Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 3.215
Filter
1.
Front Immunol ; 15: 1378359, 2024.
Article in English | MEDLINE | ID: mdl-38779662

ABSTRACT

Skin tissue-resident memory T (Trm) cells are produced by antigenic stimulation and remain in the skin for a long time without entering the peripheral circulation. In the healthy state Trm cells can play a patrolling and surveillance role, but in the disease state Trm cells differentiate into various phenotypes associated with different diseases, exhibit different localizations, and consequently have local protective or pathogenic roles, such as disease recurrence in vitiligo and maintenance of immune homeostasis in melanoma. The most common surface marker of Trm cells is CD69/CD103. However, the plasticity of tissue-resident memory T cells after colonization remains somewhat uncertain. This ambiguity is largely due to the variation in the functionality and ultimate destination of Trm cells produced from memory cells differentiated from diverse precursors. Notably, the presence of Trm cells is not stationary across numerous non-lymphoid tissues, most notably in the skin. These cells may reenter the blood and distant tissue sites during the recall response, revealing the recycling and migration potential of the Trm cell progeny. This review focuses on the origin and function of skin Trm cells, and provides new insights into the role of skin Trm cells in the treatment of autoimmune skin diseases, infectious skin diseases, and tumors.


Subject(s)
Cell Plasticity , Homeostasis , Immunologic Memory , Memory T Cells , Skin Diseases , Skin , Humans , Homeostasis/immunology , Memory T Cells/immunology , Memory T Cells/metabolism , Skin/immunology , Skin/pathology , Cell Plasticity/immunology , Animals , Skin Diseases/immunology , Antigens, CD/metabolism , Antigens, CD/immunology
2.
Front Immunol ; 15: 1343987, 2024.
Article in English | MEDLINE | ID: mdl-38690268

ABSTRACT

Autophagy is a cellular process that functions to maintain intracellular homeostasis via the degradation and recycling of defective organelles or damaged proteins. This dynamic mechanism participates in various biological processes, such as the regulation of cellular differentiation, proliferation, survival, and the modulation of inflammation and immune responses. Recent evidence has demonstrated the involvement of polymorphisms in autophagy-related genes in various skin autoimmune diseases. In addition, autophagy, along with autophagy-related proteins, also contributes to homeostasis maintenance and immune regulation in the skin, which is associated with skin autoimmune disorders. This review aims to provide an overview of the multifaceted role of autophagy in skin autoimmune diseases and shed light on the potential of autophagy-targeting therapeutic strategies in dermatology.


Subject(s)
Autoimmune Diseases , Autophagy , Skin Diseases , Humans , Autophagy/immunology , Autoimmune Diseases/immunology , Skin Diseases/immunology , Animals , Skin/immunology , Skin/pathology , Skin/metabolism , Homeostasis/immunology
3.
Acta Derm Venereol ; 104: adv40009, 2024 May 02.
Article in English | MEDLINE | ID: mdl-38698654

ABSTRACT

Immunocompromised individuals, primarily attributable to using immunosuppressants, face heightened COVID-19 risks. Despite the proven efficacy of COVID-19 vaccines, their impact on patients with immune-mediated dermatological diseases remains unclear. This study aims to thoroughly examine vaccine immunogenicity, effectiveness, and safety in immune-mediated dermatological disease patients. Clinical studies in adults that compared vaccinated immune-mediated dermatological disease patients with vaccinated healthy controls or unvaccinated immune-mediated dermatological disease patients in terms of vaccine immunogenicity, COVID-19 infection, adverse events, or exacerbation of immune-mediated dermatological diseases were searched via electronic databases. Seventeen studies (1,348,690 participants) were included. Seroconversion rates between immune-mediated dermatological disease patients and healthy controls were not different. However, among individuals aged ≤55 years, immune-mediated dermatological disease patients had lower mean anti-SARS-CoV-2 IgG levels. Immunosuppressed immune-mediated dermatological disease patients also had lower titres and were less likely to achieve T-cell response. In terms of safety, the risk of adverse events was higher in atopic dermatitis patients, but those with psoriasis had a reduced risk. Additionally, immunosuppressed patients had fewer adverse events. Vaccinated immune-mediated dermatological disease patients had a lower risk of COVID-19 infection than unvaccinated patients but a higher risk than healthy controls; however, disease exacerbation may be induced. In conclusion, immune-mediated dermatological diseases showed a reduced vaccine response in our meta-analysis, yet vaccination remained effective against COVID-19 infection and well tolerated.


Subject(s)
COVID-19 Vaccines , COVID-19 , Immunocompromised Host , Immunogenicity, Vaccine , Skin Diseases , Humans , COVID-19 Vaccines/immunology , COVID-19 Vaccines/adverse effects , COVID-19/prevention & control , COVID-19/immunology , Skin Diseases/immunology , SARS-CoV-2/immunology , Vaccine Efficacy , Middle Aged , Adult
4.
BMC Immunol ; 25(1): 30, 2024 May 11.
Article in English | MEDLINE | ID: mdl-38734636

ABSTRACT

BACKGROUND: Immune-mediated necrotizing myopathy (IMNM) is an idiopathic inflammatory myopathy (IIM). Though patients with IMNM were not considered to show skin rash, several reports have showed atypical skin conditions in patients with anti-3-hydroxy-3-methylglutaryl-coenzyme A reductase (HMGCR) antibody-positive IMNM (HMGCR-IMNM). The incidence and phenotype of skin conditions in patients with HMGCR-IMNM are not fully known. RESULTS: Among the 100 IIM patients diagnosed from April 2015 through August 2022, 34 (34%) presented some form of skin condition, with 27 having typical skin rashes; this included 13 patients with dermatomyositis (DM), 8 with anti-synthetase syndrome (ASS), and 6 with IMNM. Meanwhile, 8 of 19 patients with HMGCR-IMNM (42%) presented atypical skin lesions, but no patients with other IIMs did (p < 0.001). Skin eruption with ash-like scales was observed in four HMGCR-IMNM patients, and non-scaly red patches and lumps in the other four patients; accordingly, their skin manifestations were considered as other dermal diseases except for IIM. However, skin and muscle biopsies revealed the atypical skin conditions of patients with HMGCR-IMNM to have the same pathological background, formed by Bcl-2-positive lymphocyte infiltrations. CONCLUSIONS: HMGCR-IMNM patients frequently have atypical skin conditions of the neck and back. Skin biopsy specimens from these lesions showed the same Bcl-2-positive lymphocytic infiltrations as muscle biopsy specimens regardless of the different gross dermal findings. Thus, such atypical skin conditions may be suggestive for HMGCR-IMNM.


Subject(s)
Autoantibodies , Hydroxymethylglutaryl CoA Reductases , Myositis , Skin , Humans , Hydroxymethylglutaryl CoA Reductases/immunology , Female , Male , Middle Aged , Autoantibodies/immunology , Autoantibodies/blood , Adult , Skin/pathology , Skin/immunology , Myositis/immunology , Myositis/diagnosis , Aged , Skin Diseases/immunology , Skin Diseases/etiology , Muscular Diseases/immunology , Muscular Diseases/diagnosis , Biopsy
5.
Skin Res Technol ; 30(5): e13718, 2024 May.
Article in English | MEDLINE | ID: mdl-38700458

ABSTRACT

INTRODUCTION: Due to the increasing prevalence of immune-mediated diseases such as psoriasis, lichen planus, rheumatoid arthritis and inflammatory bowel disease, dermatologists have turned to new biologic drugs known as DMARDs (disease-modifying anti-rheumatic drugs) in recent years. AREAS COVERED: In this study, we evaluate the immune-mediated dermatological side effects of DMARDS by reviewing and analyzing previous peer-reviewed research on the effects of TNF-α inhibitors in the treatment of skin diseases, as well as adverse effects of these drugs and some of the main causes of these effects. EXPERT OPINION: DMARDs are very effective in improving control of the above diseases. TNF-α inhibitors are an important group of DMARDs that are widely used. The paradoxical adverse events (PAEs) associated with the use of TNF-α inhibitors are divided into three categories: true paradoxical, borderline paradoxical, and non-paradoxical. True PAEs include conditions for which TNF-α inhibitors are approved for treatment. Borderline PAEs are considered to occur with this class of drugs for which there is no definite approval but for which there is sufficient evidence. Although these events are rare, early recognition of the accused drug and appropriate decision-making may prevent progression of complications and irreversible side effects.


Subject(s)
Antirheumatic Agents , Tumor Necrosis Factor-alpha , Humans , Tumor Necrosis Factor-alpha/antagonists & inhibitors , Antirheumatic Agents/adverse effects , Antirheumatic Agents/therapeutic use , Skin Diseases/chemically induced , Skin Diseases/immunology , Drug Eruptions/etiology , Drug Eruptions/immunology
6.
Exp Dermatol ; 33(5): e15083, 2024 May.
Article in English | MEDLINE | ID: mdl-38794808

ABSTRACT

Interferons (IFNs) are signalling proteins primarily involved in initiating innate immune responses against pathogens and promoting the maturation of immune cells. Interferon Regulatory Factor 7 (IRF7) plays a pivotal role in the IFNs signalling pathway. The activation process of IRF7 is incited by exogenous or abnormal nucleic acids, which is followed by the identification via pattern recognition receptors (PRRs) and the ensuing signalling cascades. Upon activation, IRF7 modulates the expression of both IFNs and inflammatory gene regulation. As a multifunctional transcription factor, IRF7 is mainly expressed in immune cells, yet its presence is also detected in keratinocytes, fibroblasts, and various dermal cell types. In these cells, IRF7 is critical for skin immunity, inflammation, and fibrosis. IRF7 dysregulation may lead to autoimmune and inflammatory skin conditions, including systemic scleroderma (SSc), systemic lupus erythematosus (SLE), Atopic dermatitis (AD) and Psoriasis. This comprehensive review aims to extensively elucidate the role of IRF7 and its signalling pathways in immune cells and keratinocytes, highlighting its significance in skin-related and connective tissue diseases.


Subject(s)
Connective Tissue Diseases , Interferon Regulatory Factor-7 , Keratinocytes , Signal Transduction , Skin Diseases , Humans , Interferon Regulatory Factor-7/metabolism , Interferon Regulatory Factor-7/genetics , Skin Diseases/immunology , Skin Diseases/metabolism , Keratinocytes/metabolism , Keratinocytes/immunology , Connective Tissue Diseases/metabolism , Connective Tissue Diseases/immunology , Psoriasis/immunology , Psoriasis/metabolism , Animals , Skin/metabolism , Skin/immunology , Scleroderma, Systemic/metabolism , Scleroderma, Systemic/immunology , Scleroderma, Systemic/genetics , Immunity, Innate
7.
J Invest Dermatol ; 144(6): 1208-1216, 2024 Jun.
Article in English | MEDLINE | ID: mdl-38678465

ABSTRACT

IL-17 is widely recognized for its roles in host defense and inflammatory disorders. However, it has become clear that IL-17 is also an essential regulator of barrier tissue physiology. Steady-state microbe sensing at the skin surface induces low-level IL-17 expression that enhances epithelial integrity and resists pathogens without causing overt inflammation. Recent reports describe novel protective roles for IL-17 in wound healing and counteracting physiologic stress; however, chronic amplification of these beneficial responses contributes to skin pathologies as diverse as fibrosis, cancer, and autoinflammation. In this paper, we discuss the context-specific roles of IL-17 in skin health and disease and therapeutic opportunities.


Subject(s)
Homeostasis , Interleukin-17 , Skin , Humans , Interleukin-17/metabolism , Interleukin-17/immunology , Homeostasis/immunology , Skin/immunology , Skin/metabolism , Animals , Wound Healing/immunology , Wound Healing/physiology , Skin Diseases/immunology
9.
Medicina (Kaunas) ; 59(10)2023 Oct 08.
Article in English | MEDLINE | ID: mdl-37893505

ABSTRACT

Immuno-mediated skin diseases are a common and clinically heterogeneous group of cutaneous conditions [...].


Subject(s)
Skin Diseases , Humans , Skin Diseases/drug therapy , Skin Diseases/immunology
10.
Pediatr Rheumatol Online J ; 21(1): 38, 2023 Apr 22.
Article in English | MEDLINE | ID: mdl-37087470

ABSTRACT

BACKGROUND: This study evaluated the efficacy and safety of baricitinib (Janus kinase-1/2 inhibitor), in adult and pediatric Japanese patients with Nakajo-Nishimura syndrome/chronic atypical neutrophilic dermatosis with lipodystrophy and elevated temperature (NNS/CANDLE), stimulator of interferon genes-associated vasculopathy with onset during infancy (SAVI), or Aicardi-Goutières syndrome (AGS). METHODS: A Phase 2/3, multicenter, open-label study (NCT04517253) was conducted across 52 weeks. Primary efficacy endpoint assessed the change in mean daily diary score (DDS) from baseline to the end of primary treatment period. Other efficacy endpoints included change in mean DDS to the end of maintenance period, daily corticosteroid use, Physician's Global Assessment of Disease Activity (PGA) scores, and daily symptom-specific score (DSSS) from baseline to primary and maintenance treatment periods. All treatment-emergent adverse events (TEAEs) that occurred postdosing were recorded. RESULTS: Overall, 9 patients (5 with NNS, 3 with SAVI, and 1 with AGS) were enrolled; 55.6% were females, mean age was 26 years, and mean corticosteroid use/weight was 0.2 mg/kg. At the end of primary treatment period, mean DDS decreased from baseline in patients with NNS/CANDLE (0.22) and SAVI (0.21) and increased in the patient with AGS (0.07). At the end of maintenance treatment period, mean DDS decreased from baseline in patients with NNS/CANDLE (0.18) and SAVI (0.27) and increased in the patient with AGS (0.04). Mean percent corticosteroid use decreased by 18.4% in 3 out of 5 patients with NNS/CANDLE and 62.9% in 1 out of 3 patients with SAVI. Mean PGA score decreased from baseline in patients with NNS/CANDLE (1.60), SAVI (1.33), and AGS (1.0), and mean DSSS improved from baseline. All patients reported ≥ 1 TEAE. Frequently reported AEs included BK polyomavirus detection (3; 33.3%), increased blood creatine phosphokinase (2; 22.2%), anemia (2; 22.2%), and upper respiratory tract infection (2; 22.2%). Three (33.3%) patients reported serious adverse events, 1 of which was related to study drug. One patient with SAVI died due to intracranial hemorrhage, which was not related to study drug. CONCLUSION: Baricitinib may offer a potential therapeutic option for patients with NNS/CANDLE, SAVI, and AGS, with a positive benefit/risk profile in a vulnerable patient population with multiple comorbidities. TRIAL REGISTRATION: NLM clinicaltrials.gov, NCT04517253 . Registered 18 August 2020.


Subject(s)
East Asian People , Hereditary Autoinflammatory Diseases , Interferon Type I , Janus Kinase Inhibitors , Adult , Child , Female , Humans , Male , Autoimmune Diseases of the Nervous System/drug therapy , Autoimmune Diseases of the Nervous System/genetics , Autoimmune Diseases of the Nervous System/immunology , East Asian People/genetics , Skin Diseases/drug therapy , Skin Diseases/genetics , Skin Diseases/immunology , Treatment Outcome , Janus Kinase Inhibitors/therapeutic use , Hereditary Autoinflammatory Diseases/drug therapy , Hereditary Autoinflammatory Diseases/genetics , Hereditary Autoinflammatory Diseases/immunology , Interferon Type I/genetics , Interferon Type I/immunology , Syndrome , Lipodystrophy/drug therapy , Lipodystrophy/genetics , Lipodystrophy/immunology , Fever , Vascular Diseases/drug therapy , Vascular Diseases/genetics , Vascular Diseases/immunology , Glucocorticoids/adverse effects , Glucocorticoids/therapeutic use
11.
Int J Mol Sci ; 24(2)2023 Jan 13.
Article in English | MEDLINE | ID: mdl-36675078

ABSTRACT

The skin is exposed to environmental challenges and contains skin-resident immune cells, including mast cells (MCs) and CD8 T cells that act as sentinels for pathogens and environmental antigens. Human skin MCs and their mediators participate in the maintenance of tissue homeostasis and regulate the recruitment and activity of immune cells involved in the pathogenesis of skin diseases. The cutaneous CD8 T cell compartment is comprised of long-persisting resident memory T cells (TRM) and migratory or recirculating cells; both populations provide durable site immune surveillance. Several lines of evidence indicate that MC-derived products, such as CCL5 and TNF-α, modulate the migration and function of CD8 T cells. Conversely, activated CD8 T cells induce the upregulation of MC costimulatory molecules. Moreover, the close apposition of MCs and CD8 T cells has been recently identified in the skin of several dermatoses, such as alopecia areata. This review outlines the current knowledge about bidirectional interactions between human MCs and CD8 T cells, analyses the alteration of their communication in the context of three common skin disorders in which these cells have been found altered in number or function-psoriasis, atopic dermatitis, and vitiligo-and discusses the current unanswered questions.


Subject(s)
CD8-Positive T-Lymphocytes , Cell Communication , Mast Cells , Skin Diseases , Humans , CD8-Positive T-Lymphocytes/immunology , Cell Communication/immunology , Mast Cells/immunology , Psoriasis/immunology , Skin/immunology , Dermatitis, Atopic , Vitiligo/immunology , Skin Diseases/immunology , Inflammation/immunology
12.
Science ; 376(6596): 940-945, 2022 05 27.
Article in English | MEDLINE | ID: mdl-35617415

ABSTRACT

Human skin forms a protective barrier against the external environment and is our first line of defense against toxic, solar, and pathogenic insults. Our skin also defines our outward appearance, protects our internal tissues and organs, acts as a sensory interface, and prevents dehydration. Crucial to the skin's barrier function is the colonizing microbiota, which provides protection against pathogens, tunes immune responses, and fortifies the epithelium. Here we highlight recent advances in our understanding of how the microbiota mediates multiple facets of skin barrier function. We discuss recent insights into pathological host-microbiota interactions and implications for disorders of the skin and distant organs. Finally, we examine how microbiota-based mechanisms can be targeted to prevent or manage skin disorders and impaired wound healing.


Subject(s)
Host Microbial Interactions , Microbiota , Skin , Epithelium/immunology , Epithelium/microbiology , Hair Follicle/immunology , Hair Follicle/microbiology , Host Microbial Interactions/immunology , Humans , Skin/immunology , Skin/microbiology , Skin Diseases/immunology , Skin Diseases/therapy , Wound Healing/immunology
13.
Vet Immunol Immunopathol ; 246: 110401, 2022 Apr.
Article in English | MEDLINE | ID: mdl-35255296

ABSTRACT

A more complete understanding of canine T-lymphocyte immunity is necessary for improving diagnostic and therapeutic approaches to canine diseases, developing cell-based canine immunotherapeutics, and evaluating dogs as large mammal models for comparative immunology research. The aim of this study was to utilize CD45RA (indicating antigen inexperience) and CD62L (indicating lymph node homing capability), to quantify canine memory T-cell subsets in healthy dogs and dogs with various diseases. Peripheral blood mononuclear cells (PBMCs) were prospectively collected from dogs belonging to one of four groups:dermatologic inflammation (n = 9), solid tumors (n = 9), lymphoma (n = 9), and age-/weight-matched healthy control dogs (n = 15). Dogs receiving prednisone or any other immunomodulating medication within two weeks were excluded. Flow cytometry was performed and T-cell subsets were defined as CD4+ or CD8+, and naïve (TN), central memory (CM), effector memory (EM), or terminal effector memory re-expressing CD45RA (TEMRA). T-cell subset proportions were compared between each disease group and their healthy age-/weight-matched controls using a Mann-Whitney test. Significantly increased %CD8+ TN (P = 0.036) and decreased %CD8+ TEMRA (P = 0.045) were detected in dogs with dermatologic inflammation compared to healthy controls. Furthermore, %CD4+ TN positively correlated with Canine Atopic Dermatitis Extent and Severity Index (CADESI) score within the inflammation group (ρ = 0.817, P = 0.011). No significant differences between either cancer group and their healthy controls were detected. Taken together, these data indicate that dermatologic inflammation can alter proportions of peripheral blood T-cell subsets, possibly due to the migration of antigen-specific T-cells into tissues. Furthermore, these findings support the utility of CD45RA and CD62L in characterizing clinical canine immune responses.


Subject(s)
Dog Diseases , Immunologic Memory , Memory T Cells , Skin Diseases , Animals , CD4-Positive T-Lymphocytes , CD8-Positive T-Lymphocytes , Dog Diseases/immunology , Dogs , L-Selectin , Leukocyte Common Antigens , Leukocytes, Mononuclear , Memory T Cells/immunology , Neoplasms/immunology , Neoplasms/veterinary , Skin Diseases/immunology , Skin Diseases/veterinary
14.
J Leukoc Biol ; 111(2): 301-312, 2022 02.
Article in English | MEDLINE | ID: mdl-34730257

ABSTRACT

Deficiency of adenosine deaminase 2 (DADA2) is a monogenic vasculitis syndrome caused by autosomal-recessive loss-of-function mutations in the ADA2 gene (previously known as CECR1). Vasculitis, vasculopathy, and inflammation are dominant clinical features of this disease; the spectrum of manifestations includes immunodeficiency and lymphoproliferation as well as hematologic manifestations. ADA2 is primarily secreted by stimulated monocytes and macrophages. Aberrant monocyte differentiation to macrophages and neutrophils are important in the pathogenesis of DADA2, but little is known about T lymphocytes in this disease. We performed combined single-cell RNA sequencing and single-cell TCR sequencing in order to profile T cell repertoires in 10 patients with DADA2. Although there were no significant alterations of T cell subsets, we observed activation of both CD8+ and CD4+ T cells. There was no clonal expansion of T cells: most TCRs were expressed at basal levels in patients and healthy donors. TCR usage was private to individual patients and not disease specific, indicating as unlikely a common pathogenic background or predisposition to a common pathogen. We recognized activation of IFN pathways as a signature of T cells and STAT1 as a hub gene in the gene network of T cell activation and cytotoxicity. Overall, T cells in DADA2 patients showed distinct cell-cell interactions with monocytes, as compared with healthy donors, and many of these ligand-receptor interactions likely drove up-regulation of STAT1 in both T cells and other immune cells in patients. Our analysis reveals previously undercharacterized cell characteristics in DADA2.


Subject(s)
Adenosine Deaminase/deficiency , Biomarkers/metabolism , Gene Expression Regulation , Immunologic Deficiency Syndromes/pathology , Intercellular Signaling Peptides and Proteins/deficiency , Skin Diseases/pathology , T-Lymphocytes/pathology , Vascular Diseases/pathology , Adenosine Deaminase/genetics , Adolescent , Adult , Case-Control Studies , Cells, Cultured , Child , Female , Follow-Up Studies , Gene Expression Profiling , Humans , Immunologic Deficiency Syndromes/genetics , Immunologic Deficiency Syndromes/immunology , Intercellular Signaling Peptides and Proteins/genetics , Lymphocyte Activation , Male , Middle Aged , Mutation , Prognosis , STAT1 Transcription Factor/genetics , Single-Cell Analysis , Skin Diseases/genetics , Skin Diseases/immunology , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Vascular Diseases/genetics , Vascular Diseases/immunology , Young Adult
15.
J Cutan Med Surg ; 26(1): 33-49, 2022.
Article in English | MEDLINE | ID: mdl-34261335

ABSTRACT

Type 2 immunity, illustrated by T helper 2 lymphocytes (Th2) and downstream cytokines (IL-4, IL-13, IL-31) as well as group 2 innate lymphoid cells (ILC2), is important in host defense and wound healing.1 The hallmark of type 2 inflammation is eosinophilia and/or high IgE counts and is best recognized in atopic diathesis. Persistent eosinophilia, such as seen in hypereosinophilic syndromes, leads to fibrosis and hence therapeutic Type 2 inhibition in fibrotic diseases is of high interest. Furthermore, as demonstrated in cutaneous T cell lymphoma, advanced disease is characterized by Th1 to Th2 switch allowing cancer progression and immunosuppression. Development of targeted monoclonal antibodies against IL-4Rα (eg, dupilumab) led to a paradigm shift for the treatment of atopic dermatitis (AD) and stimulated research to better understand the role of Type 2 inflammation in other skin conditions. In this review, we summarize up to date knowledge on the role of Type 2 inflammation in skin diseases other than AD and highlight whether the use of Type 2 targeted therapies has been documented or is being investigated in clinical trials. This manuscript reviews the role of Type 2 inflammation in dermatitis, neurodermatitis, IgE-mediated dermatoses (eg, bullous pemphigoid, chronic spontaneous urticaria), sclerodermoid conditions and skin neoplasms.


Subject(s)
Immunity, Innate , Immunotherapy/methods , Skin Diseases/immunology , Skin Diseases/therapy , Wound Healing/immunology , Cytokines/immunology , Humans , Th2 Cells/immunology
16.
Front Immunol ; 13: 1000951, 2022.
Article in English | MEDLINE | ID: mdl-36865523

ABSTRACT

Shift work is associated with systemic chronic inflammation, impaired host and tumor defense and dysregulated immune responses to harmless antigens such as allergens or auto-antigens. Thus, shift workers are at higher risk to develop a systemic autoimmune disease and circadian disruption with sleep impairment seem to be the key underlying mechanisms. Presumably, disturbances of the sleep-wake cycle also drive skin-specific autoimmune diseases, but epidemiological and experimental evidence so far is scarce. This review summarizes the effects of shift work, circadian misalignment, poor sleep, and the effect of potential hormonal mediators such as stress mediators or melatonin on skin barrier functions and on innate and adaptive skin immunity. Human studies as well as animal models were considered. We will also address advantages and potential pitfalls in animal models of shift work, and possible confounders that could drive skin autoimmune diseases in shift workers such as adverse lifestyle habits and psychosocial influences. Finally, we will outline feasible countermeasures that may reduce the risk of systemic and skin autoimmunity in shift workers, as well as treatment options and highlight outstanding questions that should be addressed in future studies.


Subject(s)
Autoimmune Diseases , Shift Work Schedule , Skin Diseases , Sleep Disorders, Circadian Rhythm , Animals , Humans , Autoimmune Diseases/etiology , Autoimmune Diseases/immunology , Autoimmunity , Chronic Disease , Inflammation/etiology , Inflammation/immunology , Shift Work Schedule/adverse effects , Skin/immunology , Skin Diseases/etiology , Skin Diseases/immunology , Sleep Disorders, Circadian Rhythm/etiology , Sleep Disorders, Circadian Rhythm/immunology
17.
Front Immunol ; 12: 751533, 2021.
Article in English | MEDLINE | ID: mdl-34858408

ABSTRACT

The gasdermin (GSDM) family, a novel group of structure-related proteins, consists of GSDMA, GSDMB, GSDMC, GSDMD, GSDME/DNFA5, and PVJK/GSDMF. GSDMs possess a C-terminal repressor domain, cytotoxic N-terminal domain, and flexible linker domain (except for GSDMF). The GSDM-NT domain can be cleaved and released to form large oligomeric pores in the membrane that facilitate pyroptosis. The emerging roles of GSDMs include the regulation of various physiological and pathological processes, such as cell differentiation, coagulation, inflammation, and tumorigenesis. Here, we introduce the basic structure, activation, and expression patterns of GSDMs, summarize their biological and pathological functions, and explore their regulatory mechanisms in health and disease. This review provides a reference for the development of GSDM-targeted drugs to treat various inflammatory and tissue damage-related conditions.


Subject(s)
Neoplasm Proteins , Animals , Asthma/genetics , Asthma/immunology , Autoimmune Diseases/genetics , Autoimmune Diseases/immunology , Hearing Loss/genetics , Hearing Loss/immunology , Humans , Neoplasm Proteins/genetics , Neoplasm Proteins/immunology , Neoplasm Proteins/physiology , Neoplasms/genetics , Neoplasms/immunology , Sepsis/genetics , Sepsis/immunology , Skin Diseases/genetics , Skin Diseases/immunology
18.
Cells ; 10(12)2021 12 02.
Article in English | MEDLINE | ID: mdl-34943909

ABSTRACT

Systemic sclerosis (SSc) is a chronic connective tissue disorder characterized by immune dysregulation, chronic inflammation, vascular endothelial cell dysfunction, and progressive tissue fibrosis of the skin and internal organs. Moreover, increased cancer incidence and accelerated aging are also found. The increased cancer incidence is believed to be a result of chromosome instability. Accelerated cellular senescence has been confirmed by the shortening of telomere length due to increased DNA breakage, abnormal DNA repair response, and telomerase deficiency mediated by enhanced oxidative/nitrative stresses. The immune dysfunctions of SSc patients are manifested by excessive production of proinflammatory cytokines IL-1, IL-6, IL-17, IFN-α, and TNF-α, which can elicit potent tissue inflammation followed by tissue fibrosis. Furthermore, a number of autoantibodies including anti-topoisomerase 1 (anti-TOPO-1), anti-centromere (ACA or anti-CENP-B), anti-RNA polymerase enzyme (anti-RNAP III), anti-ribonuclear proteins (anti-U1, U2, and U11/U12 RNP), anti-nucleolar antigens (anti-Th/T0, anti-NOR90, anti-Ku, anti-RuvBL1/2, and anti-PM/Scl), and anti-telomere-associated proteins were also found. Based on these data, inflamm-aging caused by immune dysfunction-mediated inflammation exists in patients with SSc. Hence, increased cellular senescence is elicited by the interactions among excessive oxidative stress, pro-inflammatory cytokines, and autoantibodies. In the present review, we will discuss in detail the molecular basis of chromosome instability, increased oxidative stress, and functional adaptation by deranged immunome, which are related to inflamm-aging in patients with SSc.


Subject(s)
Aging/genetics , Fibrosis/genetics , Inflammation/genetics , Scleroderma, Systemic/genetics , Aging/immunology , Aging/pathology , Autoantibodies/immunology , Endothelial Cells/pathology , Fibrosis/complications , Fibrosis/immunology , Fibrosis/pathology , Humans , Immune System Diseases/complications , Immune System Diseases/genetics , Immune System Diseases/immunology , Immune System Diseases/pathology , Inflammation/complications , Inflammation/immunology , Inflammation/pathology , Inflammation Mediators , Scleroderma, Systemic/complications , Scleroderma, Systemic/immunology , Scleroderma, Systemic/pathology , Skin Diseases/complications , Skin Diseases/genetics , Skin Diseases/immunology , Skin Diseases/pathology
19.
Clin Dermatol ; 39(6): 934-965, 2021.
Article in English | MEDLINE | ID: mdl-34920833

ABSTRACT

Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2), the etiologic agent of coronavirus disease 2019 (COVID-19), is a single-stranded RNA virus whose sequence is known. COVID-19 is associated with a heterogeneous clinical phenotype ranging from asymptomatic to fatal disease. It appears that access to nasopharyngeal respiratory epithelia expressing angiotensin-converting enzyme (ACE) 2, the receptor for SARS-CoV-2, is followed by viral replication in the pulmonary alveolar septal capillary bed. We have demonstrated in earlier studies that incomplete viral particles, termed pseudovirions, dock to deep subcutaneous and other vascular beds, potentially contributing to the prothrombotic state and systemic complement activation that characterizes severe and critical COVID-19. A variety of skin eruptions have been described in the setting of SARS-CoV-2 infection and more recently, after COVID-19 vaccination. The vaccines deliver a laboratory-synthesized mRNA that encodes a protein that is identical to the spike glycoprotein of SARS-CoV-2, allowing the production of immunogenic spike glycoprotein that will then elicit T cell and B cell adaptive immune responses. In this contribution, we review an array of cutaneous manifestations of COVID-19 that provide an opportunity to study critical pathophysiologic mechanisms that underlie all clinical facets of COVID-19, ranging from asymptomatic/mild to severe and critical COVID-19. We classify cutaneous COVID-19 according to underlying pathophysiologic principles. In this regard we propose three main pathways: (1) complement mediated thrombotic vascular injury syndromes deploying the alternative and mannan binding lectin pathways and resulting in the elaboration of cytokines like interleukin 6 from endothelium in the setting of severe and critical COVID-19 and (2) the robust T cell and type I interferon-driven inflammatory and (3) humoral-driven immune complex mediated vasculitic cutaneous reactions observed with mild and moderate COVID-19. Presented are novel data on cutaneous vaccine reactions that manifest a clinical and morphologic parallel with similar eruptions observed in patients with mild and moderate COVID-19 and in some cases represent systemic eczematoid hypersensitivity reactions to a putative vaccine-based antigen versus unmasking subclinical hypersensitivity due to immune enhancing effects of the vaccine. Finally, we demonstrate for the first time the localization of human synthesized spike glycoprotein after the COVID-19 vaccine to the cutaneous and subcutaneous vasculature confirming the ability of SARS-CoV-2 spike glycoprotein to bind endothelium in the absence of intact virus.


Subject(s)
COVID-19 , Skin Diseases/virology , COVID-19/immunology , COVID-19/physiopathology , COVID-19 Vaccines , Cytokines , Humans , Skin Diseases/immunology , Spike Glycoprotein, Coronavirus
20.
Nat Cell Biol ; 23(12): 1224-1239, 2021 12.
Article in English | MEDLINE | ID: mdl-34876685

ABSTRACT

Defective silencing of retrotransposable elements has been linked to inflammageing, cancer and autoimmune diseases. However, the underlying mechanisms are only partially understood. Here we implicate the histone H3.3 chaperone Daxx, a retrotransposable element repressor inactivated in myeloid leukaemia and other neoplasms, in protection from inflammatory disease. Loss of Daxx alters the chromatin landscape, H3.3 distribution and histone marks of haematopoietic progenitors, leading to engagement of a Pu.1-dependent transcriptional programme for myelopoiesis at the expense of B-cell differentiation. This causes neutrophilia and inflammation, predisposing mice to develop an autoinflammatory skin disease. While these molecular and phenotypic perturbations are in part reverted in animals lacking both Pu.1 and Daxx, haematopoietic progenitors in these mice show unique chromatin and transcriptome alterations, suggesting an interaction between these two pathways. Overall, our findings implicate retrotransposable element silencing in haematopoiesis and suggest a cross-talk between the H3.3 loading machinery and the pioneer transcription factor Pu.1.


Subject(s)
Chromatin/pathology , Co-Repressor Proteins/genetics , Leukocyte Disorders/congenital , Molecular Chaperones/genetics , Myelopoiesis/genetics , Proto-Oncogene Proteins/metabolism , Trans-Activators/metabolism , Animals , Autoimmune Diseases/genetics , Autoimmune Diseases/pathology , B-Lymphocytes/cytology , Cell Line , Chromatin/genetics , Hematopoietic Stem Cells/cytology , Histones/metabolism , Humans , Inflammation/pathology , Leukocyte Disorders/pathology , Mice , Mice, Inbred C57BL , Mice, Knockout , Retroelements/genetics , Skin Diseases/genetics , Skin Diseases/immunology , Skin Diseases/pathology
SELECTION OF CITATIONS
SEARCH DETAIL
...