Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 66.144
Filter
Add more filters

Publication year range
1.
Cell ; 185(25): 4675-4677, 2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36493748

ABSTRACT

Wound healing in adult mammalian tissues generally involves scarring instead of tissue regeneration. A study in this issue of Cell reveals that after injury, reindeer antler skin regenerates by priming regenerative genes in wound fibroblasts instead of forming a scar through an inflammatory gene program.


Subject(s)
Reindeer , Animals , Wound Healing , Cicatrix/pathology , Skin/pathology , Fibroblasts/pathology
2.
Cell ; 185(25): 4717-4736.e25, 2022 Dec 08.
Article in English | MEDLINE | ID: mdl-36493752

ABSTRACT

Adult mammalian skin wounds heal by forming fibrotic scars. We report that full-thickness injuries of reindeer antler skin (velvet) regenerate, whereas back skin forms fibrotic scar. Single-cell multi-omics reveal that uninjured velvet fibroblasts resemble human fetal fibroblasts, whereas back skin fibroblasts express inflammatory mediators mimicking pro-fibrotic adult human and rodent fibroblasts. Consequently, injury elicits site-specific immune responses: back skin fibroblasts amplify myeloid infiltration and maturation during repair, whereas velvet fibroblasts adopt an immunosuppressive phenotype that restricts leukocyte recruitment and hastens immune resolution. Ectopic transplantation of velvet to scar-forming back skin is initially regenerative, but progressively transitions to a fibrotic phenotype akin to the scarless fetal-to-scar-forming transition reported in humans. Skin regeneration is diminished by intensifying, or enhanced by neutralizing, these pathologic fibroblast-immune interactions. Reindeer represent a powerful comparative model for interrogating divergent wound healing outcomes, and our results nominate decoupling of fibroblast-immune interactions as a promising approach to mitigate scar.


Subject(s)
Reindeer , Wound Healing , Adult , Animals , Humans , Cicatrix/pathology , Fibroblasts/pathology , Skin Transplantation , Skin/pathology , Fetus/pathology
3.
Nat Immunol ; 25(7): 1296-1305, 2024 Jul.
Article in English | MEDLINE | ID: mdl-38806708

ABSTRACT

Inflammatory pain results from the heightened sensitivity and reduced threshold of nociceptor sensory neurons due to exposure to inflammatory mediators. However, the cellular and transcriptional diversity of immune cell and sensory neuron types makes it challenging to decipher the immune mechanisms underlying pain. Here we used single-cell transcriptomics to determine the immune gene signatures associated with pain development in three skin inflammatory pain models in mice: zymosan injection, skin incision and ultraviolet burn. We found that macrophage and neutrophil recruitment closely mirrored the kinetics of pain development and identified cell-type-specific transcriptional programs associated with pain and its resolution. Using a comprehensive list of potential interactions mediated by receptors, ligands, ion channels and metabolites to generate injury-specific neuroimmune interactomes, we also uncovered that thrombospondin-1 upregulated by immune cells upon injury inhibited nociceptor sensitization. This study lays the groundwork for identifying the neuroimmune axes that modulate pain in diverse disease contexts.


Subject(s)
Nociceptors , Pain , Animals , Mice , Pain/immunology , Pain/metabolism , Nociceptors/metabolism , Transcriptome , Mice, Inbred C57BL , Inflammation/immunology , Male , Macrophages/immunology , Macrophages/metabolism , Disease Models, Animal , Thrombospondin 1/metabolism , Thrombospondin 1/genetics , Skin/immunology , Skin/metabolism , Skin/pathology , Zymosan , Single-Cell Analysis , Neuroimmunomodulation , Gene Expression Profiling , Neutrophils/immunology , Neutrophils/metabolism
4.
Annu Rev Immunol ; 32: 227-55, 2014.
Article in English | MEDLINE | ID: mdl-24655295

ABSTRACT

The skin is the front line of defense against insult and injury and contains many epidermal and immune elements that comprise the skin-associated lymphoid tissue (SALT). The reaction of these components to injury allows an effective cutaneous response to restore homeostasis. Psoriasis vulgaris is the best-understood and most accessible human disease that is mediated by T cells and dendritic cells. Inflammatory myeloid dendritic cells release IL-23 and IL-12 to activate IL-17-producing T cells, Th1 cells, and Th22 cells to produce abundant psoriatic cytokines IL-17, IFN-γ, TNF, and IL-22. These cytokines mediate effects on keratinocytes to amplify psoriatic inflammation. Therapeutic studies with anticytokine antibodies have shown the importance of the key cytokines IL-23, TNF, and IL-17 in this process. We discuss the genetic background of psoriasis and its relationship to immune function, specifically genetic mutations, key PSORS loci, single nucleotide polymorphisms, and the skin transcriptome. The association between comorbidities and psoriasis is reviewed by correlating the skin transcriptome and serum proteins. Psoriasis-related cytokine-response pathways are considered in the context of the transcriptome of different mouse models. This approach offers a model for other inflammatory skin and autoimmune diseases.


Subject(s)
Psoriasis/immunology , Animals , Comorbidity , Disease Models, Animal , Genetic Predisposition to Disease , Humans , Mice , Psoriasis/diagnosis , Psoriasis/genetics , Skin/immunology , Skin/pathology , Skin Physiological Phenomena/immunology
5.
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
6.
Nat Immunol ; 23(11): 1577-1587, 2022 Nov.
Article in English | MEDLINE | ID: mdl-36271146

ABSTRACT

Aberrant RNA splicing in keratinocytes drives inflammatory skin disorders. In the present study, we found that the RNA helicase DDX5 was downregulated in keratinocytes from the inflammatory skin lesions in patients with atopic dermatitis and psoriasis, and that mice with keratinocyte-specific deletion of Ddx5 (Ddx5∆KC) were more susceptible to cutaneous inflammation. Inhibition of DDX5 expression in keratinocytes was induced by the cytokine interleukin (IL)-17D through activation of the CD93-p38 MAPK-AKT-SMAD2/3 signaling pathway and led to pre-messenger RNA splicing events that favored the production of membrane-bound, intact IL-36 receptor (IL-36R) at the expense of soluble IL-36R (sIL-36R) and to the selective amplification of IL-36R-mediated inflammatory responses and cutaneous inflammation. Restoration of sIL-36R in Ddx5∆KC mice with experimental atopic dermatitis or psoriasis suppressed skin inflammation and alleviated the disease phenotypes. These findings indicate that IL-17D modulation of DDX5 expression controls inflammation in keratinocytes during inflammatory skin diseases.


Subject(s)
Dermatitis, Atopic , Interleukin-27 , Psoriasis , Mice , Animals , Interleukin-27/metabolism , Dermatitis, Atopic/genetics , Dermatitis, Atopic/pathology , Keratinocytes/metabolism , Skin/pathology , Psoriasis/genetics , Psoriasis/pathology , Inflammation/metabolism
7.
Nat Immunol ; 22(7): 839-850, 2021 07.
Article in English | MEDLINE | ID: mdl-34168371

ABSTRACT

Granulomas are complex cellular structures composed predominantly of macrophages and lymphocytes that function to contain and kill invading pathogens. Here, we investigated the single-cell phenotypes associated with antimicrobial responses in human leprosy granulomas by applying single-cell and spatial sequencing to leprosy biopsy specimens. We focused on reversal reactions (RRs), a dynamic process whereby some patients with disseminated lepromatous leprosy (L-lep) transition toward self-limiting tuberculoid leprosy (T-lep), mounting effective antimicrobial responses. We identified a set of genes encoding proteins involved in antimicrobial responses that are differentially expressed in RR versus L-lep lesions and regulated by interferon-γ and interleukin-1ß. By integrating the spatial coordinates of the key cell types and antimicrobial gene expression in RR and T-lep lesions, we constructed a map revealing the organized architecture of granulomas depicting compositional and functional layers by which macrophages, T cells, keratinocytes and fibroblasts can each contribute to the antimicrobial response.


Subject(s)
Leprosy, Lepromatous/immunology , Leprosy, Tuberculoid/immunology , Mycobacterium leprae/immunology , Skin/immunology , Adolescent , Adult , Aged , Female , Fibroblasts/immunology , Fibroblasts/microbiology , Fibroblasts/pathology , Gene Expression Profiling , Host-Pathogen Interactions , Humans , Keratinocytes/immunology , Keratinocytes/microbiology , Keratinocytes/pathology , Leprosy, Lepromatous/genetics , Leprosy, Lepromatous/microbiology , Leprosy, Lepromatous/pathology , Leprosy, Tuberculoid/genetics , Leprosy, Tuberculoid/microbiology , Leprosy, Tuberculoid/pathology , Macrophages/immunology , Macrophages/microbiology , Macrophages/pathology , Male , Middle Aged , Mycobacterium leprae/pathogenicity , RNA-Seq , Single-Cell Analysis , Skin/microbiology , Skin/pathology , T-Lymphocytes/immunology , T-Lymphocytes/microbiology , T-Lymphocytes/pathology , Transcriptome
8.
Immunity ; 57(7): 1451-1453, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38986439

ABSTRACT

Skin inflammation is potentiated by coordinated epithelial and immune cell metabolism. In this issue of Immunity, Subudhi and Konieczny et al. delineate how HIF1α regulates epithelial cell glycolysis during psoriasis. In turn, lactate is a byproduct that augments type 17 γδ T cell responses to sustain inflammatory skin disease.


Subject(s)
Epithelial Cells , Glycolysis , Hypoxia-Inducible Factor 1, alpha Subunit , Psoriasis , Skin , Animals , Humans , Chronic Disease , Epithelial Cells/metabolism , Epithelial Cells/immunology , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Inflammation/immunology , Inflammation/metabolism , Psoriasis/immunology , Psoriasis/metabolism , Skin/immunology , Skin/pathology , Skin/metabolism
9.
Immunity ; 57(5): 1071-1086.e7, 2024 May 14.
Article in English | MEDLINE | ID: mdl-38677291

ABSTRACT

Following tissue damage, epithelial stem cells (SCs) are mobilized to enter the wound, where they confront harsh inflammatory environments that can impede their ability to repair the injury. Here, we investigated the mechanisms that protect skin SCs within this inflammatory environment. Characterization of gene expression profiles of hair follicle SCs (HFSCs) that migrated into the wound site revealed activation of an immune-modulatory program, including expression of CD80, major histocompatibility complex class II (MHCII), and CXC motif chemokine ligand 5 (CXCL5). Deletion of CD80 in HFSCs impaired re-epithelialization, reduced accumulation of peripherally generated Treg (pTreg) cells, and increased infiltration of neutrophils in wounded skin. Importantly, similar wound healing defects were also observed in mice lacking pTreg cells. Our findings suggest that upon skin injury, HFSCs establish a temporary protective network by promoting local expansion of Treg cells, thereby enabling re-epithelialization while still kindling inflammation outside this niche until the barrier is restored.


Subject(s)
B7-1 Antigen , Hair Follicle , Inflammation , Skin , Stem Cells , T-Lymphocytes, Regulatory , Wound Healing , Animals , T-Lymphocytes, Regulatory/immunology , Mice , Wound Healing/immunology , Skin/immunology , Skin/injuries , Skin/pathology , Stem Cells/immunology , Stem Cells/metabolism , Inflammation/immunology , Hair Follicle/immunology , B7-1 Antigen/metabolism , Mice, Inbred C57BL , Mice, Knockout , Re-Epithelialization/immunology , Cell Movement/immunology , Cell Proliferation
10.
Immunity ; 57(7): 1665-1680.e7, 2024 Jul 09.
Article in English | MEDLINE | ID: mdl-38772365

ABSTRACT

Inflammatory epithelial diseases are spurred by the concomitant dysregulation of immune and epithelial cells. How these two dysregulated cellular compartments simultaneously sustain their heightened metabolic demands is unclear. Single-cell and spatial transcriptomics (ST), along with immunofluorescence, revealed that hypoxia-inducible factor 1α (HIF1α), downstream of IL-17 signaling, drove psoriatic epithelial remodeling. Blocking HIF1α in human psoriatic lesions ex vivo impaired glycolysis and phenocopied anti-IL-17 therapy. In a murine model of skin inflammation, epidermal-specific loss of HIF1α or its target gene, glucose transporter 1, ameliorated epidermal, immune, vascular, and neuronal pathology. Mechanistically, glycolysis autonomously fueled epithelial pathology and enhanced lactate production, which augmented the γδ T17 cell response. RORγt-driven genetic deletion or pharmacological inhibition of either lactate-producing enzymes or lactate transporters attenuated epithelial pathology and IL-17A expression in vivo. Our findings identify a metabolic hierarchy between epithelial and immune compartments and the consequent coordination of metabolic processes that sustain inflammatory disease.


Subject(s)
Glycolysis , Hypoxia-Inducible Factor 1, alpha Subunit , Interleukin-17 , Animals , Humans , Interleukin-17/metabolism , Interleukin-17/immunology , Mice , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Skin/immunology , Skin/pathology , Skin/metabolism , Th17 Cells/immunology , Th17 Cells/metabolism , Glucose Transporter Type 1/metabolism , Glucose Transporter Type 1/genetics , Psoriasis/immunology , Psoriasis/metabolism , Epithelium/immunology , Epithelium/metabolism , Mice, Knockout , Signal Transduction/immunology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Nuclear Receptor Subfamily 1, Group F, Member 3/genetics , Disease Models, Animal , Lactic Acid/metabolism , Chronic Disease , Inflammation/immunology , Mice, Inbred C57BL
11.
Cell ; 173(5): 1083-1097.e22, 2018 05 17.
Article in English | MEDLINE | ID: mdl-29754819

ABSTRACT

The nervous system, the immune system, and microbial pathogens interact closely at barrier tissues. Here, we find that a bacterial pathogen, Streptococcus pyogenes, hijacks pain and neuronal regulation of the immune response to promote bacterial survival. Necrotizing fasciitis is a life-threatening soft tissue infection in which "pain is out of proportion" to early physical manifestations. We find that S. pyogenes, the leading cause of necrotizing fasciitis, secretes streptolysin S (SLS) to directly activate nociceptor neurons and produce pain during infection. Nociceptors, in turn, release the neuropeptide calcitonin gene-related peptide (CGRP) into infected tissues, which inhibits the recruitment of neutrophils and opsonophagocytic killing of S. pyogenes. Botulinum neurotoxin A and CGRP antagonism block neuron-mediated suppression of host defense, thereby preventing and treating S. pyogenes necrotizing infection. We conclude that targeting the peripheral nervous system and blocking neuro-immune communication is a promising strategy to treat highly invasive bacterial infections. VIDEO ABSTRACT.


Subject(s)
Neurons/metabolism , Neutrophils/metabolism , Streptococcal Infections/pathology , Streptococcus pyogenes/pathogenicity , Animals , Bacterial Proteins/immunology , Bacterial Proteins/metabolism , Botulinum Toxins, Type A/administration & dosage , Calcitonin Gene-Related Peptide/metabolism , Caspase 1/deficiency , Caspase 1/genetics , Diterpenes/pharmacology , Fasciitis, Necrotizing/etiology , Fasciitis, Necrotizing/pathology , Fasciitis, Necrotizing/veterinary , Female , Humans , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Neurons/cytology , Neurons/drug effects , Neutrophils/immunology , Pain/etiology , Signal Transduction , Skin/metabolism , Skin/pathology , Streptococcal Infections/complications , Streptococcal Infections/veterinary , Streptococcus pyogenes/metabolism , Streptolysins/immunology , Streptolysins/metabolism , TRPV Cation Channels/deficiency , TRPV Cation Channels/genetics
12.
Nat Immunol ; 21(9): 1034-1045, 2020 09.
Article in English | MEDLINE | ID: mdl-32661363

ABSTRACT

Skin wounds heal by coordinated induction of inflammation and tissue repair, but the initiating events are poorly defined. Here we uncover a fundamental role of commensal skin microbiota in this process and show that it is mediated by the recruitment and the activation of type I interferon (IFN)-producing plasmacytoid DC (pDC). Commensal bacteria colonizing skin wounds trigger activation of neutrophils to express the chemokine CXCL10, which recruits pDC and acts as an antimicrobial protein to kill exposed microbiota, leading to the formation of CXCL10-bacterial DNA complexes. These complexes and not complexes with host-derived DNA activate pDC to produce type I IFNs, which accelerate wound closure by triggering skin inflammation and early T cell-independent wound repair responses, mediated by macrophages and fibroblasts that produce major growth factors required for healing. These findings identify a key function of commensal microbiota in driving a central innate wound healing response of the skin.


Subject(s)
Dendritic Cells/immunology , Fibroblasts/immunology , Macrophages/immunology , Microbiota/immunology , Neutrophils/immunology , Skin/immunology , Animals , Cells, Cultured , Chemokine CXCL10/metabolism , Humans , Immunity, Innate , Inflammation , Interferon Type I/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Skin/pathology , Symbiosis , Wound Healing
13.
Nat Immunol ; 20(7): 915-927, 2019 07.
Article in English | MEDLINE | ID: mdl-31110316

ABSTRACT

The molecular and cellular processes that lead to renal damage and to the heterogeneity of lupus nephritis (LN) are not well understood. We applied single-cell RNA sequencing (scRNA-seq) to renal biopsies from patients with LN and evaluated skin biopsies as a potential source of diagnostic and prognostic markers of renal disease. Type I interferon (IFN)-response signatures in tubular cells and keratinocytes distinguished patients with LN from healthy control subjects. Moreover, a high IFN-response signature and fibrotic signature in tubular cells were each associated with failure to respond to treatment. Analysis of tubular cells from patients with proliferative, membranous and mixed LN indicated pathways relevant to inflammation and fibrosis, which offer insight into their histologic differences. In summary, we applied scRNA-seq to LN to deconstruct its heterogeneity and identify novel targets for personalized approaches to therapy.


Subject(s)
Gene Expression Profiling , Interferon Type I/metabolism , Keratinocytes/metabolism , Kidney Tubules/cytology , Kidney Tubules/metabolism , Lupus Nephritis/genetics , Lupus Nephritis/metabolism , Transcriptome , Biopsy , Cell Lineage/genetics , Computational Biology/methods , Extracellular Matrix Proteins/genetics , Extracellular Matrix Proteins/metabolism , Fibrosis , Gene Expression Profiling/methods , Humans , Lupus Nephritis/pathology , Protein Binding , Signal Transduction , Single-Cell Analysis , Skin/immunology , Skin/metabolism , Skin/pathology
14.
Nat Immunol ; 20(8): 992-1003, 2019 08.
Article in English | MEDLINE | ID: mdl-31263279

ABSTRACT

Here we identify a group 2 innate lymphoid cell (ILC2) subpopulation that can convert into interleukin-17 (IL-17)-producing NKp44- ILC3-like cells. c-Kit and CCR6 define this ILC2 subpopulation that exhibits ILC3 features, including RORγt, enabling the conversion into IL-17-producing cells in response to IL-1ß and IL-23. We also report a role for transforming growth factor-ß in promoting the conversion of c-Kit- ILC2s into RORγt-expressing cells by inducing the upregulation of IL23R, CCR6 and KIT messenger RNA in these cells. This switch was dependent on RORγt and the downregulation of GATA-3. IL-4 was able to reverse this event, supporting a role for this cytokine in maintaining ILC2 identity. Notably, this plasticity has physiological relevance because a subset of RORγt+ ILC2s express the skin-homing receptor CCR10, and the frequencies of IL-17-producing ILC3s are increased at the expense of ILC2s within the lesional skin of patients with psoriasis.


Subject(s)
Interleukin-17/immunology , Lymphocytes/immunology , Psoriasis/pathology , Skin/pathology , Cells, Cultured , Humans , Interleukin-1beta/immunology , Interleukin-23 Subunit p19/immunology , Interleukin-4/immunology , Lymphocytes/cytology , Nuclear Receptor Subfamily 1, Group F, Member 3/metabolism , Proto-Oncogene Proteins c-kit/metabolism , Psoriasis/immunology , Receptors, CCR10/metabolism , Skin/immunology , Transforming Growth Factor beta/metabolism
15.
Immunity ; 54(3): 468-483.e5, 2021 03 09.
Article in English | MEDLINE | ID: mdl-33484643

ABSTRACT

Tissue resident mast cells (MCs) rapidly initiate neutrophil infiltration upon inflammatory insult, yet the molecular mechanism is still unknown. Here, we demonstrated that MC-derived tumor necrosis factor (TNF) was crucial for neutrophil extravasation to sites of contact hypersensitivity-induced skin inflammation by promoting intraluminal crawling. MC-derived TNF directly primed circulating neutrophils via TNF receptor-1 (TNFR1) while being dispensable for endothelial cell activation. The MC-derived TNF was infused into the bloodstream by directional degranulation of perivascular MCs that were part of the vascular unit with access to the vessel lumen. Consistently, intravenous administration of MC granules boosted neutrophil extravasation. Pronounced and rapid intravascular MC degranulation was also observed upon IgE crosslinking or LPs challenge indicating a universal MC potential. Consequently, the directional MC degranulation of pro-inflammatory mediators into the bloodstream may represent an important target for therapeutic approaches aimed at dampening cytokine storm syndromes or shock symptoms, or intentionally pushing immune defense.


Subject(s)
Blood Vessels/immunology , Dermatitis, Contact/immunology , Inflammation/immunology , Mast Cells/immunology , Neutrophils/immunology , Skin/pathology , Tumor Necrosis Factor-alpha/metabolism , Animals , Blood Circulation , Cell Degranulation , Cells, Cultured , Immune System Diseases , Leukocyte Disorders , Mice , Mice, Inbred C57BL , Mice, Knockout , Neutrophil Activation , Receptors, Tumor Necrosis Factor, Type I/metabolism , Secretory Vesicles/metabolism , Tumor Necrosis Factor-alpha/genetics
16.
Immunity ; 54(7): 1374-1376, 2021 07 13.
Article in English | MEDLINE | ID: mdl-34260885

ABSTRACT

In a recent issue of Nature, Hoeffel et al. describe a novel pathway of sterile tissue repair utilizing a mouse model of sunburn. This wound healing pathway is coordinated by sensory neuron-derived TAFA4 that induces IL-10 production from Tim4+ dermal macrophages to prevent sustained inflammation and the emergence of tissue fibrosis.


Subject(s)
Sensory Receptor Cells/pathology , Sunburn/pathology , Wound Healing/physiology , Animals , Cytokines/metabolism , Disease Models, Animal , Fibrosis/metabolism , Fibrosis/pathology , Inflammation/metabolism , Inflammation/pathology , Interleukin-10/metabolism , Macrophages/metabolism , Macrophages/pathology , Mice , Signal Transduction/physiology , Skin/metabolism , Skin/pathology , Sunburn/metabolism
17.
Immunity ; 54(10): 2321-2337.e10, 2021 10 12.
Article in English | MEDLINE | ID: mdl-34582748

ABSTRACT

Hair follicles (HFs) function as hubs for stem cells, immune cells, and commensal microbes, which must be tightly regulated during homeostasis and transient inflammation. Here we found that transmembrane endopeptidase ADAM10 expression in upper HFs was crucial for regulating the skin microbiota and protecting HFs and their stem cell niche from inflammatory destruction. Ablation of the ADAM10-Notch signaling axis impaired the innate epithelial barrier and enabled Corynebacterium species to predominate the microbiome. Dysbiosis triggered group 2 innate lymphoid cell-mediated inflammation in an interleukin-7 (IL-7) receptor-, S1P receptor 1-, and CCR6-dependent manner, leading to pyroptotic cell death of HFs and irreversible alopecia. Double-stranded RNA-induced ablation models indicated that the ADAM10-Notch signaling axis bolsters epithelial innate immunity by promoting ß-defensin-6 expression downstream of type I interferon responses. Thus, ADAM10-Notch signaling axis-mediated regulation of host-microbial symbiosis crucially protects HFs from inflammatory destruction, which has implications for strategies to sustain tissue integrity during chronic inflammation.


Subject(s)
ADAM10 Protein/immunology , Amyloid Precursor Protein Secretases/immunology , Dysbiosis/immunology , Hair Follicle/pathology , Lymphocytes/immunology , Membrane Proteins/immunology , Receptors, Notch/immunology , Skin/microbiology , Alopecia/immunology , Alopecia/pathology , Animals , Corynebacterium , Dysbiosis/pathology , Female , Hair Follicle/immunology , Immunity, Innate , Inflammation/immunology , Inflammation/metabolism , Inflammation/pathology , Mice , Signal Transduction/immunology , Skin/immunology , Skin/pathology
18.
Nat Immunol ; 18(2): 152-160, 2017 02.
Article in English | MEDLINE | ID: mdl-27992404

ABSTRACT

Autoimmune diseases affect 7.5% of the US population, and they are among the leading causes of death and disability. A notable feature of many autoimmune diseases is their greater prevalence in females than in males, but the underlying mechanisms of this have remained unclear. Through the use of high-resolution global transcriptome analyses, we demonstrated a female-biased molecular signature associated with susceptibility to autoimmune disease and linked this to extensive sex-dependent co-expression networks. This signature was independent of biological age and sex-hormone regulation and was regulated by the transcription factor VGLL3, which also had a strong female-biased expression. On a genome-wide level, VGLL3-regulated genes had a strong association with multiple autoimmune diseases, including lupus, scleroderma and Sjögren's syndrome, and had a prominent transcriptomic overlap with inflammatory processes in cutaneous lupus. These results identified a VGLL3-regulated network as a previously unknown inflammatory pathway that promotes female-biased autoimmunity. They demonstrate the importance of studying immunological processes in females and males separately and suggest new avenues for therapeutic development.


Subject(s)
Gene Regulatory Networks , Keratinocytes/physiology , Lupus Erythematosus, Cutaneous/genetics , Scleroderma, Systemic/genetics , Sex Factors , Sjogren's Syndrome/genetics , Skin/pathology , Transcription Factors/metabolism , Adult , Aged , Aged, 80 and over , Cells, Cultured , Female , Gene Expression Profiling , Genetic Association Studies , Genome-Wide Association Study , Humans , Male , Middle Aged , Quantitative Trait Loci , Transcription Factors/genetics , Transcriptome , Young Adult
19.
Immunity ; 53(4): 878-894.e7, 2020 10 13.
Article in English | MEDLINE | ID: mdl-33053333

ABSTRACT

High-throughput single-cell RNA-sequencing (scRNA-seq) methodologies enable characterization of complex biological samples by increasing the number of cells that can be profiled contemporaneously. Nevertheless, these approaches recover less information per cell than low-throughput strategies. To accurately report the expression of key phenotypic features of cells, scRNA-seq platforms are needed that are both high fidelity and high throughput. To address this need, we created Seq-Well S3 ("Second-Strand Synthesis"), a massively parallel scRNA-seq protocol that uses a randomly primed second-strand synthesis to recover complementary DNA (cDNA) molecules that were successfully reverse transcribed but to which a second oligonucleotide handle, necessary for subsequent whole transcriptome amplification, was not appended due to inefficient template switching. Seq-Well S3 increased the efficiency of transcript capture and gene detection compared with that of previous iterations by up to 10- and 5-fold, respectively. We used Seq-Well S3 to chart the transcriptional landscape of five human inflammatory skin diseases, thus providing a resource for the further study of human skin inflammation.


Subject(s)
High-Throughput Nucleotide Sequencing/methods , Inflammation/genetics , RNA, Small Cytoplasmic/genetics , Skin/pathology , Animals , Cell Line , DNA, Complementary/genetics , HEK293 Cells , Humans , Mice , NIH 3T3 Cells , Sequence Analysis, RNA/methods , Single-Cell Analysis/methods , Transcription, Genetic/genetics , Transcriptome/genetics
20.
Immunity ; 53(1): 204-216.e10, 2020 07 14.
Article in English | MEDLINE | ID: mdl-32553276

ABSTRACT

Psoriasis is a chronic inflammatory disease whose etiology is multifactorial. The contributions of cellular metabolism to psoriasis are unclear. Here, we report that interleukin-17 (IL-17) downregulated Protein Phosphatase 6 (PP6) in psoriatic keratinocytes, causing phosphorylation and activation of the transcription factor C/EBP-ß and subsequent generation of arginase-1. Mice lacking Pp6 in keratinocytes were predisposed to psoriasis-like skin inflammation. Accumulation of arginase-1 in Pp6-deficient keratinocytes drove polyamine production from the urea cycle. Polyamines protected self-RNA released by psoriatic keratinocytes from degradation and facilitated the endocytosis of self-RNA by myeloid dendritic cells to promote toll-like receptor-7 (TLR7)-dependent RNA sensing and IL-6 production. An arginase inhibitor improved skin inflammation in murine and non-human primate models of psoriasis. Our findings suggest that urea cycle hyperreactivity and excessive polyamine generation in psoriatic keratinocytes promote self-RNA sensation and PP6 deregulation in keratinocytes is a pivotal event that amplifies the inflammatory circuits in psoriasis.


Subject(s)
Dendritic Cells/immunology , Keratinocytes/metabolism , Phosphoprotein Phosphatases/deficiency , Polyamines/metabolism , Psoriasis/pathology , RNA/immunology , 3T3 Cells , Animals , Arginase/antagonists & inhibitors , Arginase/metabolism , Arginine/metabolism , Autoantigens/immunology , CCAAT-Enhancer-Binding Protein-beta/metabolism , Cell Line , Disease Models, Animal , HEK293 Cells , HaCaT Cells , Humans , Interleukin-17/metabolism , Macaca fascicularis , Membrane Glycoproteins/immunology , Mice , Mice, Inbred C57BL , Phosphoprotein Phosphatases/genetics , Phosphorylation , Skin/pathology , Toll-Like Receptor 7/immunology
SELECTION OF CITATIONS
SEARCH DETAIL