Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 99
Filter
Add more filters

Publication year range
1.
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
2.
Cell ; 184(8): 2151-2166.e16, 2021 04 15.
Article in English | MEDLINE | ID: mdl-33765440

ABSTRACT

Cutaneous mast cells mediate numerous skin inflammatory processes and have anatomical and functional associations with sensory afferent neurons. We reveal that epidermal nerve endings from a subset of sensory nonpeptidergic neurons expressing MrgprD are reduced by the absence of Langerhans cells. Loss of epidermal innervation or ablation of MrgprD-expressing neurons increased expression of a mast cell gene module, including the activating receptor, Mrgprb2, resulting in increased mast cell degranulation and cutaneous inflammation in multiple disease models. Agonism of MrgprD-expressing neurons reduced expression of module genes and suppressed mast cell responses. MrgprD-expressing neurons released glutamate which was increased by MrgprD agonism. Inhibiting glutamate release or glutamate receptor binding yielded hyperresponsive mast cells with a genomic state similar to that in mice lacking MrgprD-expressing neurons. These data demonstrate that MrgprD-expressing neurons suppress mast cell hyperresponsiveness and skin inflammation via glutamate release, thereby revealing an unexpected neuroimmune mechanism maintaining cutaneous immune homeostasis.


Subject(s)
Glutamic Acid/metabolism , Mast Cells/metabolism , Neurons/metabolism , Skin/metabolism , Animals , Cells, Cultured , Dermatitis/metabolism , Dermatitis/pathology , Diphtheria Toxin/pharmacology , Disease Models, Animal , Female , Integrin beta Chains/genetics , Integrin beta Chains/metabolism , Langerhans Cells/cytology , Langerhans Cells/drug effects , Langerhans Cells/metabolism , Mast Cells/cytology , Mast Cells/drug effects , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/deficiency , Receptors, G-Protein-Coupled/genetics , Receptors, G-Protein-Coupled/metabolism , Skin/pathology , beta-Alanine/chemistry , beta-Alanine/metabolism , beta-Alanine/pharmacology
3.
Cell ; 178(4): 919-932.e14, 2019 08 08.
Article in English | MEDLINE | ID: mdl-31353219

ABSTRACT

Cutaneous TRPV1+ neurons directly sense noxious stimuli, inflammatory cytokines, and pathogen-associated molecules and are required for innate immunity against some skin pathogens. Important unanswered questions are whether TRPV1+ neuron activation in isolation is sufficient to initiate innate immune responses and what is the biological function for TRPV1+ neuron-initiated immune responses. We used TRPV1-Ai32 optogenetic mice and cutaneous light stimulation to activate cutaneous neurons in the absence of tissue damage or pathogen-associated products. We found that TRPV1+ neuron activation was sufficient to elicit a local type 17 immune response that augmented host defense to C. albicans and S. aureus. Moreover, local neuron activation elicited type 17 responses and augmented host defense at adjacent, unstimulated skin through a nerve reflex arc. These data show the sufficiency of TRPV1+ neuron activation for host defense and demonstrate the existence of functional anticipatory innate immunity at sites adjacent to infection that depends on antidromic neuron activation.


Subject(s)
Immunity, Innate/immunology , Interleukin-23/metabolism , Interleukin-6/metabolism , Sensory Receptor Cells/immunology , Skin/immunology , TRPV Cation Channels/metabolism , Tumor Necrosis Factor-alpha/metabolism , Animals , Candida albicans/immunology , Inflammation/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , Optogenetics/methods , Skin/microbiology , Staphylococcus aureus/immunology , TRPV Cation Channels/genetics
4.
Cell ; 178(5): 1176-1188.e15, 2019 08 22.
Article in English | MEDLINE | ID: mdl-31442406

ABSTRACT

Adaptive immunity provides life-long protection by generating central and effector memory T cells and the most recently described tissue resident memory T (TRM) cells. However, the cellular origin of CD4 TRM cells and their contribution to host defense remain elusive. Using IL-17A tracking-fate mouse models, we found that a significant fraction of lung CD4 TRM cells derive from IL-17A-producing effector (TH17) cells following immunization with heat-killed Klebsiella pneumonia (Kp). These exTH17 TRM cells are maintained in the lung by IL-7, produced by lymphatic endothelial cells. During a memory response, neither antibodies, γδ T cells, nor circulatory T cells are sufficient for the rapid host defense required to eliminate Kp. Conversely, using parabiosis and depletion studies, we demonstrated that exTH17 TRM cells play an important role in bacterial clearance. Thus, we delineate the origin and function of airway CD4 TRM cells during bacterial infection, offering novel strategies for targeted vaccine design.


Subject(s)
Klebsiella Infections/immunology , Th17 Cells/immunology , Animals , CD4-Positive T-Lymphocytes/cytology , CD4-Positive T-Lymphocytes/immunology , CD4-Positive T-Lymphocytes/metabolism , Diphtheria Toxin/pharmacology , Disease Models, Animal , Female , Immunologic Memory , Interleukin-17/genetics , Interleukin-17/metabolism , Klebsiella Infections/pathology , Klebsiella pneumoniae/immunology , Klebsiella pneumoniae/pathogenicity , Lung/drug effects , Lung/metabolism , Lung/microbiology , Mice , Mice, Inbred C57BL , Th17 Cells/cytology , Th17 Cells/metabolism
5.
Immunity ; 57(4): 815-831, 2024 Apr 09.
Article in English | MEDLINE | ID: mdl-38599172

ABSTRACT

The sensory nervous system possesses the ability to integrate exogenous threats and endogenous signals to mediate downstream effector functions. Sensory neurons have been shown to activate or suppress host defense and immunity against pathogens, depending on the tissue and disease state. Through this lens, pro- and anti-inflammatory neuroimmune effector functions can be interpreted as evolutionary adaptations by host or pathogen. Here, we discuss recent and impactful examples of neuroimmune circuitry that regulate tissue homeostasis, autoinflammation, and host defense. Apparently paradoxical or conflicting reports in the literature also highlight the complexity of neuroimmune interactions that may depend on tissue- and microbe-specific cues. These findings expand our understanding of the nuanced mechanisms and the greater context of sensory neurons in innate immunity.


Subject(s)
Immunity, Innate , Sensory Receptor Cells , Immunity, Innate/physiology , Neuroimmunomodulation/physiology , Homeostasis
6.
Immunity ; 55(1): 11-13, 2022 01 11.
Article in English | MEDLINE | ID: mdl-35021052

ABSTRACT

Peripheral neurons and immune cells interact to modulate inflammation, but whether the brain can control this process is unknown. In a recent issue of Cell, Koren et al. (2021) show that peripheral inflammation is encoded in the insular cortex and that later re-activation of these neurons triggers inflammation.


Subject(s)
Brain , Neurons , Humans , Inflammation , Insular Cortex
7.
Nat Immunol ; 19(12): 1286-1298, 2018 12.
Article in English | MEDLINE | ID: mdl-30446754

ABSTRACT

The skin provides both a physical barrier and an immunologic barrier to external threats. The protective machinery of the skin has evolved to provide situation-specific responses to eliminate pathogens and to provide protection against physical dangers. Dysregulation of this machinery can give rise to the initiation and propagation of inflammatory loops in the epithelial microenvironment that result in inflammatory skin diseases in susceptible people. A defective barrier and microbial dysbiosis drive an interleukin 4 (IL-4) loop that underlies atopic dermatitis, while in psoriasis, disordered keratinocyte signaling and predisposition to type 17 responses drive a pathogenic IL-17 loop. Here we discuss the pathogenesis of atopic dermatitis and psoriasis in terms of the epithelial immune microenvironment-the microbiota, keratinocytes and sensory nerves-and the resulting inflammatory loops.


Subject(s)
Dermatitis, Atopic/immunology , Psoriasis/immunology , Skin/immunology , Animals , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/physiopathology , Dysbiosis/immunology , Dysbiosis/microbiology , Dysbiosis/physiopathology , Epithelium/immunology , Epithelium/microbiology , Epithelium/physiopathology , Humans , Psoriasis/microbiology , Psoriasis/physiopathology , Skin/microbiology
8.
Immunity ; 54(1): 84-98.e5, 2021 01 12.
Article in English | MEDLINE | ID: mdl-33212014

ABSTRACT

Following antigen-driven expansion in lymph node, transforming growth factor-ß (TGFß) is required for differentiation of skin-recruited CD8+ T cell effectors into epidermal resident memory T (Trm) cells and their epidermal persistence. We found that the source of TGFß -supporting Trm cells was autocrine. In addition, antigen-specific Trm cells that encountered cognate antigen in the skin, and bystander Trm cells that did not, both displayed long-term persistence in the epidermis under steady-state conditions. However, when the active-TGFß was limited or when new T cell clones were recruited into the epidermis, antigen-specific Trm cells were more efficiently retained than bystander Trm cells. Genetically enforced TGFßR signaling allowed bystander Trm cells to persist in the epidermis as efficiently as antigen-specific Trm cells in both contexts. Thus, competition between T cells for active TGFß represents an unappreciated selective pressure that promotes the accumulation and persistence of antigen-specific Trm cells in the epidermal niche.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epidermis/immunology , Keratinocytes/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/metabolism , Animals , Binding, Competitive , Bystander Effect , Cellular Microenvironment , Clone Cells , Immunologic Memory , Mice , Mice, Inbred C57BL , Organ Specificity , Receptors, Antigen, T-Cell, alpha-beta/metabolism , Signal Transduction , T-Cell Antigen Receptor Specificity
9.
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
10.
Nat Immunol ; 17(1): 48-56, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26523865

ABSTRACT

Innate lymphoid cells (ILCs) 'preferentially' localize into barrier tissues, where they function in tissue protection but can also contribute to inflammatory diseases. The mechanisms that regulate the establishment of ILCs in barrier tissues are poorly understood. Here we found that under steady-state conditions, ILCs in skin-draining lymph nodes (sLNs) were continuously activated to acquire regulatory properties and high expression of the chemokine receptor CCR10 for localization into the skin. CCR10(+) ILCs promoted the homeostasis of skin-resident T cells and, reciprocally, their establishment in the skin required T cell-regulated homeostatic environments. CD207(+) dendritic cells expressing the transcription factor Foxn1 were required for the proper generation of CCR10(+) ILCs. These observations reveal mechanisms that underlie the specific programming and priming of skin-homing CCR10(+) ILCs in the sLNs.


Subject(s)
Homeostasis/immunology , Lymph Nodes/immunology , Lymphocytes/immunology , Receptors, CCR10/immunology , Skin/immunology , Adoptive Transfer , Animals , Flow Cytometry , Immunity, Innate/immunology , Lymph Nodes/cytology , Mice , Mice, Inbred C57BL , Mice, Knockout
11.
Nat Immunol ; 17(4): 414-21, 2016 Apr.
Article in English | MEDLINE | ID: mdl-26901152

ABSTRACT

Cells of the immune system that reside in barrier epithelia provide a first line of defense against pathogens. Langerhans cells (LCs) and CD8(+) tissue-resident memory T cells (TRM cells) require active transforming growth factor-ß1 (TGF-ß) for epidermal residence. Here we found that integrins αvß6 and αvß8 were expressed in non-overlapping patterns by keratinocytes (KCs) and maintained the epidermal residence of LCs and TRM cells by activating latent TGF-ß. Similarly, the residence of dendritic cells and TRM cells in the small intestine epithelium also required αvß6. Treatment of the skin with ultraviolet irradiation decreased integrin expression on KCs and reduced the availability of active TGF-ß, which resulted in LC migration. Our data demonstrated that regulated activation of TGF-ß by stromal cells was able to directly control epithelial residence of cells of the immune system through a novel mechanism of intercellular communication.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Epidermis/immunology , Intestinal Mucosa/immunology , Keratinocytes/immunology , Langerhans Cells/immunology , Transforming Growth Factor beta/immunology , Animals , Antigens, Neoplasm/immunology , CD8-Positive T-Lymphocytes/cytology , Cell Movement , Epidermal Cells , Flow Cytometry , Fluorescent Antibody Technique , Humans , Immunity, Mucosal , Integrins/immunology , Intestinal Mucosa/cytology , Intestine, Small/cytology , Intestine, Small/immunology , Langerhans Cells/cytology , Mice , Mice, Knockout , Mink , Polymerase Chain Reaction , Stromal Cells , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , T-Lymphocytes/cytology , T-Lymphocytes/immunology , Transforming Growth Factor beta1/immunology
12.
Immunity ; 50(5): 1249-1261.e5, 2019 05 21.
Article in English | MEDLINE | ID: mdl-30952606

ABSTRACT

Regulated activation of the cytokine TGF-ß by integrins αvß6 and αvß8 expressed on keratinocytes is required for residence of epidermal-resident memory T cells, but whether skin-derived signals also affect recirculating memory cells in the skin remains unclear. Here, we show that after resolution of skin vaccinia virus (VV) infection, antigen-specific circulating memory CD8+ T cells migrated into skin. In mice lacking αvß6 and αvß8 integrins (Itgb6-/-Itgb8fl/fl-K14-cre), the absence of epidermal-activated TGF-ß resulted in a gradual loss of E- or P-selectin-binding central and peripheral memory populations, which were rescued when skin entry was inhibited. Skin recirculating memory cells were required for optimal host defense against skin VV infection. These data demonstrate that skin migration can persist after resolution of local skin infection and that the cytokine environment within this nonlymphoid tissue shapes the differentiation state and persistence of the central and peripheral memory-T-cell pool.


Subject(s)
Antigens, Neoplasm/metabolism , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Integrins/metabolism , Keratinocytes/metabolism , Transforming Growth Factor beta/metabolism , Vaccinia virus/immunology , Animals , Antigens, Neoplasm/genetics , CD8-Positive T-Lymphocytes/enzymology , Cell Differentiation/immunology , Cytokines/immunology , Enzyme Activation , Female , Integrins/genetics , Male , Mice , Mice, Inbred C57BL , Skin/cytology , Skin/immunology
13.
Nat Immunol ; 14(4): 364-71, 2013 Apr.
Article in English | MEDLINE | ID: mdl-23435120

ABSTRACT

Dendritic cells (DCs) are critical in immune responses, linking innate and adaptive immunity. We found here that DC-specific deletion of the transcription factor STAT5 was not critical for development but was required for T helper type 2 (TH2), but not TH1, allergic responses in both the skin and lungs. Loss of STAT5 in DCs led to the inability to respond to thymic stromal lymphopoietin (TSLP). STAT5 was required for TSLP-dependent DC activation, including upregulation of the expression of costimulatory molecules and chemokine production. Furthermore, TH2 responses in mice with DC-specific loss of STAT5 resembled those seen in mice deficient in the receptor for TSLP. Our results show that the TSLP-STAT5 axis in DCs is a critical component for the promotion of type 2 immunity at barrier surfaces.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , STAT5 Transcription Factor/metabolism , Th2 Cells/immunology , Animals , Cell Differentiation , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/cytology , Dermatitis, Contact/immunology , Dermatitis, Contact/metabolism , Dermis/immunology , Dermis/metabolism , Female , Homeostasis/immunology , Janus Kinases/metabolism , Lung/immunology , Lung/metabolism , Mice , Mice, Knockout , STAT5 Transcription Factor/genetics , Signal Transduction , Th1 Cells/immunology , Thymic Stromal Lymphopoietin
14.
Immunity ; 43(3): 515-26, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26377898

ABSTRACT

Innate resistance to Candida albicans in mucosal tissues requires the production of interleukin-17A (IL-17A) by tissue-resident cells early during infection, but the mechanism of cytokine production has not been precisely defined. In the skin, we found that dermal γδ T cells were the dominant source of IL-17A during C. albicans infection and were required for pathogen resistance. Induction of IL-17A from dermal γδ T cells and resistance to C. albicans required IL-23 production from CD301b(+) dermal dendritic cells (dDCs). In addition, we found that sensory neurons were directly activated by C. albicans. Ablation of sensory neurons increased susceptibility to C. albicans infection, which could be rescued by exogenous addition of the neuropeptide CGRP. These data define a model in which nociceptive pathways in the skin drive production of IL-23 by CD301b(+) dDCs resulting in IL-17A production from γδ T cells and resistance to cutaneous candidiasis.


Subject(s)
Dendritic Cells/immunology , Immunity/immunology , Interleukin-23/immunology , Sensory Receptor Cells/immunology , Skin/immunology , Animals , Candida albicans/immunology , Candida albicans/physiology , Candidiasis/genetics , Candidiasis/immunology , Candidiasis/microbiology , Cells, Cultured , Dendritic Cells/metabolism , Dermis/cytology , Flow Cytometry , Host-Pathogen Interactions/immunology , Immunity/genetics , Interleukin-17/genetics , Interleukin-17/immunology , Interleukin-17/metabolism , Interleukin-23/genetics , Interleukin-23/metabolism , Lectins, C-Type/immunology , Lectins, C-Type/metabolism , Mice, Inbred Strains , Mice, Knockout , Mice, Transgenic , Oligonucleotide Array Sequence Analysis , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/immunology , Receptors, Antigen, T-Cell, gamma-delta/metabolism , Receptors, Calcitonin Gene-Related Peptide/genetics , Receptors, Calcitonin Gene-Related Peptide/immunology , Receptors, Calcitonin Gene-Related Peptide/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Sensory Receptor Cells/metabolism , Skin/metabolism , Skin/microbiology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Transcriptome/genetics , Transcriptome/immunology
15.
Immunity ; 42(4): 756-66, 2015 Apr 21.
Article in English | MEDLINE | ID: mdl-25902485

ABSTRACT

Staphylococcus aureus skin colonization is universal in atopic dermatitis and common in cancer patients treated with epidermal growth factor receptor inhibitors. However, the causal relationship of dysbiosis and eczema has yet to be clarified. Herein, we demonstrate that Adam17(fl/fl)Sox9-(Cre) mice, generated to model ADAM17-deficiency in human, developed eczematous dermatitis with naturally occurring dysbiosis, similar to that observed in atopic dermatitis. Corynebacterium mastitidis, S. aureus, and Corynebacterium bovis sequentially emerged during the onset of eczematous dermatitis, and antibiotics specific for these bacterial species almost completely reversed dysbiosis and eliminated skin inflammation. Whereas S. aureus prominently drove eczema formation, C. bovis induced robust T helper 2 cell responses. Langerhans cells were required for eliciting immune responses against S. aureus inoculation. These results characterize differential contributions of dysbiotic flora during eczema formation, and highlight the microbiota-host immunity axis as a possible target for future therapeutics in eczematous dermatitis.


Subject(s)
Dermatitis, Atopic/immunology , Dysbiosis/immunology , Eczema/immunology , Langerhans Cells/immunology , Skin/immunology , T-Lymphocytes, Helper-Inducer/immunology , ADAM Proteins/deficiency , ADAM Proteins/genetics , ADAM Proteins/immunology , ADAM17 Protein , Animals , Anti-Bacterial Agents/pharmacology , Corynebacterium/immunology , Dermatitis, Atopic/drug therapy , Dermatitis, Atopic/genetics , Dermatitis, Atopic/microbiology , Dysbiosis/drug therapy , Dysbiosis/genetics , Dysbiosis/microbiology , Eczema/drug therapy , Eczema/genetics , Eczema/microbiology , ErbB Receptors/genetics , ErbB Receptors/immunology , Gene Expression Regulation , Humans , Immunity, Innate , Inflammation/drug therapy , Inflammation/genetics , Inflammation/immunology , Inflammation/microbiology , Integrases/genetics , Integrases/immunology , Langerhans Cells/drug effects , Langerhans Cells/microbiology , Langerhans Cells/pathology , Mice , Mice, Inbred C57BL , Mice, Transgenic , SOX9 Transcription Factor/genetics , SOX9 Transcription Factor/immunology , Signal Transduction , Skin/drug effects , Skin/microbiology , Skin/pathology , Staphylococcus aureus/immunology , T-Lymphocytes, Helper-Inducer/drug effects , T-Lymphocytes, Helper-Inducer/microbiology , T-Lymphocytes, Helper-Inducer/pathology
16.
Immunity ; 42(2): 356-366, 2015 Feb 17.
Article in English | MEDLINE | ID: mdl-25680275

ABSTRACT

Candida albicans is a dimorphic fungus responsible for chronic mucocutaneous and systemic infections. Mucocutaneous immunity to C. albicans requires T helper 17 (Th17) cell differentiation that is thought to depend on recognition of filamentous C. albicans. Systemic immunity is considered T cell independent. Using a murine skin infection model, we compared T helper cell responses to yeast and filamentous C. albicans. We found that only yeast induced Th17 cell responses through a mechanism that required Dectin-1-mediated expression of interleukin-6 (IL-6) by Langerhans cells. Filamentous forms induced Th1 without Th17 cell responses due to the absence of Dectin-1 ligation. Notably, Th17 cell responses provided protection against cutaneous infection while Th1 cell responses provided protection against systemic infection. Thus, C. albicans morphology drives distinct T helper cell responses that provide tissue-specific protection. These findings provide insight into compartmentalization of Th cell responses and C. albicans pathogenesis and have critical implications for vaccine strategies.


Subject(s)
Candidiasis, Chronic Mucocutaneous/immunology , Cell Differentiation/immunology , Dendritic Cells/immunology , Th17 Cells/cytology , Th17 Cells/immunology , Animals , Basic-Leucine Zipper Transcription Factors/genetics , Candida albicans/immunology , Candidiasis, Chronic Mucocutaneous/microbiology , Interleukin-6/biosynthesis , Interleukin-6/genetics , Interleukin-6/immunology , Langerhans Cells/immunology , Lectins, C-Type/genetics , Lectins, C-Type/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL , Mice, Knockout , Repressor Proteins/genetics , Skin/immunology , Skin/microbiology , Th1 Cells/cytology , Th1 Cells/immunology
17.
J Allergy Clin Immunol ; 151(5): 1169-1177, 2023 05.
Article in English | MEDLINE | ID: mdl-37149370

ABSTRACT

The skin is a barrier organ populated by many types of skin-resident immune cells and sensory neurons. It has become increasingly appreciated that neuroimmune interactions are an important component of inflammatory diseases such as atopic dermatitis and allergic contact dermatitis. Neuropeptides secreted from nerve terminals play an important role in mediating cutaneous immune cell function, and soluble mediators derived from immune cells interact with neurons to induce itch. In this review article, we will explore emerging research describing neuronal effector functions on skin immune cells in mouse models of atopic and contact dermatitis. We will also discuss the contributions of both specific neuronal subsets and secreted immune factors to itch induction and the associated inflammatory processes. Finally, we will explore how treatment strategies have emerged around these findings and discuss the relationship between scratching and dermatitis.


Subject(s)
Dermatitis, Allergic Contact , Dermatitis, Atopic , Mice , Animals , Neuroimmunomodulation , Pruritus , Skin , Sensory Receptor Cells
18.
J Allergy Clin Immunol ; 150(1): 114-130, 2022 07.
Article in English | MEDLINE | ID: mdl-35085664

ABSTRACT

BACKGROUND: Allergic contact dermatitis (CD) is a chronic inflammatory skin disease caused by type 1 biased adaptive immunity for which there is an unmet need for antigen (Ag)-specific immunotherapies. Exposure to skin sensitizers stimulates secretion of the proinflammatory neuropeptides substance P and hemokinin 1, which signal via the neurokinin-1 receptor (NK1R) to promote the innate and adaptive immune responses of CD. Accordingly, mice lacking the NK1R develop impaired CD. Nonetheless, the role and therapeutic opportunities of targeting the NK1R in CD remain to be elucidated. OBJECTIVE: We sought to develop an Ag-specific immunosuppressive approach to treat CD by skin codelivery of hapten and NK1R antagonists integrated in dissolvable microneedle arrays (MNA). METHODS: In vivo mouse models of contact hypersensitivity and ex vivo models of human skin were used to delineate the effects and mechanisms of NK1R signaling and the immunosuppressive effects of the contact sensitizer NK1R antagonist MNA in CD. RESULTS: We demonstrated in mice that CD requires NK1R signaling by substance P and hemokinin 1. Specific deletion of the NK1R in keratinocytes and dendritic cells, but not in mast cells, prevented CD. Skin codelivery of hapten or Ag MNA inhibited neuropeptide-mediated skin inflammation in mouse and human skin, promoted deletion of Ag-specific effector T cells, and increased regulatory T cells, which prevented CD onset and relapses locally and systemically in an Ag-specific manner. CONCLUSIONS: Immunoregulation by engineering localized skin neuroimmune networks can be used to treat cutaneous diseases that like CD are caused by type 1 immunity.


Subject(s)
Dermatitis, Allergic Contact , Neurokinin-1 Receptor Antagonists , Animals , Dermatitis, Allergic Contact/drug therapy , Haptens , Mice , Neurokinin-1 Receptor Antagonists/pharmacology , Receptors, Neurokinin-1 , Substance P
19.
J Immunol ; 204(2): 264-270, 2020 01 15.
Article in English | MEDLINE | ID: mdl-31907268

ABSTRACT

The skin is innervated by numerous sensory afferent neurons that respond to a diverse array of stimuli ranging from gentle touch to noxious pain. Various features of the immune system-pathogen recognition, secretion of soluble mediators-are shared with the nervous system. This has led to the recognition that neurons share some functions with innate immune cells and have the capacity to recognize pathogens and participate in innate immune responses. Neuroimmune interactions are bidirectional. Soluble mediators from immune cells activate neurons and soluble mediators from neurons can activate immune cells. In this review, we will focus on the interplay between neurons and innate immunity in the skin in the context of host defense and inflammation.


Subject(s)
Neuroimmunomodulation/immunology , Sensory Receptor Cells/immunology , Skin/immunology , Skin/innervation , Animals , Humans , Immunity, Innate/immunology
20.
J Immunol ; 205(3): 720-730, 2020 08 01.
Article in English | MEDLINE | ID: mdl-32601099

ABSTRACT

Oropharyngeal candidiasis (OPC) is an opportunistic infection of the oral mucosa caused by the commensal fungus Candida albicans IL-17R signaling is essential to prevent OPC in mice and humans, but the individual roles of its ligands, IL-17A, IL-17F, and IL-17AF, are less clear. A homozygous IL-17F deficiency in mice does not cause OPC susceptibility, whereas mice lacking IL-17A are moderately susceptible. In humans, a rare heterozygous mutation in IL-17F (IL-17F.S65L) was identified that causes chronic mucocutaneous candidiasis, suggesting the existence of essential antifungal pathways mediated by IL-17F and/or IL-17AF. To investigate the role of IL-17F and IL-17AF in more detail, we exploited this "experiment of nature" by creating a mouse line bearing the homologous mutation in IL-17F (Ser65Leu) by CRISPR/Cas9. Unlike Il17f-/- mice that are resistant to OPC, Il17fS65L/S65L mice showed increased oral fungal burdens similar to Il17a -/- mice. In contrast to humans, however, disease was only evident in homozygous, not heterozygous, mutant mice. The mutation was linked to modestly impaired CXC chemokine expression and neutrophil recruitment to the infected tongue but not to alterations in oral antimicrobial peptide expression. These findings suggest mechanisms by which the enigmatic cytokine IL-17F contributes to host defense against fungi. Moreover, because these mice do not phenocopy Il17f-/- mice, they may provide a valuable tool to interrogate IL-17F and IL-17AF function in vivo in other settings.


Subject(s)
Candida albicans/immunology , Candidiasis/immunology , Interleukin-17/immunology , Mouth Diseases/immunology , Animals , Candida albicans/genetics , Candidiasis/genetics , Candidiasis/pathology , Gene Knock-In Techniques , Interleukin-17/genetics , Mice , Mice, Transgenic , Mouth Diseases/genetics , Mouth Diseases/microbiology , Mouth Diseases/pathology , Mutation, Missense
SELECTION OF CITATIONS
SEARCH DETAIL