Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 152
Filtrar
Mais filtros

Base de dados
País/Região como assunto
Tipo de documento
Intervalo de ano de publicação
1.
Immunity ; 53(4): 793-804.e9, 2020 10 13.
Artigo em Inglês | MEDLINE | ID: mdl-32910906

RESUMO

Allergies are considered to represent mal-directed type 2 immune responses against mostly innocuous exogenous compounds. Immunoglobulin E (IgE) antibodies are a characteristic feature of allergies and mediate hypersensitivity against allergens through activation of effector cells, particularly mast cells (MCs). Although the physiological functions of this dangerous branch of immunity have remained enigmatic, recent evidence shows that allergic immune reactions can help to protect against the toxicity of venoms. Because bacteria are a potent alternative source of toxins, we assessed the possible role of allergy-like type 2 immunity in antibacterial host defense. We discovered that the adaptive immune response against Staphylococcus aureus (SA) skin infection substantially improved systemic host defense against secondary SA infections in mice. Moreover, this acquired protection depended on IgE effector mechanisms and MCs. Importantly, our results reveal a previously unknown physiological function of allergic immune responses, IgE antibodies, and MCs in host defense against a pathogenic bacterium.


Assuntos
Imunidade Adaptativa/imunologia , Imunoglobulina E/imunologia , Mastócitos/imunologia , Infecções Estafilocócicas/imunologia , Infecções Cutâneas Estafilocócicas/imunologia , Staphylococcus aureus/imunologia , Alérgenos/imunologia , Animais , Feminino , Hipersensibilidade/imunologia , Hipersensibilidade/microbiologia , Mastócitos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Pele/imunologia , Pele/microbiologia , Infecções Estafilocócicas/microbiologia , Infecções Cutâneas Estafilocócicas/microbiologia
2.
Proc Natl Acad Sci U S A ; 118(3)2021 01 19.
Artigo em Inglês | MEDLINE | ID: mdl-33431676

RESUMO

Pathogen interactions arising during coinfection can exacerbate disease severity, for example when the immune response mounted against one pathogen negatively affects defense of another. It is also possible that host immune responses to a pathogen, shaped by historical evolutionary interactions between host and pathogen, may modify host immune defenses in ways that have repercussions for other pathogens. In this case, negative interactions between two pathogens could emerge even in the absence of concurrent infection. Parasitic worms and tuberculosis (TB) are involved in one of the most geographically extensive of pathogen interactions, and during coinfection worms can exacerbate TB disease outcomes. Here, we show that in a wild mammal natural resistance to worms affects bovine tuberculosis (BTB) severity independently of active worm infection. We found that worm-resistant individuals were more likely to die of BTB than were nonresistant individuals, and their disease progressed more quickly. Anthelmintic treatment moderated, but did not eliminate, the resistance effect, and the effects of resistance and treatment were opposite and additive, with untreated, resistant individuals experiencing the highest mortality. Furthermore, resistance and anthelmintic treatment had nonoverlapping effects on BTB pathology. The effects of resistance manifested in the lungs (the primary site of BTB infection), while the effects of treatment manifested almost entirely in the lymph nodes (the site of disseminated disease), suggesting that resistance and active worm infection affect BTB progression via distinct mechanisms. Our findings reveal that interactions between pathogens can occur as a consequence of processes arising on very different timescales.


Assuntos
Búfalos/imunologia , Resistência à Doença , Hemoncose/microbiologia , Pulmão/imunologia , Linfonodos/imunologia , Tricostrongilose/microbiologia , Tuberculose Bovina/microbiologia , Animais , Antinematódeos/farmacologia , Búfalos/microbiologia , Búfalos/parasitologia , Bovinos , Coinfecção , Progressão da Doença , Eosinófilos/efeitos dos fármacos , Eosinófilos/imunologia , Eosinófilos/microbiologia , Eosinófilos/parasitologia , Fezes/parasitologia , Feminino , Fenbendazol/farmacologia , Hemoncose/tratamento farmacológico , Hemoncose/mortalidade , Hemoncose/parasitologia , Haemonchus/efeitos dos fármacos , Haemonchus/genética , Haemonchus/patogenicidade , Imunoglobulina A/sangue , Pulmão/efeitos dos fármacos , Pulmão/microbiologia , Pulmão/parasitologia , Linfonodos/efeitos dos fármacos , Linfonodos/microbiologia , Linfonodos/parasitologia , Mastócitos/efeitos dos fármacos , Mastócitos/imunologia , Mastócitos/microbiologia , Mastócitos/parasitologia , Mycobacterium bovis/crescimento & desenvolvimento , Mycobacterium bovis/patogenicidade , Índice de Gravidade de Doença , Análise de Sobrevida , Tricostrongilose/tratamento farmacológico , Tricostrongilose/mortalidade , Tricostrongilose/parasitologia , Trichostrongylus/efeitos dos fármacos , Trichostrongylus/genética , Trichostrongylus/patogenicidade , Tuberculose Bovina/tratamento farmacológico , Tuberculose Bovina/mortalidade , Tuberculose Bovina/parasitologia
3.
Immunity ; 39(6): 996-8, 2013 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-24332026

RESUMO

Salmonella bacteria often cause food-borne diseases. In this issue of Immunity, Choi et al. (2013) demonstrate that the Salmonella Typhimurium-secreted protein tyrosine phosphatase, SptP, suppresses mast cell degranulation, which enables bacterial dissemination.


Assuntos
Proteínas de Bactérias/metabolismo , Imunidade Inata/imunologia , Mastócitos/microbiologia , Proteínas Tirosina Fosfatases/metabolismo , Infecções por Salmonella/imunologia , Salmonella typhimurium/enzimologia , Animais , Humanos
4.
Immunity ; 39(6): 1108-20, 2013 Dec 12.
Artigo em Inglês | MEDLINE | ID: mdl-24332031

RESUMO

The virulence of Salmonella is linked to its invasive capacity and suppression of adaptive immunity. This does not explain, however, the rapid dissemination of the pathogen after it breaches the gut. In our study, S. Typhimurium suppressed degranulation of local mast cells (MCs), resulting in limited neutrophil recruitment and restricting outflow of vascular contents into infection sites, thus facilitating bacterial spread. MC suppression was mediated by secreted effector protein (SptP), which shares structural homology with Yersinia YopH. SptP functioned by dephosphorylating the vesicle fusion protein N-ethylmalemide-sensitive factor and by blocking phosphorylation of Syk. Without SptP, orally challenged S. Typhimurium failed to suppress MC degranulation and exhibited limited colonization of the mesenteric lymph nodes. Administration of SptP to sites of E. coli infection markedly enhanced its virulence. Thus, SptP-mediated inactivation of local MCs is a powerful mechanism utilized by S. Typhimurium to impede early innate immunity.


Assuntos
Proteínas de Bactérias/metabolismo , Imunidade Inata/imunologia , Mastócitos/microbiologia , Proteínas Tirosina Fosfatases/metabolismo , Infecções por Salmonella/imunologia , Salmonella typhimurium/enzimologia , Animais , Proteínas de Bactérias/genética , Degranulação Celular , Humanos , Mastócitos/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Mutação , Neutrófilos/imunologia , Fosforilação , Proteínas Tirosina Fosfatases/genética , Infecções por Salmonella/microbiologia , Salmonella typhimurium/genética , Salmonella typhimurium/imunologia , Salmonella typhimurium/patogenicidade , Yersinia pestis/enzimologia
5.
Immunity ; 38(2): 349-59, 2013 Feb 21.
Artigo em Inglês | MEDLINE | ID: mdl-23415912

RESUMO

The lower urinary tract's virtually inevitable exposure to external microbial pathogens warrants efficient tissue-specialized defenses to maintain sterility. The observation that the bladder can become chronically infected in combination with clinical observations that antibody responses after bladder infections are not detectable suggest defects in the formation of adaptive immunity and immunological memory. We have identified a broadly immunosuppressive transcriptional program specific to the bladder, but not the kidney, during infection of the urinary tract that is dependent on tissue-resident mast cells (MCs). This involves localized production of interleukin-10 and results in suppressed humoral and cell-mediated responses and bacterial persistence. Therefore, in addition to the previously described role of MCs orchestrating the early innate immunity during bladder infection, they subsequently play a tissue-specific immunosuppressive role. These findings may explain the prevalent recurrence of bladder infections and suggest the bladder as a site exhibiting an intrinsic degree of MC-maintained immune privilege.


Assuntos
Cistite/patologia , Células Dendríticas/patologia , Tolerância Imunológica , Interleucina-10/imunologia , Mastócitos/patologia , Bexiga Urinária/patologia , Escherichia coli Uropatogênica/imunologia , Animais , Doença Crônica , Cistite/imunologia , Cistite/microbiologia , Células Dendríticas/imunologia , Células Dendríticas/microbiologia , Feminino , Expressão Gênica , Humanos , Imunidade Celular , Imunidade Humoral , Imunidade Inata , Memória Imunológica , Interleucina-10/biossíntese , Rim/imunologia , Rim/microbiologia , Rim/patologia , Mastócitos/imunologia , Mastócitos/microbiologia , Camundongos , Especificidade de Órgãos , Pielonefrite/imunologia , Pielonefrite/microbiologia , Pielonefrite/patologia , Transcrição Gênica/imunologia , Bexiga Urinária/imunologia , Bexiga Urinária/microbiologia
6.
Vopr Pitan ; 91(1): 98-108, 2022.
Artigo em Russo | MEDLINE | ID: mdl-35298108

RESUMO

Helicobacter pylori induced gastritis accounts for 70% of cases in the structure of this pathology. Features of the long-term inflammatory reaction of the mucous membrane are directly related to the mechanisms of bacterial pathogenicity, and features of immunogenesis within narrow limits of the specific tissue microenvironment of organ structures. Mast cells appear to be one of the key players (promoters) in the regulation of the inflammatory mediator cascade and the formation of cytokine-induced expression. Possessing a wide arsenal of biologically active substances, mast cells are able to participate in the formation of the immune response and resistance of the gastric mucosa, modulating both pro- and anti-inflammatory effects. The antigen-presenting features of mast cells are of interest in terms of interaction with H. pylori and induction of mucosa bacterial colonization. The aim of study was to assess the mast cell tryptase profile of the gastric mucosa in the immunopathogenesis of H. pylori-associated inflammation. Material and methods. The study included 19 biopsies of the gastric mucosa with unknown status of H. pylori infection. Microslides were stained with hematoxylin and eosin, and Giemsa's dye for plain microscopy. H. pylori infection of the gastric mucosa was detected using the immunohistochemical method. Using double immunofluorescent labeling, localization of tryptase-positive mast cells and H. pylori strains was detected. Results. In patients infected with H. pylori (n=12), there was a significant increase in the number of tryptase-positive mast cells (177.99±30.55 vs 88.58±11.49; p<0.05) with activation of secretory pathways and release of protease into the extracellular matrix of the gastric mucosa. The quantitative parameters of mast cells in the group of patients with an undetected pathogen and signs of a chronic inflammation of the gastric mucosa were statistically significantly lower than in the group of infected patients. Co-localization of tryptase-positive mast cells and H. pylori strains (with the formation of areas of large free-lying granule accumulation around the glands with pronounced degree of H. pylori contamination) was detected in gastrobiopsy specimens, the fact evidencing their close involvement in the development of inflammatory reactions of the gastric mucosa. Conclusion. The study demonstrated the features of mast cells and H. pylori interaction revealing previously unknown aspects of gastritis pathophysiology. The data obtained contribute a valuable insight to choose a treatment strategy for H. pylori-associated gastritis.


