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










Base de dados
Intervalo de ano de publicação
1.
Sci Adv ; 9(13): eade7647, 2023 03 31.
Artigo em Inglês | MEDLINE | ID: mdl-37000867

RESUMO

Improved approaches to expanding the pool of donor lungs suitable for transplantation are critically needed for the growing population with end-stage lung disease. Cross-circulation (XC) of whole blood between swine and explanted human lungs has previously been reported to enable the extracorporeal recovery of donor lungs that declined for transplantation due to acute, reversible injuries. However, immunologic interactions of this xenogeneic platform have not been characterized, thus limiting potential translational applications. Using flow cytometry and immunohistochemistry, we demonstrate that porcine immune cell and immunoglobulin infiltration occurs in this xenogeneic XC system, in the context of calcineurin-based immunosuppression and complement depletion. Despite this, xenogeneic XC supported the viability, tissue integrity, and physiologic improvement of human donor lungs over 24 hours of xeno-support. These findings provide targets for future immunomodulatory strategies to minimize immunologic interactions on this organ support biotechnology.


Assuntos
Transplante de Pulmão , Pulmão , Humanos , Suínos , Animais , Terapia de Imunossupressão
2.
Gastroenterology ; 159(6): 2077-2091.e8, 2020 12.
Artigo em Inglês | MEDLINE | ID: mdl-32891625

RESUMO

BACKGROUND & AIMS: Severe injury to the lining of the stomach leads to changes in the epithelium (reprogramming) that protect and promote repair of the tissue, including development of spasmolytic polypeptide-expressing metaplasia (SPEM) and tuft and foveolar cell hyperplasia. Acute gastric damage elicits a type-2 inflammatory response that includes production of type-2 cytokines and infiltration by eosinophils and alternatively activated macrophages. Stomachs of mice that lack interleukin 33 (IL33) or interleukin 13 (IL13) did not undergo epithelial reprogramming after drug-induced injury. We investigated the role of group 2 innate lymphoid cells (ILC2s) in gastric epithelial repair. METHODS: Acute gastric injury was induced in C57BL/6J mice (wild-type and RAG1 knockout) by administration of L635. We isolated ILC2s by flow cytometry from stomachs of mice that were and were not given L635 and performed single-cell RNA sequencing. ILC2s were depleted from wild-type and RAG1-knockout mice by administration of anti-CD90.2. We assessed gastric cell lineages, markers of metaplasia, inflammation, and proliferation. Gastric tissue microarrays from patients with gastric adenocarcinoma were analyzed by immunostaining. RESULTS: There was a significant increase in the number of GATA3-positive ILC2s in stomach tissues from wild-type mice after L635-induced damage, but not in stomach tissues from IL33-knockout mice. We characterized a marker signature of gastric mucosal ILC2s and identified a transcription profile of metaplasia-associated ILC2s, which included changes in expression of Il5, Il13, Csf2, Pd1, and Ramp3; these changes were validated by quantitative polymerase chain reaction and immunocytochemistry. Depletion of ILC2s from mice blocked development of metaplasia after L635-induced injury in wild-type and RAG1-knockout mice and prevented foveolar and tuft cell hyperplasia and infiltration or activation of macrophages after injury. Numbers of ILC2s were increased in stomach tissues from patients with SPEM compared with patients with normal corpus mucosa. CONCLUSIONS: In analyses of stomach tissues from mice with gastric tissue damage and patients with SPEM, we found evidence of type 2 inflammation and increased numbers of ILC2s. Our results suggest that ILC2s coordinate the metaplastic response to severe gastric injury.


Assuntos
Mucosa Gástrica/patologia , Imunidade Inata , Subpopulações de Linfócitos/imunologia , Animais , Modelos Animais de Doenças , Mucosa Gástrica/efeitos dos fármacos , Mucosa Gástrica/imunologia , Humanos , Interleucina-33/genética , Metaplasia/induzido quimicamente , Metaplasia/genética , Metaplasia/imunologia , Camundongos , Camundongos Knockout
3.
Methods Mol Biol ; 2121: 7-22, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32147782

RESUMO

Understanding the origins and developmental trajectory of innate lymphoid cell (ILC) progenitors has been of substantial interest to the fields of ILC biology and immunology. While mature ILC are rare lymphocytes, ILC progenitors represent an even smaller fraction of cells, providing additional challenges in studying them. Moreover, though the approaches to studying these cells are conceptually straightforward, the technical nuances that underlie them can substantially affect the quality of the data. Herein, we provide a detailed protocol for assessing the frequency of ILC progenitors in the bone marrow, their phenotype, and their potential to develop into mature ILC. These methods make up the foundation of in vivo investigations into ILC development, and we hope these thorough protocols and associated notes facilitate additional, high-quality inquiries into this fascinating field.


