Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 39
Filtrar
1.
Int Immunol ; 2024 Jun 18.
Artigo em Inglês | MEDLINE | ID: mdl-38887075

RESUMO

Atopic dermatitis (AD), a prevalent Th2-dominant skin disease, involves complex genetic and environmental factors, including mutations in the Filaggrin gene and dysbiosis of skin microbiota characterized by an increased abundance of Staphylococcus aureus. Our recent findings emphasize the pivotal role of the skin barrier's integrity and microbial composition in infantile AD and allergic diseases. Early skin dysbiosis predisposes infants to AD, suggesting targeted skincare practices as a preventive strategy. The effects of skincare interventions, particularly the application of moisturizers with the appropriate molar concentration of ceramides, cholesterol, and fatty acids, play a crucial role in restoring the skin barrier. Notably, our study revealed that appropriate skincare can reduce Streptococcus abundance while supporting Cutibacterium acnes presence, thus directly linking skincare practices to microbial modulation in neonatal skin. Despite the mixed outcomes of previous Randomized Controlled Trials on the efficacy of moisturizers in AD prevention, our research points to the potential of skincare intervention as a primary preventive method against AD by minimizing the impact of genetic and environmental factors. Furthermore, our research supports the notion that early aggressive management of eczema may reduce the incidence of food allergies, highlighting the necessity for multifaceted prevention strategies that address both the skin barrier and immune sensitization. By focusing on repairing the skin barrier and adjusting the skin's microbiome from birth, we propose a novel perspective on preventing infantile AD and allergic diseases, opening new avenues for future studies and practices in allergy prevention.

2.
Nat Immunol ; 13(5): 449-56, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22484733

RESUMO

Intestinal phagocytes transport oral antigens and promote immune tolerance, but their role in innate immune responses remains unclear. Here we found that intestinal phagocytes were anergic to ligands for Toll-like receptors (TLRs) or commensals but constitutively expressed the precursor to interleukin 1ß (pro-IL-1ß). After infection with pathogenic Salmonella or Pseudomonas, intestinal phagocytes produced mature IL-1ß through the NLRC4 inflammasome but did not produce tumor necrosis factor (TNF) or IL-6. BALB/c mice deficient in NLRC4 or the IL-1 receptor were highly susceptible to orogastric but not intraperitoneal infection with Salmonella. That enhanced lethality was preceded by impaired expression of endothelial adhesion molecules, lower neutrophil recruitment and poor intestinal pathogen clearance. Thus, NLRC4-dependent production of IL-1ß by intestinal phagocytes represents a specific response that discriminates pathogenic bacteria from commensal bacteria and contributes to host defense in the intestine.


Assuntos
Proteínas Reguladoras de Apoptose/imunologia , Proteínas de Ligação ao Cálcio/imunologia , Anergia Clonal , Interações Hospedeiro-Patógeno/imunologia , Interleucina-1beta/metabolismo , Intestinos/imunologia , Intestinos/microbiologia , Fagócitos/imunologia , Animais , Proteínas Reguladoras de Apoptose/genética , Proteínas de Ligação ao Cálcio/genética , Caspase 1/metabolismo , Flagelina/imunologia , Humanos , Inflamassomos/imunologia , Inflamassomos/metabolismo , Interleucina-6/biossíntese , Interleucina-6/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monócitos/imunologia , Monócitos/metabolismo , Infiltração de Neutrófilos/genética , Infiltração de Neutrófilos/imunologia , Neutrófilos/imunologia , Neutrófilos/patologia , Fagócitos/microbiologia , Pseudomonas/imunologia , Infecções por Pseudomonas/imunologia , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/imunologia , Salmonella/genética , Salmonella/imunologia , Infecções por Salmonella/genética , Infecções por Salmonella/imunologia , Transdução de Sinais/genética , Transdução de Sinais/imunologia , Receptores Toll-Like/imunologia , Receptores Toll-Like/metabolismo , Fator de Necrose Tumoral alfa/biossíntese , Fator de Necrose Tumoral alfa/imunologia
3.
Int Immunol ; 34(8): 409-420, 2022 07 26.
Artigo em Inglês | MEDLINE | ID: mdl-35641096

