Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Allergy ; 77(3): 856-869, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34460953

RESUMO

BACKGROUND: Children with asthma have impaired production of interleukin (IL) 37; in mice, IL-37 reduces hallmarks of experimental allergic asthma (EAA). However, it remains unclear how IL-37 exerts its inhibitory properties in asthma. This study aimed to identify the mechanism(s) by which IL-37 controls allergic inflammation. METHODS: IL-37 target cells were identified by single-cell RNA-seq of IL-1R5 and IL-1R8. Airway tissues were isolated by laser-capture microdissection and examined by microarray-based gene expression analysis. Mononuclear cells (MNC) and airway epithelial cells (AECs) were isolated and stimulated with allergen, IL-1ß, or IL-33 together with recombinant human (rh) IL-37. Wild-type, IL-1R1- and IL-33-deficient mice with EAA were treated with rhIL-37. IL-1ß, IL-33, and IL-37 levels were determined in sputum and nasal secretions from adult asthma patients without glucocorticoid therapy. RESULTS: IL-37 target cells included AECs, T cells, and dendritic cells. In mice with EAA, rhIL-37 led to differential expression of >90 genes induced by IL-1ß and IL-33. rhIL-37 reduced production of Th2 cytokines in allergen-activated MNCs from wild-type but not from IL-1R1-deficient mice and inhibited IL-33-induced Th2 cytokine release. Furthermore, rhIL-37 attenuated IL-1ß- and IL-33-induced pro-inflammatory mediator expression in murine AEC cultures. In contrast to wild-type mice, hIL-37 had no effect on EAA in IL-1R1- or IL-33-deficient mice. We also observed that expression/production ratios of both IL-1ß and IL-33 to IL-37 were dramatically increased in asthma patients compared to healthy controls. CONCLUSION: IL-37 downregulates allergic airway inflammation by counterbalancing the disease-amplifying effects of IL-1ß and IL-33.


Assuntos
Asma , Interleucina-33 , Alérgenos , Animais , Asma/metabolismo , Citocinas , Modelos Animais de Doenças , Humanos , Inflamação , Pulmão/metabolismo , Camundongos , Células Th2
2.
Med Microbiol Immunol ; 209(4): 531-543, 2020 Aug.
Artigo em Inglês | MEDLINE | ID: mdl-32507938

RESUMO

Mast cells (MCs) are long-living immune cells highly specialized in the storage and release of different biologically active compounds and are involved in the regulation of innate and adaptive immunity. MC degranulation and replacement of MC granules are accompanied by active membrane remodelling. Tetraspanins represent an evolutionary conserved family of transmembrane proteins. By interacting with lipids and other membrane and intracellular proteins, they are involved in organisation of membrane protein complexes and act as "molecular facilitators" connecting extracellular and cytoplasmic signaling elements. MCs express different tetraspanins and MC degranulation is accompanied by changes in membrane organisation. Therefore, tetraspanins are very likely involved in the regulation of MC exocytosis and membrane reorganisation after degranulation. Antiviral response and production of exosomes are further aspects of MC function characterized by dynamic changes of membrane organization. In this review, we pay a particular attention to tetraspanin gene expression in different human and murine MC populations, discuss tetraspanin involvement in regulation of key MC signaling complexes, and analyze the potential contribution of tetraspanins to MC antiviral response and exosome production. In-depth knowledge of tetraspanin-mediated molecular mechanisms involved in different aspects of the regulation of MC response will be beneficial for patients with allergies, characterized by overwhelming MC reactions.


Assuntos
Mastócitos/imunologia , Mastócitos/metabolismo , Tetraspaninas/imunologia , Tetraspaninas/metabolismo , Animais , Degranulação Celular , Exossomos/metabolismo , Humanos , Camundongos , Transdução de Sinais , Tetraspaninas/genética , Viroses/imunologia
3.
Front Immunol ; 10: 1174, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31191542