Assuntos
Transferência Adotiva/métodos , Células da Medula Óssea , Células Matadoras Naturais/citologia , Fígado/citologia , Linfócitos/citologia , Células Progenitoras Linfoides/citologia , Linfopoese/imunologia , Animais , Medula Óssea , Células da Medula Óssea/citologia , Linhagem da Célula , Feminino , Citometria de Fluxo , Células Matadoras Naturais/imunologia , Fígado/imunologia , Linfócitos/imunologia , Células Progenitoras Linfoides/imunologia , Células Progenitoras Linfoides/metabolismo , Camundongos , Camundongos Endogâmicos BALB C
4.
J Immunol ; 203(6): 1457-1467, 2019 09 15.
Artigo em Inglês | MEDLINE | ID: mdl-31391233

RESUMO

IL-33 is an IL-1 family member protein that is a potent driver of inflammatory responses in both allergic and nonallergic disease. This proinflammatory effect is mediated primarily by extracellular release of IL-33 from stromal cells and binding of the C-terminal domain of IL-33 to its receptor ST2 on targets such as CD4+ Th2 cells, ILC2, and mast cells. Notably, IL-33 has a distinct N-terminal domain that mediates nuclear localization and chromatin binding. However, a defined in vivo cell-intrinsic role for IL-33 has not been established. We identified IL-33 expression in the nucleus of progenitor B (pro-B) and large precursor B cells in the bone marrow, an expression pattern unique to B cells among developing lymphocytes. The IL-33 receptor ST2 was not expressed within the developing B cell lineage at either the transcript or protein level. RNA sequencing analysis of wild-type and IL-33-deficient pro-B and large precursor B cells revealed a unique, IL-33-dependent transcriptional profile wherein IL-33 deficiency led to an increase in E2F targets, cell cycle genes, and DNA replication and a decrease in the p53 pathway. Using mixed bone marrow chimeric mice, we demonstrated that IL-33 deficiency resulted in an increased frequency of developing B cells via a cell-intrinsic mechanism starting at the pro-B cell stage paralleling IL-33 expression. Finally, IL-33 was detectable during early B cell development in humans and IL33 mRNA expression was decreased in B cell chronic lymphocytic leukemia samples compared with healthy controls. Collectively, these data establish a cell-intrinsic, ST2-independent role for IL-33 in early B cell development.


Assuntos
Linfócitos B/imunologia , Interleucina-33/imunologia , Adulto , Animais , Replicação do DNA/imunologia , Feminino , Humanos , Leucemia Linfocítica Crônica de Células B/imunologia , Masculino , Mastócitos/imunologia , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Endogâmicos C57BL , RNA Mensageiro/imunologia , Transdução de Sinais/imunologia , Células Th2/imunologia , Proteína Supressora de Tumor p53/imunologia
5.
Ann Am Thorac Soc ; 15(Suppl 3): S205-S209, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-30431348

RESUMO

Respiratory syncytial virus (RSV) is a leading cause of hospitalization of infants worldwide each year. Both host and viral factors host factors predispose a subset of what appear to be healthy infants to severe RSV-induced disease. In this review, we outline many genetic and immunologic factors that contribute to airway obstruction that contributes to the severity of RSV infection.


Assuntos
Infecções por Vírus Respiratório Sincicial/etiologia , Vírus Sincicial Respiratório Humano/patogenicidade , Humanos , Lactente , Mucinas/metabolismo , Muco/fisiologia , Infecções por Vírus Respiratório Sincicial/metabolismo , Infecções por Vírus Respiratório Sincicial/patologia
7.
J Allergy Clin Immunol ; 142(5): 1515-1528.e8, 2018 11.
Artigo em Inglês | MEDLINE | ID: mdl-29331643

RESUMO

BACKGROUND: IL-33 is one of the most consistently associated gene candidates for asthma identified by using a genome-wide association study. Studies in mice and in human cells have confirmed the importance of IL-33 in inducing type 2 cytokine production from both group 2 innate lymphoid cells (ILC2s) and TH2 cells. However, there are no pharmacologic agents known to inhibit IL-33 release from airway cells. OBJECTIVE: We sought to determine the effect of glucagon-like peptide 1 receptor (GLP-1R) signaling on aeroallergen-induced airway IL-33 production and release and on innate type 2 airway inflammation. METHODS: BALB/c mice were challenged intranasally with Alternaria extract for 4 consecutive days. GLP-1R agonist or vehicle was administered starting either 2 days before the first Alternaria extract challenge or 1 day after the first Alternaria extract challenge. RESULTS: GLP-1R agonist treatment starting 2 days before the first Alternaria extract challenge decreased IL-33 release in the bronchoalveolar lavage fluid and dual oxidase 1 (Duox1) mRNA expression 1 hour after the first Alternaria extract challenge and IL-33 expression in lung epithelial cells 24 hours after the last Alternaria extract challenge. Furthermore, GLP-1R agonist significantly decreased the number of ILC2s expressing IL-5 and IL-13, lung protein expression of type 2 cytokines and chemokines, the number of perivascular eosinophils, mucus production, and airway responsiveness compared with vehicle treatment. GLP-1R agonist treatment starting 1 day after the first Alternaria extract challenge also significantly decreased eosinophilia and type 2 cytokine and chemokine expression in the airway after 4 days of Alternaria extract challenge. CONCLUSION: These results reveal that GLP-1R signaling might be a therapy to reduce IL-33 release and inhibit the ILC2 response to protease-containing aeroallergens, such as Alternaria.


Assuntos
Asma/imunologia , Peptídeo 1 Semelhante ao Glucagon/imunologia , Receptor do Peptídeo Semelhante ao Glucagon 1/imunologia , Interleucina-33/imunologia , Alérgenos/imunologia , Alternaria/imunologia , Animais , Citocinas/imunologia , Dermatophagoides pteronyssinus/imunologia , Eosinofilia/imunologia , Feminino , Receptor do Peptídeo Semelhante ao Glucagon 1/agonistas , Imunidade Inata , Pulmão/citologia , Pulmão/imunologia , Linfócitos/imunologia , Camundongos Endogâmicos BALB C , Camundongos Transgênicos , Muco/imunologia , Transdução de Sinais
8.
J Exp Med ; 215(1): 263-281, 2018 01 02.
Artigo em Inglês | MEDLINE | ID: mdl-29222107

RESUMO

Group 2 innate lymphoid cells (ILC2s) are effector cells within the mucosa and key participants in type 2 immune responses in the context of allergic inflammation and infection. ILC2s develop in the bone marrow from common lymphoid progenitor cells, but little is known about how ILC2s egress from the bone marrow for hematogenous trafficking. In this study, we identified a critical role for IL-33, a hallmark peripheral ILC2-activating cytokine, in promoting the egress of ILC2 lineage cells from the bone marrow. Mice lacking IL-33 signaling had normal development of ILC2s but retained significantly more ILC2 progenitors in the bone marrow via augmented expression of CXCR4. Intravenous injection of IL-33 or pulmonary fungal allergen challenge mobilized ILC2 progenitors to exit the bone marrow. Finally, IL-33 enhanced ILC2 trafficking to the lungs in a parabiosis mouse model of tissue disruption and repopulation. Collectively, these data demonstrate that IL-33 plays a critical role in promoting ILC2 egress from the bone marrow.


Assuntos
Células da Medula Óssea/metabolismo , Imunidade Inata , Interleucina-33/metabolismo , Subpopulações de Linfócitos/metabolismo , Alérgenos/imunologia , Animais , Antígenos de Fungos/imunologia , Medula Óssea , Células da Medula Óssea/citologia , Células da Medula Óssea/imunologia , Contagem de Células , Diferenciação Celular , Proteína 1 Semelhante a Receptor de Interleucina-1/deficiência , Interleucina-33/genética , Subpopulações de Linfócitos/citologia , Subpopulações de Linfócitos/imunologia , Camundongos , Camundongos Knockout , Receptores CXCR4/genética , Receptores CXCR4/metabolismo , Transdução de Sinais
10.
Cell Rep ; 21(9): 2487-2499, 2017 11 28.
Artigo em Inglês | MEDLINE | ID: mdl-29186686

RESUMO

Sex hormones regulate many autoimmune and inflammatory diseases, including asthma. As adults, asthma prevalence is 2-fold greater in women compared to men. The number of group 2 innate lymphoid cells (ILC2) is increased in patients with asthma, and we investigate how testosterone attenuates ILC2 function. In patients with moderate to severe asthma, we determine that women have an increased number of circulating ILC2 compared to men. ILC2 from adult female mice have increased IL-2-mediated ILC2 proliferation versus ILC2 from adult male mice, as well as pre-pubescent females and males. Further, 5α-dihydrotestosterone, a hormone downstream of testosterone, decreases lung ILC2 numbers and IL-5 and IL-13 expression from ILC2. In vivo, testosterone attenuated Alternaria-extract-induced IL-5+ and IL-13+ ILC2 numbers and lung eosinophils by intrinsically decreasing lung ILC2 numbers, as well as by decreasing expression of IL-33 and thymic stromal lymphopoietin (TSLP), ILC2-stimulating cytokines. Collectively, these findings provide a foundational understanding of sexual dimorphism in ILC2 function.


Assuntos
Inflamação/tratamento farmacológico , Linfócitos/efeitos dos fármacos , Linfócitos/metabolismo , Testosterona/uso terapêutico , Adolescente , Adulto , Animais , Asma/tratamento farmacológico , Asma/imunologia , Proliferação de Células/efeitos dos fármacos , Feminino , Citometria de Fluxo , Humanos , Inflamação/imunologia , Interleucina-13/metabolismo , Interleucina-2/metabolismo , Interleucina-33/metabolismo , Linfócitos/imunologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Pessoa de Meia-Idade , Fosforilação/efeitos dos fármacos , Fator de Transcrição STAT5/metabolismo , Adulto Jovem
11.
Cell Host Microbe ; 22(4): 531-542.e8, 2017 Oct 11.
Artigo em Inglês | MEDLINE | ID: mdl-28943329

RESUMO

Diet, and specifically dietary metals, can modify the risk of infection. However, the mechanisms by which manganese (Mn), a common dietary supplement, alters infection remain unexplored. We report that dietary Mn levels dictate the outcome of systemic infections caused by Staphylococcus aureus, a leading cause of bacterial endocarditis. Mice fed a high Mn diet display alterations in Mn levels and localization within infected tissues, and S. aureus virulence and infection of the heart are enhanced. Although the canonical mammalian Mn-sequestering protein calprotectin surrounds staphylococcal heart abscesses, calprotectin is not released into the abscess nidus and does not limit Mn in this organ. Consequently, excess Mn is bioavailable to S. aureus in the heart. Bioavailable Mn is utilized by S. aureus to detoxify reactive oxygen species and protect against neutrophil killing, enhancing fitness within the heart. Therefore, a single dietary modification overwhelms vital host antimicrobial strategies, leading to fatal staphylococcal infection.


Assuntos
Endocardite Bacteriana/microbiologia , Coração/microbiologia , Manganês/metabolismo , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/metabolismo , Abscesso , Animais , Dieta , Modelos Animais de Doenças , Coração/fisiopatologia , Humanos , Complexo Antígeno L1 Leucocitário/metabolismo , Fígado/microbiologia , Fígado/fisiopatologia , Manganês/análise , Camundongos , Camundongos Congênicos , Camundongos Endogâmicos C57BL , Neutrófilos/metabolismo , Espécies Reativas de Oxigênio/metabolismo , Staphylococcus aureus/patogenicidade
12.
J Immunol ; 199(2): 510-519, 2017 07 15.
Artigo em Inglês | MEDLINE | ID: mdl-28576981

RESUMO

The appropriate orchestration of different arms of the immune response is critical during viral infection to promote efficient viral clearance while limiting immunopathology. However, the signals and mechanisms that guide this coordination are not fully understood. IFNs are produced at high levels during viral infection and have convergent signaling through STAT1. We hypothesized that STAT1 signaling during viral infection regulates the balance of innate lymphoid cells (ILC), a diverse class of lymphocytes that are poised to respond to environmental insults including viral infections with the potential for both antiviral or immunopathologic functions. During infection with respiratory syncytial virus (RSV), STAT1-deficient mice had reduced numbers of antiviral IFN-γ+ ILC1 and increased numbers of immunopathologic IL-5+ and IL-13+ ILC2 and IL-17A+ ILC3 compared with RSV-infected wild-type mice. Using bone marrow chimeric mice, we found that both ILC-intrinsic and ILC-extrinsic factors were responsible for this ILC dysregulation during viral infection in STAT1-deficient mice. Regarding ILC-extrinsic mechanisms, we found that STAT1-deficient mice had significantly increased expression of IL-33 and IL-23, cytokines that promote ILC2 and ILC3, respectively, compared with wild-type mice during RSV infection. Moreover, disruption of IL-33 or IL-23 signaling attenuated cytokine-producing ILC2 and ILC3 responses in STAT1-deficient mice during RSV infection. Collectively, these data demonstrate that STAT1 is a key orchestrator of cytokine-producing ILC responses during viral infection via ILC-extrinsic regulation of IL-33 and IL-23.


Assuntos
Imunidade Inata , Linfócitos/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Fator de Transcrição STAT1/metabolismo , Animais , Citocinas/biossíntese , Regulação da Expressão Gênica , Interferon gama/biossíntese , Interferon gama/genética , Interferon gama/imunologia , Interleucina-13/genética , Interleucina-13/imunologia , Interleucina-17/genética , Interleucina-17/imunologia , Interleucina-23/genética , Interleucina-23/imunologia , Interleucina-33/genética , Interleucina-33/imunologia , Interleucina-5/genética , Interleucina-5/imunologia , Linfócitos/classificação , Camundongos , Infecções por Vírus Respiratório Sincicial/virologia , Fator de Transcrição STAT1/deficiência , Fator de Transcrição STAT1/genética , Transdução de Sinais
13.
J Virol ; 91(6)2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28053109

RESUMO

Mouse adenovirus type 1 (MAV-1) infection causes encephalitis in susceptible strains of mice and alters the permeability of infected brains to small molecules, which indicates disruption of the blood-brain barrier (BBB). Under pathological conditions, matrix metalloproteinases (MMPs) can disrupt the BBB through their proteolytic activity on basement membrane and tight junction proteins. We examined whether MAV-1 infection alters MMP activity in vivo and in vitro Infected MAV-1-susceptible SJL mice had higher MMP2 and MMP9 activity in brains, measured by gelatin zymography, than mock-infected mice. Infected MAV-1-resistant BALB/c mice had MMP activity levels equivalent to those in mock infection. Primary SJL mouse brain endothelial cells (a target of MAV-1 in vivo) infected ex vivo with MAV-1 had no difference in activities of secreted MMP2 and MMP9 from mock cells. We show for the first time that astrocytes and microglia are also infected in vivo by MAV-1. Infected mixed primary cultures of astrocytes and microglia had higher levels of MMP2 and MMP9 activity than mock-infected cells. These results indicate that increased MMP activity in the brains of MAV-1-infected susceptible mice may be due to MMP activity produced by endothelial cells, astrocytes, and microglia, which in turn may contribute to BBB disruption and encephalitis in susceptible mice.IMPORTANCE RNA and DNA viruses can cause encephalitis; in some cases, this is accompanied by MMP-mediated disruption of the BBB. Activated MMPs degrade extracellular matrix and cleave tight-junction proteins and cytokines, modulating their functions. MAV-1 infection of susceptible mice is a tractable small-animal model for encephalitis, and the virus causes disruption of the BBB. We showed that MAV-1 infection increases enzymatic activity of two key MMPs known to be secreted and activated in neuroinflammation, MMP2 and MMP9, in brains of susceptible mice. MAV-1 infects endothelial cells, astrocytes, and microglia, cell types in the neurovascular unit that can secrete MMPs. Ex vivo MAV-1 infection of these cell types caused higher MMP activity than mock infection, suggesting that they may contribute to the higher MMP activity seen in vivo To our knowledge, this provides the first evidence of an encephalitic DNA virus in its natural host causing increased MMP activity in brains.


Assuntos
Infecções por Adenoviridae/patologia , Encefalite Viral/patologia , Mastadenovirus/patogenicidade , Metaloproteinase 2 da Matriz/análise , Metaloproteinase 9 da Matriz/análise , Infecções por Adenoviridae/virologia , Animais , Astrócitos/enzimologia , Astrócitos/virologia , Encéfalo/patologia , Células Cultivadas , Modelos Animais de Doenças , Encefalite Viral/virologia , Células Endoteliais/enzimologia , Células Endoteliais/virologia , Camundongos , Microglia/enzimologia , Microglia/virologia
14.
J Allergy Clin Immunol ; 138(3): 814-824.e11, 2016 09.
Artigo em Inglês | MEDLINE | ID: mdl-27156176

RESUMO

BACKGROUND: Respiratory syncytial virus (RSV) is a major health care burden with a particularly high worldwide morbidity and mortality rate among infants. Data suggest that severe RSV-associated illness is in part caused by immunopathology associated with a robust type 2 response. OBJECTIVE: We sought to determine the capacity of RSV infection to stimulate group 2 innate lymphoid cells (ILC2s) and the associated mechanism in a murine model. METHODS: Wild-type (WT) BALB/c, thymic stromal lymphopoietin receptor (TSLPR) knockout (KO), or WT mice receiving an anti-TSLP neutralizing antibody were infected with the RSV strain 01/2-20. During the first 4 to 6 days of infection, lungs were collected for evaluation of viral load, protein concentration, airway mucus, airway reactivity, or ILC2 numbers. Results were confirmed with 2 additional RSV clinical isolates, 12/11-19 and 12/12-6, with known human pathogenic potential. RESULTS: RSV induced a 3-fold increase in the number of IL-13-producing ILC2s at day 4 after infection, with a concurrent increase in total lung IL-13 levels. Both thymic stromal lymphopoietin (TSLP) and IL-33 levels were increased 12 hours after infection. TSLPR KO mice did not mount an IL-13-producing ILC2 response to RSV infection. Additionally, neutralization of TSLP significantly attenuated the RSV-induced IL-13-producing ILC2 response. TSLPR KO mice displayed reduced lung IL-13 protein levels, decreased airway mucus and reactivity, attenuated weight loss, and similar viral loads as WT mice. Both 12/11-19 and 12/12-6 similarly induced IL-13-producing ILC2s through a TSLP-dependent mechanism. CONCLUSION: These data demonstrate that multiple pathogenic strains of RSV induce IL-13-producing ILC2 proliferation and activation through a TSLP-dependent mechanism in a murine model and suggest the potential therapeutic targeting of TSLP during severe RSV infection.


Assuntos
Citocinas/imunologia , Interleucina-13/imunologia , Linfócitos/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Animais , Citocinas/genética , Feminino , Interleucina-33/imunologia , Pulmão/imunologia , Pulmão/metabolismo , Pulmão/virologia , Camundongos Endogâmicos BALB C , Camundongos Knockout , Muco/metabolismo , Infecções por Vírus Respiratório Sincicial/virologia , Carga Viral , Linfopoietina do Estroma do Timo
15.
Thorax ; 71(7): 633-45, 2016 07.
Artigo em Inglês | MEDLINE | ID: mdl-27071418

RESUMO

BACKGROUND: Group 2 innate lymphoid cells (ILC2) are an important source of the type 2 cytokines interleukin (IL)-5 and IL-13 that are critical to the allergic airway phenotype. Previous studies reported that histone deacetylase (HDAC) inhibition by trichostatin A (TSA) downregulated adaptive allergic immune responses; however, the effect of HDAC inhibition on the early innate allergic immune response is unknown. Therefore, we investigated the effect of TSA on innate airway inflammation mediated by ILC2 activation. METHODS: BALB/c mice were challenged intranasally with Alternaria extract, exogenous recombinant mouse IL-33 (rmIL-33) or the respective vehicles for four consecutive days following TSA or vehicle treatment. Bronchoalveolar lavage (BAL) fluids and lungs were harvested 24 h after the last challenge. RESULTS: We found that TSA treatment significantly decreased the number of ILC2 expressing IL-5 and IL-13 in the lungs challenged with Alternaria extract or rmIL-33 compared with vehicle treatment (p<0.05). TSA treatment significantly decreased protein expression of IL-5, IL-13, CCL11 and CCL24 in the lung homogenates from Alternaria extract-challenged mice or rmIL-33-challenged mice compared with vehicle treatment (p<0.05). Further, TSA treatment significantly decreased the number of perivascular eosinophils and mucus production in the large airways that are critical components of the asthma phenotype (p<0.05). TSA did not change early IL-33 release in the BAL fluids; however, TSA decreased lung IL-33 expression from epithelial cells 24 h after last Alternaria extract challenge compared with vehicle treatment (p<0.05). CONCLUSIONS: These results reveal that TSA reduces allergen-induced ILC2 activation and the early innate immune responses to an inhaled protease-containing aeroallergen.


Assuntos
Asma/imunologia , Inibidores de Histona Desacetilases/farmacologia , Ácidos Hidroxâmicos/farmacologia , Imunidade Inata , Linfócitos/imunologia , Alérgenos/imunologia , Alternaria , Animais , Lavagem Broncoalveolar , Quimiocina CCL11/metabolismo , Quimiocina CCL24/metabolismo , Interleucina-13/metabolismo , Interleucina-33/farmacologia , Interleucina-5/metabolismo , Pulmão/imunologia , Camundongos , Camundongos Endogâmicos BALB C
16.
Infect Immun ; 84(5): 1548-1555, 2016 May.
Artigo em Inglês | MEDLINE | ID: mdl-26953325

RESUMO

γδ T cells are prevalent at mucosal and epithelial surfaces and are a critical first line of defense against bacterial and fungal pathogens. γδ17 cells are a subset of γδ T cells which, in the presence of IL-23 and IL-1ß, produce large quantities of interleukin-17A (IL-17A), a cytokine crucial to these cells' antibacterial and antifungal function. STAT6, an important transcription factor in Th2 differentiation and inhibition of Th1 differentiation, is expressed at high levels in the T cells of people with parasitic infections and asthma. Our group and others have shown that STAT6 attenuates IL-17A protein expression by CD4(+) T cells. By extension, we hypothesized that STAT6 activation also inhibits innate γδ17 cell cytokine secretion. We show here that γδ17 cells expressed the type I IL-4 receptor (IL-4R), and IL-4 increased STAT6 phosphorylation in γδ T cells. IL-4 inhibited γδ17 cell production of IL-17A. IL-4 also decreased γδ17 cell expression of IL-23R as well as Sgk1. To determine whether STAT6 signaling regulates γδ17 cell numbers in vivo, we used a model of Klebsiella pneumoniae in mice deficient in STAT6. We chose K. pneumoniae for our in vivo model, since K. pneumoniae increases IL-17A expression and γδ17 numbers. K. pneumoniae infection of STAT6 knockout mice resulted in a statistically significant increase in the number of γδ17 cells compared to that of wild-type mice. These studies are the first to demonstrate that γδ17 cells express the type I IL-4R and that STAT6 signaling negatively regulates γδ17 cells, a cell population that plays a front-line role in mucosal immunity.


Assuntos
Interleucina-17/metabolismo , Infecções por Klebsiella/patologia , Klebsiella pneumoniae/imunologia , Fator de Transcrição STAT6/metabolismo , Transdução de Sinais , Subpopulações de Linfócitos T/imunologia , Células Th17/imunologia , Animais , Modelos Animais de Doenças , Feminino , Imunidade nas Mucosas , Interleucina-4/metabolismo , Camundongos Endogâmicos BALB C , Camundongos Knockout , Receptores de Superfície Celular/metabolismo
17.
Am J Respir Crit Care Med ; 193(1): 31-42, 2016 Jan 01.
Artigo em Inglês | MEDLINE | ID: mdl-26378386

RESUMO

RATIONALE: Group 2 innate lymphoid cells (ILC2s) robustly produce IL-5 and IL-13, cytokines central to the asthma phenotype; however, the effect of prostaglandin (PG) I2 on ILC2 function is unknown. OBJECTIVES: To determine the effect of PGI2 on mouse and human ILC2 cytokine expression in vitro and the effect of endogenous PGI2 and the PGI2 analog cicaprost on lung ILC2s in vivo. METHODS: Flow-sorted bone marrow ILC2s of wild-type (WT) and PGI2 receptor-deficient (IP(-/-)) mice were cultured with IL-33 and treated with the PGI2 analog cicaprost. WT and IP(-/-) mice were challenged intranasally with Alternaria alternata extract for 4 consecutive days to induce ILC2 responses, and these were quantified. Prior to A. alternata extract, challenged WT mice were treated with cicaprost. Human flow-sorted peripheral blood ILC2s were cultured with IL-33 and IL-2 and treated with the PGI2 analog cicaprost. MEASUREMENT AND MAIN RESULTS: We demonstrate that PGI2 inhibits IL-5 and IL-13 protein expression by IL-33-stimulated ILC2s purified from mouse bone marrow in a manner that was dependent on signaling through the PGI2 receptor IP. In a mouse model of 4 consecutive days of airway challenge with an extract of A. alternata, a fungal aeroallergen associated with severe asthma exacerbations, endogenous PGI2 signaling significantly inhibited lung IL-5 and IL-13 protein expression, and reduced the number of lung IL-5- and IL-13-expressing ILC2s, as well as the mean fluorescence intensity of IL-5 and IL-13 staining. In addition, exogenous administration of a PGI2 analog inhibited Alternaria extract-induced lung IL-5 and IL-13 protein expression, and reduced the number of lung IL-5- and IL-13-expressing ILC2s and the mean fluorescence intensity of IL-5 and IL-13 staining. Finally, a PGI2 analog inhibited IL-5 and IL-13 expression by human ILC2s that were stimulated with IL-2 and IL-33. CONCLUSIONS: These results suggest that PGI2 may be a potential therapy to reduce the ILC2 response to protease-containing aeroallergens, such as Alternaria.


Assuntos
Epoprostenol/fisiologia , Linfócitos/fisiologia , Transdução de Sinais/fisiologia , Alternaria/imunologia , Animais , Epoprostenol/análogos & derivados , Epoprostenol/farmacologia , Humanos , Técnicas In Vitro , Interleucina-13/fisiologia , Interleucina-33/farmacologia , Interleucina-5/fisiologia , Pulmão/citologia , Pulmão/imunologia , Linfócitos/efeitos dos fármacos , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Transdução de Sinais/efeitos dos fármacos
18.
Mamm Genome ; 23(3-4): 250-8, 2012 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-22101863

RESUMO

Strain-specific differences in susceptibility to mouse adenovirus type 1 (MAV-1) are linked to the quantitative trait locus Msq1 on mouse chromosome 15. This region contains 14 Ly6 or Ly6-related genes, many of which are known to be expressed on the surface of immune cells, suggesting a possible role in host defense. We analyzed these genes for polymorphisms between MAV-1-susceptible and MAV-1-resistant inbred mouse strains. Sequencing of cDNAs identified 12 coding-region polymorphisms in 2010109I03Rik, Ly6e, Ly6a, Ly6c1, and Ly6c2, six of which were nonsynonymous and five of which were previously unlisted in dbSNP Build 132. We also clarified sequence discrepancies in GenBank for the coding regions of I830127L07Rik and Ly6g. Additionally, Southern blotting revealed size polymorphisms within the DNA regions of Ly6e, Ly6a, and Ly6g. Collectively, these genetic variations have implications for the structure, function, and/or expression of Ly6 and Ly6-related genes that may contribute to the observed strain-specific differences in susceptibility to MAV-1.


Assuntos
Infecções por Adenoviridae/veterinária , Adenoviridae/fisiologia , Antígenos Ly/genética , Cromossomos de Mamíferos/genética , Predisposição Genética para Doença , Camundongos/genética , Polimorfismo Genético , Doenças dos Roedores/genética , Infecções por Adenoviridae/genética , Infecções por Adenoviridae/virologia , Sequência de Aminoácidos , Animais , Antígenos Ly/química , Feminino , Masculino , Camundongos/virologia , Camundongos Endogâmicos , Dados de Sequência Molecular , Doenças dos Roedores/virologia , Alinhamento de Sequência
19.
J Allergy Clin Immunol ; 127(1): 254-61, 261.e1-6, 2011 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21126757

RESUMO

BACKGROUND: Asthma is a major public health burden worldwide. Studies from our group and others have demonstrated that SERPINB3 and SERPINB4 are induced in patients with asthma; however, their mechanistic role in asthma has yet to be determined. OBJECTIVE: To evaluate the role of Serpin3a, the murine homolog of SERPINB3 and SERPINB4, in asthma. METHODS: We studied wild-type Balb/c and Serpinb3a-null mice in house dust mite or IL-13-induced asthma models and evaluated airway hyperresponsiveness, inflammation, and goblet cell hyperplasia. RESULTS: Airway hyperresponsiveness and goblet cell hyperplasia were markedly attenuated in the Serpinb3a-null mice compared with the wild-type mice after allergen challenge, with minimal effects on inflammation. Expression of sterile alpha motif pointed domain containing v-ets avian erythroblastosis virus E26 oncogene homolog transcription factor (SPDEF), a transcription factor that mediates goblet cell hyperplasia, was decreased in the absence of Serpinb3a. IL-13-treated Serpinb3a-null mice showed attenuated airway hyperresponsiveness, inflammation, and mucus production. CONCLUSION: Excessive mucus production and mucus plugging are key pathologic features of asthma, yet the mechanisms responsible for mucus production are not well understood. Our data reveal a novel nonredundant role for Serpinb3a in mediating mucus production through regulation of SPDEF expression. This pathway may be used to target mucus hypersecretion effectively.


Assuntos
Asma/imunologia , Muco/imunologia , Proteínas Proto-Oncogênicas c-ets/imunologia , Serpinas/imunologia , Animais , Asma/metabolismo , Asma/patologia , Líquido da Lavagem Broncoalveolar , Separação Celular , Ensaio de Imunoadsorção Enzimática , Citometria de Fluxo , Expressão Gênica , Regulação da Expressão Gênica/imunologia , Células Caliciformes/imunologia , Células Caliciformes/metabolismo , Imunoglobulina E/sangue , Imunoglobulina G/sangue , Imuno-Histoquímica , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Muco/metabolismo , Proteínas Proto-Oncogênicas c-ets/biossíntese , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Serpinas/metabolismo
20.
J Immunol ; 183(12): 7870-6, 2009 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-20007572

RESUMO

Although mice have nanogram per milliliter serum levels of soluble (s) IL-13Ralpha2, humans lack sIL-13Ralpha2 in serum. Our data provide a mechanism for this biological divergence. In mice, discrete transcripts encoding soluble and membrane forms of IL-13Ralpha2 are generated by alternative splicing. We used small interfering RNA to specifically deplete the transcript encoding membrane (mem) IL-13Ralpha2 (full-length) or sIL-13Ralpha2 (DeltaEx10) in murine cells. Depletion of the full-length transcript decreased memIL-13Ralpha2 but had no effect on the level of sIL-13Ralpha2 in cell supernatants at baseline or following cytokine stimulation. Depletion of the DeltaEx10 transcript decreased sIL-13Ralpha2 in supernatants at baseline and following stimulation. In contrast to mice, we were unable to find a transcript encoding sIL-13Ralpha2 in humans and siRNA-mediated depletion of full-length IL-13Ralpha2 decreased both sIL-13Ralpha2 and memIL-13Ralpha2 in human cells. Inhibition of matrix metalloproteinases (MMP)/MMP-8 abolished production of sIL-13Ralpha2 from human cells. Thus, sIL-13Ralpha2 is derived exclusively from the memIL-13Ralpha2 transcript in humans through MMPs/MMP-8 cleavage of memIL-13Ralpha2, supporting a limited role for sIL-13Ralpha2 in humans and highlighting the potential importance of memIL-13Ralpha2 in human immunity. These observations require consideration when results of murine IL-13 studies are applied to humans.


Assuntos
Subunidade alfa2 de Receptor de Interleucina-13 , Proteínas de Membrana , Processamento Alternativo/imunologia , Animais , Linhagem Celular , Humanos , Subunidade alfa2 de Receptor de Interleucina-13/sangue , Subunidade alfa2 de Receptor de Interleucina-13/deficiência , Subunidade alfa2 de Receptor de Interleucina-13/genética , Subunidade alfa2 de Receptor de Interleucina-13/isolamento & purificação , Proteínas de Membrana/sangue , Proteínas de Membrana/deficiência , Proteínas de Membrana/genética , Proteínas de Membrana/isolamento & purificação , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Knockout , Dados de Sequência Molecular , Isoformas de Proteínas/biossíntese , Isoformas de Proteínas/deficiência , Isoformas de Proteínas/genética , Isoformas de Proteínas/isolamento & purificação , Solubilidade , Células U937
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...