RESUMO

IL-17 plays important roles in host defense against Candida albicans at barrier surfaces and during invasive infection. However, the role of IL-17 in host defense after colonization of the epidermis, a main site of C. albicans infection, remains poorly understood. Using a murine model of epicutaneous candidiasis without skin abrasion, we found that skin inflammation triggered by epidermal C. albicans colonization was self-limiting with fungal clearance completed by day 7 after inoculation in wild-type mice or animals deficient in IL-17A or IL-17F. In contrast, marked neutrophilic inflammation in the epidermis and impaired fungal clearance were observed in mice lacking both IL-17A and IL-17F. Clearance of C. albicans was independent of Dectin-1, Dectin-2, CARD9 (caspase-recruitment domain family, member 9), TLR2 (Toll-like receptor 2) and MyD88 in the epidermal colonization model. We found that group 3 innate lymphoid cells (ILC3s) and γδT cells were the major IL-17 producers in the epicutaneous candidiasis model. Analyses of Rag2-/- mice and Rag2-/-Il2rg-/- mice revealed that production of IL-17A and IL-17F by ILC3s was sufficient for C. albicans clearance. Finally, we found that depletion of neutrophils impaired C. albicans clearance in the epidermal colonization model. Taken together, these findings indicate a critical and redundant function of IL-17A and IL-17F produced by ILC3s in host defense against C. albicans in the epidermis. The results also suggest that epidermal C. albicans clearance is independent of innate immune receptors or that these receptors act redundantly in fungal recognition and clearance.


Assuntos
Candida albicans , Candidíase , Interleucina-17/imunologia , Animais , Proteínas Adaptadoras de Sinalização CARD , Epiderme/metabolismo , Imunidade Inata , Inflamação , Linfócitos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout
4.
Allergol Int ; 71(3): 310-317, 2022 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-35662539

RESUMO

In October 2021, researchers from the German Society of Allergy and Clinical Immunology (DGAKI) and from the Japanese Society of Allergology (JSA) focused their attention on the pathological conditions and modifiers of various allergic diseases. Topics included 1) the pathophysiology of IgE/mast cell-mediated allergic diseases; 2) the diagnosis and prevention of IgE/mast cell-mediated diseases; 3) the pathophysiology, diagnosis, and treatment of eosinophilic airway diseases; and 4) host-pathogen interaction and allergic diseases. This report summarizes the panel discussions, which highlighted the importance of recognizing the diversity of genetics, immunological mechanisms, and modifying factors underlying allergic diseases.


Assuntos
Hipersensibilidade , Imunoglobulina E , Humanos , Hipersensibilidade/tratamento farmacológico , Hipersensibilidade/terapia
5.
J Infect Dis ; 223(10): 1753-1765, 2021 05 28.
Artigo em Inglês | MEDLINE | ID: mdl-33837391

RESUMO

BACKGROUND: Among skin commensal fungi, lipophilic Malassezia species exist on nearly all human skin surfaces. The pathophysiology of Malassezia-associated skin diseases remains poorly understood due in part to the lack of appropriate animal models. Our objective was to investigate the mechanisms underlying Malassezia-induced skin inflammation using a novel murine model that physiologically recapitulates Malassezia skin infection. METHODS: Mice were inoculated epicutaneously with Malassezia yeasts without barrier disruption and in the absence of external lipid supplementation. Skin inflammation, lesional fungal loads, and expression of cytokines and antimicrobial peptides were evaluated in wild-type and mutant mouse strains. RESULTS: Malassezia-induced skin inflammation and epidermal thickening were observed on day 4 after inoculation in wild-type mice. High fungal burdens were detected in the cornified layer on day 2 and decreased thereafter with near complete clearance by day 7 after inoculation. Malassezia-induced skin inflammation and fungal clearance by the host were interleukin-17 (IL-17) dependent with contribution of group 3 innate lymphoid cells. Moreover, IL-17-dependent skin inflammation was mediated through IL-36 receptor and keratinocyte MyD88 signaling. CONCLUSION: Using a new skin infection model, it is shown that Malassezia-induced IL-17- dependent skin inflammation and control of fungal infection are mediated via keratinocyte IL-36 receptor/MyD88 signaling.


