Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 13 de 13
Filtrar
1.
J Allergy Clin Immunol ; 153(4): 879-893, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-37634890

RESUMO

Type 2 inflammation is characterized by overexpression and heightened activity of type 2 cytokines, mediators, and cells that drive neuroimmune activation and sensitization to previously subthreshold stimuli. The consequences of altered neuroimmune activity differ by tissue type and disease; they include skin inflammation, sensitization to pruritogens, and itch amplification in atopic dermatitis and prurigo nodularis; airway inflammation and/or hyperresponsiveness, loss of expiratory volume, airflow obstruction and increased mucus production in asthma; loss of sense of smell in chronic rhinosinusitis with nasal polyps; and dysphagia in eosinophilic esophagitis. We describe the neuroimmune interactions that underlie the various sensory and autonomic pathologies in type 2 inflammatory diseases and present recent advances in targeted treatment approaches to reduce type 2 inflammation and its associated symptoms in these diseases. Further research is needed to better understand the neuroimmune mechanisms that underlie chronic, sustained inflammation and its related sensory pathologies in diseases associated with type 2 inflammation.


Assuntos
Asma , Dermatite Atópica , Sinusite , Humanos , Inflamação , Prurido/tratamento farmacológico , Sinusite/patologia
2.
J Neurosci ; 35(30): 10821-30, 2015 Jul 29.
Artigo em Inglês | MEDLINE | ID: mdl-26224864

RESUMO

Patients with systemic inflammatory diseases (e.g., rheumatoid arthritis, inflammatory bowel disease, chronic liver disease) commonly develop debilitating symptoms (i.e., sickness behaviors) that arise from changes in brain function. The microbiota-gut-brain axis alters brain function and probiotic ingestion can influence behavior. However, how probiotics do this remains unclear. We have previously described a novel periphery-to-brain communication pathway in the setting of peripheral organ inflammation whereby monocytes are recruited to the brain in response to systemic TNF-α signaling, leading to microglial activation and subsequently driving sickness behavior development. Therefore, we investigated whether probiotic ingestion (i.e., probiotic mixture VSL#3) alters this periphery-to-brain communication pathway, thereby reducing subsequent sickness behavior development. Using a well characterized mouse model of liver inflammation, we now show that probiotic (VSL#3) treatment attenuates sickness behavior development in mice with liver inflammation without affecting disease severity, gut microbiota composition, or gut permeability. Attenuation of sickness behavior development was associated with reductions in microglial activation and cerebral monocyte infiltration. These events were paralleled by changes in markers of systemic immune activation, including decreased circulating TNF-α levels. Our observations highlight a novel pathway through which probiotics mediate cerebral changes and alter behavior. These findings allow for the potential development of novel therapeutic interventions targeted at the gut microbiome to treat inflammation-associated sickness behaviors in patients with systemic inflammatory diseases. SIGNIFICANCE STATEMENT: This research shows that probiotics, when eaten, can improve the abnormal behaviors (including social withdrawal and immobility) that are commonly associated with inflammation. Probiotics are able to cause this effect within the body by changing how the immune system signals the brain to alter brain function. These findings broaden our understanding of how probiotics may beneficially affect brain function in the context of inflammation occurring within the body and may open potential new therapeutic alternatives for the treatment of these alterations in behavior that can greatly affect patient quality of life.


Assuntos
Encéfalo/efeitos dos fármacos , Comportamento de Doença/efeitos dos fármacos , Sistema Imunitário/efeitos dos fármacos , Neuroimunomodulação/efeitos dos fármacos , Probióticos/farmacologia , Animais , Comportamento Animal , Encéfalo/imunologia , Modelos Animais de Doenças , Inflamação/complicações , Masculino , Camundongos , Camundongos Endogâmicos C57BL
3.
Am J Physiol Gastrointest Liver Physiol ; 311(3): G466-79, 2016 09 01.
Artigo em Inglês | MEDLINE | ID: mdl-27492333

