Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 10.176
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 38: 421-453, 2020 04 26.
Article in English | MEDLINE | ID: mdl-31990619

ABSTRACT

Foxp3-expressing CD4+ regulatory T (Treg) cells play key roles in the prevention of autoimmunity and the maintenance of immune homeostasis and represent a major barrier to the induction of robust antitumor immune responses. Thus, a clear understanding of the mechanisms coordinating Treg cell differentiation is crucial for understanding numerous facets of health and disease and for developing approaches to modulate Treg cells for clinical benefit. Here, we discuss current knowledge of the signals that coordinate Treg cell development, the antigen-presenting cell types that direct Treg cell selection, and the nature of endogenous Treg cell ligands, focusing on evidence from studies in mice. We also highlight recent advances in this area and identify key unanswered questions.


Subject(s)
Cell Differentiation/immunology , Lymphopoiesis/immunology , T-Lymphocytes, Regulatory/immunology , T-Lymphocytes, Regulatory/metabolism , Animals , Antigen Presentation/immunology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Biomarkers , Cell Differentiation/genetics , Clonal Deletion , Clonal Selection, Antigen-Mediated , Humans , Lymphocyte Activation/genetics , Lymphocyte Activation/immunology , Lymphopoiesis/genetics , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes, Regulatory/cytology , Thymus Gland/cytology , Thymus Gland/immunology , Thymus Gland/metabolism
2.
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
3.
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
4.
Annu Rev Immunol ; 34: 151-72, 2016 05 20.
Article in English | MEDLINE | ID: mdl-26772212

ABSTRACT

Butyrophilin molecules (commonly contracted to BTN), collectively take their name from the eponymous protein in cow's milk. They are considered to be members of the B7 family of costimulatory receptors, which includes B7.1 (CD80), B7.2 (CD86), and related molecules, such as PD-L1 (B7-H1, CD274), ICOS-L (CD275), and B7-H3 (CD276). These coreceptors modulate T cell responses upon antigen presentation by major histocompatibility complex and cognate αß T cell receptor engagement. Molecules such as BTN3A1 (CD277), myelin oligodendrocyte glycoprotein, and mouse Skint1 and Btnl2, all members of the butyrophilin family, show greater structural and functional diversity than the canonical B7 receptors. Some butyrophilins mediate complex interactions between antigen-presenting cells and conventional αß T cells, and others regulate the immune responses of specific γδ T cell subsets by mechanisms that have characteristics of both innate and adaptive immunity.


Subject(s)
Adaptive Immunity , Antigen-Presenting Cells/immunology , B7 Antigens/metabolism , Butyrophilins/metabolism , Immunity, Innate , Milk/metabolism , T-Lymphocytes/immunology , Animals , Butyrophilins/immunology , Cattle , Humans , Lymphocyte Activation , Mice , Receptors, Antigen, T-Cell/metabolism , Signal Transduction
5.
Annu Rev Immunol ; 33: 445-74, 2015.
Article in English | MEDLINE | ID: mdl-25622193

ABSTRACT

The observation that a subset of cancer patients show evidence for spontaneous CD8+ T cell priming against tumor-associated antigens has generated renewed interest in the innate immune pathways that might serve as a bridge to an adaptive immune response to tumors. Manipulation of this endogenous T cell response with therapeutic intent-for example, using blocking antibodies inhibiting PD-1/PD-L1 (programmed death-1/programmed death ligand 1) interactions-is showing impressive clinical results. As such, understanding the innate immune mechanisms that enable this T cell response has important clinical relevance. Defined innate immune interactions in the cancer context include recognition by innate cell populations (NK cells, NKT cells, and γδ T cells) and also by dendritic cells and macrophages in response to damage-associated molecular patterns (DAMPs). Recent evidence has indicated that the major DAMP driving host antitumor immune responses is tumor-derived DNA, sensed by the stimulator of interferon gene (STING) pathway and driving type I IFN production. A deeper knowledge of the clinically relevant innate immune pathways involved in the recognition of tumors is leading toward new therapeutic strategies for cancer treatment.