Assuntos
Dermatomicoses/imunologia , Interleucina-17/imunologia , Queratinócitos , Fator 88 de Diferenciação Mieloide , Receptores de Interleucina-1/imunologia , Animais , Peptídeos Antimicrobianos , Imunidade Inata , Inflamação/microbiologia , Linfócitos , Malassezia/patogenicidade , Camundongos , Fator 88 de Diferenciação Mieloide/genética , Fator 88 de Diferenciação Mieloide/metabolismo , Pele
6.
Immunity ; 37(1): 85-95, 2012 Jul 27.
Artigo em Inglês | MEDLINE | ID: mdl-22819042

RESUMO

Cryopyrin-associated periodic syndromes (CAPS) are caused by aberrant interleukin-1ß (IL-1ß) production induced by mutations in the NLRP3 protein in humans, but the mechanisms involved remain poorly understood. Using a mouse model, we show a role for the indigenous microbiota and mast cells (MCs) in skin disease associated with mutant Nlrp3 protein. Unlike normal cells, MCs expressing mutant Nlrp3 produced IL-1ß in response to lipopolysaccharide or tumor necrosis factor-α (TNF-α). In neonatal mice, the microbiota induced TNF-α and IL-1ß and promoted skin disease. MC deficiency greatly reduced disease in Nlrp3 mutant mice, and reconstitution of MC-deficient mice with mutant MCs restored skin disease, which required the expression of IL-1ß in MCs. Surprisingly, neutralization of TNF-α abrogated IL-1ß production and skin disease in neonatal Nlrp3 mutant mice, but not in affected adult mice. Thus, the microbiota and MCs initiate cellular events leading to dysregulated IL-1ß production and skin inflammation in neonatal mice with the CAPS-associated Nlrp3 mutation.


Assuntos
Proteínas de Transporte/genética , Dermatite/genética , Dermatite/imunologia , Interleucina-1beta/metabolismo , Mastócitos/imunologia , Mastócitos/metabolismo , Mutação , Animais , Caspase 1/metabolismo , Dermatite/microbiologia , Feminino , Interleucina-1beta/imunologia , Lipopolissacarídeos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteína 3 que Contém Domínio de Pirina da Família NLR , Dermatopatias/induzido quimicamente , Dermatopatias/genética , Dermatopatias/imunologia , Fator de Necrose Tumoral alfa/administração & dosagem , Fator de Necrose Tumoral alfa/imunologia
7.
J Immunol ; 201(1): 167-182, 2018 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-29794016

RESUMO

IL-36α (gene symbol Il1f6), a member of the IL-36 family, is closely associated with inflammatory diseases, including colitis and psoriasis. In this study, we found that Il1f6-/- mice developed milder psoriasiform dermatitis upon treatment with imiquimod, a ligand for TLR ligand 7 (TLR7) and TLR8, whereas Il1f6-/- mice showed similar susceptibility to dextran sodium sulfate-induced colitis to wild-type mice. These effects were observed in both cohoused and separately housed conditions, and antibiotic treatment did not cancel the resistance of Il1f6-/- mice to imiquimod-induced dermatitis. Bone marrow (BM) cell transfer revealed that IL-36α expression in skin-resident cells is important for the pathogenesis of dermatitis in these mice. Following stimulation with IL-36α, the expression of Il1f6 and Il1f9 (IL-36γ), but not Il1f8 (IL-36ß), was enhanced in murine BM-derived Langerhans cells (BMLCs) and murine primary keratinocytes but not in fibroblasts from mice. Upon stimulation with agonistic ligands of TLRs and C-type lectin receptors (CLRs), Il1f6 expression was induced in BMLCs and BM-derived dendritic cells. Furthermore, IL-36α stimulation resulted in significantly increased gene expression of psoriasis-associated Th17-related cytokines and chemokines such as IL-1α, IL-1ß, IL-23, CXCL1, and CXCL2 in BMLCs and fibroblasts, and IL-1α, IL-1ß, IL-17C, and CXCL2 in keratinocytes. Collectively, these results suggest that TLR/CLR signaling-induced IL-36α plays an important role for the development of psoriasiform dermatitis by enhancing Th17-related cytokine/chemokine production in skin-resident cells via a local autoamplification loop.


Assuntos
Adjuvantes Imunológicos/toxicidade , Quimiocinas/biossíntese , Colite/patologia , Imiquimode/toxicidade , Interleucina-1/metabolismo , Queratinócitos/metabolismo , Psoríase/patologia , Pele/patologia , Células Th17/imunologia , Animais , Células da Medula Óssea/citologia , Transplante de Medula Óssea , Células Cultivadas , Colite/induzido quimicamente , Células Dendríticas/metabolismo , Sulfato de Dextrana/toxicidade , Fibroblastos/metabolismo , Interleucina-1/genética , Células de Langerhans/metabolismo , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Psoríase/tratamento farmacológico , Psoríase/genética , Pele/citologia , Receptor 7 Toll-Like/metabolismo , Receptor 8 Toll-Like/metabolismo
8.
J Infect Dis ; 220(5): 892-901, 2019 07 31.
Artigo em Inglês | MEDLINE | ID: mdl-31107940

RESUMO

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.


Assuntos
Interleucina-17/farmacologia , Lactococcus lactis/fisiologia , Pele/imunologia , Infecções Estafilocócicas/tratamento farmacológico , Infecções Cutâneas Estafilocócicas/imunologia , Staphylococcus aureus/efeitos dos fármacos , Animais , Peptídeos Catiônicos Antimicrobianos/genética , Linfócitos T CD8-Positivos/metabolismo , Calgranulina A/metabolismo , Proliferação de Células , Claudina-1/genética , Claudina-1/metabolismo , Citocinas/genética , Citocinas/metabolismo , Células Dendríticas/imunologia , Modelos Animais de Doenças , Feminino , Homeostase , Interleucina-17/metabolismo , Linfonodos , Ativação Linfocitária , Camundongos Endogâmicos BALB C , Propionibacterium acnes , Pele/microbiologia , Pele/patologia , Infecções Cutâneas Estafilocócicas/microbiologia , Staphylococcus epidermidis , Proteína da Zônula de Oclusão-1/genética , Proteína da Zônula de Oclusão-1/metabolismo , beta-Defensinas/metabolismo
10.
Nature ; 503(7476): 397-401, 2013 Nov 21.
Artigo em Inglês | MEDLINE | ID: mdl-24172897

RESUMO

Atopic dermatitis is a chronic inflammatory skin disease that affects 15-30% of children and approximately 5% of adults in industrialized countries. Although the pathogenesis of atopic dermatitis is not fully understood, the disease is mediated by an abnormal immunoglobulin-E immune response in the setting of skin barrier dysfunction. Mast cells contribute to immunoglobulin-E-mediated allergic disorders including atopic dermatitis. Upon activation, mast cells release their membrane-bound cytosolic granules leading to the release of several molecules that are important in the pathogenesis of atopic dermatitis and host defence. More than 90% of patients with atopic dermatitis are colonized with Staphylococcus aureus in the lesional skin whereas most healthy individuals do not harbour the pathogen. Several staphylococcal exotoxins can act as superantigens and/or antigens in models of atopic dermatitis. However, the role of these staphylococcal exotoxins in disease pathogenesis remains unclear. Here we report that culture supernatants of S. aureus contain potent mast-cell degranulation activity. Biochemical analysis identified δ-toxin as the mast cell degranulation-inducing factor produced by S. aureus. Mast cell degranulation induced by δ-toxin depended on phosphoinositide 3-kinase and calcium (Ca(2+)) influx; however, unlike that mediated by immunoglobulin-E crosslinking, it did not require the spleen tyrosine kinase. In addition, immunoglobulin-E enhanced δ-toxin-induced mast cell degranulation in the absence of antigen. Furthermore, S. aureus isolates recovered from patients with atopic dermatitis produced large amounts of δ-toxin. Skin colonization with S. aureus, but not a mutant deficient in δ-toxin, promoted immunoglobulin-E and interleukin-4 production, as well as inflammatory skin disease. Furthermore, enhancement of immunoglobulin-E production and dermatitis by δ-toxin was abrogated in Kit(W-sh/W-sh) mast-cell-deficient mice and restored by mast cell reconstitution. These studies identify δ-toxin as a potent inducer of mast cell degranulation and suggest a mechanistic link between S. aureus colonization and allergic skin disease.


Assuntos
Toxinas Bacterianas/metabolismo , Degranulação Celular , Dermatite Atópica/microbiologia , Mastócitos/citologia , Staphylococcus aureus/patogenicidade , Animais , Toxinas Bacterianas/farmacologia , Sinalização do Cálcio/efeitos dos fármacos , Degranulação Celular/efeitos dos fármacos , Meios de Cultivo Condicionados/farmacologia , Dermatite Atópica/imunologia , Dermatite Atópica/metabolismo , Dermatite Atópica/patologia , Feminino , Imunoglobulina E/biossíntese , Imunoglobulina E/imunologia , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/microbiologia , Inflamação/patologia , Interleucina-4/imunologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Masculino , Mastócitos/efeitos dos fármacos , Camundongos , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Tirosina Quinases/metabolismo , Proteínas Proto-Oncogênicas c-kit/genética , Proteínas Proto-Oncogênicas c-kit/metabolismo , Staphylococcus aureus/metabolismo , Quinase Syk
11.
J Infect Dis ; 218(6): 1009-1013, 2018 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-29733353

RESUMO

Atopic dermatitis (AD) is a chronic inflammatory skin disease where more than 90% of patients affected are colonized with Staphylococcus aureus. In AD, S. aureus δ-toxin is a major virulence factor causing cutaneous inflammation via mast cell degranulation. δ-toxin is controlled by the S. aureus agr quorum sensing system, and thus we addressed whether interference with agr signaling would limit skin inflammation. Indeed, treatment of S. aureus with the agr-inhibitor solonamide B (SolB) abolished δ-toxin production and reduced skin inflammation in a mouse model of inflammatory skin disease, demonstrating the potential of antivirulence therapy in treating S. aureus-induced skin disorders.


Assuntos
Proteínas de Bactérias/metabolismo , Depsipeptídeos/administração & dosagem , Dermatite Atópica/tratamento farmacológico , Infecções Estafilocócicas/tratamento farmacológico , Staphylococcus aureus/patogenicidade , Transativadores/metabolismo , Animais , Proteínas de Bactérias/genética , Linhagem Celular , Depsipeptídeos/farmacologia , Dermatite Atópica/microbiologia , Modelos Animais de Doenças , Regulação Bacteriana da Expressão Gênica/efeitos dos fármacos , Proteínas Hemolisinas/genética , Humanos , Camundongos , Mutação , Transdução de Sinais , Staphylococcus aureus/efeitos dos fármacos , Staphylococcus aureus/metabolismo , Resultado do Tratamento , Virulência/efeitos dos fármacos
12.
Allergol Int ; 66(4): 539-544, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28882556

RESUMO

Atopic dermatitis (AD) is a chronic inflammatory skin disease that affects 15-20% of children and 2-5% of adults in industrialized countries. The pathogen Staphylococcus aureus selectively colonizes the lesional skin of AD patients while this bacterium is absent in the skin of the majority of healthy individuals. However, the role of S. aureus in the pathogenesis of AD remains poorly understood. In addition to S. aureus, recent studies show a contribution of the skin microbiota to the regulation of immune responses in the skin as well as to the development of inflammatory skin disease. This review summarizes current knowledge about the role of the microbiota in skin immune responses and the role of S. aureus virulent factors in the pathogenesis of AD.


Assuntos
Dermatite Atópica/etiologia , Interações Hospedeiro-Patógeno/imunologia , Microbiota , Pele/imunologia , Pele/microbiologia , Animais , Biomarcadores , Dermatite Atópica/metabolismo , Dermatite Atópica/patologia , Modelos Animais de Doenças , Humanos , Imunomodulação , Fenótipo , Índice de Gravidade de Doença , Transdução de Sinais , Staphylococcus aureus/patogenicidade , Staphylococcus aureus/fisiologia , Virulência , Fatores de Virulência/imunologia
13.
J Biol Chem ; 289(2): 1142-50, 2014 Jan 10.
Artigo em Inglês | MEDLINE | ID: mdl-24265316

RESUMO

The NLRP3 inflammasome is a critical component of the innate immune system. NLRP3 activation is induced by diverse stimuli associated with bacterial infection or tissue damage, but its inappropriate activation is involved in the pathogenesis of inherited and acquired inflammatory diseases. However, the mechanism by which NLRP3 is activated remains poorly understood. In this study, we explored the role of kinases in NLRP3 inflammasome activation by screening a kinase inhibitor library and identified 3,4-methylenedioxy-ß-nitrostyrene (MNS) as an inhibitor for NLRP3 inflammasome activation. Notably, MNS did not affect the activation of the NLRC4 or AIM2 (absent in melanoma 2) inflammasome. Mechanistically, MNS specifically prevented NLRP3-mediated ASC speck formation and oligomerization without blocking potassium efflux induced by NLRP3 agonists. Surprisingly, Syk kinase, the reported target of MNS, did not mediate the inhibitory activity of MNS on NLRP3 inflammasome activation. We also found that the nitrovinyl group of MNS is essential for the inhibitory activity of MNS. Immunoprecipitation, mass spectrometry, and mutation studies suggest that both the nucleotide binding oligomerization domain and the leucine-rich repeat domain of NLRP3 were the intracellular targets of MNS. Administration of MNS also inhibited NLRP3 ATPase activity in vitro, suggesting that MNS blocks the NLRP3 inflammasome by directly targeting NLRP3 or NLRP3-associated complexes. These studies identified a novel chemical probe for studying the molecular mechanism of NLRP3 inflammasome activation which may advance the development of novel strategies to treat diseases associated with abnormal activation of NLRP3 inflammasome.


Assuntos
Proteínas de Transporte/metabolismo , Dioxolanos/farmacologia , Inflamassomos/metabolismo , Macrófagos/efeitos dos fármacos , Adenosina Trifosfatases/antagonistas & inibidores , Adenosina Trifosfatases/genética , Adenosina Trifosfatases/metabolismo , Trifosfato de Adenosina/metabolismo , Trifosfato de Adenosina/farmacologia , Animais , Western Blotting , Proteínas de Transporte/genética , Células Cultivadas , Dioxolanos/química , Inibidores Enzimáticos/farmacologia , Células HEK293 , Humanos , Inflamassomos/genética , Interleucina-1beta/metabolismo , Peptídeos e Proteínas de Sinalização Intracelular/antagonistas & inibidores , Peptídeos e Proteínas de Sinalização Intracelular/genética , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/citologia , Macrófagos/metabolismo , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Microscopia de Fluorescência , Estrutura Molecular , Proteína 3 que Contém Domínio de Pirina da Família NLR , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/genética , Proteínas Tirosina Quinases/metabolismo , Quinase Syk , Fator de Necrose Tumoral alfa/metabolismo
14.
Inflamm Regen ; 44(1): 9, 2024 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-38429810

RESUMO

BACKGROUND: Staphylococcus aureus is a leading cause of human bacterial infections worldwide. It is the most common causative agent of skin and soft tissue infections, and can also cause various other infections, including pneumonia, osteomyelitis, as well as life-threatening infections, such as sepsis and infective endocarditis. The pathogen can also asymptomatically colonize human skin, nasal cavity, and the intestine. S. aureus colonizes approximately 20-30% of human nostrils, being an opportunistic pathogen for subsequent infection. Its strong ability to silently spread via human contact makes it difficult to eradicate S. aureus. A major concern with S. aureus is its capacity to develop antibiotic resistance and adapt to diverse environmental conditions. The variability in the accessory gene regulator (Agr) region of the genome contributes to a spectrum of phenotypes within the bacterial population, enhancing the likelihood of survival in different environments. Agr functions as a central quorum sensing (QS) system in S. aureus, allowing bacteria to adjust gene expression in response to population density. Depending on Agr expression, S. aureus secretes various toxins, contributing to virulence in infectious diseases. Paradoxically, expressing Agr may be disadvantageous in certain situations, such as in hospitals, causing S. aureus to generate Agr mutants responsible for infections in healthcare settings. MAIN BODY: This review aims to demonstrate the molecular mechanisms governing the diverse phenotypes of S. aureus, ranging from a harmless colonizer to an organism capable of infecting various human organs. Emphasis will be placed on QS and its role in orchestrating S. aureus behavior across different contexts. SHORT CONCLUSION: The pathophysiology of S. aureus infection is substantially influenced by phenotypic changes resulting from factors beyond Agr. Future studies are expected to give the comprehensive understanding of S. aureus overall profile in various settings.

15.
Front Pharmacol ; 15: 1365815, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38659576

RESUMO

The virulence of Staphylococcus aureus, including methicillin-resistant S. aureus (MRSA), depends on the expression of toxins and virulence factors controlled by the quorum-sensing (QS) system, encoded on the virulence accessory gene regulator (agr) locus. The aim of this study was to identify a phytochemical that inhibits Agr-QS function and to elucidate its mechanism. We screened 577 compounds and identified physalin H, physalin B, and isophysalin B--phytochemicals belonging to physalins found in plants of the Solanaceae family--as novel Agr-QS modulators. Biological analyses and in vitro protein-DNA binding assays suggested that these physalins suppress gene expression related to the Agr-QS system by inhibiting binding of the key response regulator AgrA to the agr promoters, reducing the function of hemolytic toxins downstream of these genes in MRSA. Furthermore, although physalin F suppressed gene expression in the Agr-QS system, its anti-hemolytic activity was lower than that of physalins H, B, and isophysalin B. Conversely, five physalins isolated from the same plant with the ability to suppress Agr-QS did not reduce bacterial Agr-QS activity but inhibited AgrA binding to DNA in vitro. A docking simulation revealed that physalin interacts with the DNA-binding site of AgrA in three docking states. The carbonyl oxygens at C-1 and C-18 of physalins, which can suppress Agr-QS, were directed to residues N201 and R198 of AgrA, respectively, whereas these carbonyl oxygens of physalins, without Agr-QS suppression activity, were oriented in different directions. Next, 100-ns molecular dynamics simulations revealed that the hydrogen bond formed between the carbonyl oxygen at C-15 of physalins and L186 of AgrA functions as an anchor, sustaining the interaction between the carbonyl oxygen at C-1 of physalins and N201 of AgrA. Thus, these results suggest that physalin H, physalin B, and isophysalin B inhibit the interaction of AgrA with the agr promoters by binding to the DNA-binding site of AgrA, suppressing the Agr-QS function of S. aureus. Physalins that suppress the Agr-QS function are proposed as potential lead compounds in the anti-virulence strategy for MRSA infections.

16.
Front Cell Infect Microbiol ; 13: 1178650, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37124047

RESUMO

The skin is home to various bacteria, archaea, fungi, and viruses, collectively referred to as the skin microbiota. Patients with certain skin diseases reportedly have unique skin "dysbiosis," a condition involving imbalanced microbiota, suggesting that dysbiosis in the skin may be either causal or a consequence of specific skin diseases. Atopic dermatitis (AD) is the most common allergic skin disease that affects 15-20% of children and 2-10% of adults worldwide. Both intrinsic genetic factors, such as susceptibility to type 2 inflammation or skin barrier dysfunction, and extrinsic environmental factors, such as air pollen and skin microbiota, contribute to AD. Staphylococcus aureus, which does not often colonize the skin of healthy individuals, is commonly identified in the lesional skin of patients with AD and is correlated with the disease flare. However, the role of S. aureus in the pathogenesis of AD has not been elucidated. Here, we discuss the pathological behavior of S. aureus, focusing on accessory gene regulator (Agr) quorum sensing, which is a fundamental bacterial cell-to-cell interaction mechanism that affects the behavior of S. aureus and other members of the microbial community. Importantly, beyond bacteria-bacteria interactions, the Agr quorum sensing system also regulates various virulence factors, which induce type 2 and IL-17-dependent skin inflammation in the host. Furthermore, the colonization of Agr-positive S. aureus in early life accelerates the development of pediatric AD. Finally, we aim to highlight the current efforts to establish novel therapeutic methods to ameliorate or prevent AD through Agr-targeted intervention.


Assuntos
Dermatite Atópica , Infecções Estafilocócicas , Adulto , Humanos , Criança , Staphylococcus aureus , Percepção de Quorum , Disbiose , Staphylococcus , Inflamação/patologia , Infecções Estafilocócicas/microbiologia , Bactérias
17.
Micromachines (Basel) ; 14(5)2023 May 11.
Artigo em Inglês | MEDLINE | ID: mdl-37241650

RESUMO

In this study, we developed a comb-shaped microfluidic device that can efficiently trap and culture a single cell (bacterium). Conventional culture devices have difficulty in trapping a single bacterium and often use a centrifuge to push the bacterium into the channel. The device developed in this study can store bacteria in almost all growth channels using the flowing fluid. In addition, chemical replacement can be performed in a few seconds, making this device suitable for culture experiments with resistant bacteria. The storage efficiency of microbeads that mimic bacteria was significantly improved from 0.2% to 84%. We used simulations to investigate the pressure loss in the growth channel. The pressure in the growth channel of the conventional device was more than 1400 PaG, whereas that of the new device was less than 400 PaG. Our microfluidic device was easily fabricated by a soft microelectromechanical systems method. The device was highly versatile and can be applied to various bacteria, such as Salmonella enterica serovar Typhimurium and Staphylococcus aureus.

18.
J Diabetes Investig ; 14(9): 1136-1139, 2023 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-37293689

RESUMO

Psoriasis is a chronic inflammatory skin disease that is associated with obesity and myocardial infarction. Obesity-induced changes in lipid metabolism promote T helper 17 (Th17) cell differentiation, which in turn promotes chronic inflammation. Th17 cells have central roles in many inflammatory diseases, including psoriasis and atherosclerosis; however, whether treatment of obesity attenuates Th17 cells and chronic inflammatory diseases has been unknown. In this study, we found an increase in Th17 cells in a patient with obesity, type 2 diabetes and psoriasis. Furthermore, weight loss with diet and exercise resulted in a decrease in Th17 cells and improvement of psoriasis. This case supports the hypothesis that obesity leads to an increase in Th17 cells and chronic inflammation of the skin and blood vessel walls, thereby promoting psoriasis and atherosclerosis.


Assuntos
Aterosclerose , Diabetes Mellitus Tipo 2 , Psoríase , Humanos , Células Th17/metabolismo , Diabetes Mellitus Tipo 2/metabolismo , Psoríase/complicações , Inflamação/complicações , Inflamação/metabolismo , Obesidade/complicações , Obesidade/metabolismo , Redução de Peso
20.
Microorganisms ; 9(3)2021 Mar 09.
Artigo em Inglês | MEDLINE | ID: mdl-33803200

RESUMO

(1) Background: Lactococcus lactis strain Plasma (LC-Plasma) is a unique strain which directly activates plasmacytoid dendritic cells, resulting in the prevention against broad spectrum of viral infection. Additionally, we found that LC-Plasma intake stimulated skin immunity and prevents Staphylococcus aureus epicutaneous infection. The aim of this study was to investigate the effect of LC-Plasma dietary supplementation on skin microbiome, gene expression in the skin, and skin conditions in healthy subjects. (2) Method: A randomized, double-blind, placebo-controlled, parallel-group trial was conducted. Seventy healthy volunteers were enrolled and assigned into two groups receiving either placebo or LC-Plasma capsules (approximately 1 × 1011 cells/day) for 8 weeks. The skin microbiome was analyzed by NGS and qPCR. Gene expression was analyzed by qPCR and skin conditions were diagnosed by dermatologists before and after intervention. (3) Result: LC-Plasma supplementation prevented the decrease of Staphylococcus epidermidis and Staphylococcus pasteuri and overgrowth of Propionibacterium acnes. In addition, LC-Plasma supplementation suggested to increase the expression of antimicrobial peptide genes but not tight junction genes. Furthermore, the clinical scores of skin conditions were ameliorated by LC-Plasma supplementation. (4) Conclusions: Our findings provided the insights that the dietary supplementation of LC-Plasma might have stabilizing effects on seasonal change of skin microbiome and skin conditions in healthy subjects.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA