Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
Add more filters

Publication year range
1.
Cell ; 184(14): 3794-3811.e19, 2021 07 08.
Article in English | MEDLINE | ID: mdl-34166614

ABSTRACT

The microbiota plays a fundamental role in regulating host immunity. However, the processes involved in the initiation and regulation of immunity to the microbiota remain largely unknown. Here, we show that the skin microbiota promotes the discrete expression of defined endogenous retroviruses (ERVs). Keratinocyte-intrinsic responses to ERVs depended on cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes protein (STING) signaling and promoted the induction of commensal-specific T cells. Inhibition of ERV reverse transcription significantly impacted these responses, resulting in impaired immunity to the microbiota and its associated tissue repair function. Conversely, a lipid-enriched diet primed the skin for heightened ERV- expression in response to commensal colonization, leading to increased immune responses and tissue inflammation. Together, our results support the idea that the host may have co-opted its endogenous virome as a means to communicate with the exogenous microbiota, resulting in a multi-kingdom dialog that controls both tissue homeostasis and inflammation.


Subject(s)
Endogenous Retroviruses/physiology , Homeostasis , Inflammation/microbiology , Inflammation/pathology , Microbiota , Animals , Bacteria/metabolism , Chromosomes, Bacterial/genetics , Diet, High-Fat , Inflammation/immunology , Inflammation/virology , Interferon Type I/metabolism , Keratinocytes/metabolism , Membrane Proteins/metabolism , Mice, Inbred C57BL , Nucleotidyltransferases/metabolism , Retroelements/genetics , Signal Transduction , Skin/immunology , Skin/microbiology , T-Lymphocytes/immunology , Transcription, Genetic
2.
Cell ; 172(4): 784-796.e18, 2018 02 08.
Article in English | MEDLINE | ID: mdl-29358051

ABSTRACT

Mammalian barrier surfaces are constitutively colonized by numerous microorganisms. We explored how the microbiota was sensed by the immune system and the defining properties of such responses. Here, we show that a skin commensal can induce T cell responses in a manner that is restricted to non-classical MHC class I molecules. These responses are uncoupled from inflammation and highly distinct from pathogen-induced cells. Commensal-specific T cells express a defined gene signature that is characterized by expression of effector genes together with immunoregulatory and tissue-repair signatures. As such, non-classical MHCI-restricted commensal-specific immune responses not only promoted protection to pathogens, but also accelerated skin wound closure. Thus, the microbiota can induce a highly physiological and pleiotropic form of adaptive immunity that couples antimicrobial function with tissue repair. Our work also reveals that non-classical MHC class I molecules, an evolutionarily ancient arm of the immune system, can promote homeostatic immunity to the microbiota.


Subject(s)
Adaptive Immunity , Bacteria/immunology , Histocompatibility Antigens Class I/immunology , Microbiota/immunology , Skin/immunology , T-Lymphocytes/immunology , Animals , Gene Expression Regulation/immunology , Histocompatibility Antigens Class I/genetics , Mice , Mice, Transgenic
3.
Trends Immunol ; 44(4): 256-265, 2023 04.
Article in English | MEDLINE | ID: mdl-36964020

ABSTRACT

Malaria is caused by Plasmodium protozoa that are transmitted by anopheline mosquitoes. Plasmodium sporozoites are released with saliva when an infected female mosquito takes a blood meal on a vertebrate host. Sporozoites deposited into the skin must enter a blood vessel to start their journey towards the liver. After migration out of the mosquito, sporozoites are associated with, or in proximity to, many components of vector saliva in the skin. Recent work has elucidated how Anopheles saliva, and components of saliva, can influence host-pathogen interactions during the early stage of Plasmodium infection in the skin. Here, we discuss how components of Anopheles saliva can modulate local host responses and affect Plasmodium infectivity. We hypothesize that therapeutic strategies targeting mosquito salivary proteins can play a role in controlling malaria and other vector-borne diseases.