Subject(s)
Immunity, Innate , Neoplasms/immunology , Neoplasms/metabolism , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Complement System Proteins/immunology , Complement System Proteins/metabolism , Cytotoxicity, Immunologic , Dendritic Cells/immunology , Dendritic Cells/metabolism , Humans , Immune System/cytology , Immunotherapy , Ligands , Macrophage Activation , Macrophages/immunology , Macrophages/metabolism , Microbiota , Neoplasms/microbiology , Neoplasms/therapy , Signal Transduction
6.
Annu Rev Immunol ; 32: 579-608, 2014.
Article in English | MEDLINE | ID: mdl-24499272

ABSTRACT

This paper reviews the presentation of peptides by major histocompatibility complex (MHC) class II molecules in the autoimmune diabetes of the nonobese diabetic (NOD) mouse. Islets of Langerhans contain antigen-presenting cells that capture the proteins and peptides of the beta cells' secretory granules. Peptides bound to I-A(g7), the unique MHC class II molecule of NOD mice, are presented in islets and in pancreatic lymph nodes. The various beta cell-derived peptides interact with selected CD4 T cells to cause inflammation and beta cell demise. Many autoreactive T cells are found in NOD mice, but not all have a major role in the initiation of the autoimmune process. I emphasize here the evidence pointing to insulin autoreactivity as a seminal component in the diabetogenic process.


Subject(s)
Antigen Presentation/immunology , Diabetes Mellitus, Type 1/immunology , Animals , Antigen-Presenting Cells/immunology , Autoantigens/immunology , Disease Models, Animal , Epitopes, T-Lymphocyte , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/immunology , Humans , Insulin/immunology , Islets of Langerhans/cytology , Islets of Langerhans/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred NOD , Peptides/immunology , T-Cell Antigen Receptor Specificity/immunology , T-Lymphocyte Subsets/immunology
7.
Cell ; 184(15): 3998-4015.e19, 2021 07 22.
Article in English | MEDLINE | ID: mdl-34157302

ABSTRACT

Foxp3+ T regulatory (Treg) cells promote immunological tumor tolerance, but how their immune-suppressive function is regulated in the tumor microenvironment (TME) remains unknown. Here, we used intravital microscopy to characterize the cellular interactions that provide tumor-infiltrating Treg cells with critical activation signals. We found that the polyclonal Treg cell repertoire is pre-enriched to recognize antigens presented by tumor-associated conventional dendritic cells (cDCs). Unstable cDC contacts sufficed to sustain Treg cell function, whereas T helper cells were activated during stable interactions. Contact instability resulted from CTLA-4-dependent downregulation of co-stimulatory B7-family proteins on cDCs, mediated by Treg cells themselves. CTLA-4-blockade triggered CD28-dependent Treg cell hyper-proliferation in the TME, and concomitant Treg cell inactivation was required to achieve tumor rejection. Therefore, Treg cells self-regulate through a CTLA-4- and CD28-dependent feedback loop that adjusts their population size to the amount of local co-stimulation. Its disruption through CTLA-4-blockade may off-set therapeutic benefits in cancer patients.


Subject(s)
CTLA-4 Antigen/metabolism , Feedback, Physiological , Neoplasms/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Antigen-Presenting Cells/immunology , CD28 Antigens/metabolism , Cell Proliferation , Dendritic Cells/immunology , Green Fluorescent Proteins/metabolism , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Inhibitors/therapeutic use , Immunotherapy , Interleukin-2/metabolism , Ligands , Lymph Nodes/metabolism , Lymphocyte Activation/immunology , Mice, Inbred BALB C , Mice, Inbred C57BL , NFATC Transcription Factors/metabolism , Neoplasms/pathology , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes, Helper-Inducer/immunology , Tumor Microenvironment
8.
Cell ; 184(6): 1401, 2021 03 18.
Article in English | MEDLINE | ID: mdl-33740443

ABSTRACT

The first two vaccines proven to be effective for inhibiting COVID-19 illness were both mRNA, achieving 95% efficacy (and safety) among 74,000 participants (half receiving placebo) after intramuscular delivery of two shots, 3-4 weeks apart. To view this Bench to Bedside, open or download the PDF.


Subject(s)
COVID-19 Vaccines/immunology , COVID-19 Vaccines/therapeutic use , COVID-19/immunology , COVID-19/prevention & control , SARS-CoV-2/immunology , Vaccines, Synthetic/immunology , Vaccines, Synthetic/therapeutic use , Antigen-Presenting Cells/immunology , B-Lymphocytes/immunology , COVID-19/virology , Drug Delivery Systems/methods , Humans , Liposomes , Nanoparticles , Spike Glycoprotein, Coronavirus/immunology , T-Lymphocytes/immunology , Treatment Outcome , mRNA Vaccines
9.
Cell ; 183(4): 1142-1142.e1, 2020 11 12.
Article in English | MEDLINE | ID: mdl-33186524

ABSTRACT

Immune checkpoints are key regulatory mechanisms integral to the maintenance of self-tolerance and execution of antigen-specific immune responses. In recent years, they have been leveraged to treat both autoimmune disease and various forms of cancer with much success. This SnapShot illustrates known immune checkpoint interactions in APC-mediated T cell modulation. To view this SnapShot, open or download the PDF.


Subject(s)
Antigen-Presenting Cells/immunology , Immune Checkpoint Proteins/metabolism , T-Lymphocytes/immunology , Antigen-Presenting Cells/drug effects , Humans , Immune Checkpoint Inhibitors/pharmacology , Immune Checkpoint Proteins/chemistry , Protein Domains , Protein Interaction Maps , T-Lymphocytes/drug effects
10.
Annu Rev Immunol ; 30: 733-58, 2012.
Article in English | MEDLINE | ID: mdl-22224762

ABSTRACT

Regulatory T lymphocytes are essential to maintain homeostasis of the immune system, limiting the magnitude of effector responses and allowing the establishment of immunological tolerance. Two main types of regulatory T cells have been identified--natural and induced (or adaptive)-and both play significant roles in tuning down effector immune responses. Adaptive CD4(+)Foxp3(+) regulatory T (iTreg) cells develop outside the thymus under a variety of conditions. These include not only antigen presentation under subimmunogenic or noninflammatory conditions, but also chronic inflammation and infections. We speculate that the different origin of iTreg cells (noninflammatory versus inflammatory) results in distinct properties, including their stability. iTreg cells are also generated during homeostasis of the gut and in cancer, although some cancers also favor expansion of natural regulatory T (nTreg) cells. Here we review how iTreg cells develop and how they participate in immunological tolerance, contrasting, when possible, iTreg cells with nTreg cells.


Subject(s)
Immune Tolerance/immunology , T-Lymphocytes, Regulatory/immunology , Adoptive Transfer , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , CD4 Antigens/metabolism , Forkhead Transcription Factors/metabolism , Humans , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , T-Lymphocytes, Regulatory/metabolism
11.
Annu Rev Immunol ; 30: 39-68, 2012.
Article in English | MEDLINE | ID: mdl-22136167

ABSTRACT

mTOR is an evolutionarily conserved serine/threonine kinase that plays a central role in integrating environmental cues in the form of growth factors, amino acids, and energy. In the study of the immune system, mTOR is emerging as a critical regulator of immune function because of its role in sensing and integrating cues from the immune microenvironment. With the greater appreciation of cellular metabolism as an important regulator of immune cell function, mTOR is proving to be a vital link between immune function and metabolism. In this review, we discuss the ability of mTOR to direct the adaptive immune response. Specifically, we focus on the role of mTOR in promoting differentiation, activation, and function in T cells, B cells, and antigen-presenting cells.


Subject(s)
Immunity , TOR Serine-Threonine Kinases/metabolism , Animals , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Cell Differentiation/immunology , Enzyme Activation , Humans , Immunosuppressive Agents/pharmacology , Lymphocyte Activation/immunology , Protein Kinase Inhibitors/pharmacology , Signal Transduction , T-Lymphocytes/cytology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , T-Lymphocytes, Regulatory/immunology , TOR Serine-Threonine Kinases/antagonists & inhibitors
12.
Cell ; 175(4): 1059-1073.e21, 2018 11 01.
Article in English | MEDLINE | ID: mdl-30270039

ABSTRACT

Motivated by the clinical observation that interruption of the mevalonate pathway stimulates immune responses, we hypothesized that this pathway may function as a druggable target for vaccine adjuvant discovery. We found that lipophilic statin drugs and rationally designed bisphosphonates that target three distinct enzymes in the mevalonate pathway have potent adjuvant activities in mice and cynomolgus monkeys. These inhibitors function independently of conventional "danger sensing." Instead, they inhibit the geranylgeranylation of small GTPases, including Rab5 in antigen-presenting cells, resulting in arrested endosomal maturation, prolonged antigen retention, enhanced antigen presentation, and T cell activation. Additionally, inhibiting the mevalonate pathway enhances antigen-specific anti-tumor immunity, inducing both Th1 and cytolytic T cell responses. As demonstrated in multiple mouse cancer models, the mevalonate pathway inhibitors are robust for cancer vaccinations and synergize with anti-PD-1 antibodies. Our research thus defines the mevalonate pathway as a druggable target for vaccine adjuvants and cancer immunotherapies.


Subject(s)
Adjuvants, Immunologic/pharmacology , Cancer Vaccines/immunology , Diphosphonates/pharmacology , Hydroxymethylglutaryl-CoA Reductase Inhibitors/pharmacology , Mevalonic Acid/metabolism , rab5 GTP-Binding Proteins/antagonists & inhibitors , Animals , Antigen Presentation , Antigen-Presenting Cells/drug effects , Antigen-Presenting Cells/immunology , Cell Line, Tumor , Endosomes/drug effects , Female , Macaca fascicularis , Male , Mice , Mice, Inbred C57BL , Protein Prenylation , rab5 GTP-Binding Proteins/metabolism
13.
Nat Immunol ; 20(12): 1681-1691, 2019 12.
Article in English | MEDLINE | ID: mdl-31636462

ABSTRACT

Much attention has focused on commensal bacteria in health and disease, but the role of commensal viruses is understudied. Although metagenomic analysis shows that the intestine of healthy humans and animals harbors various commensal viruses and the dysbiosis of these viruses can be associated with inflammatory diseases, there is still a lack of causal data and underlying mechanisms to understand the physiological role of commensal viruses in intestinal homeostasis. In the present study, we show that commensal viruses are essential for the homeostasis of intestinal intraepithelial lymphocytes (IELs). Mechanistically, the cytosolic viral RNA-sensing receptor RIG-I in antigen-presenting cells can recognize commensal viruses and maintain IELs via a type I interferon-independent, but MAVS-IRF1-IL-15 axis-dependent, manner. The recovery of IELs by interleukin-15 administration reverses the susceptibility of commensal virus-depleted mice to dextran sulfate sodium-induced colitis. Collectively, our results indicate that commensal viruses maintain the IELs and consequently sustain intestinal homeostasis via noncanonical RIG-I signaling.


Subject(s)
Antigen-Presenting Cells/immunology , Caliciviridae Infections/immunology , Colitis/immunology , DEAD Box Protein 58/metabolism , Intestines/immunology , Intraepithelial Lymphocytes/immunology , Norovirus/physiology , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Caliciviridae Infections/virology , Cells, Cultured , Colitis/chemically induced , Colitis/virology , DEAD Box Protein 58/genetics , Dextran Sulfate , Disease Susceptibility , Homeostasis , Interferon Regulatory Factor-1/genetics , Interferon Regulatory Factor-1/metabolism , Interleukin-15/metabolism , Intestines/virology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Signal Transduction , Symbiosis/immunology
14.
Nat Immunol ; 20(3): 301-312, 2019 03.
Article in English | MEDLINE | ID: mdl-30664737

ABSTRACT

The fetus is thought to be protected from exposure to foreign antigens, yet CD45RO+ T cells reside in the fetal intestine. Here we combined functional assays with mass cytometry, single-cell RNA sequencing and high-throughput T cell antigen receptor (TCR) sequencing to characterize the CD4+ T cell compartment in the human fetal intestine. We identified 22 CD4+ T cell clusters, including naive-like, regulatory-like and memory-like subpopulations, which were confirmed and further characterized at the transcriptional level. Memory-like CD4+ T cells had high expression of Ki-67, indicative of cell division, and CD5, a surrogate marker of TCR avidity, and produced the cytokines IFN-γ and IL-2. Pathway analysis revealed a differentiation trajectory associated with cellular activation and proinflammatory effector functions, and TCR repertoire analysis indicated clonal expansions, distinct repertoire characteristics and interconnections between subpopulations of memory-like CD4+ T cells. Imaging mass cytometry indicated that memory-like CD4+ T cells colocalized with antigen-presenting cells. Collectively, these results provide evidence for the generation of memory-like CD4+ T cells in the human fetal intestine that is consistent with exposure to foreign antigens.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Fetus/immunology , Immunologic Memory/immunology , Intestines/immunology , Antigen-Presenting Cells/cytology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/metabolism , CD5 Antigens/genetics , CD5 Antigens/immunology , CD5 Antigens/metabolism , Cells, Cultured , Fetus/cytology , Fetus/metabolism , Flow Cytometry , Gene Expression Profiling/methods , Gene Expression Regulation, Developmental/immunology , High-Throughput Nucleotide Sequencing , Humans , Immunologic Memory/genetics , Immunophenotyping , Intestines/cytology , Intestines/embryology , Ki-67 Antigen/genetics , Ki-67 Antigen/immunology , Ki-67 Antigen/metabolism
15.
Immunity ; 54(4): 721-736.e10, 2021 04 13.
Article in English | MEDLINE | ID: mdl-33725478

ABSTRACT

Hyperglycemia and hyperlipidemia are often observed in individuals with type II diabetes (T2D) and related mouse models. One dysmetabolic biochemical consequence is the non-enzymatic reaction between sugars, lipids, and proteins, favoring protein glycation, glycoxidation, and lipoxidation. Here, we identified oxidative alterations in key components of the major histocompatibility complex (MHC) class II molecule antigen processing and presentation machinery in vivo under conditions of hyperglycemia-induced metabolic stress. These modifications were linked to epitope-specific changes in endosomal processing efficiency, MHC class II-peptide binding, and DM editing activity. Moreover, we observed some quantitative and qualitative changes in the MHC class II immunopeptidome of Ob/Ob mice on a high-fat diet compared with controls, including changes in the presentation of an apolipoprotein B100 peptide associated previously with T2D and metabolic syndrome-related clinical complications. These findings highlight a link between glycation reactions and altered MHC class II antigen presentation that may contribute to T2D complications.


Subject(s)
Antigen Presentation/immunology , Histocompatibility Antigens Class II/immunology , Stress, Physiological/immunology , Animals , Antigen-Presenting Cells/immunology , Diabetes Mellitus, Experimental/immunology , Diabetes Mellitus, Type 2/immunology , Disease Models, Animal , Epitopes/immunology , Female , Male , Mice , Mice, Inbred C57BL , Peptides/immunology , Protein Binding/immunology
16.
Nat Immunol ; 18(8): 851-860, 2017 Jul 19.
Article in English | MEDLINE | ID: mdl-28722709

ABSTRACT

The study of the intestinal microbiota has begun to shift from cataloging individual members of the commensal community to understanding their contributions to the physiology of the host organism in health and disease. Here, we review the effects of the microbiome on innate and adaptive immunological players from epithelial cells and antigen-presenting cells to innate lymphoid cells and regulatory T cells. We discuss recent studies that have identified diverse microbiota-derived bioactive molecules and their effects on inflammation within the intestine and distally at sites as anatomically remote as the brain. Finally, we highlight new insights into how the microbiome influences the host response to infection, vaccination and cancer, as well as susceptibility to autoimmune and neurodegenerative disorders.


Subject(s)
Gastrointestinal Microbiome/immunology , Infections/immunology , Inflammation/immunology , Neoplasms/immunology , Adaptive Immunity/immunology , Antigen-Presenting Cells/immunology , Autoimmune Diseases/immunology , Humans , Immunity, Innate/immunology , Immunity, Mucosal/immunology , Lymphocytes/immunology , Neurodegenerative Diseases/immunology , Symbiosis , T-Lymphocytes, Regulatory/immunology , Vaccination
17.
Nat Immunol ; 18(10): 1068-1075, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-28926543

ABSTRACT

Langerhans cells (LCs) are epidermis-resident antigen-presenting cells that share a common ontogeny with macrophages but function as dendritic cells (DCs). Their development, recruitment and retention in the epidermis is orchestrated by interactions with keratinocytes through multiple mechanisms. LC and dermal DC subsets often show functional redundancy, but LCs are required for specific types of adaptive immune responses when antigen is concentrated in the epidermis. This Review will focus on those developmental and functional properties that are unique to LCs.


Subject(s)
Epidermis/immunology , Epidermis/metabolism , Langerhans Cells/immunology , Langerhans Cells/metabolism , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Biomarkers , Cell Communication , Cell Differentiation , Cross-Priming , Dendritic Cells/immunology , Dendritic Cells/metabolism , Disease Susceptibility , Keratinocytes/metabolism , Lymphocyte Activation , Mice , Phagocytes/immunology , Phagocytes/metabolism , Phenotype , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism
18.
Cell ; 158(3): 492-505, 2014 Jul 31.
Article in English | MEDLINE | ID: mdl-25083865

ABSTRACT

To mount an immune response, T lymphocytes must successfully search for foreign material bound to the surface of antigen-presenting cells. How T cells optimize their chances of encountering and responding to these antigens is unknown. T cell motility in tissues resembles a random or Levy walk and is regulated in part by external factors including chemokines and lymph-node topology, but motility parameters such as speed and propensity to turn may also be cell intrinsic. Here we found that the unconventional myosin 1g (Myo1g) motor generates membrane tension, enforces cell-intrinsic meandering search, and enhances T-DC interactions during lymph-node surveillance. Increased turning and meandering motility, as opposed to ballistic motility, is enhanced by Myo1g. Myo1g acts as a "turning motor" and generates a form of cellular "flânerie." Modeling and antigen challenges show that these intrinsically programmed elements of motility search are critical for the detection of rare cognate antigen-presenting cells.


Subject(s)
Immunologic Surveillance , Myosins/metabolism , T-Lymphocytes/cytology , Animals , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Cell Membrane/metabolism , Cell Movement , Lymph Nodes/immunology , Mice , Minor Histocompatibility Antigens , Myosins/genetics , Receptors, Antigen, T-Cell/metabolism , T-Lymphocytes/immunology , T-Lymphocytes/metabolism
19.
Nature ; 618(7967): 1033-1040, 2023 Jun.
Article in English | MEDLINE | ID: mdl-37316667

ABSTRACT

Most clinically applied cancer immunotherapies rely on the ability of CD8+ cytolytic T cells to directly recognize and kill tumour cells1-3. These strategies are limited by the emergence of major histocompatibility complex (MHC)-deficient tumour cells and the formation of an immunosuppressive tumour microenvironment4-6. The ability of CD4+ effector cells to contribute to antitumour immunity independently of CD8+ T cells is increasingly recognized, but strategies to unleash their full potential remain to be identified7-10. Here, we describe a mechanism whereby a small number of CD4+ T cells is sufficient to eradicate MHC-deficient tumours that escape direct CD8+ T cell targeting. The CD4+ effector T cells preferentially cluster at tumour invasive margins where they interact with MHC-II+CD11c+ antigen-presenting cells. We show that T helper type 1 cell-directed CD4+ T cells and innate immune stimulation reprogramme the tumour-associated myeloid cell network towards interferon-activated antigen-presenting and iNOS-expressing tumouricidal effector phenotypes. Together, CD4+ T cells and tumouricidal myeloid cells orchestrate the induction of remote inflammatory cell death that indirectly eradicates interferon-unresponsive and MHC-deficient tumours. These results warrant the clinical exploitation of this ability of CD4+ T cells and innate immune stimulators in a strategy to complement the direct cytolytic activity of CD8+ T cells and natural killer cells and advance cancer immunotherapies.


Subject(s)
CD4-Positive T-Lymphocytes , Cell Death , Immunotherapy , Inflammation , Neoplasms , Tumor Microenvironment , Humans , Antigen-Presenting Cells/immunology , CD11c Antigen/immunology , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Cell Death/immunology , Histocompatibility Antigens Class II/immunology , Immunity, Innate , Inflammation/immunology , Interferons/immunology , Major Histocompatibility Complex/immunology , Neoplasms/immunology , Neoplasms/pathology , Neoplasms/therapy , Tumor Microenvironment/immunology , Immunotherapy/methods , Killer Cells, Natural/immunology , Myeloid Cells/immunology , Th1 Cells/cytology , Th1 Cells/immunology
20.
Nature ; 624(7992): 630-638, 2023 Dec.
Article in English | MEDLINE | ID: mdl-38093012

ABSTRACT

The COVID-19 pandemic has fostered major advances in vaccination technologies1-4; however, there are urgent needs for vaccines that induce mucosal immune responses and for single-dose, non-invasive administration4-6. Here we develop an inhalable, single-dose, dry powder aerosol SARS-CoV-2 vaccine that induces potent systemic and mucosal immune responses. The vaccine encapsulates assembled nanoparticles comprising proteinaceous cholera toxin B subunits displaying the SARS-CoV-2 RBD antigen within microcapsules of optimal aerodynamic size, and this unique nano-micro coupled structure supports efficient alveoli delivery, sustained antigen release and antigen-presenting cell uptake, which are favourable features for the induction of immune responses. Moreover, this vaccine induces strong production of IgG and IgA, as well as a local T cell response, collectively conferring effective protection against SARS-CoV-2 in mice, hamsters and nonhuman primates. Finally, we also demonstrate a mosaic iteration of the vaccine that co-displays ancestral and Omicron antigens, extending the breadth of antibody response against co-circulating strains and transmission of the Omicron variant. These findings support the use of this inhaled vaccine as a promising multivalent platform for fighting COVID-19 and other respiratory infectious diseases.


Subject(s)
COVID-19 Vaccines , Immunity, Mucosal , Animals , Cricetinae , Humans , Mice , Administration, Inhalation , Aerosols , Antibodies, Viral/immunology , Antigen-Presenting Cells/immunology , Antigen-Presenting Cells/metabolism , Antigens, Viral/immunology , Cholera Toxin , COVID-19/immunology , COVID-19/prevention & control , COVID-19 Vaccines/administration & dosage , Immunity, Mucosal/immunology , Immunoglobulin A/immunology , Immunoglobulin G/immunology , Nanoparticles , Powders , Primates/virology , SARS-CoV-2/classification , SARS-CoV-2/immunology , T-Lymphocytes/immunology , Vaccination , Capsules
SELECTION OF CITATIONS
SEARCH DETAIL