RESUMO

Mast cells (MCs) have long been mainly regarded as effector cells in IgE-associated allergic disorders with potential immunoregulatory roles. Located close to the allergen entry sites in the skin and mucosa, MCs can capture foreign substances such as allergens, toxins, or noxious substances and are exposed to the danger signals produced by epithelial cells. MC reactivity shaped by tissue-specific factors is crucial for allergic responses ranging from local skin reactions to anaphylactic shock. Development of Th2 response leading to allergen-specific IgE production is a prerequisite for MC sensitization and induction of FcεRI-mediated MC degranulation. Up to now, IgE production has been mainly associated with proteins, whereas lipids present in plant pollen grains, mite fecal particles, insect venoms, or food have been largely overlooked regarding their immunostimulatory and immunomodulatory properties. Recent studies, however, have now demonstrated that lipids affect the sensitization process by modulating innate immune responses of epithelial cells, dendritic cells, and NK-T cells and thus crucially contribute to the outcome of sensitization. Whether and how lipids affect also MC effector functions in allergic reactions has not yet been fully clarified. Here, we discuss how lipids can affect MC responses in the context of allergic inflammation. Direct effects of immunomodulatory lipids on MC degranulation, changes in local lipid composition induced by allergens themselves and changes in lipid transport affecting MC reactivity are possible mechanisms by which the function of MC might be modulated.


Assuntos
Imunomodulação , Lipídeos/imunologia , Mastócitos/imunologia , Mastócitos/metabolismo , Alérgenos/química , Alérgenos/imunologia , Animais , Degranulação Celular/imunologia , Suscetibilidade a Doenças , Humanos , Hipersensibilidade/etiologia , Hipersensibilidade/metabolismo , Metabolismo dos Lipídeos , Lipídeos/química , Lipídeos de Membrana/imunologia , Lipídeos de Membrana/metabolismo , Relação Estrutura-Atividade
4.
Front Immunol ; 10: 974, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31134071

RESUMO

Plant pollen are an important source of antigens that evoke allergic responses. Protein antigens have been the focus of studies aiming to elucidate the mechanisms responsible for allergic reactions to pollen. However, proteins are not the sole active agent present in pollen. It is known that pollen grains contain lipids essential for its reproduction and bioactive lipid mediators. These small molecular compounds are co-delivered with the allergens and hence have the potential to modulate the immune response of subjects by activating their innate immune cells. Previous reports showed that pollen associated lipid mediators exhibited neutrophil- and eosinophil-chemotactic activity and induced polarization of dendritic cells (DCs) toward a Th2-inducing phenotype. In our study we performed chemical analyses of the pollen associated lipids, that are rapidly released upon hydration. As main components we have identified different types of phytoprostanes (PhytoPs), and for the first time phytofurans (PhytoFs), with predominating 16-F1t-PhytoPs (PPF1-I), 9-F1t-PhytoPs (PPF1-II), 16-E1t-PhytoPs (PPE1-I) and 9-D1t-PhytoPs (PPE1-II), and 16(RS)-9-epi-ST-Δ14-10-PhytoFs. Interestingly 16-E1t-PhytoP and 9-D1t-PhytoPs were found to be bound to glycerol. Lipid-containing samples (aqueous pollen extract, APE) induced murine mast cell chemotaxis and IL-6 release, and enhanced their IgE-dependent degranulation, demonstrating a role for these lipids in the immediate effector phase of allergic inflammation. Noteworthy, mast cell degranulation seems to be dependent on glycerol-bound, but not free phytoprostanes. On murine dendritic cells, APE selectively induced the upregulation of CD1d, likely preparing lipid-antigen presentation to iNKT cells. Our report contributes to the understanding of the activity of lipid mediators in the immediate effector phase of allergic reactions but identifies a yet undescribed pathway for the recognition of pollen-derived glycolipids by iNKT cells.


Assuntos
Alérgenos/imunologia , Células Dendríticas/imunologia , Glicolipídeos/imunologia , Hipersensibilidade/imunologia , Lipídeos/imunologia , Phleum/imunologia , Alérgenos/análise , Alérgenos/isolamento & purificação , Animais , Antígenos CD1d/genética , Antígenos CD1d/metabolismo , Degranulação Celular/imunologia , Quimiotaxia de Leucócito/imunologia , Células Dendríticas/metabolismo , Ácidos Graxos Insaturados/imunologia , Ácidos Graxos Insaturados/isolamento & purificação , Furanos/imunologia , Furanos/isolamento & purificação , Glicolipídeos/metabolismo , Humanos , Lipídeos/análise , Lipídeos/isolamento & purificação , Mastócitos/imunologia , Mastócitos/fisiologia , Camundongos Endogâmicos C57BL , Camundongos Knockout , Phleum/química , Pólen/química , Pólen/imunologia
5.
Allergy ; 74(4): 767-779, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30341777

RESUMO

BACKGROUND: Food-induced anaphylaxis is a serious allergic reaction caused by Fcε-receptor activation on mast cells (MCs). The exact mechanisms breaking oral tolerance and the effector pathways driving food allergy remain elusive. As complement is activated in food-induced anaphylaxis, we aimed to assess the role of C5a in disease pathogenesis. METHODS: Oral antigen-induced food-induced anaphylaxis was induced in BALB/c wild-type (wt) and C5ar1-/- mice. Readouts included diarrhea development, changes in rectal temperature, hematocrit, antigen-specific serum IgE, MCPT-1, and intestinal MC numbers, as well as FcεR1-mediated MC functions including C5a receptor 1 (C5aR1) regulation. Further, histamine-mediated hypothermia and regulation of endothelial tight junctions were determined. RESULTS: Repeated oral OVA challenge resulted in diarrhea, hypothermia, increased hematocrit, high OVA-specific serum IgE, and MCPT-1 levels in wt mice. Male C5ar1-/- mice were completely whereas female C5ar1-/- mice were partially protected from anaphylaxis development. Serum MCPT-1 levels were reduced gender-independent, whereas IgE levels were reduced in male but not in female C5ar1-/- mice. Mechanistically, IgE-mediated degranulation and IL-6 production from C5ar1-/- BMMCs of both sexes were significantly reduced. Importantly, FcεR1 cross-linking strongly upregulated C5aR1 MC expression in vitro and in vivo. Finally, C5ar1-/- male mice were largely protected from histamine-induced hypovolemic shock, which was associated with protection from histamine-induced barrier dysfunction in vitro following C5aR targeting. CONCLUSIONS: Our findings identify C5aR1 activation as an important driver of IgE-mediated food allergy through regulation of allergen-specific IgE production, FcεR1-mediated MC degranulation, and histamine-driven effector functions preferentially in male mice.


Assuntos
Hipersensibilidade Alimentar/etiologia , Imunoglobulina E/sangue , Receptor da Anafilatoxina C5a/genética , Fatores Sexuais , Anafilaxia , Animais , Degranulação Celular , Quimases/sangue , Feminino , Masculino , Mastócitos/metabolismo , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Receptor da Anafilatoxina C5a/deficiência , Receptores de IgE/imunologia
6.
J Allergy Clin Immunol ; 144(4S): S4-S18, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-30468774

RESUMO

Mast cells (MCs), which are well known for their effector functions in TH2-skewed allergic and also autoimmune inflammation, have become increasingly acknowledged for their role in protection of health. It is now clear that they are also key modulators of immune responses at interface organs, such as the skin or gut. MCs can prime tissues for adequate inflammatory responses and cooperate with dendritic cells in T-cell activation. They also regulate harmful immune responses in trauma and help to successfully orchestrate pregnancy. This review focuses on the beneficial effects of MCs on tissue homeostasis and elimination of toxins or venoms. MCs can enhance pathogen clearance in many bacterial, viral, and parasitic infections, such as through Toll-like receptor 2-triggered degranulation, secretion of antimicrobial cathelicidins, neutrophil recruitment, or provision of extracellular DNA traps. The role of MCs in tumors is more ambiguous; however, encouraging new findings show they can change the tumor microenvironment toward antitumor immunity when adequately triggered. Uterine tissue remodeling by α-chymase (mast cell protease [MCP] 5) is crucial for successful embryo implantation. MCP-4 and the tryptase MCP-6 emerge to be protective in central nervous system trauma by reducing inflammatory damage and excessive scar formation, thereby protecting axon growth. Last but not least, proteases, such as carboxypeptidase A, released by FcεRI-activated MCs detoxify an increasing number of venoms and endogenous toxins. A better understanding of the plasticity of MCs will help improve these advantageous effects and hint at ways to cut down detrimental MC actions.


Assuntos
Imunidade Inata , Infecções/imunologia , Mastócitos/imunologia , Animais , Catelicidinas/metabolismo , Degranulação Celular , Implantação do Embrião , Feminino , Homeostase , Humanos , Gravidez , Receptor 2 Toll-Like/metabolismo
7.
Sci Rep ; 8(1): 8636, 2018 06 05.
Artigo em Inglês | MEDLINE | ID: mdl-29872093

RESUMO

IL-9 is a cytokine with pleiotropic function that mediates allergic inflammation and immunity to intestinal helminth parasites. Accumulating evidence suggests that IL-9 acts via both, initiation and regulation of adaptive immune responses and direct activation of intestinal effector pathways. Here we use IL-9 receptor deficient mice on BALB/c and C57BL/6 genetic background to dissect effector and regulatory functions of IL-9 during infection with the parasitic nematode Strongyloides ratti. IL-9 receptor-deficient mice displayed increased intestinal parasite burden and prolonged infection irrespective of the genetic background of the mice. Increased parasite burden was correlated to a reciprocally reduced early degranulation of mucosal mast cells, reduced intestinal IL-13 expression and caused by IL-9 receptor deficiency on hematopoietic cells. We observed additional significant changes in the adaptive immune response to S. ratti infection in the absence of the IL-9 receptor that depended on the mouse strain. However, the generation of protective memory to a second infection was intact in IL-9 receptor-deficient mice, irrespective of the genetic background. In summary, our results support a central role for IL-9 as an early mast cell activating effector cytokine during intestinal helminth infection while non-redundant functions in the initiation and amplification of adaptive immune responses were not apparent.


Assuntos
Interleucina-9/metabolismo , Mastócitos/imunologia , Strongyloides ratti/imunologia , Estrongiloidíase/imunologia , Animais , Modelos Animais de Doenças , Fatores Imunológicos/metabolismo , Memória Imunológica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , Camundongos Knockout , Receptores de Interleucina-9/deficiência
8.
Front Immunol ; 7: 514, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27917175

RESUMO

Suppressor of cytokine signaling 1 (SOCS1) is a negative feedback inhibitor of cytoplasmic Janus kinase and signal transducer and activator of transcription (STAT) signaling. SOCS1 also contains a nuclear localization sequence (NLS), yet, the in vivo importance of nuclear translocation is unknown. We generated transgenic mice containing mutated Socs1ΔNLS that fails to translocate in the cell nucleus (MGLtg mice). Whereas mice fully deficient for SOCS1 die within the first 3 weeks due to excessive interferon signaling and multiorgan inflammation, mice expressing only non-nuclear Socs1ΔNLS (Socs1-/-MGLtg mice) were rescued from early lethality. Canonical interferon gamma signaling was still functional in Socs1-/-MGLtg mice as shown by unaltered tyrosine phosphorylation of STAT1 and whole genome expression analysis. However, a subset of NFκB inducible genes was dysregulated. Socs1-/-MGLtg mice spontaneously developed low-grade inflammation in the lung and had elevated Th2-type cytokines. Upon ovalbumin sensitization and challenge, airway eosinophilia was increased in Socs1-/-MGLtg mice. Decreased transepithelial electrical resistance in trachea epithelial cells from Socs1-/-MGLtg mice suggests disrupted epithelial cell barrier. The results indicate that nuclear SOCS1 is a regulator of local immunity in the lung and unravel a so far unrecognized function for SOCS1 in the cell nucleus.

9.
Sci Rep ; 6: 19699, 2016 Jan 29.
Artigo em Inglês | MEDLINE | ID: mdl-26822794

RESUMO

The common gamma (γc)-chain cytokine interleukin 15 (IL15) is a multifunctional immune-modulator which impacts the generation, maturation and activity of many cell types of the innate, as well as the adaptive immune system, including natural killer (NK) and CD8(+) T cells. Using a new series of transgenic mice, we analyzed the in vivo potential of IL15 as an immune-regulator when available at different concentrations or delivery modes, i.e. soluble monomer or complexed to its specific receptor α (Rα)-chain. We have identified distinct effects on selected IL15-responsive populations. While CD8(+) T cells required complexed forms of IL15/IL15Rα for full functionality, mature NK populations were rescued in an IL15/IL15Rα-deficient environment by high levels of CD11c-restricted IL15. These IL15-conditions were sufficient to limit tumor formation in a lung metastasis model indicating that the NK cell populations were fully functional. These data underline the potential of "free" IL15 in the absence of Rα-complex as a powerful and specific immuno-modulator, which may be beneficial where selective immune-activation is desired.


Assuntos
Apresentação de Antígeno , Antígeno CD11c/imunologia , Antígeno CD11c/metabolismo , Citotoxicidade Imunológica , Interleucina-15/metabolismo , Células Matadoras Naturais/fisiologia , Animais , Antígeno CD11c/genética , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Diferenciação Celular , Linhagem Celular Tumoral , Interleucina-15/genética , Células Matadoras Naturais/citologia , Melanoma Experimental , Camundongos , Camundongos Knockout , Camundongos Transgênicos , Metástase Neoplásica , Neoplasias/imunologia , Neoplasias/metabolismo , Neoplasias/patologia , Nódulos Linfáticos Agregados/citologia , Nódulos Linfáticos Agregados/imunologia , Regiões Promotoras Genéticas , Receptores de Interleucina-15/genética , Timo/citologia , Timo/imunologia
10.
Eur J Immunol ; 46(2): 432-9, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26564814

RESUMO

The interaction between tissue-resident mast cells (MCs) and recruited immune cells contributes to tissue immunosurveillance. However, the cells, mechanisms, and receptors involved in this crosstalk remain ill defined. Invariant natural killer T (iNKT) cells are CD1d-restricted innate lymphocytes that recognize glycolipid antigens and have emerged as critical players in immunity. Here, we show that primary mouse peritoneal MCs express surface CD1d, which is upregulated in vivo following administration of alpha-galactosylceramide. In contrast, in BM-derived MCs CD1d was found to be stored intracellularly and to relocate at the cell surface upon IgE-mediated degranulation. Activated BM-derived MCs expressing surface CD1d and loaded with alpha-galactosylceramide were found to induce iNKT-cell proliferation and the release of IFN-γ, IL-13, and IL-4 in a CD1d-restricted manner. Moreover, the costimulatory molecules CD48, CD137L, CD252, CD274, and CD275 affected MC-induced IFN-γ release and iNKT-cell proliferation. Interestingly, among the costimulatory molecules, CD48 and CD252 exhibited a distinctly regulatory activity on iNKT-cell release of both IFN-γ and IL-13. In conclusion, we demonstrate that the crosstalk between MCs and iNKT cells may regulate inflammatory immune responses.


Assuntos
Antígenos CD1d/metabolismo , Ativação Linfocitária , Mastócitos/imunologia , Células T Matadoras Naturais/imunologia , Ligante OX40/metabolismo , Animais , Antígenos CD1d/genética , Comunicação Celular , Diferenciação Celular , Células Cultivadas , Citocinas/metabolismo , Galactosilceramidas/administração & dosagem , Regulação da Expressão Gênica , Imunidade Inata/efeitos dos fármacos , Imunomodulação , Ativação Linfocitária/efeitos dos fármacos , Mastócitos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Monitorização Imunológica , Ligante OX40/genética
11.
Eur J Immunol ; 43(4): 979-88, 2013 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-23404570

RESUMO

Mast cells are important cellular constituents of epithelial-mesenchymal interactions, densely located at sites of microbial entry into the host where they are continuously exposed to products from commensals. In order to avoid excessive activation and the associated pathology, mast cell responses to TLR agonists must be tightly regulated. Here, we show that exposure in vitro to subactivating levels of the epithelial cell product, IL-33, renders mast cells insensitive to bacterial cell wall products. Mast cell responsiveness to Ag, cytoplasmic dsDNA, and TLR7/8 agonists is unaffected or enhanced by IL-33. The IL-33-induced mast cell selective tolerance requires the IL-33 receptor ST2 and peritoneal mast cells from St2(-/-) mice display a constitutively activated phenotype, demonstrated by increased expression of activation markers including CD11b and CD28. IL-33 exposure neither affects the levels of TLR4, MyD88, TIRAP, IL-1R associated kinase 2 (IRAK2), or IRAK4, nor induces persistent A20 or Tollip expression, but potently causes ST2-dependent IRAK1 degradation. We show that while IRAK2 is redundant for TLR4 signaling, IRAK1 is essential for TLR4 signaling in mast cells. We suggest that IL-33 produced during homeostasis retains mast cells in an unresponsive state to bacterial cell wall products via IRAK1 degradation, thus preventing chronic inflammation and tissue destruction.


Assuntos
Tolerância Imunológica/efeitos dos fármacos , Quinases Associadas a Receptores de Interleucina-1/metabolismo , Interleucinas/farmacologia , Lipopolissacarídeos/imunologia , Mastócitos/imunologia , Mastócitos/metabolismo , Animais , Células Cultivadas , Endotoxinas/imunologia , Tolerância Imunológica/genética , Quinases Associadas a Receptores de Interleucina-1/genética , Proteína 1 Semelhante a Receptor de Interleucina-1 , Interleucina-33 , Mastócitos/efeitos dos fármacos , Camundongos , Camundongos Knockout , Modelos Biológicos , Proteólise , Receptores de Interleucina/genética , Receptores de Interleucina/metabolismo , Transdução de Sinais
12.
Eur J Immunol ; 43(1): 194-208, 2013 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-23042080

RESUMO

Syntaxin 11 (STX11) controls vesicular trafficking and is a key player in exocytosis. Since Stx11 mutations are causally associated with a familial hemophagocytic lymphohistio-cytosis, we wanted to clarify whether STX11 is functionally important for key immune cell populations. This was studied in primary cells obtained from newly generated Stx11(-/-) mice. Our data revealed that STX11 is not only widely expressed in different immune cells, but also induced upon LPS or IFN-γ treatment. However, Stx11 deficiency does not affect macrophage phagocytic function and cytokine secretion, mast cell activation, or antigen presentation by DCs. Instead, STX11 selectively controls lymphocyte cytotoxicity in NK and activated CD8(+) T cells and degranulation in neutrophils. Stx11(-/-) NK cells and CTLs show impaired degranulation, despite a comparable activation, maturation and expression of the complex-forming partners MUNC18-2 and VTI1B. In addition, Stx11(-/-) CTLs and NK cells produce abnormal levels of IFN-γ. Since functional reconstitution rescues the defective phenotype of Stx11(-/-) CTLs, we suggest a direct, specific and key role of STX11 in controlling lymphocyte cytotoxicity, cytokine production and secretion. Finally, we show that these mice are a very useful tool for dissecting the role of STX11 in vesicular trafficking and secretion.


Assuntos
Linfócitos T CD8-Positivos/imunologia , Células Matadoras Naturais/imunologia , Neutrófilos/imunologia , Proteínas Qa-SNARE/imunologia , Animais , Degranulação Celular/genética , Linhagem Celular , Citotoxicidade Imunológica/genética , Humanos , Interferon gama/imunologia , Lipopolissacarídeos/imunologia , Linfo-Histiocitose Hemofagocítica/genética , Linfo-Histiocitose Hemofagocítica/imunologia , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Knockout , Proteínas Munc18/imunologia , Mutação/genética , Proteínas Qa-SNARE/genética , Proteínas Qb-SNARE/imunologia
13.
PLoS One ; 7(9): e45234, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23028866

RESUMO

Regulatory CD8(+) T cells are critical for self-tolerance and restricting excessive immune responses. The variety of immune functions they fulfill, the heterogeneity of their phenotype, and the mechanism of action are still poorly understood. Here we describe that regulatory CD8(+) T cells exhibiting immunosuppressive actions in vitro and in vivo are recognized as CD38(high) T cells and present in naive mice. CD38 is a glycosylated membrane protein with ectonucleotidase properties. CD8(+)CD38(high) (CD44(+)CD122(+)CD62L(high)) lymphocytes suppress CD4(+) effector T-cell proliferation in an antigen-non specific manner via IFN-γ. While direct cell-to-cell contact is needed for this suppressor activity, it is independent of membrane-bound TGF-ß and granzyme B release. IL-15 potentiates the suppressive activity of CD8(+)CD38(high) T cells and controls their survival and expansion. In humans CD8(+)CD38(high) T cells inhibit CD4(+) effector T cell proliferation. In vivo, CD8(+)CD38(high), but not CD8(+)CD38(-) T cells mitigate murine experimental autoimmune encephalomyelitis (EAE) by reducing the clinical score and delaying disease occurrence. EAE suppression is enhanced by pre-treatment of CD8(+)CD38(high) T cells with IL-15. These findings add evidence that the expression of ectoenzyme receptor family members positively correlates with suppressor functions and identifies CD8(+)CD38(high) T cells as potential inhibitors of excessive immune responses.


Assuntos
ADP-Ribosil Ciclase 1/imunologia , Linfócitos T CD8-Positivos/patologia , Encefalomielite Autoimune Experimental/patologia , Interferon gama/imunologia , Glicoproteínas de Membrana/imunologia , Nucleotidases/imunologia , Linfócitos T Reguladores/patologia , ADP-Ribosil Ciclase 1/metabolismo , Animais , Antígenos CD/imunologia , Antígenos CD/metabolismo , Linfócitos T CD8-Positivos/imunologia , Linfócitos T CD8-Positivos/metabolismo , Comunicação Celular/imunologia , Proliferação de Células , Sobrevivência Celular , Células Cultivadas , Encefalomielite Autoimune Experimental/imunologia , Encefalomielite Autoimune Experimental/metabolismo , Feminino , Citometria de Fluxo , Memória Imunológica , Imunofenotipagem , Interleucina-15/imunologia , Ativação Linfocitária , Glicoproteínas de Membrana/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , Nucleotidases/metabolismo , Linfócitos T Reguladores/imunologia , Linfócitos T Reguladores/metabolismo
14.
Int Arch Allergy Immunol ; 158(1): 43-53, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22212634

RESUMO

BACKGROUND: Allergen-specific immunoglobulin (Ig) E initiates the effector cascade of allergic asthma and has been identified as a valuable target for therapeutic treatment of this disease. The proteasome inhibitor bortezomib was previously shown to deplete Ig-secreting plasma cells and to efficiently suppress Ig serum titers. The present study aimed at evaluating the therapeutic potential of the proteasome inhibitor bortezomib in allergic bronchial asthma. METHODS: To address this question, a chronic experimental asthma mouse model was used in a therapeutic setting. Mice were sensitized to ovalbumin (OVA) and challenged with OVA aerosol for 12 weeks. After 6 weeks of challenge, bortezomib treatment was started and continued for 1 week (short-term) or 6 weeks (long-term) with a dosage of 0.75 mg/kg body weight twice a week. Lung function, lung histology, Ig serum titers and plasma cell numbers were assessed. RESULTS: Whereas short-term treatment lowered bronchoalveolar lavage eosinophils, long-term treatment considerably reduced serum titers of anti-OVA IgE in mice with chronic experimental asthma. However, neither short-term nor long-term treatment significantly reduced plasma cell numbers, anti-OVA IgG1 serum titers or allergic airway inflammation or ablated airway hyperresponsiveness. CONCLUSION: Our results suggest that bortezomib treatment has only limited value as plasma cell-depleting therapy against allergic bronchial asthma.


Assuntos
Alérgenos/imunologia , Antiasmáticos/uso terapêutico , Asma/tratamento farmacológico , Ácidos Borônicos/uso terapêutico , Imunoglobulina E/imunologia , Pirazinas/uso terapêutico , Animais , Asma/imunologia , Asma/fisiopatologia , Bortezomib , Doença Crônica , Modelos Animais de Doenças , Eosinófilos/efeitos dos fármacos , Eosinófilos/imunologia , Feminino , Imunoglobulina E/sangue , Imunoglobulina G/sangue , Imunoglobulina G/imunologia , Pulmão/imunologia , Pulmão/patologia , Pulmão/fisiopatologia , Camundongos , Camundongos Endogâmicos BALB C , Plasmócitos/efeitos dos fármacos , Plasmócitos/imunologia , Falha de Tratamento
15.
J Exp Med ; 208(9): 1777-87, 2011 Aug 29.
Artigo em Inglês | MEDLINE | ID: mdl-21844203

RESUMO

Mast cell (MC) activation via aggregation of the high affinity IgE receptor (FcεRI) causes degranulation and release of proinflammatory mediators in a process that involves the reorganization of the actin cytoskeleton. However, the regulatory pathways and the molecular links between cytoskeletal changes and MC function are incompletely understood. In this study, we provide genetic evidence for a critical role of the actin-regulatory proteins Coronin1a (Coro1a) and Coro1b on exocytic pathways in MCs: Coro1a(-/-) bone marrow-derived MCs exhibit increased FcεRI-mediated degranulation of secretory lysosomes but significantly reduced secretion of cytokines. Hyperdegranulation of Coro1a(-/-) MCs is further augmented by the additional loss of Coro1b. In vivo, Coro1a(-/-)Coro1b(-/-) mice displayed enhanced passive cutaneous anaphylaxis. Functional reconstitution assays revealed that the inhibitory effect of Coro1a on MC degranulation strictly correlates with cortical localization of Coro1a, requires its filamentous actin-binding activity, and is regulated by phosphorylation of Ser2 of Coro1a. Thus, coronin proteins, and in turn the actin cytoskeleton, exhibit a functional dichotomy as differential regulators of degranulation versus cytokine secretion in MC biology.


Assuntos
Degranulação Celular/fisiologia , Citocinas/metabolismo , Citoesqueleto/metabolismo , Mastócitos/metabolismo , Proteínas dos Microfilamentos/metabolismo , Animais , Citocinas/genética , Citoesqueleto/genética , Camundongos , Camundongos Mutantes , Proteínas dos Microfilamentos/genética , Anafilaxia Cutânea Passiva/genética , Fosforilação/fisiologia
17.
Eur J Cell Biol ; 90(6-7): 476-83, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21439673

RESUMO

Since cytokines are potent immunoregulators that can determine the fate of an immune response, their expression is tightly regulated at the transcriptional level. Recent research, however, has also revealed complex post-translational mechanisms through which cytokine secretion, and thereby cytokine activity, is regulated. Here, we review the progress in our understanding of the portfolio of pathways that regulate cytokine intracellular storage, transport, and release. Like other secreted proteins, cytokines utilize canonical and non-canonical secretory pathways for extracellular release. Illustrated by IL-1ß, IL-2, TNF-α, IL-12 and IL-15 secretion as selected examples, we discuss common and alternative cytokine secretion pathways and relate them to the consequences these distinct pathways have for cytokine function, mode of action and stability.


Assuntos
Citocinas/metabolismo , Animais , Citocinas/biossíntese , Humanos
20.
Blood ; 116(15): 2665-75, 2010 Oct 14.
Artigo em Inglês | MEDLINE | ID: mdl-20595514

RESUMO

Mast cell (MC) differentiation, survival, and activation are controlled by the membrane tyrosine kinase c-Kit upon interaction with stem cell factor (SCF). Here we describe a single point mutation induced by N-ethyl-N-nitrosurea (ENU) mutagenesis in C57BL/6J mice-an A to T transversion at position 2388 (exon 17) of the c-Kit gene, resulting in the isoleucine 787 substitution by phenylalanine (787F), and analyze the consequences of this mutation for ligand binding, signaling, and MC development. The Kit(787F/787F) mice carrying the single amino acid exchange of c-Kit lacks both mucosal and connective tissue-type MCs. In bone marrow-derived mast cells (BMMCs), the 787F mutation does not affect SCF binding and c-Kit receptor shedding, but strongly impairs SCF-induced cytokine production, degranulation enhancement, and apoptosis rescue. Interestingly, c-Kit downstream signaling in 787F BMMCs is normally initiated (Erk1/2 and p38 activation as well as c-Kit autophosphorylation) but fails to be sustained thereafter. In addition, 787F c-Kit does not efficiently mediate Cbl activation, leading to the absence of subsequent receptor ubiquitination and impaired c-Kit internalization. Thus, I787 provides nonredundant signals for c-Kit internalization and functionality.


Assuntos
Diferenciação Celular/fisiologia , Mastócitos/citologia , Mastócitos/metabolismo , Proteínas Proto-Oncogênicas c-kit/química , Proteínas Proto-Oncogênicas c-kit/metabolismo , Sequência de Aminoácidos , Substituição de Aminoácidos , Animais , Sequência de Bases , Diferenciação Celular/efeitos dos fármacos , Diferenciação Celular/genética , Linhagem Celular , Sobrevivência Celular/genética , Sobrevivência Celular/fisiologia , Primers do DNA/genética , Técnicas In Vitro , Interleucina-3/farmacologia , Isoleucina/química , Mastócitos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos C3H , Camundongos Endogâmicos C57BL , Camundongos Mutantes , Mutagênese Sítio-Dirigida , Proteínas Mutantes/química , Proteínas Mutantes/genética , Proteínas Mutantes/metabolismo , Mutação Puntual , Proteínas Proto-Oncogênicas c-cbl/metabolismo , Proteínas Proto-Oncogênicas c-kit/genética , Transdução de Sinais , Fator de Células-Tronco/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...