Subject(s)
Anopheles , Malaria , Humans , Animals , Female , Anopheles/parasitology , Anopheles/physiology , Saliva , Mosquito Vectors/parasitology , Sporozoites
4.
Proc Natl Acad Sci U S A ; 120(34): e2219932120, 2023 08 22.
Article in English | MEDLINE | ID: mdl-37579158

ABSTRACT

Tissue-resident memory CD8+ T cells (TRM) reside at sites of previous infection, providing protection against reinfection with the same pathogen. In the skin, TRM patrol the epidermis, where keratinocytes are the entry site for many viral infections. Epidermal TRM react rapidly to cognate antigen encounter with the secretion of cytokines and differentiation into cytotoxic effector cells, constituting a first line of defense against skin reinfection. Despite the important protective role of skin TRM, it has remained unclear, whether their reactivation requires a professional antigen-presenting cell (APC). We show here, using a model system that allows antigen targeting selectively to keratinocytes in a defined area of the skin, that limited antigen expression by keratinocytes results in rapid, antigen-specific reactivation of skin TRM. Our data identify epidermal Langerhans cells that cross-present keratinocyte-derived antigens, as the professional APC indispensable for the early reactivation of TRM in the epidermal layer of the skin.


Subject(s)
CD8-Positive T-Lymphocytes , Langerhans Cells , Humans , Memory T Cells , Reinfection/metabolism , Epidermis , Antigens , Immunologic Memory
5.
Allergy ; 79(6): 1531-1547, 2024 06.
Article in English | MEDLINE | ID: mdl-38385963

ABSTRACT

BACKGROUND: The skin barrier is vital for protection against environmental threats including insults caused by skin-resident microbes. Dysregulation of this barrier is a hallmark of atopic dermatitis (AD) and ichthyosis, with variable consequences for host immune control of colonizing commensals and opportunistic pathogens. While Malassezia is the most abundant commensal fungus of the skin, little is known about the host control of this fungus in inflammatory skin diseases. METHODS: In this experimental study, MC903-treated mice were colonized with Malassezia spp. to assess the host-fungal interactions in atopic dermatitis. Additional murine models of AD and ichthyosis, including tape stripping, K5-Nrf2 overexpression and flaky tail mice, were employed to confirm and expand the findings. Skin fungal counts were enumerated. High parameter flow cytometry was used to characterize the antifungal response in the AD-like skin. Structural and functional alterations in the skin barrier were determined by histology and transcriptomics of bulk skin. Finally, differential expression of metabolic genes in Malassezia in atopic and control skin was quantified. RESULTS: Malassezia grows excessively in AD-like skin. Fungal overgrowth could, however, not be explained by the altered immune status of the atopic skin. Instead, we found that by upregulating key metabolic genes in the altered cutaneous niche, Malassezia acquired enhanced fitness to efficiently colonise the impaired skin barrier. CONCLUSIONS: This study provides evidence that structural and metabolic changes in the dysfunctional epidermal barrier environment provide increased accessibility and an altered lipid profile, to which the lipid-dependent yeast adapts for enhanced nutrient assimilation. Our findings reveal fundamental insights into the implication of the mycobiota in the pathogenesis of common skin barrier disorders.


Subject(s)
Dermatitis, Atopic , Disease Models, Animal , Malassezia , Skin , Animals , Malassezia/immunology , Mice , Dermatitis, Atopic/microbiology , Dermatitis, Atopic/immunology , Skin/microbiology , Skin/immunology , Epidermis/microbiology , Epidermis/immunology , Epidermis/metabolism , Disease Susceptibility , Hypersensitivity/immunology , Hypersensitivity/microbiology , Female
6.
J Virol ; 96(17): e0099922, 2022 09 14.
Article in English | MEDLINE | ID: mdl-36000846

ABSTRACT

Arthritogenic alphaviruses are mosquito-borne arboviruses that include several re-emerging human pathogens, including the chikungunya (CHIKV), Ross River (RRV), Mayaro (MAYV), and o'nyong-nyong (ONNV) virus. Arboviruses are transmitted via a mosquito bite to the skin. Herein, we describe intradermal RRV infection in a mouse model that replicates the arthritis and myositis seen in humans with Ross River virus disease (RRVD). We show that skin infection with RRV results in the recruitment of inflammatory monocytes and neutrophils, which together with dendritic cells migrate to draining lymph nodes (LN) of the skin. Neutrophils and monocytes are productively infected and traffic virus from the skin to LN. We show that viral envelope N-linked glycosylation is a key determinant of skin immune responses and disease severity. RRV grown in mammalian cells elicited robust early antiviral responses in the skin, while RRV grown in mosquito cells stimulated poorer early antiviral responses. We used glycan mass spectrometry to characterize the glycan profile of mosquito and mammalian cell-derived RRV, showing deglycosylation of the RRV E2 glycoprotein is associated with curtailed skin immune responses and reduced disease following intradermal infection. Altogether, our findings demonstrate skin infection with an arthritogenic alphavirus leads to musculoskeletal disease and envelope glycoprotein glycosylation shapes disease outcome. IMPORTANCE Arthritogenic alphaviruses are transmitted via mosquito bites through the skin, potentially causing debilitating diseases. Our understanding of how viral infection starts in the skin and how virus systemically disseminates to cause disease remains limited. Intradermal arbovirus infection described herein results in musculoskeletal pathology, which is dependent on viral envelope N-linked glycosylation. As such, intradermal infection route provides new insights into how arboviruses cause disease and could be extended to future investigations of skin immune responses following infection with other re-emerging arboviruses.


Subject(s)
Alphavirus Infections , Arthritis , Myositis , Polysaccharides , Ross River virus , Skin , Alphavirus Infections/complications , Alphavirus Infections/immunology , Animals , Antiviral Agents/immunology , Arthritis/complications , Arthritis/immunology , Culicidae/virology , Dendritic Cells , Disease Models, Animal , Glycosylation , Humans , Mass Spectrometry , Mice , Monocytes , Myositis/complications , Myositis/immunology , Neutrophils , Polysaccharides/chemistry , Polysaccharides/immunology , Ross River virus/immunology , Skin/immunology , Skin/virology , Viral Envelope Proteins/chemistry , Viral Envelope Proteins/immunology
7.
Trends Immunol ; 41(2): 100-112, 2020 02.
Article in English | MEDLINE | ID: mdl-31948873

ABSTRACT

Skin is the largest barrier organ and an important interface between the body and the outside environment. Immune surveillance and homeostatic regulation of skin function are governed by complex interactions between resident lymphoid and myeloid cells and their communications with the surrounding parenchyma. Recent studies have provided exciting insights about the unique characteristics of skin-resident innate lymphoid cells (ILCs). Here, we discuss advances demonstrating how skin ILCs contribute to tissue homeostasis by regulating microbiome balance in steady-state and how their dysregulation can trigger and promote inflammatory skin diseases such as atopic dermatitis and psoriasis. We review the phenotypic and functional similarities and differences of ILCs between the skin and other organs and highlight future areas of investigation for this field.


Subject(s)
Immunity, Innate , Lymphocytes , Skin , Dermatitis, Atopic/immunology , Humans , Immunity, Innate/immunology , Lymphocytes/immunology , Psoriasis/immunology , Skin/cytology , Skin/immunology
8.
Parasite Immunol ; 45(2): e12948, 2023 02.
Article in English | MEDLINE | ID: mdl-36047038

ABSTRACT

Fungi represent an integral part of the skin microbiota. Their complex interaction network with the host shapes protective immunity during homeostasis. If host defences are breached, skin-resident fungi including Malassezia and Candida, and environmental fungi such as dermatophytes can cause cutaneous infections. In addition, fungi are associated with diverse non-infectious skin disorders. Despite their multiple roles in health and disease, fungi remain elusive and understudied, and the mechanisms underlying the emergence of pathological conditions linked to fungi are largely unclear. The identification of IL-17 as an important antifungal effector mechanism represents a milestone for understanding homeostatic antifungal immunity. At the same time, host-adverse, disease-promoting roles of IL-17 have been delineated, as in psoriasis. Fungal dysbiosis represents another feature of many pathological skin conditions with an unknown causal link of intra- and interkingdom interactions to disease pathogenesis. The emergence of new fungal pathogens such as Candida auris highlights the need for more research into fungal immunology to understand how antifungal responses shape health and diseases. Recent technological advances for genetically manipulating fungi to target immunomodulatory fungal determinants, multi-omics approaches for studying immune cells in the human skin, and novel experimental models open up a promising future for skin fungal immunity.


Subject(s)
Malassezia , Microbiota , Humans , Interleukin-17 , Antifungal Agents , Skin , Fungi/physiology
9.
Microsc Microanal ; 29(2): 762-776, 2023 04 05.
Article in English | MEDLINE | ID: mdl-37749732

ABSTRACT

Allergic contact dermatitis (ACD) is an occupation-dependent skin disease that afflicts humans with recurrent, non-specific episodes. Telocyte (TC) is a novel interstitial cell discovered in recent years and, together with fibroblasts, constitutes the predominant interstitial cell population in the skin. The purpose of this study was to investigate the morphodynamic changes of interstitial cells, especially TCs, in the skin during the development and treatment of ACD by histological and microscopic scientific methods. Hematoxylin-eosin staining, Masson staining, immunohistochemistry (IHC), immunofluorescence (IF), scanning electron microscopy (SEM), and transmission electron microscopy (TEM) were used to track morphodynamic changes in interstitial cells during the development and treatment in the ACD-involved skin induced by 2,4-dinitrochlorobenzene (DNCB). The results demonstrated that TCs were mainly present around dermal collagen fibers, perivascular (except dermal papillary vascular loop), and skin appendages, which expressed CD34+, Vimentin+, PDGFR-α+, and α-SMA-. The absence of TCs during ACD development and after ACD recovery causes dermal interstitial cell dysregulation. The special anatomical relationships between TCs, immune cells, and follicular stem cells were also revealed, suggesting their potential dermatitis-regulating function. In a nutshell, our results provide morphodynamic evidence for the process of ACD development and recovery and offer potential cytological ideas for ACD treatment.


Subject(s)
Dermatitis, Allergic Contact , Interstitial Cells of Cajal , Telocytes , Humans , Telocytes/ultrastructure , Skin/pathology , Dermatitis, Allergic Contact/pathology , Immunohistochemistry
10.
J Allergy Clin Immunol ; 147(2): 439-455, 2021 02.
Article in English | MEDLINE | ID: mdl-32560971

ABSTRACT

Research into the pathophysiology of psoriasis remains challenging, because this disease does not occur naturally in laboratory animals. However, specific aspects of its complex immune-pathology can be illuminated through transgenic, knockout, xenotransplantation, immunological reconstitution, drug-induced, or spontaneous mutation models in rodents. Although some of these approaches have already been pursued for more than 5 decades and even more models have been described in recent times, they have surprisingly not yet been systematically validated. As a consequence, researchers regularly examine specific aspects that only partially reflect the complex overall picture of the human disease. Nonetheless, animal models are of great utility to investigate inflammatory mediators, the communication between cells of the innate and the adaptive immune systems, the role of resident cells as well as new therapies. Of note, various manipulations in experimental animals resulted in rather similar phenotypes. These were called "psoriasiform", "psoriasis-like" or even "psoriasis" usually on the basis of some similarities with the human disorder. Xenotransplantation of human skin onto immunocompromised animals can overcome this limitation only in part. In this review, we elucidate approaches for the generation of animal models of psoriasis and assess their strengths and limitations with a certain focus on more recently developed models.


Subject(s)
Disease Models, Animal , Psoriasis , Animals , Humans
11.
Int J Mol Sci ; 21(14)2020 Jul 19.
Article in English | MEDLINE | ID: mdl-32707732

ABSTRACT

The skin is an important organ that acts as a physical barrier to the outer environment. It is rich in immune cells such as keratinocytes, Langerhans cells, mast cells, and T cells, which provide the first line of defense mechanisms against numerous pathogens by activating both the innate and adaptive response. Cutaneous immunological processes may be stimulated or suppressed by numerous plant extracts via their immunomodulatory properties. Several plants are rich in bioactive molecules; many of these exert antimicrobial, antiviral, and antifungal effects. The present study describes the impact of plant extracts on the modulation of skin immunity, and their antimicrobial effects against selected skin invaders. Plant products remain valuable counterparts to modern pharmaceuticals and may be used to alleviate numerous skin disorders, including infected wounds, herpes, and tineas.


Subject(s)
Anti-Infective Agents/administration & dosage , Plant Extracts/administration & dosage , Skin Diseases, Infectious/drug therapy , Skin Diseases, Infectious/microbiology , Dermatomycoses/drug therapy , Dermatomycoses/immunology , Dermatomycoses/microbiology , Drug Synergism , Humans , Immunologic Factors/administration & dosage , Plants, Medicinal/chemistry , Skin/drug effects , Skin/immunology , Skin/microbiology , Skin Diseases, Bacterial/drug therapy , Skin Diseases, Bacterial/immunology , Skin Diseases, Bacterial/microbiology , Skin Diseases, Infectious/immunology , Virus Diseases/drug therapy , Virus Diseases/immunology , Virus Diseases/virology
12.
Yale J Biol Med ; 93(1): 133-143, 2020 03.
Article in English | MEDLINE | ID: mdl-32226343

ABSTRACT

The skin serves as a front line of defense against harmful environmental elements and thus is vital for organismal survival. This barrier is comprised of a water-tight epithelial structure reinforced by an arsenal of immune cells. The epithelial and immune components of the skin are interdependent and actively dialogue to maintain health and combat infectious, injurious, and noxious stimuli. Here, we discuss the molecular mediators of this crosstalk that establish tissue homeostasis and their dynamic adaptations to various stress conditions. In particular, we focus on immune-epithelial interactions in homeostatic tissue regeneration, during natural cycling of the hair follicle, and following skin injury. We also highlight the epithelial derived factors that orchestrate immunity. A comprehensive and mechanistic understanding of dynamic interactions between cutaneous immune cells and the epithelium can be leveraged to develop novel therapies to treat of range of skin diseases and boost skin health.


Subject(s)
Epithelium/immunology , Immunity , Skin , Environmental Exposure , Homeostasis/immunology , Humans , Skin/immunology , Skin/pathology
13.
J Infect Dis ; 220(5): 892-901, 2019 07 31.
Article in English | MEDLINE | ID: mdl-31107940

ABSTRACT

BACKGROUND: Lactococcus lactis strain Plasma (LC-Plasma) was revealed to stimulate plasmacytoid dendritic cells and induce antiviral immunity in vitro and in vivo. In this study, we assessed the effects of LC-Plasma on skin immunity. METHODS: To evaluate the effect of LC-Plasma on skin immunity and Staphylococcus aureus epicutaneous infection, lymphocyte activities in skin-draining lymph nodes (SLNs) and gene expression in skin were analyzed after 2 weeks of oral administration of LC-Plasma. To evaluate the mechanisms of interleukin 17A production, SLN lymphocytes were cultured with or without LC-Plasma, and the interleukin 17A concentrations in supernatants were measured. RESULTS: Oral administration of LC-Plasma activated plasma dendritic cells in SLNs, augmented skin homeostasis, and elicited suppression of Staphylococcus aureus, Staphylococcus epidermidis, and Propionibacterium acnes proliferation. In addition, significant suppression of the S. aureus burden and reduced skin inflammation were observed following oral administration of LC-Plasma. Furthermore, a subsequent in vitro study revealed that LC-Plasma could elicit interleukin 17A production from CD8+ T cells and that its induction mechanism depended on the Toll-like receptor 9 signaling pathway, with type I interferon partially involved. CONCLUSIONS: Our results suggest that LC-Plasma oral administration enhances skin homeostasis via plasma dendritic cell activation in SLNs, resulting in suppression of S. aureus epicutaneous infection and skin inflammation.


Subject(s)
Interleukin-17/pharmacology , Lactococcus lactis/physiology , Skin/immunology , Staphylococcal Infections/drug therapy , Staphylococcal Skin Infections/immunology , Staphylococcus aureus/drug effects , Animals , Antimicrobial Cationic Peptides/genetics , CD8-Positive T-Lymphocytes/metabolism , Calgranulin A/metabolism , Cell Proliferation , Claudin-1/genetics , Claudin-1/metabolism , Cytokines/genetics , Cytokines/metabolism , Dendritic Cells/immunology , Disease Models, Animal , Female , Homeostasis , Interleukin-17/metabolism , Lymph Nodes , Lymphocyte Activation , Mice, Inbred BALB C , Propionibacterium acnes , Skin/microbiology , Skin/pathology , Staphylococcal Skin Infections/microbiology , Staphylococcus epidermidis , Zonula Occludens-1 Protein/genetics , Zonula Occludens-1 Protein/metabolism , beta-Defensins/metabolism
14.
Exp Dermatol ; 28(6): 656-661, 2019 06.
Article in English | MEDLINE | ID: mdl-30636075

ABSTRACT

The understanding of the immune mechanisms of vitiligo has profoundly improved over the past years. The recent discovery of a new population of antigen-experienced memory T cells called resident memory T cells (TRM ) has changed the concept of immune surveillance in peripheral tissue as skin, and the presence of melanocyte-specific TRM is clearly demonstrated in vitiligo, a disease that could be now seen such as a memory skin disease. This review summarizes the recent knowledge on skin TRM and their role in vitiligo. Future management or therapies for this disease will have the goal to block their migration/differentiation, to dampen their activation and/or their accumulation in the vitiligo skin to prevent flare-up or to promote repigmentation.


Subject(s)
Immunologic Memory , Immunomodulation , Skin/immunology , T-Lymphocytes/immunology , Vitiligo/immunology , Vitiligo/therapy , Adult , Cell Differentiation , Cell Movement , Cell Proliferation , Female , Humans , Hypopigmentation , Immunologic Factors , Male , Melanocytes/physiology , Methotrexate/administration & dosage , Middle Aged , Oxidative Stress , Skin Pigmentation , Translational Research, Biomedical , Ultraviolet Rays , Vitiligo/diagnosis
15.
Exp Dermatol ; 26(9): 827-829, 2017 09.
Article in English | MEDLINE | ID: mdl-28094872

ABSTRACT

Antimicrobial peptides (AMPs) form a part of the skin's innate immune system. Their primary activity is to provide antimicrobial benefits and hence protect from infections. AMPs that are present on human skin include psoriasin (S100A7), RNase 7, lysozyme, LL-37 and defensins. Niacinamide is a well-known cosmetic ingredient that has been used traditionally for multiple skin benefits. Recent data indicate that niacinamide treatment can boost AMPs in human gut epithelial cells and in neutrophils. Treatment with niacinamide in mice also provided protection from skin infections by enhancing AMPs. In this article, we find that treatment with niacinamide formulation provides long-lasting protection against bacteria, potentially through the activation of an AMP response.


Subject(s)
Escherichia coli/drug effects , Niacinamide/pharmacology , Skin/drug effects , Staphylococcus aureus/drug effects , Vitamin B Complex/pharmacology , Adult , Healthy Volunteers , Humans , Skin/microbiology
16.
Exp Dermatol ; 26(11): 989-998, 2017 11.
Article in English | MEDLINE | ID: mdl-28191680

ABSTRACT

Host defense peptides/proteins (HDPs), also known as antimicrobial peptides/proteins (AMPs), are key molecules in the cutaneous innate immune system. AMPs/HDPs historically exhibit broad-spectrum killing activity against bacteria, enveloped viruses, fungi and several parasites. Recently, AMPs/HDPs were shown to have important biological functions, including inducing cell proliferation, migration and differentiation; regulating inflammatory responses; controlling the production of various cytokines/chemokines; promoting wound healing; and improving skin barrier function. Despite the fact that AMPs/HDPs protect our body, several studies have hypothesized that these molecules actively contribute to the pathogenesis of various skin diseases. For example, AMPs/HDPs play crucial roles in the pathological processes of psoriasis, atopic dermatitis, rosacea, acne vulgaris, systemic lupus erythematosus and systemic sclerosis. Thus, AMPs/HDPs may be a double-edged sword, promoting cutaneous immunity while simultaneously initiating the pathogenesis of some skin disorders. This review will describe the most common skin-derived AMPs/HDPs (defensins, cathelicidins, S100 proteins, ribonucleases and dermcidin) and discuss the biology and both the positive and negative aspects of these AMPs/HDPs in skin inflammatory/infectious diseases. Understanding the regulation, functions and mechanisms of AMPs/HDPs may offer new therapeutic opportunities in the treatment of various skin disorders.


Subject(s)
Antimicrobial Cationic Peptides/immunology , Antimicrobial Cationic Peptides/metabolism , Skin Diseases/metabolism , Skin/immunology , Skin/metabolism , Cathelicidins/immunology , Cathelicidins/metabolism , Defensins/immunology , Defensins/metabolism , Humans , Immunity, Innate , Peptides/immunology , Peptides/metabolism , Ribonucleases/immunology , Ribonucleases/metabolism , S100 Proteins/immunology , S100 Proteins/metabolism , Skin Physiological Phenomena , Wound Healing
17.
J Infect Dis ; 213(7): 1189-97, 2016 Apr 01.
Article in English | MEDLINE | ID: mdl-26610735

ABSTRACT

Leprosy is a chronic disease characterized by skin and peripheral nerve pathology and immune responses that fail to control Mycobacterium leprae. Toll-interacting protein (TOLLIP) regulates Toll-like receptor (TLR) and interleukin 1 receptor (IL-1R) signaling against mycobacteria. We analyzed messenger RNA (mRNA) expression of candidate immune genes in skin biopsy specimens from 85 individuals with leprosy. TOLLIP mRNA was highly and specifically correlated with IL-1R antagonist (IL-1Ra). In a case-control gene-association study with 477 cases and 1021 controls in Nepal, TOLLIP single-nucleotide polymorphism rs3793964 TT genotype was associated with increased susceptibility to leprosy (recessive, P = 1.4 × 10(-3)) and with increased skin expression of TOLLIP and IL-1Ra. Stimulation of TOLLIP-deficient monocytes with M. leprae produced significantly less IL-1Ra (P < .001), compared with control. These data suggest that M. leprae upregulates IL-1Ra by a TOLLIP-dependent mechanism. Inhibition of TOLLIP may decrease an individual's susceptibility to leprosy and offer a novel therapeutic target for IL-1-dependent diseases.


Subject(s)
Gene Expression Regulation, Bacterial/physiology , Genetic Predisposition to Disease , Interleukin 1 Receptor Antagonist Protein/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , Leprosy/genetics , Polymorphism, Single Nucleotide , Adult , Case-Control Studies , Genotype , Humans , Interleukin 1 Receptor Antagonist Protein/genetics , Intracellular Signaling Peptides and Proteins/genetics , Leprosy/epidemiology , Nepal , Prospective Studies , Skin/metabolism
18.
Fish Shellfish Immunol ; 39(2): 206-14, 2014 Aug.
Article in English | MEDLINE | ID: mdl-24821425

ABSTRACT

The presence and production of IgM in the skin of the Antarctic teleost Trematomus bernacchii were investigated in this study. Immunoglobulins purified from cutaneous mucus and analysed by SDS-PAGE run under non-reducing and reducing conditions, were composed of heavy and light chains of 78 kDa and 25 kDa respectively, with a relative molecular mass of 830 kDa indicating that mucus IgM are tetramers as the serum IgM. Mature transcripts encoding the constant domains of both the secretory and membrane-bound Igµ chain were seen in T. bernacchii skin using a PCR strategy and the expression of the secretory Igµ chain in the skin was compared with that in other tissues by Real-time PCR. Cytological investigations revealed the presence of either immunoglobulins or their transcripts in occasional lymphocytes distributed close to the basal membrane. IgM once produced here, enters the filament-containing cells and is released into the mucus when these cells degenerate and detach from the epidermis. Our findings indicate that a cutaneous defence mechanism, functioning as anatomical and physiological barrier under subzero conditions, is present in this Antarctic species as an important component of the immune system.


Subject(s)
Acclimatization/immunology , Immunoglobulin M/immunology , Perciformes/immunology , Skin/immunology , Animals , Antarctic Regions , Cold Temperature , DNA Primers/genetics , Electrophoresis, Polyacrylamide Gel/veterinary , Immunohistochemistry/veterinary , In Situ Hybridization/veterinary , Lymphocytes/immunology , Real-Time Polymerase Chain Reaction/veterinary
19.
Oxf Open Immunol ; 5(1): iqae003, 2024.
Article in English | MEDLINE | ID: mdl-38737941

ABSTRACT

Dengue virus (DENV) poses a global health threat, affecting millions individuals annually with no specific therapy and limited vaccines. Mosquitoes, mainly Aedes aegypti and Aedes albopictus worldwide, transmit DENV through their saliva during blood meals. In this study, we aimed to understand how Aedes mosquito saliva modulate skin immune responses during DENV infection in individuals living in mosquito-endemic regions. To accomplish this, we dissociated skin cells from Cambodian volunteers and incubated them with salivary gland extract (SGE) from three different mosquito strains: Ae. aegypti USDA strain, Ae. aegypti and Ae. albopictus wild type (WT) in the presence/absence of DENV. We observed notable alterations in skin immune cell phenotypes subsequent to exposure to Aedes salivary gland extract (SGE). Specifically, exposure lead to an increase in the frequency of macrophages expressing chemokine receptor CCR2, and neutrophils expressing CD69. Additionally, we noted a substantial increase in the percentage of macrophages that became infected with DENV in the presence of Aedes SGE. Differences in cellular responses were observed when Aedes SGE of three distinct mosquito strains were compared. Our findings deepen the understanding of mosquito saliva's role in DENV infection and skin immune responses in individuals regularly exposed to mosquito bites. This study provides insights into skin immune cell dynamics that could guide strategies to mitigate DENV transmission and other arbovirus diseases.

20.
Immunol Med ; 46(2): 62-68, 2023 Jun.
Article in English | MEDLINE | ID: mdl-36705253

ABSTRACT

Epigenetics has been well understood for its role in cell development; however, it is now known to regulate many processes involved in immune cell activation in a variety of cells. The skin maintains homeostasis via crosstalk between immune and non-immune cells. Disruption of normal epigenetic regulation in these cells may alter the transcription of immune-regulatory factors and affect the immunological balance in the skin. This review summarizes recent evidence for the epigenetic regulation of skin immunity. Much of what is known about epigenetic involvement in skin immunity is associated with DNA methylation. This review focuses on epigenetic regulation of histone modification and chromatin remodeling and describes their role in the transcriptional regulation of immune-regulatory factors. While much is still unknown regarding the regulation of skin immunity via histone modification or chromatin remodeling, these processes may underlie the pathogenesis of chronic cutaneous immune disorders.


Subject(s)
DNA Methylation , Epigenesis, Genetic , Gene Expression Regulation , Skin , Chromatin Assembly and Disassembly
SELECTION OF CITATIONS
SEARCH DETAIL