RESUMO

Barrier dysfunction is a characteristic of the inflammatory bowel diseases (IBD), Crohn's disease and ulcerative colitis. Understanding how the tight junction is modified to maintain barrier function may provide avenues for treatment of IBD. We have previously shown that the apical addition of serine proteases to intestinal epithelial cell lines causes a rapid and sustained increase in transepithelial electrical resistance (TER), but the mechanisms are unknown. We hypothesized that serine proteases increase barrier function through trafficking and insertion of tight junction proteins into the membrane, and this could enhance recovery of a disrupted monolayer after calcium switch or cytokine treatment. In the canine epithelial cell line, SCBN, we showed that matriptase, an endogenous serine protease, could potently increase TER. Using detergent solubility-based cell fractionation, we found that neither trypsin nor matriptase treatment changed levels of tight junction proteins at the membrane. In a fast calcium switch assay, serine proteases did not enhance the rate of recovery of the junction. In addition, serine proteases could not reverse barrier disruption induced by IFNγ and TNFα. We knocked down occludin in our cells using siRNA and found this prevented the serine protease-induced increase in TER. Using fluorescence recovery after photobleaching (FRAP), we found serine proteases induce a greater mobile fraction of occludin in the membrane. These data suggest that a functional tight junction is needed for serine proteases to have an effect on TER, and that occludin is a crucial tight junction protein in this mechanism.


Assuntos
Células Epiteliais/enzimologia , Mucosa Intestinal/citologia , Ocludina/metabolismo , Junções Íntimas/fisiologia , Animais , Linhagem Celular , Cães , Impedância Elétrica , Fenômenos Eletrofisiológicos , Células Epiteliais/citologia , Células Epiteliais/fisiologia , Ocludina/genética , Transporte Proteico , Serina Endopeptidases/farmacologia , Serina Proteases , Proteínas de Junções Íntimas/metabolismo , Tripsina/farmacologia
4.
J Immunol ; 182(8): 4854-64, 2009 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-19342664

RESUMO

Human rhinovirus (HRV) infections can trigger exacerbations of lower airway diseases. Infection of airway epithelial cells induces production of a number of proinflammatory chemokines that may exacerbate airway inflammation, including CXCL10, a chemoattractant for type 1 lymphocytes and NK cells. Primary human bronchial epithelial cells and the BEAS-2B human bronchial epithelial cell line were used to examine the role of MAPK pathways in HRV-16-induced production of CXCL10. Surprisingly, PD98059 and U0126, two inhibitors of the MEK1/2-ERK MAPK pathway, significantly enhanced HRV-16-induced CXCL10 mRNA and protein. This enhancement was not seen with IFN-beta-induced production of CXCL10. Studies using small interfering RNA revealed that knockdown of MEK1, but not MEK2, was associated with enhanced HRV-induced CXCL10 production. Promoter construct studies revealed that PD98059 and U0126 enhanced HRV-16-induced transcriptional activation of CXCL10. HRV-16-induced promoter activation was regulated by two NF-kappaB binding sites, kappaB1 and kappaB2, and by an IFN-stimulated response element. Inhibitors of the MEK1/2-ERK pathway did not alter HRV-16-induced activation of tandem repeat kappaB1 or kappaB2 constructs, nor did they alter HRV-16-induced nuclear translocation/binding of NF-kappaB to either kappaB1 or kappaB2 recognition sequences. Furthermore, PD98059 and U0126 did not alter phosphorylation or degradation of IkappaBalpha. In contrast, inhibitors of the MEK1/2-ERK pathway, and small interfering RNA knockdown of MEK1, enhanced nuclear translocation/binding of IFN regulatory factor (IRF)-1 to the IFN-stimulated response element recognition sequence in HRV-16 infected cells. We conclude that activation of MEK1 selectively down-regulates HRV-16-induced expression of CXCL10 via modulation of IRF-1 interactions with the gene promoter in human airway epithelial cells.


Assuntos
Brônquios/imunologia , Quimiocina CXCL10/biossíntese , Células Epiteliais/enzimologia , Células Epiteliais/imunologia , MAP Quinase Quinase 1/metabolismo , Rhinovirus/imunologia , Transcrição Gênica/genética , Brônquios/enzimologia , Linhagem Celular , Quimiocina CCL5/metabolismo , Quimiocina CXCL10/genética , Quimiocina CXCL10/imunologia , Regulação para Baixo/efeitos dos fármacos , Regulação para Baixo/genética , Humanos , Interferon beta/farmacologia , MAP Quinase Quinase 1/genética , MAP Quinase Quinase 2/genética , MAP Quinase Quinase 2/metabolismo , Sistema de Sinalização das MAP Quinases , NF-kappa B/metabolismo , Fosforilação , RNA Mensageiro/genética , RNA Interferente Pequeno/genética
5.
Am J Respir Cell Mol Biol ; 43(4): 413-21, 2010 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-19880820

RESUMO

Human rhinovirus (HRV) infections are associated with exacerbations of lower-airway diseases. HRV-induced production of proinflammatory chemokines, such as CXCL10, from infected airway epithelial cells may play a role in the pathogenesis of exacerbations. We have previously shown that the MAP/ERK kinase (MEK) pathway selectively down-regulates HRV-16-induced epithelial production of CXCL10 by modulating nuclear translocation and/or binding of IFN regulatory factor (IRF)-1 with the CXCL10 promoter. Using primary human bronchial epithelial cells (HBEs) and the BEAS-2B bronchial epithelial cell line, we have further evaluated the role of IRF-1 in HRV-16-induced epithelial CXCL10 production. We demonstrate that HRV-16 induced the expression of both IRF-1 mRNA and protein in a time-dependent manner. Interestingly, MEK1 pathway inhibition with PD98059 or U0126 significantly enhanced HRV-16-induced IRF-1 mRNA levels in BEAS-2B cells and HBEs, although IRF-1 protein expression was only enhanced in HBEs. Using short interfering RNA (siRNA), we both inhibited HRV-16-induced IRF-1 expression and reduced nuclear translocation and/or binding of IRF-1 to the CXCL10 promoter. Knockdown of IRF-1 also led to a significant reduction in HRV-16-induced CXCL10 production, confirming that IRF-1 is directly involved in HRV-16-induced CXCL10 expression in epithelial cells. Moreover, pronounced IRF-1 knockdown abrogated the enhancement of CXCL10 normally induced by inhibitors of the MEK1 pathway. Phosphatase experiments indicate that IRF-1 binding to the CXCL10 promoter is not dependent upon its phosphorylation state. We conclude that HRV-16-induced CXCL10 production is dependent upon IRF-1, and that the MEK1 pathway-dependent suppression of CXCL10 expression is also mediated via effects on IRF-1.


Assuntos
Quimiocina CXCL10/biossíntese , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Fator Regulador 1 de Interferon/metabolismo , Infecções por Picornaviridae/imunologia , Rhinovirus/fisiologia , Fosfatase Alcalina/metabolismo , Linhagem Celular , DNA/metabolismo , Células Epiteliais/enzimologia , Células Epiteliais/patologia , Regulação da Expressão Gênica/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Humanos , Fator Regulador 1 de Interferon/genética , Pulmão/efeitos dos fármacos , Pulmão/imunologia , Pulmão/patologia , Pulmão/virologia , MAP Quinase Quinase 1/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Fosforilação/efeitos dos fármacos , Infecções por Picornaviridae/genética , Ligação Proteica/efeitos dos fármacos , Inibidores de Proteínas Quinases/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , RNA Interferente Pequeno/metabolismo , Rhinovirus/efeitos dos fármacos
6.
J Allergy Clin Immunol ; 124(3): 551-7, 2009 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-19541350

RESUMO

BACKGROUND: Nitric oxide (NO) has previously been shown to inhibit human rhinovirus (HRV) replication in airway epithelial cells and to inhibit rhinovirus-induced epithelial cytokine and chemokine production independently of its effects on viral replication by modulating nuclear translocation and binding of transcription factors. OBJECTIVE: To define the molecular mechanisms by which NO inhibits HRV-16-induced epithelial production of CXCL10 by affecting nuclear translocation and binding of nuclear factor-kappaB (NF-kappaB) and IFN regulatory factor 1 (IRF-1). METHODS: Cultured human airway epithelial cells were infected with HRV-16 in the absence or presence of a NO donor, or were preincubated with 2 highly selective inhibitors of inhibitor of kappaB kinase (IKK)beta and then infected with HRV-16. Effects on the NF-kappaB and IRF-1 pathways were examined by using electrophoretic mobility shift assays, Western blotting, and real-time RT-PCR. RESULTS: Nitric oxide directly inhibited the binding of both recombinant NF-kappaB p50 protein and recombinant IRF-1 to their recognition sequences from the CXCL10 promoter. NO also inhibited phosphorylation of the NF-kappaB inhibitor, IkappaBalpha, in HRV-16-infected cells. In addition, both NO and inhibitors of IKKbeta inhibited viral induction of IRF-1 mRNA and protein. CONCLUSIONS: Nitric oxide blocks rhinovirus-mediated activation and nuclear translocation of both NF-kappaB and IRF-1. NO also directly inhibits the binding of each of these transcription factors to their respective recognition sites in the CXCL10 promoter. In addition, the ability of HRV-16 to induce epithelial expression of IRF-1 is dependent, at least in part, on viral activation of NF-kappaB.


Assuntos
Células Epiteliais/imunologia , Fator Regulador 1 de Interferon/antagonistas & inibidores , NF-kappa B/antagonistas & inibidores , Óxido Nítrico/metabolismo , Infecções por Picornaviridae/imunologia , Rhinovirus , Carbolinas/farmacologia , Células Cultivadas , Quimiocina CXCL10/antagonistas & inibidores , Quimiocina CXCL10/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/virologia , Compostos Heterocíclicos com 3 Anéis/farmacologia , Humanos , Quinase I-kappa B/antagonistas & inibidores , Niacinamida/análogos & derivados , Niacinamida/farmacologia , Doadores de Óxido Nítrico/farmacologia , Piridinas/farmacologia
7.
J Allergy Clin Immunol ; 123(1): 201-208.e9, 2009 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-18986693

RESUMO

BACKGROUND: Human rhinovirus (HRV) infections trigger exacerbations of asthma and chronic obstructive pulmonary disease. Nitric oxide (NO) inhibits HRV replication in human airway epithelial cells and suppresses HRV-induced epithelial production of several cytokines and chemokines. OBJECTIVE: We sought to delineate the mechanisms by which NO inhibits HRV-induced epithelial production of CXCL10, a chemoattractant for type 1 T cells and natural killer cells. METHODS: Primary human bronchial epithelial cells or cells of the BEAS-2B human bronchial epithelial cell line were exposed to HRV-16 in the presence or absence of the NO donor 3-(2-hydroxy-2-nitroso-1-propylhydrazino)-1-propanamine (PAPA NONOate). A cGMP analogue and an inhibitor of soluble guanylyl cyclase were used to examine the role of the cyclic guanosine monophosphate (cGMP) pathway in the actions of NO. BEAS-2B cells were transfected with CXCL10 promoter-luciferase constructs and the effects of PAPA NONOate were examined to study mechanisms of transcriptional regulation. Electrophoretic mobility shift assays were also used. RESULTS: PAPA NONOate inhibited HRV-16-induced increases in CXCL10 mRNA and protein. Inhibition of CXCL10 production occurred through a cGMP-independent pathway. PAPA NONOate inhibited HRV-16-induced CXCL10 transcription by blocking nuclear translocation, binding, or both of both nuclear factor kappaB and IFN response factors (IRFs) to their respective recognition elements in the CXCL10 promoter. CONCLUSIONS: NO inhibits HRV-16-induced production of CXCL10 by inhibiting viral activation of nuclear factor kappaB and of IRFs, including IRF-1, through a cGMP-independent pathway. The broad-ranging inhibition of HRV-induced epithelial cytokine and chemokine production by NO suggests a potential therapeutic utility of NO donors in viral exacerbations of asthma and chronic obstructive pulmonary disease.


Assuntos
Quimiocina CXCL10/imunologia , Células Epiteliais/imunologia , Infecções por Picornaviridae/imunologia , Mucosa Respiratória/imunologia , Rhinovirus/imunologia , Ativação Transcricional/imunologia , Transporte Ativo do Núcleo Celular/efeitos dos fármacos , Transporte Ativo do Núcleo Celular/imunologia , Asma/imunologia , Linhagem Celular , Núcleo Celular/imunologia , Núcleo Celular/metabolismo , Quimiocina CXCL10/biossíntese , GMP Cíclico/imunologia , GMP Cíclico/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Humanos , Hidrazinas/farmacologia , Hidrazinas/uso terapêutico , Fator Regulador 1 de Interferon/imunologia , Fator Regulador 1 de Interferon/metabolismo , NF-kappa B/imunologia , NF-kappa B/metabolismo , Óxido Nítrico/biossíntese , Óxido Nítrico/imunologia , Óxido Nítrico/farmacologia , Óxido Nítrico/uso terapêutico , Doadores de Óxido Nítrico/farmacologia , Doadores de Óxido Nítrico/uso terapêutico , Infecções por Picornaviridae/tratamento farmacológico , Infecções por Picornaviridae/metabolismo , Mucosa Respiratória/virologia , Elementos de Resposta/imunologia , Rhinovirus/metabolismo , Transcrição Gênica/efeitos dos fármacos , Transcrição Gênica/imunologia , Ativação Transcricional/efeitos dos fármacos , Ativação Viral/efeitos dos fármacos , Ativação Viral/imunologia
8.
J Allergy Clin Immunol ; 121(5): 1238-1245.e4, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18355907

RESUMO

BACKGROUND: Childhood human rhinovirus (HRV) infections are associated with an increased risk of asthma. We reasoned that HRV infections might be important in the pathogenesis of airway remodeling, thereby providing a mechanism by which these children are at risk of asthma. OBJECTIVE: We sought to determine whether HRV infection of airway epithelial cells regulates production of growth factors associated with airway remodeling and to determine whether vascular endothelial growth factor (VEGF) was upregulated in airways during HRV-induced natural colds. METHODS: Cultured human airway epithelial cells were infected with HRV. Amphiregulin, activin A, and VEGF protein levels were assayed by means of ELISA, and VEGF mRNA was quantified by using real-time RT-PCR. Pharmacologic inhibitors were used to assess the role of mitogen-activated protein kinase and nuclear factor kappaB pathways. Nasal lavage samples from subjects with confirmed natural HRV infections were assayed for VEGF protein and compared with baseline levels and with control levels. RESULTS: HRV infection upregulated amphiregulin, activin A, and VEGF protein levels compared with control media (P < .05). VEGF gene expression was maximally induced 3 hours after infection. HRV-induced generation of VEGF was regulated by p38 mitogen-activated protein kinase and extracellular signal-regulated kinase 1/2 pathways but did not depend on nuclear factor kappaB activation. In subjects with HRV infections, VEGF levels during peak cold symptoms were significantly higher than at baseline (P = .005) or in control subjects (P < .01). CONCLUSION: HRV-16 infection upregulates amphiregulin, activin A, and VEGF in airway epithelial cells, and HRV infections in vivo upregulate airway VEGF production.


Assuntos
Células Epiteliais/virologia , Peptídeos e Proteínas de Sinalização Intercelular/biossíntese , Infecções por Picornaviridae/metabolismo , Mucosa Respiratória/virologia , Adulto , Western Blotting , Linhagem Celular , Ensaio de Imunoadsorção Enzimática , Células Epiteliais/metabolismo , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Expressão Gênica , Humanos , Líquido da Lavagem Nasal/química , Infecções por Picornaviridae/fisiopatologia , RNA Mensageiro/análise , RNA Viral/análise , Mucosa Respiratória/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Rhinovirus , Fator A de Crescimento do Endotélio Vascular/biossíntese
9.
Physiol Rep ; 5(19)2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-29038361

RESUMO

Inflammatory diseases of the gut are associated with altered electrolyte and water transport, leading to the development of diarrhea. Epithelially expressed aquaporins (AQPs) are downregulated in inflammation, although the mechanisms involved are not known. We hypothesized that AQP3 expression in intestinal epithelial cells is altered in intestinal inflammation and that these changes are driven by tumor necrosis factor (TNF) α Human colonic adenocarcinoma (HT-29) cells were treated with TNFα to investigate signaling mechanisms in vitro. AQP3 expression was assessed by real-time PCR and radiolabeled glycerol uptake, with select inhibitors and a luciferase reporter construct used to further elucidate intracellular signaling. AQP3 expression was downregulated in HT-29 cells treated with TNFα Luciferase reporter construct experiments revealed that TNFα downregulated constitutive transcriptional activity of the AQP3 promoter, and inhibition of MEK/ERK and nuclear factor κB (NF-κB) signaling prevented the decrease in AQP3 mRNA expression. Constitutive AQP3 expression was suppressed by specificity protein (Sp) 3, and knockdown of this transcription factor bound to the AQP3 promoter was able to partially prevent the TNFα-induced downregulation of AQP3. TNFα signals through MEK/ERK and NF-κB to enhance the negative transcriptional control of AQP3 expression exerted by Sp3. Similar mechanisms regulate numerous ion channels, suggesting a common mechanism by which both ion and water transport are altered in inflammation.


Assuntos
Aquaporina 3/metabolismo , Enterócitos/metabolismo , Fator de Necrose Tumoral alfa/farmacologia , Aquaporina 3/genética , Enterócitos/efeitos dos fármacos , Células HT29 , Humanos , Sistema de Sinalização das MAP Quinases , NF-kappa B/metabolismo , Regiões Promotoras Genéticas , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Transcrição Sp3/metabolismo
10.
PLoS One ; 10(3): e0118713, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-25793528

RESUMO

Inflammatory bowel diseases are associated with dysregulated electrolyte and water transport and resultant diarrhea. Aquaporins are transmembrane proteins that function as water channels in intestinal epithelial cells. We investigated the effect of the inflammatory cytokine, interferon-γ, which is a major player in inflammatory bowel diseases, on aquaporin-1 expression in a mouse colonic epithelial cell line, CMT93. CMT93 monolayers were exposed to 10 ng/mL interferon-γ and aquaporin-1 mRNA and protein expressions were measured by real-time PCR and western blot, respectively. In other experiments, CMT93 cells were pretreated with inhibitors or were transfected with siRNA to block the effects of Janus kinases, STATs 1 and 3, or interferon regulatory factor 2, prior to treatment with interferon-γ. Interferon-γ decreased aquaporin-1 expression in mouse intestinal epithelial cells in a manner that did not depend on the classical STAT1/JAK2/IRF-1 pathway, but rather, on an alternate Janus kinase (likely JAK1) as well as on STAT3. The pro-inflammatory cytokine, interferon-γ may contribute to diarrhea associated with intestinal inflammation in part through regulation of the epithelial aquaporin-1 water channel via a non-classical JAK/STAT receptor signalling pathway.


Assuntos
Aquaporina 1/genética , Células Epiteliais/metabolismo , Interferon gama/farmacologia , Intestinos/citologia , Janus Quinases/metabolismo , Fator de Transcrição STAT3/metabolismo , Animais , Aquaporina 1/metabolismo , Linhagem Celular , Células Epiteliais/efeitos dos fármacos , Técnicas de Silenciamento de Genes , Fator Regulador 1 de Interferon/metabolismo , Fator Regulador 2 de Interferon/metabolismo , Camundongos , Modelos Biológicos , Inibidores de Proteínas Quinases/farmacologia , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Fator de Transcrição STAT1/metabolismo , Fatores de Tempo
11.
PLoS One ; 7(7): e40762, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-22808255

RESUMO

Human rhinovirus (HRV) infections trigger acute exacerbations of chronic obstructive pulmonary disease (COPD) and asthma. The human airway epithelial cell is the primary site of HRV infection and responds to infection with altered expression of multiple genes, the products of which could regulate the outcome to infection. Cigarette smoking aggravates asthma symptoms, and is also the predominant risk factor for the development and progression of COPD. We, therefore, examined whether cigarette smoke extract (CSE) modulates viral responses by altering HRV-induced epithelial gene expression. Primary cultures of human bronchial epithelial cells were exposed to medium alone, CSE alone, purified HRV-16 alone or to HRV-16+ CSE. After 24 h, supernatants were collected and total cellular RNA was isolated. Gene array analysis was performed to examine mRNA expression. Additional experiments, using real-time RT-PCR, ELISA and/or western blotting, validated altered expression of selected gene products. CSE and HRV-16 each induced groups of genes that were largely independent of each other. When compared to gene expression in response to CSE alone, cells treated with HRV+CSE showed no obvious differences in CSE-induced gene expression. By contrast, compared to gene induction in response to HRV-16 alone, cells exposed to HRV+CSE showed marked suppression of expression of a number of HRV-induced genes associated with various functions, including antiviral defenses, inflammation, viral signaling and airway remodeling. These changes were not associated with altered expression of type I or type III interferons. Thus, CSE alters epithelial responses to HRV infection in a manner that may negatively impact antiviral and host defense outcomes.


Assuntos
Brônquios/patologia , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Interações Hospedeiro-Patógeno/genética , Infecções por Picornaviridae/genética , Rhinovirus/fisiologia , Fumar/efeitos adversos , Adulto , Antivirais/metabolismo , Quimiocinas/genética , Quimiocinas/metabolismo , Regulação para Baixo/genética , Células Epiteliais/patologia , Feminino , Humanos , Interferons/genética , Interferons/metabolismo , Masculino , Pessoa de Meia-Idade , Análise de Sequência com Séries de Oligonucleotídeos , Infecções por Picornaviridae/imunologia , Infecções por Picornaviridae/virologia , Reprodutibilidade dos Testes , Transdução de Sinais/genética
12.
Am J Physiol Cell Physiol ; 296(5): C1140-50, 2009 May.
Artigo em Inglês | MEDLINE | ID: mdl-19279234

RESUMO

To investigate the potential role of the local expression of alternative complement factor B (hBf) in human sepsis, we examined the induction of Bf gene expression in human peripheral blood monocytes (PBMCs) from patients with septic shock and the mechanisms of hBf gene regulation by tumor necrosis factor (TNF)-alpha, interferon (IFN)-gamma, and lipopolysaccharide (LPS) in human monocytes. PBMCs from septic shock patients showed increased hBf mRNA expression when compared with control patients. Costimulation with TNF-alpha and IFN-gamma or stimulation with LPS demonstrated a time- and dose-dependent induction of hBf mRNA expression in human PBMCs. A region of the hBf promoter between -735 and +128 bp was found to mediate IFN-gamma, TNF-alpha, and LPS responsiveness as well as the synergistic effect of IFN-gamma/TNF-alpha on hBf promoter activity. Site-directed mutagenesis of a IFN-gamma-activation site (GAS) cis element (-90 to -82 bp) abrogated IFN-gamma responsiveness. Mutagenesis of a nuclear factor (NF)-kappaB cis element at -466 to -456 bp abrogated TNF-alpha and LPS responsiveness of the Bf promoter. Thus hBf gene expression is induced in PBMCs from septic shock patients, and the induction of hBf by IFN-gamma, TNF-alpha, and LPS is through GAS and NF-kappaB cis-binding sites on the hBf promoter. Furthermore, activated protein C (APC) inhibited LPS-stimulated hBf promoter activity and protein expression in human monocytes suggesting that the beneficial effect of APC therapy in sepsis may in part be due to inhibition of complement induction and/or activation via the alternative pathway.


Assuntos
Fator B do Complemento/genética , Macrófagos/imunologia , Monócitos/imunologia , Proteína C/metabolismo , Sepse/imunologia , Sepse/fisiopatologia , Sequência de Bases , Fator B do Complemento/imunologia , Fator B do Complemento/metabolismo , Via Alternativa do Complemento/imunologia , Humanos , Interferon gama/imunologia , Interferon gama/metabolismo , Lipopolissacarídeos/farmacologia , Macrófagos/metabolismo , Dados de Sequência Molecular , Monócitos/metabolismo , Regiões Promotoras Genéticas/genética , Regiões Promotoras Genéticas/imunologia , Proteína C/imunologia , RNA Mensageiro/metabolismo , Sepse/metabolismo , Ativação Transcricional/efeitos dos fármacos , Ativação Transcricional/imunologia , Fator de Necrose Tumoral alfa/imunologia , Fator de Necrose Tumoral alfa/metabolismo
13.
Am J Physiol Lung Cell Mol Physiol ; 289(1): L85-95, 2005 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-15764644

RESUMO

Human rhinovirus (HRV) infections trigger exacerbations of asthma and chronic obstructive pulmonary disease (COPD) and are associated with lymphocytic infiltration of the airways. We demonstrate that infection of primary cultures of human airway epithelial cells, or of the BEAS-2B human bronchial epithelial cell line, with human rhinovirus type 16 (HRV-16) induces expression of CXCL10 [IFN-gamma-inducible protein 10 (IP-10)], a ligand for the CXCR3 receptor found on activated type 1 T lymphocytes and natural killer cells. IP-10 mRNA reached maximal levels 24 h after HRV-16 infection then declined, whereas protein levels peaked 48 h after infection with no subsequent new synthesis. Cytosolic levels of AU-rich factor 1, a protein associated with mRNA destabilization, increased beginning 24 h after HRV-16 infection. Generation of IP-10 required virus capable of replication but was not dependent on prior induction of type 1 interferons. Transfection of synthetic double-stranded RNA into epithelial cells induced robust production of IP-10, whereas transfection of single-stranded RNA had no effect. Induction of IP-10 gene expression by HRV-16 depended upon activation of NF-kappaB, as well as other transcription factor recognition sequences further upstream in the IP-10 promoter. In vivo infection of human volunteers with HRV-16 strikingly increased IP-10 protein in nasal lavages during symptomatic colds. Levels of IP-10 correlated with symptom severity, viral titer, and numbers of lymphocytes in airway secretions. Thus IP-10 may play a role in the pathogenesis of HRV-induced colds and in HRV-induced exacerbations of COPD and asthma.


Assuntos
Quimiocinas CXC/biossíntese , Células Epiteliais/metabolismo , Regulação da Expressão Gênica , Infecções por Picornaviridae/metabolismo , Sistema Respiratório/metabolismo , Rhinovirus , Asma/metabolismo , Asma/virologia , Linhagem Celular , Quimiocina CXCL10 , Quimiocinas CXC/genética , Células Epiteliais/virologia , Ribonucleoproteína Nuclear Heterogênea D0 , Ribonucleoproteínas Nucleares Heterogêneas Grupo D/metabolismo , Humanos , Linfócitos/metabolismo , NF-kappa B/metabolismo , Infecções por Picornaviridae/virologia , Regiões Promotoras Genéticas , Doença Pulmonar Obstrutiva Crônica/metabolismo , Doença Pulmonar Obstrutiva Crônica/virologia , RNA de Cadeia Dupla/genética , RNA Mensageiro/biossíntese , RNA Mensageiro/genética , Sistema Respiratório/citologia , Transfecção
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA