Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 756
Filtrar
Mais filtros

Tipo de documento
Intervalo de ano de publicação
1.
Annu Rev Immunol ; 37: 377-403, 2019 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-31026410

RESUMO

The gut-associated lymphoid tissue (GALT) faces a considerable challenge. It encounters antigens derived from an estimated 1014 commensal microbes and greater than 30 kg of food proteins yearly. It must distinguish these harmless antigens from potential pathogens and mount the appropriate host immune response. Local and systemic hyporesponsiveness to dietary antigens, classically referred to as oral tolerance, comprises a distinct complement of adaptive cellular and humoral immune responses. It is increasingly evident that a functional epithelial barrier engaged in intimate interplay with innate immune cells and the resident microbiota is critical to establishing and maintaining oral tolerance. Moreover, innate immune cells serve as a bridge between the microbiota, epithelium, and the adaptive immune system, parlaying tonic microbial stimulation into signals critical for mucosal homeostasis. Dysregulation of gut homeostasis and the subsequent disruption of tolerance therefore have clinically significant consequences for the development of food allergy.


Assuntos
Disbiose/imunologia , Hipersensibilidade Alimentar/imunologia , Microbioma Gastrointestinal/imunologia , Mucosa Intestinal/imunologia , Administração Oral , Alérgenos/imunologia , Animais , Alimentos , Hipersensibilidade Alimentar/microbiologia , Homeostase , Humanos , Tolerância Imunológica , Imunidade Inata , Mucosa Intestinal/microbiologia
2.
Annu Rev Immunol ; 34: 31-64, 2016 05 20.
Artigo em Inglês | MEDLINE | ID: mdl-27168239

RESUMO

Inflammatory bowel disease (IBD), including Crohn disease and ulcerative colitis, is characterized by chronic intestinal inflammation due to a complex interaction of genetic determinants, disruption of mucosal barriers, aberrant inflammatory signals, loss of tolerance, and environmental triggers. Importantly, the incidence of pediatric IBD is rising, particularly in children younger than 10 years. In this review, we discuss the clinical presentation of these patients and highlight environmental exposures that may affect disease risk, particularly among people with a background genetic risk. With regard to both children and adults, we review advancements in understanding the intestinal epithelium, the mucosal immune system, and the resident microbiota, describing how dysfunction at any level can lead to diseases like IBD. We conclude with future directions for applying advances in IBD genetics to better understand pathogenesis and develop therapeutics targeting key pathogenic nodes.


Assuntos
Disbiose/imunologia , Microbioma Gastrointestinal/imunologia , Imunidade nas Mucosas , Inflamação/imunologia , Doenças Inflamatórias Intestinais/imunologia , Mucosa Intestinal/imunologia , Adulto , Animais , Criança , Pré-Escolar , Exposição Ambiental/efeitos adversos , Interação Gene-Ambiente , Predisposição Genética para Doença , Humanos , Doenças Inflamatórias Intestinais/genética , Doenças Inflamatórias Intestinais/terapia , Terapia de Alvo Molecular
3.
Cell ; 167(4): 1125-1136.e8, 2016 11 03.
Artigo em Inglês | MEDLINE | ID: mdl-27814509

RESUMO

Gut microbial dysbioses are linked to aberrant immune responses, which are often accompanied by abnormal production of inflammatory cytokines. As part of the Human Functional Genomics Project (HFGP), we investigate how differences in composition and function of gut microbial communities may contribute to inter-individual variation in cytokine responses to microbial stimulations in healthy humans. We observe microbiome-cytokine interaction patterns that are stimulus specific, cytokine specific, and cytokine and stimulus specific. Validation of two predicted host-microbial interactions reveal that TNFα and IFNγ production are associated with specific microbial metabolic pathways: palmitoleic acid metabolism and tryptophan degradation to tryptophol. Besides providing a resource of predicted microbially derived mediators that influence immune phenotypes in response to common microorganisms, these data can help to define principles for understanding disease susceptibility. The three HFGP studies presented in this issue lay the groundwork for further studies aimed at understanding the interplay between microbial, genetic, and environmental factors in the regulation of the immune response in humans. PAPERCLIP.


Assuntos
Citocinas/imunologia , Microbioma Gastrointestinal , Inflamação/imunologia , Microbiota , Adolescente , Adulto , Idoso , Bactérias/classificação , Bactérias/imunologia , Sangue/imunologia , Disbiose/imunologia , Disbiose/microbiologia , Fezes/microbiologia , Feminino , Fungos/classificação , Fungos/imunologia , Interação Gene-Ambiente , Projeto Genoma Humano , Humanos , Infecções/imunologia , Infecções/microbiologia , Leucócitos Mononucleares/imunologia , Masculino , Pessoa de Meia-Idade
4.
Nat Immunol ; 19(12): 1286-1298, 2018 12.
Artigo em Inglês | MEDLINE | ID: mdl-30446754

RESUMO

The skin provides both a physical barrier and an immunologic barrier to external threats. The protective machinery of the skin has evolved to provide situation-specific responses to eliminate pathogens and to provide protection against physical dangers. Dysregulation of this machinery can give rise to the initiation and propagation of inflammatory loops in the epithelial microenvironment that result in inflammatory skin diseases in susceptible people. A defective barrier and microbial dysbiosis drive an interleukin 4 (IL-4) loop that underlies atopic dermatitis, while in psoriasis, disordered keratinocyte signaling and predisposition to type 17 responses drive a pathogenic IL-17 loop. Here we discuss the pathogenesis of atopic dermatitis and psoriasis in terms of the epithelial immune microenvironment-the microbiota, keratinocytes and sensory nerves-and the resulting inflammatory loops.


Assuntos
Dermatite Atópica/imunologia , Psoríase/imunologia , Pele/imunologia , Animais , Dermatite Atópica/microbiologia , Dermatite Atópica/fisiopatologia , Disbiose/imunologia , Disbiose/microbiologia , Disbiose/fisiopatologia , Epitélio/imunologia , Epitélio/microbiologia , Epitélio/fisiopatologia , Humanos , Psoríase/microbiologia , Psoríase/fisiopatologia , Pele/microbiologia
5.
Immunity ; 54(6): 1137-1153.e8, 2021 06 08.
Artigo em Inglês | MEDLINE | ID: mdl-34051146

RESUMO

Alterations in the cGAS-STING DNA-sensing pathway affect intestinal homeostasis. We sought to delineate the functional role of STING in intestinal inflammation. Increased STING expression was a feature of intestinal inflammation in mice with colitis and in humans afflicted with inflammatory bowel disease. Mice bearing an allele rendering STING constitutively active exhibited spontaneous colitis and dysbiosis, as well as progressive chronic intestinal inflammation and fibrosis. Bone marrow chimera experiments revealed STING accumulation in intestinal macrophages and monocytes as the initial driver of inflammation. Depletion of Gram-negative bacteria prevented STING accumulation in these cells and alleviated intestinal inflammation. STING accumulation occurred at the protein rather than transcript level, suggesting post-translational stabilization. We found that STING was ubiquitinated in myeloid cells, and this K63-linked ubiquitination could be elicited by bacterial products, including cyclic di-GMP. Our findings suggest a positive feedback loop wherein dysbiosis foments the accumulation of STING in intestinal myeloid cells, driving intestinal inflammation.


Assuntos
Colite/imunologia , Disbiose/imunologia , Imunidade Inata/imunologia , Proteínas de Membrana/imunologia , Células Mieloides/imunologia , Ubiquitinação/imunologia , Animais , Estudos de Casos e Controles , Feminino , Humanos , Inflamação/imunologia , Intestinos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Monócitos/imunologia
6.
Immunity ; 54(10): 2321-2337.e10, 2021 10 12.
Artigo em Inglês | MEDLINE | ID: mdl-34582748

RESUMO

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


Assuntos
Proteína ADAM10/imunologia , Secretases da Proteína Precursora do Amiloide/imunologia , Disbiose/imunologia , Folículo Piloso/patologia , Linfócitos/imunologia , Proteínas de Membrana/imunologia , Receptores Notch/imunologia , Pele/microbiologia , Alopecia/imunologia , Alopecia/patologia , Animais , Corynebacterium , Disbiose/patologia , Feminino , Folículo Piloso/imunologia , Imunidade Inata , Inflamação/imunologia , Inflamação/metabolismo , Inflamação/patologia , Camundongos , Transdução de Sinais/imunologia , Pele/imunologia , Pele/patologia
7.
Immunity ; 52(2): 241-255, 2020 02 18.
Artigo em Inglês | MEDLINE | ID: mdl-32075727

RESUMO

Asthma is a common chronic respiratory disease affecting more than 300 million people worldwide. Clinical features of asthma and its immunological and molecular etiology vary significantly among patients. An understanding of the complexities of asthma has evolved to the point where precision medicine approaches, including microbiome analysis, are being increasingly recognized as an important part of disease management. Lung and gut microbiota play several important roles in the development, regulation, and maintenance of healthy immune responses. Dysbiosis and subsequent dysregulation of microbiota-related immunological processes affect the onset of the disease, its clinical characteristics, and responses to treatment. Bacteria and viruses are the most extensively studied microorganisms relating to asthma pathogenesis, but other microbes, including fungi and even archaea, can potently influence airway inflammation. This review focuses on recently discovered connections between lung and gut microbiota, including bacteria, fungi, viruses, and archaea, and their influence on asthma.


Assuntos
Asma/imunologia , Asma/microbiologia , Trato Gastrointestinal , Pulmão , Microbiota/imunologia , Animais , Asma/patologia , Asma/fisiopatologia , Disbiose/imunologia , Trato Gastrointestinal/imunologia , Trato Gastrointestinal/microbiologia , Trato Gastrointestinal/parasitologia , Trato Gastrointestinal/virologia , Humanos , Pulmão/imunologia , Pulmão/microbiologia , Pulmão/parasitologia , Pulmão/virologia , Sistema Respiratório/imunologia , Sistema Respiratório/microbiologia , Sistema Respiratório/parasitologia , Sistema Respiratório/virologia
8.
Cell ; 158(5): 1000-1010, 2014 Aug 28.
Artigo em Inglês | MEDLINE | ID: mdl-25171403

RESUMO

Specific members of the intestinal microbiota dramatically affect inflammatory bowel disease (IBD) in mice. In humans, however, identifying bacteria that preferentially affect disease susceptibility and severity remains a major challenge. Here, we used flow-cytometry-based bacterial cell sorting and 16S sequencing to characterize taxa-specific coating of the intestinal microbiota with immunoglobulin A (IgA-SEQ) and show that high IgA coating uniquely identifies colitogenic intestinal bacteria in a mouse model of microbiota-driven colitis. We then used IgA-SEQ and extensive anaerobic culturing of fecal bacteria from IBD patients to create personalized disease-associated gut microbiota culture collections with predefined levels of IgA coating. Using these collections, we found that intestinal bacteria selected on the basis of high coating with IgA conferred dramatic susceptibility to colitis in germ-free mice. Thus, our studies suggest that IgA coating identifies inflammatory commensals that preferentially drive intestinal disease. Targeted elimination of such bacteria may reduce, reverse, or even prevent disease development.


Assuntos
Colite Ulcerativa/imunologia , Doença de Crohn/imunologia , Imunoglobulina A/imunologia , Microbiota , Animais , Colite Ulcerativa/microbiologia , Colite Ulcerativa/patologia , Doença de Crohn/microbiologia , Doença de Crohn/patologia , DNA Bacteriano/análise , Disbiose/imunologia , Disbiose/microbiologia , Humanos , Inflamassomos/imunologia , Inflamação/imunologia , Inflamação/microbiologia , Intestinos/imunologia , Intestinos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , RNA Ribossômico 16S/análise , Organismos Livres de Patógenos Específicos
9.
Cell ; 156(1-2): 109-22, 2014 Jan 16.
Artigo em Inglês | MEDLINE | ID: mdl-24439372

RESUMO

Interactions between commensals and the host impact the metabolic and immune status of metazoans. Their deregulation is associated with age-related pathologies like chronic inflammation and cancer, especially in barrier epithelia. Maintaining a healthy commensal population by preserving innate immune homeostasis in such epithelia thus promises to promote health and longevity. Here, we show that, in the aging intestine of Drosophila, chronic activation of the transcription factor Foxo reduces expression of peptidoglycan recognition protein SC2 (PGRP-SC2), a negative regulator of IMD/Relish innate immune signaling, and homolog of the anti-inflammatory molecules PGLYRP1-4. This repression causes deregulation of Rel/NFkB activity, resulting in commensal dysbiosis, stem cell hyperproliferation, and epithelial dysplasia. Restoring PGRP-SC2 expression in enterocytes of the intestinal epithelium, in turn, prevents dysbiosis, promotes tissue homeostasis, and extends lifespan. Our results highlight the importance of commensal control for lifespan of metazoans and identify SC-class PGRPs as longevity-promoting factors.


Assuntos
Proteínas de Transporte/metabolismo , Drosophila melanogaster/microbiologia , Drosophila melanogaster/fisiologia , Imunidade Inata , Longevidade/imunologia , Modelos Animais , Animais , Citocinas/imunologia , Proteínas de Drosophila/metabolismo , Drosophila melanogaster/imunologia , Disbiose/imunologia , Disbiose/microbiologia , Fatores de Transcrição Forkhead/metabolismo , Homeostase , Intestinos/imunologia , Intestinos/microbiologia , Transcriptoma
11.
Nat Immunol ; 17(3): 230-40, 2016 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-26882261

RESUMO

Inflammation is emerging as one of the hallmarks of cancer, yet its role in most tumors remains unclear. Whereas a minority of solid tumors are associated with overt inflammation, long-term treatment with non-steroidal anti-inflammatory drugs is remarkably effective in reducing cancer rate and death. This indicates that inflammation might have many as-yet-unrecognized facets, among which an indolent course might be far more prevalent than previously appreciated. In this Review, we explore the various inflammatory processes underlying the development and progression of colorectal cancer and discuss anti-inflammatory means for its prevention and treatment.


Assuntos
Adenocarcinoma/imunologia , Adenoma/imunologia , Carcinogênese/imunologia , Neoplasias Colorretais/imunologia , Disbiose/imunologia , Microbioma Gastrointestinal/imunologia , Inflamação , Doenças Inflamatórias Intestinais/imunologia , Mucosa Intestinal/imunologia , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/prevenção & controle , Animais , Anti-Inflamatórios não Esteroides/uso terapêutico , Neoplasias Colorretais/tratamento farmacológico , Neoplasias Colorretais/prevenção & controle , Dano ao DNA/imunologia , Progressão da Doença , Humanos , Imunidade Inata/imunologia , Imunidade nas Mucosas/imunologia , Interleucina-1beta/antagonistas & inibidores , Janus Quinases/antagonistas & inibidores , Receptor de Morte Celular Programada 1/antagonistas & inibidores , Receptores de Interleucina-6/antagonistas & inibidores , Fatores de Transcrição STAT/antagonistas & inibidores , Fator de Necrose Tumoral alfa/antagonistas & inibidores
12.
Immunity ; 51(2): 214-224, 2019 08 20.
Artigo em Inglês | MEDLINE | ID: mdl-31433969

RESUMO

In ecological terms, the microbiome is defined as the microbiota and its environment, a definition that encompasses the human host. The size, species composition, and biogeography of microbial communities is shaped by host interactions, and, in turn, the microbiota influences many aspects of human health. Here we discuss the concept of microbiota-nourishing immunity, a host-microbe chimera composed of the microbiota and host factors that shape the microbial ecosystem, which functions in conferring colonization resistance against pathogens. We propose that dysbiosis is a biomarker of a weakening in microbiota-nourishing immunity and that homeostasis can be defined as a state of immune competence. Microbiota-nourishing immunity thus provides a conceptual framework to further examine the mechanisms that preserve a healthy microbiome and the drivers and consequences of dysbiosis.


Assuntos
Disbiose/imunologia , Interações Hospedeiro-Patógeno , Microbiota/imunologia , Animais , Autoantígenos/imunologia , Homeostase , Humanos , Imunidade , Imunocompetência , Tolerância a Antígenos Próprios
13.
Immunity ; 49(3): 504-514.e4, 2018 09 18.
Artigo em Inglês | MEDLINE | ID: mdl-30231984

RESUMO

The adaptor protein CARD9 links detection of fungi by surface receptors to the activation of the NF-κB pathway. Mice deficient in CARD9 exhibit dysbiosis and are more susceptible to colitis. Here we examined the impact of Card9 deficiency in the development of colitis-associated colon cancer (CAC). Treatment of Card9-/- mice with AOM-DSS resulted in increased tumor loads as compared to WT mice and in the accumulation of myeloid-derived suppressor cells (MDSCs) in tumor tissue. The impaired fungicidal functions of Card9-/- macrophages led to increased fungal loads and variation in the overall composition of the intestinal mycobiota, with a notable increase in C. tropicalis. Bone marrow cells incubated with C. tropicalis exhibited MDSC features and suppressive functions. Fluconazole treatment suppressed CAC in Card9-/- mice and was associated with decreased MDSC accumulation. The frequency of MDSCs in tumor tissues of colon cancer patients correlated positively with fungal burden, pointing to the relevance of this regulatory axis in human disease.


Assuntos
Proteínas Adaptadoras de Sinalização CARD/metabolismo , Colite/imunologia , Neoplasias do Colo/imunologia , Disbiose/imunologia , Microbioma Gastrointestinal/imunologia , Células Supressoras Mieloides/fisiologia , Animais , Proteínas Adaptadoras de Sinalização CARD/genética , Proliferação de Células , Células Cultivadas , Técnicas de Cocultura , Colite/induzido quimicamente , Colite/genética , Neoplasias do Colo/genética , Disbiose/genética , Humanos , Interferon gama/genética , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Células Supressoras Mieloides/microbiologia , Regiões Promotoras Genéticas/genética
14.
Immunity ; 47(2): 339-348.e4, 2017 08 15.
Artigo em Inglês | MEDLINE | ID: mdl-28801232

RESUMO

The gut microbiota regulate susceptibility to multiple human diseases. The Nlrp6-ASC inflammasome is widely regarded as a hallmark host innate immune axis that shapes the gut microbiota composition. This notion stems from studies reporting dysbiosis in mice lacking these inflammasome components when compared with non-littermate wild-type animals. Here, we describe microbial analyses in inflammasome-deficient mice while minimizing non-genetic confounders using littermate-controlled Nlrp6-deficient mice and ex-germ-free littermate-controlled ASC-deficient mice that were all allowed to shape their gut microbiota naturally after birth. Careful microbial phylogenetic analyses of these cohorts failed to reveal regulation of the gut microbiota composition by the Nlrp6- and ASC-dependent inflammasomes. Our results obtained in two geographically separated animal facilities dismiss a generalizable impact of Nlrp6- and ASC-dependent inflammasomes on the composition of the commensal gut microbiota and highlight the necessity for littermate-controlled experimental design in assessing the influence of host immunity on gut microbial ecology.


Assuntos
Proteínas Reguladoras de Apoptose/metabolismo , Bactérias/genética , Colite/imunologia , Disbiose/imunologia , Microbioma Gastrointestinal/imunologia , Inflamassomos/metabolismo , Receptores de Superfície Celular/metabolismo , Animais , Proteínas Adaptadoras de Sinalização CARD , Células Cultivadas , Colite/induzido quimicamente , Colite/microbiologia , Disbiose/microbiologia , Feminino , Patrimônio Genético , Imunidade Inata , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Microbiota , RNA Ribossômico 16S/análise , Receptores de Superfície Celular/genética , Dodecilsulfato de Sódio
15.
Gastroenterology ; 166(6): 995-1019, 2024 06.
Artigo em Inglês | MEDLINE | ID: mdl-38342195

RESUMO

Autoimmune liver diseases include primary biliary cholangitis, primary sclerosing cholangitis, and autoimmune hepatitis, a family of chronic immune-mediated disorders that target hepatocytes and cholangiocytes. Treatments remain nonspecific, variably effective, and noncurative, and the need for liver transplantation is disproportionate to their rarity. Development of effective therapies requires better knowledge of pathogenic mechanisms, including the roles of genetic risk, and how the environment and gut dysbiosis cause immune cell dysfunction and aberrant bile acid signaling. This review summarizes key etiologic and pathogenic concepts and themes relevant for clinical practice and how such learning can guide the development of new therapies for people living with autoimmune liver diseases.


Assuntos
Colangite Esclerosante , Hepatite Autoimune , Cirrose Hepática Biliar , Humanos , Colangite Esclerosante/imunologia , Hepatite Autoimune/imunologia , Cirrose Hepática Biliar/imunologia , Cirrose Hepática Biliar/etiologia , Animais , Autoimunidade , Microbioma Gastrointestinal/imunologia , Fatores de Risco , Predisposição Genética para Doença , Disbiose/imunologia , Ácidos e Sais Biliares/metabolismo
16.
Int Immunol ; 36(7): 365-371, 2024 Jun 08.
Artigo em Inglês | MEDLINE | ID: mdl-38442194

RESUMO

The intestinal barrier consists of mucosal, epithelial, and immunological barriers and serves as a dynamic interface between the host and its environment. Disruption of the intestinal barrier integrity is a leading cause of various gastrointestinal diseases, such as inflammatory bowel disease. The homeostasis of the intestinal barrier is tightly regulated by crosstalk between gut microbes and the immune system; however, the implication of the immune system on the imbalance of gut microbes that disrupts barrier integrity remains to be fully elucidated. An inhibitory immunoglobulin-like receptor, Allergin-1, is expressed on mast cells and dendritic cells and inhibits Toll-like receptor (TLR)-2 and TLR-4 signaling in these cells. Since TLRs are major sensors of microbiota and are involved in local epithelial homeostasis, we investigated the role of Allergin-1 in maintaining intestinal homeostasis. Allergin-1-deficient (Milr1-/-) mice exhibited more severe dextran sulfate sodium (DSS)-induced colitis than did wild-type (WT) mice. Milr1-/- mice showed an enhanced intestinal permeability compared with WT mice even before DSS administration. Treatment of Milr1-/- mice with neomycin, but not ampicillin, restored intestinal barrier integrity. The 16S rRNA gene sequencing analysis demonstrated that Bifidobacterium pseudolongum was the dominant bacterium in Milr1-/- mice after treatment with ampicillin. Although the transfer of B. pseudolongum to germ-free WT mice had no effect on intestinal permeability, its transfer into ampicillin-treated WT mice enhanced intestinal permeability. These results demonstrated that Allergin-1 deficiency enhanced intestinal dysbiosis with expanded B. pseudolongum, which contributes to intestinal barrier dysfunction in collaboration with neomycin-sensitive and ampicillin-resistant microbiota.


Assuntos
Disbiose , Mucosa Intestinal , Camundongos Endogâmicos C57BL , Camundongos Knockout , Animais , Disbiose/imunologia , Camundongos , Mucosa Intestinal/imunologia , Mucosa Intestinal/microbiologia , Mucosa Intestinal/metabolismo , Sulfato de Dextrana , Microbioma Gastrointestinal/imunologia , Colite/imunologia , Colite/microbiologia , Receptores Imunológicos/metabolismo , Receptores Imunológicos/genética , Receptores Imunológicos/imunologia , Neomicina/farmacologia , Permeabilidade
17.
Int Arch Allergy Immunol ; 185(5): 489-502, 2024.
Artigo em Inglês | MEDLINE | ID: mdl-38253027

RESUMO

BACKGROUND: Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection can result in a prolonged multisystem disorder termed long COVID, which may affect up to 10% of people following coronavirus disease 2019 (COVID-19). It is currently unclear why certain individuals do not fully recover following SARS-CoV-2 infection. SUMMARY: In this review, we examine immunological mechanisms that may underpin the pathophysiology of long COVID. These mechanisms include an inappropriate immune response to acute SARS-CoV-2 infection, immune cell exhaustion, immune cell metabolic reprogramming, a persistent SARS-CoV-2 reservoir, reactivation of other viruses, inflammatory responses impacting the central nervous system, autoimmunity, microbiome dysbiosis, and dietary factors. KEY MESSAGES: Unfortunately, the currently available diagnostic and treatment options for long COVID are inadequate, and more clinical trials are needed that match experimental interventions to underlying immunological mechanisms.


Assuntos
COVID-19 , Síndrome de COVID-19 Pós-Aguda , SARS-CoV-2 , Humanos , COVID-19/imunologia , SARS-CoV-2/imunologia , Disbiose/imunologia , Autoimunidade
18.
Immunity ; 42(4): 692-703, 2015 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-25888259

RESUMO

The interleukin-17 (IL-17) family of cytokines has emerged as a critical player in inflammatory diseases. Among them, IL-25 has been shown to be important in allergic inflammation and protection against parasitic infection. Here we have demonstrated that IL-17B, a poorly understood cytokine, functions to inhibit IL-25-driven inflammation. IL-17B and IL-25, both binding to the interleukin-17 receptor B (IL-17RB), were upregulated in their expression after acute colonic inflammation. Individual inhibition of these cytokines revealed opposing functions in colon inflammation: IL-25 was pathogenic but IL-17B was protective. Similarly opposing phenotypes were observed in Citrobacter rodentium infection and allergic asthma. Moreover, IL-25 was found to promote IL-6 production from colon epithelial cells, which was inhibited by IL-17B. Therefore, our data demonstrate that IL-17B is an anti-inflammatory cytokine in the IL-17 family.


Assuntos
Asma/imunologia , Colite/imunologia , Disbiose/imunologia , Infecções por Enterobacteriaceae/imunologia , Interleucina-17/imunologia , Interleucinas/imunologia , Mucosa Intestinal/imunologia , Animais , Antibacterianos , Asma/induzido quimicamente , Asma/genética , Asma/patologia , Linhagem Celular , Citrobacter rodentium/imunologia , Colite/induzido quimicamente , Colite/genética , Colite/patologia , Disbiose/induzido quimicamente , Disbiose/genética , Disbiose/patologia , Infecções por Enterobacteriaceae/genética , Infecções por Enterobacteriaceae/microbiologia , Infecções por Enterobacteriaceae/patologia , Células Epiteliais/imunologia , Células Epiteliais/patologia , Regulação da Expressão Gênica , Interleucina-17/deficiência , Interleucina-17/genética , Interleucina-6/genética , Interleucina-6/imunologia , Interleucinas/deficiência , Interleucinas/genética , Mucosa Intestinal/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Ovalbumina , Ligação Proteica , Receptores de Interleucina-17/genética , Receptores de Interleucina-17/imunologia , Transdução de Sinais , Dodecilsulfato de Sódio
19.
Immunity ; 42(4): 756-66, 2015 Apr 21.
Artigo em Inglês | MEDLINE | ID: mdl-25902485

RESUMO

Staphylococcus aureus skin colonization is universal in atopic dermatitis and common in cancer patients treated with epidermal growth factor receptor inhibitors. However, the causal relationship of dysbiosis and eczema has yet to be clarified. Herein, we demonstrate that Adam17(fl/fl)Sox9-(Cre) mice, generated to model ADAM17-deficiency in human, developed eczematous dermatitis with naturally occurring dysbiosis, similar to that observed in atopic dermatitis. Corynebacterium mastitidis, S. aureus, and Corynebacterium bovis sequentially emerged during the onset of eczematous dermatitis, and antibiotics specific for these bacterial species almost completely reversed dysbiosis and eliminated skin inflammation. Whereas S. aureus prominently drove eczema formation, C. bovis induced robust T helper 2 cell responses. Langerhans cells were required for eliciting immune responses against S. aureus inoculation. These results characterize differential contributions of dysbiotic flora during eczema formation, and highlight the microbiota-host immunity axis as a possible target for future therapeutics in eczematous dermatitis.


Assuntos
Dermatite Atópica/imunologia , Disbiose/imunologia , Eczema/imunologia , Células de Langerhans/imunologia , Pele/imunologia , Linfócitos T Auxiliares-Indutores/imunologia , Proteínas ADAM/deficiência , Proteínas ADAM/genética , Proteínas ADAM/imunologia , Proteína ADAM17 , Animais , Antibacterianos/farmacologia , Corynebacterium/imunologia , Dermatite Atópica/tratamento farmacológico , Dermatite Atópica/genética , Dermatite Atópica/microbiologia , Disbiose/tratamento farmacológico , Disbiose/genética , Disbiose/microbiologia , Eczema/tratamento farmacológico , Eczema/genética , Eczema/microbiologia , Receptores ErbB/genética , Receptores ErbB/imunologia , Regulação da Expressão Gênica , Humanos , Imunidade Inata , Inflamação/tratamento farmacológico , Inflamação/genética , Inflamação/imunologia , Inflamação/microbiologia , Integrases/genética , Integrases/imunologia , Células de Langerhans/efeitos dos fármacos , Células de Langerhans/microbiologia , Células de Langerhans/patologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Fatores de Transcrição SOX9/genética , Fatores de Transcrição SOX9/imunologia , Transdução de Sinais , Pele/efeitos dos fármacos , Pele/microbiologia , Pele/patologia , Staphylococcus aureus/imunologia , Linfócitos T Auxiliares-Indutores/efeitos dos fármacos , Linfócitos T Auxiliares-Indutores/microbiologia , Linfócitos T Auxiliares-Indutores/patologia
20.
Fish Shellfish Immunol ; 149: 109618, 2024 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-38729251

RESUMO

An eight-week feeding trial was designed to assess which component of commensal Bacillus siamensis LF4 can mitigate SBM-induced enteritis and microbiota dysbiosis in spotted seabass (Lateolabrax maculatus) based on TLRs-MAPKs/NF-кB signaling pathways. Fish continuously fed low SBM (containing 16 % SBM) and high SBM (containing 40 % SBM) diets were used as positive (FM group) and negative (SBM group) control, respectively. After feeding high SBM diet for 28 days, fish were supplemented with B. siamensis LF4-derived whole cell wall (CW), cell wall protein (CWP), lipoteichoic acid (LTA) or peptidoglycan (PGN) until 56 days. The results showed that a high inclusion of SBM in the diet caused enteritis, characterized with significantly (P < 0.05) decreased muscular thickness, villus height, villus width, atrophied and loosely arranged microvillus. Moreover, high SBM inclusion induced an up-regulation of pro-inflammatory cytokines and a down-regulation of occludin, E-cadherin, anti-inflammatory cytokines, apoptosis related genes and antimicrobial peptides. However, dietary supplementation with CW, LTA, and PGN of B. siamensis LF4 could effectively alleviate enteritis caused by a high level of dietary SBM. Additionally, CWP and PGN administration increased beneficial Cetobacterium and decreased pathogenic Plesiomonas and Brevinema, while dietary LTA decreased Plesiomonas and Brevinema, suggesting that CWP, LTA and PGN positively modulated intestinal microbiota in spotted seabass. Furthermore, CW, LTA, and PGN application significantly stimulated TLR2, TLR5 and MyD88 expressions, and inhibited the downstream p38 and NF-κB signaling. Taken together, these results suggest that LTA and PGN from B. siamensis LF4 could alleviate soybean meal-induced enteritis and microbiota dysbiosis in L. maculatus, and p38 MAPK/NF-κB pathways might be involved in those processes.


Assuntos
Ração Animal , Bacillus , Dieta , Disbiose , Enterite , Doenças dos Peixes , Microbioma Gastrointestinal , Glycine max , Lipopolissacarídeos , Peptidoglicano , Ácidos Teicoicos , Animais , Doenças dos Peixes/imunologia , Ração Animal/análise , Enterite/veterinária , Enterite/imunologia , Enterite/microbiologia , Disbiose/veterinária , Disbiose/imunologia , Bacillus/fisiologia , Bacillus/química , Microbioma Gastrointestinal/efeitos dos fármacos , Dieta/veterinária , Glycine max/química , Lipopolissacarídeos/farmacologia , Ácidos Teicoicos/farmacologia , Peptidoglicano/farmacologia , Peptidoglicano/administração & dosagem , Bass/imunologia , Probióticos/farmacologia , Probióticos/administração & dosagem , Suplementos Nutricionais/análise , Distribuição Aleatória
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA