Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 28
Filter
1.
Sci Immunol ; 9(91): eadh0152, 2024 Jan 05.
Article in English | MEDLINE | ID: mdl-38181095

ABSTRACT

Immune tolerance is maintained in lymphoid organs (LOs). Despite the presence of complex immune cell networks in non-LOs, it is unknown whether self-tolerance is maintained in these tissues. We developed a technique to restrict genetic recombination to regulatory T cells (Tregs) only in skin. Selective depletion of skin Tregs resulted in T cell-mediated inflammation of hair follicles (HFs). Suppression did not rely on CTLA-4, but instead on high-affinity interleukin-2 (IL-2) receptor expression by skin Tregs, functioning exclusively in a cell-extrinsic manner. In a novel model of HF stem cell (HFSC)-driven autoimmunity, we reveal that skin Tregs immunologically protect the HFSC niche. Finally, we used spatial transcriptomics to identify aberrant IL-2 signaling at stromal-HF interfaces in a rare form of human alopecia characterized by HFSC destruction and alopecia areata. Collectively, these results reveal the fundamental biology of Tregs in skin uncoupled from the systemic pool and elucidate a mechanism of self-tolerance.


Subject(s)
Immune Privilege , T-Lymphocytes, Regulatory , Humans , Hair Follicle , Interleukin-2 , Stem Cell Niche
2.
JCI Insight ; 9(3)2024 Feb 08.
Article in English | MEDLINE | ID: mdl-38113104

ABSTRACT

Hidradenitis suppurativa (HS) is a chronic skin condition affecting approximately 1% of the US population. HS skin lesions are highly inflammatory and characterized by a large immune infiltrate. While B cells and plasma cells comprise a major component of this immune milieu, the biology and the contribution of these cells in HS pathogenesis are unclear. We aimed to investigate the dynamics and microenvironmental interactions of B cells within cutaneous HS lesions. Combining histological analysis, single-cell RNA sequencing, and spatial transcriptomics profiling of HS lesions, we defined the tissue microenvironment relative to B cell activity within this disease. Our findings identified tertiary lymphoid structures (TLSs) within HS lesions and described organized interactions among T cells, B cells, antigen-presenting cells, and skin stroma. We found evidence that B cells within HS TLSs actively underwent maturation, including participation in germinal center reactions and class switch recombination. Moreover, skin stroma and accumulating T cells were primed to support the formation of TLSs and facilitate B cell recruitment during HS. Our data definitively demonstrated the presence of TLSs in lesional HS skin and point to ongoing cutaneous B cell maturation through class switch recombination and affinity maturation during disease progression in this inflamed nonlymphoid tissue.


Subject(s)
Hidradenitis Suppurativa , Tertiary Lymphoid Structures , Humans , Hidradenitis Suppurativa/pathology , Tertiary Lymphoid Structures/pathology , Skin/pathology , B-Lymphocytes/pathology , T-Lymphocytes/pathology
3.
bioRxiv ; 2023 Feb 15.
Article in English | MEDLINE | ID: mdl-36824918

ABSTRACT

Background: Hidradenitis suppurativa (HS) skin lesions are highly inflammatory and characterized by a large immune infiltrate. While B cells and plasma cells comprise a major component of this immune milieu the biology and contribution of these cells in HS pathogenesis is unclear. Objective: We aimed to investigate the dynamics and microenvironmental interactions of B cells within cutaneous HS lesions. Methods: We combined histological analysis, single-cell RNA-sequencing (scRNAseq), and spatial transcriptomic profiling of HS lesions to define the tissue microenvironment relative to B cell activity within this disease. Results: Our findings identify tertiary lymphoid structures (TLS) within HS lesions and describe organized interactions between T cells, B cells, antigen presenting cells and skin stroma. We find evidence that B cells within HS TLS actively undergo maturation, including participation in germinal center reactions and class switch recombination. Moreover, skin stroma and accumulating T cells are primed to support the formation of TLS and facilitate B cell recruitment during HS. Conclusion: Our data definitively demonstrate the presence of TLS in lesional HS skin and point to ongoing cutaneous B cell maturation through class switch recombination and affinity maturation during disease progression in this inflamed non-lymphoid tissue.

5.
J Vis Exp ; (184)2022 06 29.
Article in English | MEDLINE | ID: mdl-35848826

ABSTRACT

The human skin xenograft model, in which human donor skin is transplanted onto an immunodeficient mouse host, is an important option for translational research in skin immunology. Murine and human skin differ substantially in anatomy and immune cell composition. Therefore, traditional mouse models have limitations for dermatological research and drug discovery. However, successful xenotransplants are technically challenging and require optimal specimen and mouse graft site preparation for graft and host survival. The present protocol provides an optimized technique for transplanting human skin onto mice and discusses necessary considerations for downstream experimental aims. This report describes the appropriate preparation of a human donor skin sample, assembly of a surgical setup, mouse and surgical site preparation, skin transplantation, and post-surgical monitoring. Adherence to these methods allows for maintenance of xenografts for over 6 weeks post-surgery. The techniques outlined below allow maximum grafting efficiency due to the development of engineering controls, sterile technique, and pre- and post-surgical conditioning. Appropriate performance of the xenograft model results in long-lived human skin graft samples for experimental characterization of human skin and preclinical testing of compounds in vivo.


Subject(s)
Skin Transplantation , Skin , Animals , Disease Models, Animal , Heterografts , Humans , Immunity , Mice , Skin Transplantation/methods , Transplantation, Heterologous
6.
Nature ; 599(7886): 667-672, 2021 11.
Article in English | MEDLINE | ID: mdl-34707292

ABSTRACT

Inflammation early in life can prime the local immune milieu of peripheral tissues, which can cause lasting changes in immunological tone that confer disease protection or susceptibility1. The cellular and molecular mechanisms that prompt changes in immune tone in many nonlymphoid tissues remain largely unknown. Here we find that time-limited neonatal inflammation induced by a transient reduction in neonatal regulatory T cells causes a dysregulation of subcutaneous tissue in mouse skin. This is accompanied by the selective accumulation of type 2 helper T (TH2) cells within a distinct microanatomical niche. TH2 cells are maintained into adulthood through interactions with a fibroblast population in skin fascia that we refer to as TH2-interacting fascial fibroblasts (TIFFs), which expand in response to TH2 cytokines to form subcutaneous fibrous bands. Activation of the TH2-TIFF niche due to neonatal inflammation primes the skin for altered reparative responses to wounding. Furthermore, we identify fibroblasts in healthy human skin that express the TIFF transcriptional signature and detect these cells at high levels in eosinophilic fasciitis, an orphan disease characterized by inflammation and fibrosis of the skin fascia. Taken together, these data define a previously unidentified TH2 cell niche in skin and functionally characterize a disease-associated fibroblast population. The results also suggest a mechanism of immunological priming whereby inflammation early in life creates networks between adaptive immune cells and stromal cells to establish an immunological set-point in tissues that is maintained throughout life.


Subject(s)
Fibroblasts/cytology , Inflammation/pathology , Skin/cytology , Stem Cell Niche , Th2 Cells/cytology , Animals , Animals, Newborn , Cytokines/immunology , Eosinophilia/pathology , Fasciitis/pathology , Fibrosis/pathology , Health , Humans , Interleukin-13 Receptor alpha1 Subunit/metabolism , Male , Mice , Skin/pathology , T-Lymphocytes, Regulatory/cytology , Wound Healing
7.
J Immunol ; 207(7): 1763-1775, 2021 10 01.
Article in English | MEDLINE | ID: mdl-34470859

ABSTRACT

Regulatory T cells (Tregs) reside in nonlymphoid tissues where they carry out unique functions. The molecular mechanisms responsible for Treg accumulation and maintenance in these tissues are relatively unknown. Using an unbiased discovery approach, we identified LAYN (layilin), a C-type lectin-like receptor, to be preferentially and highly expressed on a subset of activated Tregs in healthy and diseased human skin. Expression of layilin on Tregs was induced by TCR-mediated activation in the presence of IL-2 or TGF-ß. Mice with a conditional deletion of layilin in Tregs had reduced accumulation of these cells in tumors. However, these animals somewhat paradoxically had enhanced immune regulation in the tumor microenvironment, resulting in increased tumor growth. Mechanistically, layilin expression on Tregs had a minimal effect on their activation and suppressive capacity in vitro. However, expression of this molecule resulted in a cumulative anchoring effect on Treg dynamic motility in vivo. Taken together, our results suggest a model whereby layilin facilitates Treg adhesion in skin and, in doing so, limits their suppressive capacity. These findings uncover a unique mechanism whereby reduced Treg motility acts to limit immune regulation in nonlymphoid organs and may help guide strategies to exploit this phenomenon for therapeutic benefit.


Subject(s)
Carrier Proteins/metabolism , Membrane Glycoproteins/metabolism , Receptors, Lymphocyte Homing/metabolism , Skin/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Carrier Proteins/genetics , Cell Movement , Cells, Cultured , Humans , Immune Tolerance , Lymphocyte Activation , Membrane Glycoproteins/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Models, Immunological , Organ Specificity , Receptors, Lymphocyte Homing/genetics , Transforming Growth Factor beta/metabolism
8.
Sci Immunol ; 6(62)2021 08 27.
Article in English | MEDLINE | ID: mdl-34452925

ABSTRACT

Regulatory T cells (Tregs) use multiple mechanisms to attenuate inflammation and prevent autoimmunity. Tregs residing in peripheral (i.e., nonlymphoid) tissues have specialized functions; specifically, skin Tregs promote wound healing, suppress dermal fibrosis, facilitate epidermal regeneration, and augment hair follicle cycling. Here, we demonstrated that skin Tregs were transcriptionally attuned to interact with their tissue environment through increased expression of integrin and TGF-ß pathway genes that influence epithelial cell biology. We identified a molecular pathway where skin Tregs license keratinocytes to promote innate inflammation after skin barrier breach. Using a single-cell discovery approach, we identified preferential expression of the integrin αvß8 on skin Tregs Upon skin injury, Tregs used this integrin to activate latent TGF-ß, which acted directly on epithelial cells to promote CXCL5 production and neutrophil recruitment. Induction of this circuit delayed epidermal regeneration but provided protection from Staphylococcus aureus infection across a compromised barrier. Thus, αvß8-expressing Tregs in the skin, somewhat paradoxical to their canonical immunosuppressive functions, facilitated inflammation acutely after loss of barrier integrity to promote host defense against infection.


Subject(s)
Immunity, Innate/immunology , Inflammation/immunology , Skin/immunology , T-Lymphocytes, Regulatory/immunology , Transforming Growth Factor beta/immunology , Animals , Mice , Mice, Congenic , Mice, Inbred C57BL , Mice, Transgenic
9.
J Invest Dermatol ; 141(8): 1885-1887, 2021 08.
Article in English | MEDLINE | ID: mdl-34303469

ABSTRACT

B cells were long presumed to be a minor and functionally unimportant component of cutaneous immunobiology. However, it is now clear that these lymphocytes are present in healthy skin and accumulate during inflammatory disease. Aira and Debes (2021) identify ⍺4ꞵ1 integrin-mediated recruitment of IL-10+ B cells as a key pathway in attenuating skin inflammation. Their work provides valuable insight into the potential for B cells to regulate skin pathology.


Subject(s)
Dermatitis , Integrins , Animals , B-Lymphocytes , Disease Models, Animal , Inflammation
10.
J Exp Med ; 218(4)2021 04 05.
Article in English | MEDLINE | ID: mdl-33651880

ABSTRACT

The ability to monitor anti-tumor CD8+ T cell responses in the blood has tremendous therapeutic potential. Here, we used paired single-cell RNA and TCR sequencing to detect and characterize "tumor-matching" (TM) CD8+ T cells in the blood of mice with MC38 tumors or melanoma patients using the TCR as a molecular barcode. TM cells showed increased activation compared with nonmatching T cells in blood and were less exhausted than matching cells in tumors. Importantly, PD-1, which has been used to identify putative circulating anti-tumor CD8+ T cells, showed poor sensitivity for identifying TM cells. By leveraging the transcriptome, we identified candidate cell surface markers for TM cells in mice and patients and validated NKG2D, CD39, and CX3CR1 in mice. These data show that the TCR can be used to identify tumor-relevant cells for characterization, reveal unique transcriptional properties of TM cells, and develop marker panels for tracking and analysis of these cells.


Subject(s)
Adenocarcinoma/immunology , CD8-Positive T-Lymphocytes/immunology , Colonic Neoplasms/immunology , Melanoma/blood , Melanoma/immunology , Single-Cell Analysis/methods , Skin Neoplasms/blood , Skin Neoplasms/immunology , Adenocarcinoma/pathology , Animals , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Cell Line, Tumor , Colonic Neoplasms/pathology , Female , Humans , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor/genetics , Programmed Cell Death 1 Receptor/metabolism , Receptors, Antigen, T-Cell/genetics , Receptors, Antigen, T-Cell/metabolism , Transcriptome
11.
Cell Rep Med ; 1(8): 100132, 2020 11 17.
Article in English | MEDLINE | ID: mdl-33294857

ABSTRACT

Lymphocytes in barrier tissues play critical roles in host defense and homeostasis. These cells take up residence in tissues during defined developmental windows, when they may demonstrate distinct phenotypes and functions. Here, we utilized mass and flow cytometry to elucidate early features of human skin immunity. Although most conventional αß T (Tconv) cells in fetal skin have a naive, proliferative phenotype, a subset of CD4+ Tconv and CD8+ cells demonstrate memory-like features and a propensity for interferon (IFN)γ production. Skin regulatory T cells dynamically accumulate over the second trimester in temporal and regional association with hair follicle development. These fetal skin regulatory T cells (Tregs) demonstrate an effector memory phenotype while differing from their adult counterparts in expression of key effector molecules. Thus, we identify features of prenatal skin lymphocytes that may have key implications for understanding antigen and allergen encounters in utero and in infancy.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/immunology , Immunologic Memory/immunology , Interferon-gamma/immunology , Skin/immunology , Flow Cytometry/methods , Humans , Lymphocyte Activation/immunology , T-Lymphocytes, Regulatory/immunology
12.
JCI Insight ; 5(19)2020 10 02.
Article in English | MEDLINE | ID: mdl-32841223

ABSTRACT

Hidradenitis suppurativa (HS) is a highly prevalent, morbid inflammatory skin disease with limited treatment options. The major cell types and inflammatory pathways in skin of patients with HS are poorly understood, and which patients will respond to TNF-α blockade is currently unknown. We discovered that clinically and histologically healthy appearing skin (i.e., nonlesional skin) is dysfunctional in patients with HS with a relative loss of immune regulatory pathways. HS skin lesions were characterized by quantitative and qualitative dysfunction of type 2 conventional dendritic cells, relatively reduced regulatory T cells, an influx of memory B cells, and a plasma cell/plasmablast infiltrate predominantly in end-stage fibrotic skin. At the molecular level, there was a relative bias toward the IL-1 pathway and type 1 T cell responses when compared with both healthy skin and psoriatic patient skin. Anti-TNF-α therapy markedly attenuated B cell activation with minimal effect on other inflammatory pathways. Finally, we identified an immune activation signature in skin before anti-TNF-α treatment that correlated with subsequent lack of response to this modality. Our results reveal the fundamental immunopathogenesis of HS and provide a molecular foundation for future studies focused on stratifying patients based on likelihood of clinical response to TNF-α blockade.


Subject(s)
Biomarkers/analysis , Gene Expression Regulation , Hidradenitis Suppurativa/drug therapy , T-Lymphocytes, Regulatory/immunology , Transcriptome/drug effects , Tumor Necrosis Factor-alpha/pharmacology , Case-Control Studies , Gene Regulatory Networks , Hidradenitis Suppurativa/immunology , Hidradenitis Suppurativa/pathology , Humans , Signal Transduction , Single-Cell Analysis/methods , T-Lymphocytes, Regulatory/drug effects
13.
JCI Insight ; 5(19)2020 10 02.
Article in English | MEDLINE | ID: mdl-32853177

ABSTRACT

Hidradenitis suppurativa (HS) is a debilitating chronic inflammatory skin disease characterized by chronic abscess formation and development of multiple draining sinus tracts in the groin, axillae, and perineum. Using proteomic and transcriptomic approaches, we characterized the inflammatory responses in HS in depth, revealing immune responses centered on IFN-γ, IL-36, and TNF, with lesser contribution from IL-17A. We further identified B cells and plasma cells, with associated increases in immunoglobulin production and complement activation, as pivotal players in HS pathogenesis, with Bruton's tyrosine kinase (BTK) and spleen tyrosine kinase (SYK) pathway activation as a central signal transduction network in HS. These data provide preclinical evidence to accelerate the path toward clinical trials targeting BTK and SYK signaling in moderate-to-severe HS.


Subject(s)
B-Lymphocytes/immunology , Biomarkers/analysis , Gene Expression Regulation , Hidradenitis Suppurativa/pathology , Plasma Cells/immunology , Proteome/metabolism , Transcriptome , Agammaglobulinaemia Tyrosine Kinase/genetics , Agammaglobulinaemia Tyrosine Kinase/metabolism , B-Lymphocytes/metabolism , B-Lymphocytes/pathology , Case-Control Studies , Gene Regulatory Networks , Hidradenitis Suppurativa/genetics , Hidradenitis Suppurativa/immunology , Hidradenitis Suppurativa/metabolism , Humans , Plasma Cells/metabolism , Plasma Cells/pathology , Proteome/analysis , Signal Transduction , Single-Cell Analysis , Syk Kinase/genetics , Syk Kinase/metabolism
14.
J Exp Med ; 217(9)2020 09 07.
Article in English | MEDLINE | ID: mdl-32539073

ABSTRACT

Tumor-infiltrating CD8+ T cells mediate antitumor immune responses. However, the mechanisms by which T cells remain poised to kill cancer cells despite expressing high levels of inhibitory receptors are unknown. Here, we report that layilin, a C-type lectin domain-containing membrane glycoprotein, is selectively expressed on highly activated, clonally expanded, but phenotypically exhausted CD8+ T cells in human melanoma. Lineage-specific deletion of layilin on murine CD8+ T cells reduced their accumulation in tumors and increased tumor growth in vivo. Congruently, gene editing of LAYN in human CD8+ T cells reduced direct tumor cell killing ex vivo. On a molecular level, layilin colocalized with integrin αLß2 (LFA-1) on T cells, and cross-linking layilin promoted the activated state of this integrin. Accordingly, LAYN deletion resulted in attenuated LFA-1-dependent cellular adhesion. Collectively, our results identify layilin as part of a molecular pathway in which exhausted or "dysfunctional" CD8+ T cells enhance cellular adhesiveness to maintain their cytotoxic potential.


Subject(s)
Carrier Proteins/metabolism , Immunity , Integrins/metabolism , Lectins, C-Type/metabolism , Membrane Glycoproteins/metabolism , Neoplasms/immunology , Animals , CD8-Positive T-Lymphocytes/immunology , Cell Adhesion , Cell Proliferation , Clone Cells , Cytokines/biosynthesis , Cytotoxicity, Immunologic , Gene Editing , Humans , Lymphocyte Activation/immunology , Lymphocyte Function-Associated Antigen-1/metabolism , Lymphocytes, Tumor-Infiltrating/immunology , Melanoma/immunology , Melanoma/pathology , Mice, Inbred C57BL , Neoplasm Metastasis , Neoplasms/pathology , Protein Binding , Talin/metabolism
15.
Biomaterials ; 230: 119626, 2020 02.
Article in English | MEDLINE | ID: mdl-31753473

ABSTRACT

Systemic cytokine therapy is limited by toxicity due to activation of unwanted immune cells in off-target tissues. Injectable nanomaterials that interact with the immune system have potential to offer improved pharmacokinetics and cell specificity compared to systemic cytokine therapy by instead capturing and potentiating endogenous cytokine. Here we demonstrate the use of high aspect ratio polycaprolactone nanowires conjugated to cytokine-binding antibodies that assemble into porous matrices when injected into the subcutaneous space. Nanowires are well tolerated in vivo over several weeks, incite minimal foreign body response and resist clearance. Nanowires conjugated with JES6-1, an anti-interleukin-2 (IL-2) antibody, were designed to capture endogenous IL-2 and selectively activate tissue resident regulatory T cells (Tregs). Together these nanowire-antibody matrices were capable of sequestering endogenous IL-2 in the skin and were successful in rebalancing local immune compartments to a more suppressive, Treg-mediated phenotype in both wild type and transgenic murine autoimmune disease models.


Subject(s)
Autoimmune Diseases , Cytokines , Animals , Antibodies , Autoimmune Diseases/drug therapy , Interleukin-2 , Mice , T-Lymphocytes, Regulatory
16.
JCI Insight ; 4(24)2019 12 19.
Article in English | MEDLINE | ID: mdl-31852848

ABSTRACT

Distinct subsets of Tregs reside in nonlymphoid tissues where they mediate unique functions. To interrogate the biology of tissue Tregs in human health and disease, we phenotypically and functionally compared healthy skin Tregs with those in peripheral blood, inflamed psoriatic skin, and metastatic melanoma. The mitochondrial enzyme, arginase 2 (ARG2), was preferentially expressed in Tregs in healthy skin, increased in Tregs in metastatic melanoma, and reduced in Tregs from psoriatic skin. ARG2 enhanced Treg suppressive capacity in vitro and conferred a selective advantage for accumulation in inflamed tissues in vivo. CRISPR-mediated deletion of this gene in primary human Tregs was sufficient to skew away from a tissue Treg transcriptional signature. Notably, the inhibition of ARG2 increased mTOR signaling, whereas the overexpression of this enzyme suppressed it. Taken together, our results suggest that Tregs express ARG2 in human tissues to both regulate inflammation and enhance their metabolic fitness.


Subject(s)
Arginase/metabolism , Skin/pathology , T-Lymphocytes, Regulatory/metabolism , Adoptive Transfer , Adult , Aged , Aged, 80 and over , Animals , Arginase/genetics , Cells, Cultured , Dendritic Cells , Gene Knockout Techniques , Humans , Keratinocytes , Male , Melanoma/immunology , Melanoma/pathology , Mice , Middle Aged , Primary Cell Culture , Psoriasis/immunology , Psoriasis/pathology , RNA-Seq , Signal Transduction/immunology , Skin/cytology , Skin/immunology , T-Lymphocytes, Regulatory/immunology , TOR Serine-Threonine Kinases/immunology , TOR Serine-Threonine Kinases/metabolism
17.
Cell Host Microbe ; 26(6): 795-809.e5, 2019 12 11.
Article in English | MEDLINE | ID: mdl-31784259

ABSTRACT

The host must develop tolerance to commensal microbes and protective responses to infectious pathogens, yet the mechanisms enabling a privileged relationship with commensals remain largely unknown. Skin colonization by commensal Staphylococcus epidermidis facilitates immune tolerance preferentially in neonates via induction of antigen-specific regulatory T cells (Tregs). Here, we demonstrate that this tolerance is not indiscriminately extended to all bacteria encountered in this early window. Rather, neonatal colonization by Staphylococcus aureus minimally enriches for antigen-specific Tregs and does not prevent skin inflammation upon later-life exposure. S. aureus α-toxin contributes to this response by stimulating myeloid cell production of IL-1ß, which limits S. aureus-specific Tregs. Loss of α-toxin or the IL-1 receptor increases Treg enrichment, whereas topical application of IL-1ß or α-toxin diminishes tolerogenic responses to S. epidermidis. Thus, the preferential activation of a key alarmin pathway facilitates early discrimination of microbial "foe" from "friend," thereby preventing tolerance to a common skin pathogen.


Subject(s)
Bacterial Toxins/immunology , Receptors, Interleukin-1/metabolism , Skin/microbiology , Staphylococcal Infections/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Animals, Newborn , Bacterial Toxins/metabolism , Host Microbial Interactions/immunology , Immune Tolerance , Mice , Receptors, Interleukin-1/immunology , Signal Transduction/immunology , Staphylococcus aureus/immunology , Staphylococcus epidermidis/immunology , Symbiosis/immunology , Virulence/immunology
18.
Sci Immunol ; 4(39)2019 09 06.
Article in English | MEDLINE | ID: mdl-31492709

ABSTRACT

At the center of fibrosing diseases is the aberrant activation of tissue fibroblasts. The cellular and molecular mechanisms of how the immune system augments fibroblast activation have been described; however, little is known about how the immune system controls fibroblast function in tissues. Here, we identify regulatory T cells (Tregs) as important regulators of fibroblast activation in skin. Bulk cell and single-cell analysis of Tregs in murine skin and lungs revealed that Tregs in skin are transcriptionally distinct and skewed toward T helper 2 (TH2) differentiation. When compared with Tregs in lung, skin Tregs preferentially expressed high levels of GATA3, the master TH2 transcription factor. Genes regulated by GATA3 were highly enriched in skin "TH2 Treg" subsets. In functional experiments, Treg depletion resulted in a preferential increase in TH2 cytokine production in skin. Both acute depletion and chronic reduction of Tregs resulted in spontaneous skin fibroblast activation, profibrotic gene expression, and dermal fibrosis, all of which were exacerbated in a bleomycin-induced murine model of skin sclerosis. Lineage-specific deletion of Gata3 in Tregs resulted in an exacerbation of TH2 cytokine secretion that was preferential to skin, resulting in enhanced fibroblast activation and dermal fibrosis. Together, we demonstrate that Tregs play a critical role in regulating fibroblast activation in skin and do so by expressing a unique tissue-restricted transcriptional program that is mediated, at least in part, by GATA3.


Subject(s)
Fibroblasts/immunology , Skin/immunology , T-Lymphocytes, Regulatory/immunology , Animals , Mice , Mice, Inbred C57BL , Mice, Transgenic
19.
Immunity ; 50(3): 655-667.e4, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30893588

ABSTRACT

Restoration of barrier-tissue integrity after injury is dependent on the function of immune cells and stem cells (SCs) residing in the tissue. In response to skin injury, hair-follicle stem cells (HFSCs), normally poised for hair generation, are recruited to the site of injury and differentiate into cells that repair damaged epithelium. We used a SC fate-mapping approach to examine the contribution of regulatory T (Treg) cells to epidermal-barrier repair after injury. Depletion of Treg cells impaired skin-barrier regeneration and was associated with a Th17 inflammatory response and failed HFSC differentiation. In this setting, damaged epithelial cells preferentially expressed the neutrophil chemoattractant CXCL5, and blockade of CXCL5 or neutrophil depletion restored barrier function and SC differentiation after epidermal injury. Thus, Treg-cell regulation of localized inflammation enables HFSC differentiation and, thereby, skin-barrier regeneration, with implications for the maintenance and repair of other barrier tissues.


Subject(s)
Cell Differentiation/physiology , Chemokine CXCL5/metabolism , Epidermis/metabolism , Hair Follicle/metabolism , Interleukin-17/metabolism , Regeneration/physiology , T-Lymphocytes, Regulatory/metabolism , Animals , Epidermal Cells/metabolism , Epithelial Cells/metabolism , Hair/metabolism , Mice , Mice, Inbred C57BL , Stem Cells/metabolism
20.
Sci Immunol ; 3(30)2018 12 21.
Article in English | MEDLINE | ID: mdl-30578350

ABSTRACT

Regulatory T cells (Tregs) are closely related to TH17 cells and use aspects of the TH17-differentiation program for optimal immune regulation. In several chronic inflammatory human diseases, Tregs express IL-17A, suggesting that dysregulation of TH17-associated pathways in Tregs may result in either loss of suppressive function and/or conversion into pathogenic cells. The pathways that regulate the TH17 program in Tregs are poorly understood. We have identified two TNF receptor superfamily (TNFRSF) members, CD27 and OX40, that are preferentially expressed by skin-resident Tregs Both CD27 and OX40 signaling suppressed the expression of TH17-associated genes from Tregs in a cell-intrinsic manner in vitro and in vivo. However, only OX40 played a nonredundant role in promoting Treg accumulation. Tregs that lacked both CD27 and OX40 were defective in controlling skin inflammation and expressed high levels of IL-17A, as well as the master TH17 transcription factor, RORγt. Last, we found that CD27 expression was inversely correlated with Treg IL-17 production in skin of patients with psoriasis and hidradenitis suppurativa. Together, our results suggest that TNFRSF members play both redundant and distinct roles in regulating Treg plasticity in tissues.


Subject(s)
Membrane Glycoproteins/immunology , T-Lymphocytes, Regulatory/immunology , Th17 Cells/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Tumor Necrosis Factors/immunology , Animals , Cell Differentiation/immunology , Membrane Glycoproteins/deficiency , Mice , Mice, Inbred C57BL , Mice, Knockout , OX40 Ligand , Tumor Necrosis Factor Receptor Superfamily, Member 7/deficiency , Tumor Necrosis Factors/deficiency
SELECTION OF CITATIONS
SEARCH DETAIL
...