Assuntos
Gastrite , Infecções por Helicobacter , Helicobacter pylori , Mucosa Gástrica/metabolismo , Mucosa Gástrica/microbiologia , Mucosa Gástrica/patologia , Gastrite/metabolismo , Gastrite/microbiologia , Gastrite/patologia , Infecções por Helicobacter/metabolismo , Infecções por Helicobacter/patologia , Humanos , Inflamação/metabolismo , Mastócitos/metabolismo , Mastócitos/microbiologia , Mastócitos/patologia , Triptases/metabolismo
7.
J Allergy Clin Immunol ; 145(1): 147-159, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31254531

RESUMO

BACKGROUND: Chronic rhinosinusitis (CRS) with nasal polyps is a common chronic condition. The exact cause of nasal polyps remains unknown. Recently, we made the novel observation of intracellular localization of Staphylococcus aureus within mast cells in nasal polyps. OBJECTIVE: This follow-up study aimed to further characterize interactions between S aureus and mast cells in this setting and elucidate potential internalization mechanisms with particular emphasis on the role of staphylococcal enterotoxin B (SEB). METHODS: A prospective study was performed using an explant tissue model with ex vivo inferior turbinate mucosa obtained from patients with chronic rhinosinusitis with nasal polyps (n = 7) and patients without CRS (n = 5). Immunohistochemistry was used to characterize S aureus uptake into mast cells and investigate the effects of SEB on this process. An in vitro cell-culture model was used to investigate mast cell-S aureus interactions by using a combination of fluorescent in situ hybridization, confocal laser scanning microscopy, scanning electron microscopy, transmission electron microscopy, and proliferation assays. RESULTS: S aureus was captured by extracellular traps and entered mast cells through phagocytosis. Proliferating intracellular S aureus led to the expansion and eventual rupture of mast cells, resulting in release of viable S aureus into the extracellular space. The presence of SEB appeared to promote internalization of S aureus into mast cells. CONCLUSION: This study provides new insights into the interactions between S aureus and mast cells, including the internalization process, and demonstrates a prominent role for SEB in promoting uptake of the bacteria into these cells.


Assuntos
Enterotoxinas/imunologia , Mastócitos , Pólipos Nasais , Fagocitose , Staphylococcus aureus , Adulto , Idoso , Linhagem Celular , Feminino , Humanos , Masculino , Mastócitos/imunologia , Mastócitos/microbiologia , Mastócitos/ultraestrutura , Pessoa de Meia-Idade , Pólipos Nasais/imunologia , Pólipos Nasais/microbiologia , Pólipos Nasais/ultraestrutura , Estudos Prospectivos , Staphylococcus aureus/imunologia , Staphylococcus aureus/patogenicidade , Técnicas de Cultura de Tecidos
8.
Cell Microbiol ; 21(3): e12972, 2019 03.
Artigo em Inglês | MEDLINE | ID: mdl-30423602

RESUMO

Interleukin (IL)-31 is important for innate immunity in mucosal tissues and skin, and increased IL-31 expression participates in the pathogenesis of chronic inflammatory diseases affecting the skin, airways, lungs, and intestines. We investigated the contribution of mast cells to the induction of IL-31 production following infection with the periodontal pathogen, Porphyromonas gingivalis. We found that oral infection with P. gingivalis increased IL-31 expression in the gingival tissues of wild-type mice but not in those of mast cell-deficient mice. The P. gingivalis-induced IL-31 production by human mast cells occurred through the activation of the JNK and NF-κB signalling pathways and was dependent on the P. gingivalis lysine-specific protease gingipain-K. P. gingivalis infection induced IL-31 receptor α and oncostatin M receptor ß expression in human gingival epithelial cells. Notably, the P. gingivalis-induced IL-31 production by mast cells led to the downregulation of claudin-1, a tight junction molecule, in gingival epithelial cells, resulting in an IL-31-dependent increase in the paracellular permeability of the gingival epithelial barrier. These findings suggest that IL-31 produced by mast cells in response to P. gingivalis infection causes gingival epithelial barrier dysfunction, which may contribute to the chronic inflammation observed in periodontitis.


Assuntos
Interações Hospedeiro-Patógeno , Imunidade Inata , Interleucinas/metabolismo , Mastócitos/imunologia , Mastócitos/microbiologia , Porphyromonas gingivalis/crescimento & desenvolvimento , Porphyromonas gingivalis/imunologia , Animais , Infecções por Bacteroidaceae/imunologia , Infecções por Bacteroidaceae/microbiologia , Células Cultivadas , Modelos Animais de Doenças , Células Epiteliais/patologia , Humanos , Camundongos , Periodontite/microbiologia , Periodontite/patologia , Transdução de Sinais
9.
Cell Microbiol ; 21(9): e13064, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31155820

RESUMO

Mast cells are implicated in the innate proinflammatory immune defence against bacterial insult, but the mechanisms through which mast cells respond to bacterial encounter are poorly defined. Here, we addressed this issue and show that mast cells respond vividly to wild type Streptococcus equi by up-regulating a panel of proinflammatory genes and by secreting proinflammatory cytokines. However, this response was completely abrogated when the bacteria lacked expression of sagA, whereas the lack of a range of other potential virulence genes (seeH, seeI, seeL, seeM, hasA, seM, aroB, pyrC, and recA) had no effect on the amplitude of the mast cell responses. The sagA gene encodes streptolysin S, a lytic toxin, and we next showed that the wild type strain but not a sagA-deficient mutant induced lysis of mast cells. To investigate whether host cell membrane perturbation per se could play a role in the activation of the proinflammatory response, we evaluated the effects of detergent- and pneumolysin-dependent lysis on mast cells. Indeed, exposure of mast cells to sublytic concentrations of all these agents resulted in cytokine responses of similar amplitudes as those caused by wild type streptococci. This suggests that sublytic membrane perturbation is sufficient to trigger full-blown proinflammatory signalling in mast cells. Subsequent analysis showed that the p38 and Erk1/2 signalling pathways had central roles in the proinflammatory response of mast cells challenged by either sagA-expressing streptococci or detergent. Altogether, these findings suggest that sagA-dependent mast cell membrane perturbation is a mechanism capable of activating the innate immune response upon bacterial challenge.


