. This review summarizes recent work characterizing commensal microbes that contribute to the antimicrobial defense/inflammation axis.


Subject(s)
Disease Resistance/immunology , Gastroenteritis/immunology , Gastroenteritis/microbiology , Gastrointestinal Microbiome/immunology , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Adaptive Immunity , Animals , Autoimmune Diseases/immunology , Autoimmune Diseases/metabolism , Autoimmune Diseases/microbiology , Computational Biology , Diet , Disease Susceptibility , Gastroenteritis/metabolism , Host-Pathogen Interactions/immunology , Humans , Immunity, Innate , Immunity, Mucosal , Intestinal Mucosa/metabolism , Metabolome , Neoplasms/etiology , Vitamins/metabolism
15.
Annu Rev Immunol ; 33: 563-606, 2015.
Article in English | MEDLINE | ID: mdl-25665078

ABSTRACT

In the 40 years since their discovery, dendritic cells (DCs) have been recognized as central players in immune regulation. DCs sense microbial stimuli through pathogen-recognition receptors (PRRs) and decode, integrate, and present information derived from such stimuli to T cells, thus stimulating immune responses. DCs can also regulate the quality of immune responses. Several functionally specialized subsets of DCs exist, but DCs also display functional plasticity in response to diverse stimuli. In addition to sensing pathogens via PRRs, emerging evidence suggests that DCs can also sense stress signals, such as amino acid starvation, through ancient stress and nutrient sensing pathways, to stimulate adaptive immunity. Here, I discuss these exciting advances in the context of a historic perspective on the discovery of DCs and their role in immune regulation. I conclude with a discussion of emerging areas in DC biology in the systems immunology era and suggest that the impact of DCs on immunity can be usefully contextualized in a hierarchy-of-organization model in which DCs, their receptors and signaling networks, cell-cell interactions, tissue microenvironment, and the host macroenvironment represent different levels of the hierarchy. Immunity or tolerance can then be represented as a complex function of each of these hierarchies.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Animals , Cell Communication/immunology , Cell Differentiation/immunology , Clonal Selection, Antigen-Mediated , Disease Resistance , Disease Susceptibility , Host-Pathogen Interactions/immunology , Humans , Lymphocytes/cytology , Lymphocytes/immunology , Lymphocytes/metabolism , Stress, Physiological
16.
Cell ; 185(10): 1694-1708.e19, 2022 05 12.
Article in English | MEDLINE | ID: mdl-35447074

ABSTRACT

Immunotherapy is a promising treatment for triple-negative breast cancer (TNBC), but patients relapse, highlighting the need to understand the mechanisms of resistance. We discovered that in primary breast cancer, tumor cells that resist T cell attack are quiescent. Quiescent cancer cells (QCCs) form clusters with reduced immune infiltration. They also display superior tumorigenic capacity and higher expression of chemotherapy resistance and stemness genes. We adapted single-cell RNA-sequencing with precise spatial resolution to profile infiltrating cells inside and outside the QCC niche. This transcriptomic analysis revealed hypoxia-induced programs and identified more exhausted T cells, tumor-protective fibroblasts, and dysfunctional dendritic cells inside clusters of QCCs. This uncovered differential phenotypes in infiltrating cells based on their intra-tumor location. Thus, QCCs constitute immunotherapy-resistant reservoirs by orchestrating a local hypoxic immune-suppressive milieu that blocks T cell function. Eliminating QCCs holds the promise to counteract immunotherapy resistance and prevent disease recurrence in TNBC.


Subject(s)
Triple Negative Breast Neoplasms , Humans , Immunosuppressive Agents/therapeutic use , Immunotherapy , Neoplasm Recurrence, Local , T-Lymphocytes/pathology , Triple Negative Breast Neoplasms/pathology , Tumor Microenvironment
17.
Cell ; 184(3): 792-809.e23, 2021 02 04.
Article in English | MEDLINE | ID: mdl-33545035

ABSTRACT

Tumor-infiltrating myeloid cells (TIMs) are key regulators in tumor progression, but the similarity and distinction of their fundamental properties across different tumors remain elusive. Here, by performing a pan-cancer analysis of single myeloid cells from 210 patients across 15 human cancer types, we identified distinct features of TIMs across cancer types. Mast cells in nasopharyngeal cancer were found to be associated with better prognosis and exhibited an anti-tumor phenotype with a high ratio of TNF+/VEGFA+ cells. Systematic comparison between cDC1- and cDC2-derived LAMP3+ cDCs revealed their differences in transcription factors and external stimulus. Additionally, pro-angiogenic tumor-associated macrophages (TAMs) were characterized with diverse markers across different cancer types, and the composition of TIMs appeared to be associated with certain features of somatic mutations and gene expressions. Our results provide a systematic view of the highly heterogeneous TIMs and suggest future avenues for rational, targeted immunotherapies.


Subject(s)
Myeloid Cells/pathology , Neoplasms/genetics , Neoplasms/pathology , Single-Cell Analysis , Transcription, Genetic , Cell Line, Tumor , Cell Lineage , Dendritic Cells/metabolism , Female , Humans , Lysosomal Membrane Proteins/metabolism , Macrophages/metabolism , Male , Mast Cells/pathology , Monocytes/metabolism , Neoplasm Proteins/metabolism , Transcriptome/genetics
18.
Cell ; 184(15): 4016-4031.e22, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34081922

ABSTRACT

Cross-presentation of antigens from dead tumor cells by type 1 conventional dendritic cells (cDC1s) is thought to underlie priming of anti-cancer CD8+ T cells. cDC1 express high levels of DNGR-1 (a.k.a. CLEC9A), a receptor that binds to F-actin exposed by dead cell debris and promotes cross-presentation of associated antigens. Here, we show that secreted gelsolin (sGSN), an extracellular protein, decreases DNGR-1 binding to F-actin and cross-presentation of dead cell-associated antigens by cDC1s. Mice deficient in sGsn display increased DNGR-1-dependent resistance to transplantable tumors, especially ones expressing neoantigens associated with the actin cytoskeleton, and exhibit greater responsiveness to cancer immunotherapy. In human cancers, lower levels of intratumoral sGSN transcripts, as well as presence of mutations in proteins associated with the actin cytoskeleton, are associated with signatures of anti-cancer immunity and increased patient survival. Our results reveal a natural barrier to cross-presentation of cancer antigens that dampens anti-tumor CD8+ T cell responses.


Subject(s)
Cross-Priming/immunology , Gelsolin/metabolism , Immunity , Lectins, C-Type/metabolism , Neoplasms/immunology , Receptors, Immunologic/metabolism , Receptors, Mitogen/metabolism , Actins/metabolism , Amino Acid Sequence , Animals , Antigens, Neoplasm/metabolism , CD8-Positive T-Lymphocytes/drug effects , CD8-Positive T-Lymphocytes/immunology , Cell Movement/drug effects , Cell Proliferation/drug effects , Cross-Priming/drug effects , Cytoskeleton/drug effects , Cytoskeleton/metabolism , Dendritic Cells/drug effects , Dendritic Cells/immunology , Gelsolin/chemistry , Gelsolin/deficiency , Gene Expression Regulation, Neoplastic/drug effects , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunity/drug effects , Mice, Inbred C57BL , Mutation/genetics , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/pathology , Protein Binding/drug effects , Survival Analysis
19.
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
20.
Cell ; 184(17): 4512-4530.e22, 2021 08 19.
Article in English | MEDLINE | ID: mdl-34343496

ABSTRACT

Cytotoxic T lymphocyte (CTL) responses against tumors are maintained by stem-like memory cells that self-renew but also give rise to effector-like cells. The latter gradually lose their anti-tumor activity and acquire an epigenetically fixed, hypofunctional state, leading to tumor tolerance. Here, we show that the conversion of stem-like into effector-like CTLs involves a major chemotactic reprogramming that includes the upregulation of chemokine receptor CXCR6. This receptor positions effector-like CTLs in a discrete perivascular niche of the tumor stroma that is densely occupied by CCR7+ dendritic cells (DCs) expressing the CXCR6 ligand CXCL16. CCR7+ DCs also express and trans-present the survival cytokine interleukin-15 (IL-15). CXCR6 expression and IL-15 trans-presentation are critical for the survival and local expansion of effector-like CTLs in the tumor microenvironment to maximize their anti-tumor activity before progressing to irreversible dysfunction. These observations reveal a cellular and molecular checkpoint that determines the magnitude and outcome of anti-tumor immune responses.


Subject(s)
Receptors, CXCR6/metabolism , T-Lymphocytes, Cytotoxic/immunology , Tumor Microenvironment , Animals , B7-H1 Antigen/metabolism , Cell Communication , Cell Movement , Cell Proliferation , Cell Survival , Chemokine CXCL16 , Dendritic Cells/metabolism , Interleukin-12/metabolism , Interleukin-15/metabolism , Ligands , Lymph Nodes/metabolism , Melanoma/immunology , Melanoma/pathology , Mice, Inbred C57BL
SELECTION OF CITATIONS
SEARCH DETAIL