Assuntos
Proteínas de Bactérias/metabolismo , Inflamação/metabolismo , Mastócitos/imunologia , Streptococcus equi/genética , Streptococcus equi/patogenicidade , Estreptolisinas/metabolismo , Animais , Proteínas de Bactérias/genética , Proteínas de Bactérias/farmacologia , Membrana Celular/efeitos dos fármacos , Membrana Celular/genética , Membrana Celular/metabolismo , Citocinas/metabolismo , Sistema de Sinalização das MAP Quinases/genética , Mastócitos/efeitos dos fármacos , Mastócitos/metabolismo , Mastócitos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/genética , Estreptolisinas/genética , Estreptolisinas/farmacologia , Virulência/genética , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
10.
Biochem Biophys Res Commun ; 511(2): 350-355, 2019 04 02.
Artigo em Inglês | MEDLINE | ID: mdl-30795864

RESUMO

Staphylococcal superantigen-like (SSL) protein is a family of exotoxins that consists of 14 SSLs, and the roles of several SSLs in immune evasion of the cocci have been revealed. However little is known whether they act as immune activators and are involved in inflammatory disorders such as atopic dermatitis. In this study we examined whether SSLs activate mast cells, the key player of local inflammation. SSL12 evoked the release of a granule enzyme ß-hexosaminidase from bone marrow derived mast cells (BMMCs) in the absence of IgE. The release of the granule enzyme caused by SSL12 was not accompanied with the leakage of a cytosolic enzyme lactate dehydrogenase (LDH), unlike staphylococcal δ-toxin that was reported to induce both the release of ß-hexosaminidase and the leakage of LDH from the cells, suggesting that SSL12 evokes the degranulation of mast cells without cell membrane damage. Furthermore SSL12 induced IL-6 and IL-13 in both mRNA and protein levels indicating that SSL12 induces de novo synthesis of the cytokines. Evans blue extravasation was elevated by the intradermal injection of SSL12, suggesting that SSL12 is also able to evoke local inflammation in vivo. These findings indicate the novel mast cell activating activity of SSLs, and SSL12 is likely an important factor in both initiation phase and effector phase of allergic and immune responses.


Assuntos
Mastócitos/microbiologia , Staphylococcus/imunologia , Superantígenos/imunologia , Animais , Degranulação Celular , Células Cultivadas , Citocinas/imunologia , Interações Hospedeiro-Patógeno , Mastócitos/imunologia , Mastócitos/fisiologia , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Infecções Estafilocócicas/imunologia , Infecções Estafilocócicas/microbiologia
11.
Gastroenterology ; 153(4): 948-960.e3, 2017 10.
Artigo em Inglês | MEDLINE | ID: mdl-28711627

RESUMO

BACKGROUND & AIMS: Irritable bowel syndrome (IBS) is associated with intestinal dysbiosis and symptoms of IBS develop following gastroenteritis. We aimed to study the passage of live bacteria through the colonic epithelium, and determine the role of mast cells (MCs) and vasoactive intestinal polypeptide (VIP) in barrier regulation in IBS and healthy individuals. METHODS: Colon biopsies from 32 women with IBS and 15 age-matched healthy women (controls) were mounted in Ussing chambers; we measured numbers of fluorescently labeled Escherichia coli HS and Salmonella typhimurium that passed through from the mucosal side to the serosal side of the tissue. Some biopsies were exposed to agents that block the VIP receptors (VPAC1 and VPAC2) or MCs. Levels of VIP and tryptase were measured in plasma and biopsy lysates. Number of MCs and MCs that express VIP or VIP receptors were quantified by immunofluorescence. Biopsies from an additional 5 patients with IBS and 4 controls were mounted in chambers and Salmonella were added; we studied passage routes through the epithelium by transmission electron microscopy and expression of tight junctions by confocal microscopy. RESULTS: In colon biopsies from patients with IBS, larger numbers of E coli HS and S typhimurium passed through the epithelium than in biopsies from controls (P < .0005). In transmission electron microscopy analyses, bacteria were found to cross the epithelium via only the transcellular route. Bacterial passage was reduced in biopsies from patients with IBS and controls after addition of antibodies against VPACs or ketotifen, which inhibits MCs. Plasma samples from patients with IBS had higher levels of VIP than plasma samples from controls. Biopsies from patients with IBS had higher levels of tryptase, larger numbers of MCs, and a higher percentage of MCs that express VPAC1 than biopsies from controls. In biopsies from patients with IBS, addition of Salmonella significantly reduced levels of occludin; subsequent addition of ketotifen significantly reversed this effect. CONCLUSIONS: We found that colonic epithelium tissues from patients with IBS have increased translocation of commensal and pathogenic live bacteria compared with controls. The mechanisms of increased translocation include MCs and VIP.


Assuntos
Translocação Bacteriana , Colo/microbiologia , Escherichia coli/fisiologia , Mucosa Intestinal/microbiologia , Síndrome do Intestino Irritável/microbiologia , Mastócitos/microbiologia , Salmonella typhimurium/fisiologia , Peptídeo Intestinal Vasoativo/metabolismo , Adulto , Biópsia , Estudos de Casos e Controles , Colo/ultraestrutura , Disbiose , Impedância Elétrica , Escherichia coli/patogenicidade , Feminino , Imunofluorescência , Microbioma Gastrointestinal , Humanos , Mucosa Intestinal/ultraestrutura , Síndrome do Intestino Irritável/diagnóstico , Síndrome do Intestino Irritável/metabolismo , Mastócitos/metabolismo , Mastócitos/ultraestrutura , Microscopia Confocal , Microscopia Eletrônica de Transmissão , Pessoa de Meia-Idade , Receptores Tipo II de Peptídeo Intestinal Vasoativo/metabolismo , Receptores Tipo I de Polipeptídeo Intestinal Vasoativo/metabolismo , Salmonella typhimurium/patogenicidade , Simbiose , Junções Íntimas/microbiologia , Junções Íntimas/ultraestrutura , Adulto Jovem
12.
Immunity ; 31(4): 665-76, 2009 Oct 16.
Artigo em Inglês | MEDLINE | ID: mdl-19818652

RESUMO

The characteristics, importance, and molecular requirements for interactions between mast cells (MCs) and CD8(+) T cells have not been elucidated. Here, we demonstrated that MCs induced antigen-specific CD8(+) T cell activation and proliferation. This process required direct cell contact and MHC class I-dependent antigen cross-presentation by MCs and induced the secretion of interleukin-2, interferon-gamma, and macrophage inflammatory protein-1alpha by CD8(+) T cells. MCs regulated antigen-specific CD8(+) T cell cytotoxicity by increasing granzyme B expression and by promoting CD8(+) T cell degranulation. Because MCs also upregulated their expression of costimulatory molecules (4-1BB) and released osteopontin upon direct T cell contact, MC-T cell interactions probably are bidirectional. In vivo, adoptive transfer of antigen-pulsed MCs induced MHC class I-dependent, antigen-specific CD8(+) T cell proliferation, and MCs regulated CD8(+) T cell-specific priming in experimental autoimmune encephalomyelitis. Thus, MCs are important players in antigen-specific regulation of CD8(+) T cells.


Assuntos
Apresentação de Antígeno/imunologia , Linfócitos T CD8-Positivos/imunologia , Apresentação Cruzada/imunologia , Encefalomielite Autoimune Experimental/imunologia , Mastócitos/imunologia , Transferência Adotiva , Animais , Linfócitos T CD8-Positivos/metabolismo , Linfócitos T CD8-Positivos/microbiologia , Degranulação Celular/imunologia , Quimiocina CCL3/biossíntese , Quimiocina CCL3/imunologia , Técnicas de Cocultura , Encefalomielite Autoimune Experimental/metabolismo , Encefalomielite Autoimune Experimental/patologia , Glicoproteínas/imunologia , Granzimas/imunologia , Granzimas/metabolismo , Antígenos de Histocompatibilidade Classe I/imunologia , Antígenos de Histocompatibilidade Classe I/metabolismo , Interferon gama/biossíntese , Interferon gama/imunologia , Interleucina-2/biossíntese , Interleucina-2/imunologia , Listeria monocytogenes/imunologia , Listeriose/imunologia , Listeriose/microbiologia , Mastócitos/metabolismo , Mastócitos/microbiologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Glicoproteína Mielina-Oligodendrócito , Osteopontina/imunologia , Osteopontina/metabolismo , Ovalbumina/imunologia , Fragmentos de Peptídeos/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/imunologia , Membro 9 da Superfamília de Receptores de Fatores de Necrose Tumoral/metabolismo
13.
Gastroenterology ; 151(5): 923-932, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27436071

RESUMO

BACKGROUND & AIMS: The gut microbiota affects intestinal permeability and mucosal mast cells (MMCs) responses. Activation of MMCs has been associated with absorption of dietary fat. We investigated whether the gut microbiota contributes to the fat-induced activation of MMCs in rats, and how antibiotics might affect this process. METHODS: Adult male Sprague-Dawley rats were given streptomycin and penicillin for 4 days (n = 6-8) to reduce the abundance of their gut flora, or normal drinking water (controls, n = 6-8). They underwent lymph fistula surgery and after an overnight recovery were given an intraduodenal bolus of intralipid. We collected intestinal tissues and lymph fluid and assessed activation of MMCs, intestinal permeability, and fat transport parameters. RESULTS: Compared with controls, intestinal lymph from rats given antibiotics had reduced levels of mucosal mast cell protease II (produced by MMCs) and decreased activity of diamine oxidase (produced by enterocytes) (P < .05). Rats given antibiotics had reduced intestinal permeability in response to dietary lipid compared with controls (P < .01). Unexpectedly, antibiotics also reduced lymphatic transport of triacylglycerol and phospholipid (P < .01), concomitant with decreased levels of mucosal apolipoproteins B, A-I, and A-IV (P < .01). No differences were found in intestinal motility or luminal pancreatic lipase activity between rats given antibiotics and controls. These effects were not seen with an acute dose of antibiotics or 4 weeks after the antibiotic regimen ended. CONCLUSIONS: The intestinal microbiota appears to activate MMCs after the ingestion of fat in rats; this contributes to fat-induced intestinal permeability. We found that the gut microbiome promotes absorption of lipid, probably by intestinal production of apolipoproteins and secretion of chylomicrons.


Assuntos
Antibacterianos/farmacologia , Gorduras na Dieta/metabolismo , Absorção Intestinal/efeitos dos fármacos , Mucosa Intestinal/efeitos dos fármacos , Mastócitos/efeitos dos fármacos , Penicilinas/farmacologia , Estreptomicina/farmacologia , Animais , Antibacterianos/administração & dosagem , Microbioma Gastrointestinal/efeitos dos fármacos , Microbioma Gastrointestinal/fisiologia , Absorção Intestinal/imunologia , Mucosa Intestinal/imunologia , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiologia , Masculino , Mastócitos/metabolismo , Mastócitos/microbiologia , Penicilinas/administração & dosagem , Permeabilidade , Ratos , Ratos Sprague-Dawley , Estreptomicina/administração & dosagem
14.
Brain Behav Immun ; 65: 68-89, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28676349

RESUMO

When related to central nervous system (CNS) health and disease, brain mast cells (MCs) can be a source of either beneficial or deleterious signals acting on neural cells. We review the current state of knowledge about molecular interactions between MCs and glia in neurodegenerative diseases such as Multiple Sclerosis, Alzheimer's disease, Amyotrophic Lateral Sclerosis, Parkinson's disease, Epilepsy. We also discuss the influence on MC actions evoked by the host microbiota, which has a profound effect on the host immune system, inducing important consequences in neurodegenerative disorders. Gut dysbiosis, reduced intestinal motility and increased intestinal permeability, that allow bacterial products to circulate and pass through the blood-brain barrier, are associated with neurodegenerative disease. There are differences between the microbiota of neurologic patients and healthy controls. Distinguishing between cause and effect is a challenging task, and the molecular mechanisms whereby remote gut microbiota can alter the brain have not been fully elucidated. Nevertheless, modulation of the microbiota and MC activation have been shown to promote neuroprotection. We review this new information contributing to a greater understanding of MC-microbiota-neural cells interactions modulating the brain, behavior and neurodegenerative processes.


Assuntos
Mastócitos/imunologia , Mastócitos/fisiologia , Doenças Neurodegenerativas/imunologia , Barreira Hematoencefálica/imunologia , Barreira Hematoencefálica/microbiologia , Barreira Hematoencefálica/fisiologia , Encéfalo/imunologia , Comunicação Celular , Permeabilidade da Membrana Celular/imunologia , Sistema Nervoso Central/imunologia , Microbioma Gastrointestinal/imunologia , Microbioma Gastrointestinal/fisiologia , Motilidade Gastrointestinal/imunologia , Humanos , Intestinos/microbiologia , Mastócitos/microbiologia , Microbiota , Doenças Neurodegenerativas/microbiologia , Neuroglia/imunologia
15.
Cell Microbiol ; 18(6): 807-19, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26595647

RESUMO

Mast cells (MCs) are important sentinels of the host defence against invading pathogens. We previously reported that Staphylococcus aureus evaded the extracellular antimicrobial activities of MCs by promoting its internalization within these cells via ß1 integrins. Here, we investigated the molecular mechanisms governing this process. We found that S. aureus responded to the antimicrobial mediators released by MCs by up-regulating the expression of α-hemolysin (Hla), fibronectin-binding protein A and several regulatory systems. We also found that S. aureus induced the up-regulation of ß1 integrin expression on MCs and that this effect was mediated by Hla-ADAM10 (a disintegrin and metalloproteinase 10) interaction. Thus, deletion of Hla or inhibition of Hla-ADAM10 interaction significantly impaired S. aureus internalization within MCs. Furthermore, purified Hla but not the inactive HlaH35L induced up-regulation of ß1 integrin expression in MCs in a dose-dependent manner. Our data support a model in which S. aureus counter-reacts the extracellular microbicidal mechanisms of MCs by increasing expression of fibronectin-binding proteins and by inducing Hla-ADAM10-mediated up-regulation of ß1 integrin in MCs. The up-regulation of bacterial fibronectin-binding proteins, concomitantly with the increased expression of its receptor ß1 integrin on the MCs, resulted in enhanced S. aureus internalization through the binding of fibronectin-binding proteins to integrin ß1 via fibronectin.


Assuntos
Toxinas Bacterianas/metabolismo , Proteínas Hemolisinas/metabolismo , Integrina beta1/metabolismo , Mastócitos/microbiologia , Staphylococcus aureus/fisiologia , Staphylococcus aureus/patogenicidade , Proteína ADAM10/metabolismo , Adesinas Bacterianas/genética , Adesinas Bacterianas/metabolismo , Secretases da Proteína Precursora do Amiloide/metabolismo , Animais , Toxinas Bacterianas/genética , Células Cultivadas , Feminino , Regulação Bacteriana da Expressão Gênica , Proteínas Hemolisinas/genética , Interações Hospedeiro-Patógeno , Mastócitos/metabolismo , Proteínas de Membrana/metabolismo , Camundongos Endogâmicos C57BL , Infecções Cutâneas Estafilocócicas/metabolismo , Infecções Cutâneas Estafilocócicas/microbiologia , Regulação para Cima
16.
J Allergy Clin Immunol ; 137(2): 507-516.e8, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26433560

RESUMO

BACKGROUND: The incidence of food allergies has increased dramatically during the last decade. Recently, probiotics have been studied for the prevention and treatment of allergic disease. OBJECTIVE: We examined whether Bifidobacterium longum KACC 91563 and Enterococcus faecalis KACC 91532 have the capacity to suppress food allergies. METHODS: B longum KACC 91563 and E faecalis KACC 91532 were administered to BALB/c wild-type mice, in which food allergy was induced by using ovalbumin and alum. Food allergy symptoms and various immune responses were assessed. RESULTS: B longum KACC 91563, but not E faecalis KACC 91532, alleviated food allergy symptoms. Extracellular vesicles of B longum KACC 91563 bound specifically to mast cells and induced apoptosis without affecting T-cell immune responses. Furthermore, injection of family 5 extracellular solute-binding protein, a main component of extracellular vesicles, into mice markedly reduced the occurrence of diarrhea in a mouse food allergy model. CONCLUSION: B longum KACC 91563 induces apoptosis of mast cells specifically and alleviates food allergy symptoms. Accordingly, B longum KACC 91563 and family 5 extracellular solute-binding protein exhibit potential as therapeutic approaches for food allergies.


Assuntos
Proteínas de Bactérias/imunologia , Bifidobacterium/imunologia , Vesículas Extracelulares/imunologia , Hipersensibilidade Alimentar/imunologia , Hipersensibilidade Alimentar/microbiologia , Imunomodulação , Mastócitos/imunologia , Animais , Apoptose/imunologia , Proteínas de Bactérias/metabolismo , Bifidobacterium/metabolismo , Contagem de Células , Citocinas/biossíntese , Modelos Animais de Doenças , Endocitose/imunologia , Vesículas Extracelulares/metabolismo , Hipersensibilidade Alimentar/metabolismo , Intestino Delgado/imunologia , Intestino Delgado/metabolismo , Intestino Delgado/microbiologia , Intestino Delgado/patologia , Mastócitos/metabolismo , Mastócitos/microbiologia , Camundongos , Probióticos , Subpopulações de Linfócitos T/imunologia , Subpopulações de Linfócitos T/metabolismo
17.
Prostate ; 76(15): 1431-44, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27325623

RESUMO

BACKGROUND: Chronic inflammation has a role in the pathogenesis of benign prostatic hyperplasia (BPH) and prostate cancer. Mast cells have been detected in chronic inflammatory infiltrate of the prostate, and it is possible that the interaction between prostate epithelial cells and Trichomonas vaginalis influences the activity of mast cells in the prostate stroma. Activated mast cells might influence the biological functions of nearby tissues and cells. In this study, we investigated whether mast cells reacted with the culture supernatant of BPH epithelial cells infected with T. vaginalis may induce the proliferation of prostate stromal cells. METHODS: To measure the proliferation of prostate stromal cells in response to chronic inflammation caused by the infection of BPH-1 cells with T. vaginalis, the CCK-8 assay and wound healing assay were used. ELISAs, quantitative real-time PCR, western blotting and immunofluorescence were used to measure the production and expression of inflammatory cytokine and cytokine receptor. RESULTS: BPH-1 cells incubated with live trichomonads produced increased levels of CCL2, IL-1ß, IL-6, and CXCL8, and induced the migration of mast cells and monocytes. When the culture supernatant of BPH-1 cells stimulated with trichomonads (TCM) was added to mast cells, they became activated, as confirmed by release of ß-hexosaminidase and CXCL8. Prostate stromal cells incubated with the culture supernatant of mast cells activated with TCM (M-TCM) proliferated and expressed increased levels of CXCL8, CCL2, and the cytokine receptors CXCR1 and CCR2. Blocking the chemokine receptors reduced the proliferation of stromal cells and also decreased the production of CXCL8 and CCL2. Moreover, the expression of FGF2, cyclin D1, and Bcl-2 was increased in the proliferated stromal cells stimulated with M-TCM. Additionally, the M-TCM-treated stromal cells were more invasive than control cells. CONCLUSIONS: The inflammatory mediators released by BPH epithelial cells in response to infection by trichomonads induce the migration and activation of mast cells. The activated mast cells induce the proliferation of prostate stromal cells via CXCL8-CXCR1 and CCL2-CCR2 signaling. Our results therefore show that the inflammatory response by BPH epithelial cells stimulated with T. vaginalis induce the proliferation of prostate stromal cells via crosstalk with mast cells. Prostate 76:1431-1444, 2016. © 2016 Wiley Periodicals, Inc.


Assuntos
Mastócitos/microbiologia , Próstata/imunologia , Hiperplasia Prostática/imunologia , Receptor Cross-Talk/imunologia , Células Estromais/imunologia , Trichomonas vaginalis/imunologia , Proliferação de Células , Células Cultivadas , Células Epiteliais/imunologia , Células Epiteliais/patologia , Humanos , Inflamação , Masculino , Mastócitos/patologia , Próstata/patologia , Hiperplasia Prostática/patologia , Células Estromais/patologia , Trichomonas vaginalis/patogenicidade
18.
J Immunol ; 193(10): 5171-80, 2014 Nov 15.
Artigo em Inglês | MEDLINE | ID: mdl-25326026

RESUMO

The commensal microbiota has a high impact on health and disease by modulating the development and homeostasis of host immune system. Immune cells are involved in virtually every aspect of the wound repair process; however, the impact of commensal microbiota on skin wound healing is largely unknown. In this study, we evaluated the influence of commensal microbiota on tissue repair of excisional skin wounds by using germ-free (GF) Swiss mice. We observed that macroscopic wound closure rate is accelerated in the absence of commensal microbiota. Accordantly, histologically assessed wound epithelization was accelerated in GF in comparison with conventional (CV) Swiss mice. The wounds of GF mice presented a significant decrease in neutrophil accumulation and an increase in mast cell and macrophage infiltration into wounds. Interestingly, alternatively activated healing macrophage-related genes were highly expressed in the wound tissue of GF mice. Moreover, levels of the anti-inflammatory cytokine IL-10, the angiogenic growth factor VEGF and angiogenesis were higher in the wound tissue of those mice. Conversely, scarring and levels of the profibrogenic factor TGF-ß1 were greatly reduced in GF mice wounded skin when compared with CV mice. Of note, conventionalization of GF mice with CV microbiota restored wound closure rate, neutrophil and macrophage accumulation, cytokine production, and scarring to the same extent as CV mice. Overall, our findings suggest that, in the absence of any contact with microbiota, skin wound healing is accelerated and scarless, partially because of reduced accumulation of neutrophils, increased accumulation of alternatively activated healing macrophages, and better angiogenesis at wound sites.


Assuntos
Cicatriz/prevenção & controle , Vida Livre de Germes/imunologia , Reepitelização/fisiologia , Pele/imunologia , Animais , Movimento Celular/imunologia , Cicatriz/imunologia , Feminino , Regulação da Expressão Gênica , Interleucina-10/genética , Interleucina-10/imunologia , Macrófagos/citologia , Macrófagos/imunologia , Macrófagos/microbiologia , Masculino , Mastócitos/citologia , Mastócitos/imunologia , Mastócitos/microbiologia , Camundongos , Microbiota/imunologia , Neovascularização Fisiológica , Neutrófilos/citologia , Neutrófilos/imunologia , Neutrófilos/microbiologia , Pele/irrigação sanguínea , Pele/lesões , Pele/microbiologia , Simbiose/imunologia , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/imunologia , Fator A de Crescimento do Endotélio Vascular/genética , Fator A de Crescimento do Endotélio Vascular/imunologia
19.
J Dairy Sci ; 99(9): 6889-6901, 2016 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-27289147

RESUMO

Interest is increasing in the potentially beneficial role of probiotics in the prevention and treatment of atopic diseases. In this study, we investigated the protective effects of Lactococcus chungangensis CAU 28(T) against atopic dermatitis using murine macrophage RAW 264.7 cells, human keratinocyte HaCaT cells, human mast cell line HMC-1 cells, and a 2,4-dinitrochlorobenzene-induced atopic dermatitis model (NC/Nga mice). The results showed that L. chungangensis CAU 28(T) exhibited potent antiinflammatory activity by inhibiting the production of the proinflammatory mediators nitric oxide and prostaglandin E2 in lipopolysaccharide-stimulated RAW 264.7 cells. Treatment with L. chungangensis CAU 28(T) reduced the release of ß-hexosaminidase and histamine in HMC-1 cells stimulated with mast cell activator compound 48/80. In addition, the back skin and ears of NC/Nga mice exhibited reduced histological manifestations of atopic skin lesions such as erosion, hyperplasia of the epidermis and dermis, and inflammatory cell infiltration. Oral administration of L. chungangensis CAU 28(T) suppressed the production of IL-4, IL-5, IL-12, IFN-γ, tumor necrosis factor-α, and thymus- and activation-regulated chemokine (TARC) in skin lesions, indicating that it strongly drives the local immune system with efficacy comparable to that of tacrolimus, a topical immunomodulatory drug used for the treatment of atopic dermatitis. The findings indicate that L. chungangensis CAU 28(T) could be a novel probiotic candidate for controlling the symptoms of atopic dermatitis.


Assuntos
Dermatite Atópica/terapia , Dinitroclorobenzeno/toxicidade , Lactococcus , Probióticos/administração & dosagem , Administração Oral , Animais , Linhagem Celular Tumoral , Sobrevivência Celular , Quimiocina CCL17/metabolismo , Dermatite Atópica/induzido quimicamente , Dinoprostona/metabolismo , Feminino , Histamina/metabolismo , Humanos , Imunoglobulina E/sangue , Interferon gama/metabolismo , Interleucina-12/metabolismo , Interleucina-4/metabolismo , Interleucina-5/metabolismo , Lipopolissacarídeos/toxicidade , Mastócitos/microbiologia , Camundongos , Óxido Nítrico/metabolismo , Células RAW 264.7 , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Pele/microbiologia , Pele/patologia , Tacrolimo/farmacologia , Fator de Necrose Tumoral alfa/antagonistas & inibidores , Fator de Necrose Tumoral alfa/metabolismo , beta-N-Acetil-Hexosaminidases/metabolismo
20.
Immunology ; 146(3): 470-85, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26288256

RESUMO

Human mast cells (huMCs) are involved in both innate and adaptive immune responses where they release mediators including amines, reactive oxygen species (ROS), eicosanoids and cytokines. We have reported that interferon-γ (IFN-γ) enhances FcγR-dependent ROS production. The aim of this study was to extend these observations by investigating the effect of IFN-γ on the biological responses of huMCs to Staphylococcus aureus. We found that exposure of huMCs to S. aureus generated intracellular and extracellular ROS, which were enhanced in the presence of IFN-γ. IFN-γ also promoted bacteria killing, ß-hexosaminidase release and eicosanoid production. Interferon-γ similarly increased expression of mRNAs encoding CCL1 to CCL4, granulocyte-macrophage colony-stimulating factor (GM-CSF), tumour necrosis factor-α and CXCL8 in S. aureus-stimulated huMCs. The ability of IFN-γ to increase CXCL8 and GM-CSF protein levels was confirmed by ELISA. Fibronectin or a ß1 integrin blocking antibody completely abrogated IFN-γ-dependent S. aureus binding and reduced S. aureus-dependent CXCL8 secretion. These data demonstrate that IFN-γ primes huMCs for enhanced anti-bacterial and pro-inflammatory responses to S. aureus, partially mediated by ß1 integrin.


Assuntos
Interferon gama/imunologia , Mastócitos/imunologia , Mastócitos/microbiologia , Staphylococcus aureus/imunologia , Imunidade Adaptativa , Animais , Células Cultivadas , Quimiocinas CC/genética , Quimiocinas CC/metabolismo , Citocinas/genética , Citocinas/metabolismo , Fator Estimulador de Colônias de Granulócitos e Macrófagos/genética , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Imunidade Inata , Mediadores da Inflamação/metabolismo , Integrina beta1/metabolismo , Interferon gama/farmacologia , Interleucina-8/genética , Interleucina-8/metabolismo , Mastócitos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Staphylococcus aureus/patogenicidade , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA