Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
1.
Cancer Nurs ; 45(3): 224-235, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-33654011

RESUMO

BACKGROUND: The breast cancer diagnosis causes a high level of suffering and distress in patients who experience difficulties in coping. There is a need to improve knowledge of emotional and spiritual coping in response to the stressful situation of women who must face this diagnosis. OBJECTIVES: The aims of this study were to map women's spiritual and emotional coping experiences reported after a breast cancer diagnosis and examine the proposed interventions and suggestions for clinical practice. METHODS: A scoping review was performed by searching the Scientific Electronic Library Online, Scopus, Cumulative Index to Nursing and Allied Health Literature, Latin American & Caribbean Health Sciences Literature, Medical Literature Analyses and Retrieval System Online, Spanish Bibliographic Index of Health Sciences, PSYCINFO, and Google Scholar databases using Medical Subject Headings terms. Additional pertinent studies were identified by reviewing the bibliographies of the included studies. Twenty articles were included according to the recommendations for scoping reviews. RESULTS: Study findings regarding emotional and spiritual coping with the diagnosis and proposed interventions were synthesized. A thematic list of interventions and recommendations for clinical practice is also provided. CONCLUSIONS: The studies demonstrated that women with breast cancer are challenged by their emotions and experiences. The review highlights the importance of spiritual coping for redefining women's meaning in life. In clinical practice, caring for women's inherent needs when they are coping with a diagnosis is important to establish integral care. IMPLICATIONS FOR PRACTICE: Nurses can evaluate coping strategies, offer support for adaptation to the disease, provide qualified listening, help women in their search for significance while coping with cancer, and help them identify ways to overcome this stressful situation. Similarly, they can encourage patients to find spiritual comfort and emotional support.


Assuntos
Neoplasias da Mama , Adaptação Psicológica , Neoplasias da Mama/diagnóstico , Neoplasias da Mama/psicologia , Emoções , Feminino , Humanos , Espiritualidade
2.
J Investig Med ; 65(6): 984-990, 2017 08.
Artigo em Inglês | MEDLINE | ID: mdl-28363939

RESUMO

RATIONALE: The nasopharyngeal (NP) microbiota of newborns and infants plays a key role in modulating airway inflammation and respiratory symptoms during viral infections. Premature (PM) birth modifies the early NP environment and is a major risk factor for severe viral respiratory infections. However, it is currently unknown if the NP microbiota of PM infants is altered relative to full-term (FT) individuals. OBJECTIVES: To characterize the NP microbiota differences in preterm and FT infants during rhinovirus (RV) infection. METHODS: We determined the NP microbiota of infants 6 months to ≤2 years of age born FT (n=6) or severely PM<32 weeks gestation (n=7). We compared microbiota composition in healthy NP samples and performed a longitudinal analysis during naturally occurring RV infections to contrast the microbiota dynamics in PM versus FT infants. RESULTS: We observed significant differences in the NP bacterial community of PM versus FT. NP from PM infants had higher within-group dissimilarity (heterogeneity) relative to FT infants. Bacterial composition of NP samples from PM infants showed increased Proteobacteria and decreased in Firmicutes. There were also differences in the major taxonomic groups identified, including Streptococcus, Moraxella, and Haemophilus. Longitudinal data showed that these prematurity-related microbiota features persisted during RV infection. CONCLUSIONS: PM is associated with NP microbiota changes beyond the neonatal stage. PM infants have an NP microbiota with high heterogeneity relative to FT infants. These prematurity-related microbiota features persisted during RV infection, suggesting that the NP microbiota of PM may play an important role in modulating airway inflammatory and immune responses in this vulnerable group.


Assuntos
Recém-Nascido Prematuro/fisiologia , Microbiota , Nasofaringe/microbiologia , Infecções por Picornaviridae/microbiologia , Infecções por Picornaviridae/virologia , Rhinovirus/fisiologia , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Análise de Componente Principal , Estações do Ano
3.
Children (Basel) ; 3(4)2016 Oct 20.
Artigo em Inglês | MEDLINE | ID: mdl-27775602

RESUMO

Premature children are prone to severe viral respiratory infections in early life, but the age at which susceptibility peaks and disappears for each pathogen is unclear. Methods: A retrospective analysis was performed of the age distribution and clinical features of acute viral respiratory infections in full-term and premature children, aged zero to seven years. Results: The study comprised of a total of 630 hospitalizations (n = 580 children). Sixty-seven percent of these hospitalizations occurred in children born full-term (>37 weeks), 12% in preterm (32-37 weeks) and 21% in severely premature children (<32 weeks). The most common viruses identified were rhinovirus (RV; 60%) and respiratory syncytial virus (RSV; 17%). Age-distribution analysis of each virus identified that severely premature children had a higher relative frequency of RV and RSV in their first three years, relative to preterm or full-term children. Additionally, the probability of RV- or RSV-induced wheezing was higher overall in severely premature children less than three years old. Conclusions: Our results indicate that the vulnerability to viral infections in children born severely premature is more specific for RV and RSV and persists during the first three years of age. Further studies are needed to elucidate the age-dependent molecular mechanisms that underlie why premature infants develop RV- and RSV-induced wheezing in early life.

4.
J Microbiol Methods ; 130: 95-99, 2016 11.
Artigo em Inglês | MEDLINE | ID: mdl-27609714

RESUMO

BACKGROUND: Cystic fibrosis (CF) is an autosomal recessive disease characterized by recurrent lung infections. Studies of the lung microbiome have shown an association between decreasing diversity and progressive disease. 454 pyrosequencing has frequently been used to study the lung microbiome in CF, but will no longer be supported. We sought to identify the benefits and drawbacks of using two state-of-the-art next generation sequencing (NGS) platforms, MiSeq and PacBio RSII, to characterize the CF lung microbiome. Each has its advantages and limitations. METHODS: Twelve samples of extracted bacterial DNA were sequenced on both MiSeq and PacBio NGS platforms. DNA was amplified for the V4 region of the 16S rRNA gene and libraries were sequenced on the MiSeq sequencing platform, while the full 16S rRNA gene was sequenced on the PacBio RSII sequencing platform. Raw FASTQ files generated by the MiSeq and PacBio platforms were processed in mothur v1.35.1. RESULTS: There was extreme discordance in alpha-diversity of the CF lung microbiome when using the two platforms. Because of its depth of coverage, sequencing of the 16S rRNA V4 gene region using MiSeq allowed for the observation of many more operational taxonomic units (OTUs) and higher Chao1 and Shannon indices than the PacBio RSII. Interestingly, several patients in our cohort had Escherichia, an unusual pathogen in CF. Also, likely because of its coverage of the complete 16S rRNA gene, only PacBio RSII was able to identify Burkholderia, an important CF pathogen. CONCLUSION: When comparing microbiome diversity in clinical samples from CF patients using 16S sequences, MiSeq and PacBio NGS platforms may generate different results in microbial community composition and structure. It may be necessary to use different platforms when trying to correctly identify dominant pathogens versus measuring alpha-diversity estimates, and it would be important to use the same platform for comparisons to minimize errors in interpretation.


Assuntos
Bactérias/classificação , Bactérias/genética , Fibrose Cística/microbiologia , Sequenciamento de Nucleotídeos em Larga Escala/métodos , Pulmão/microbiologia , Microbiota/genética , Escarro/microbiologia , Bactérias/patogenicidade , Sequência de Bases , Biodiversidade , Classificação , Biologia Computacional/métodos , DNA Bacteriano/genética , Humanos , Metagenoma , Filogenia , RNA Ribossômico 16S/genética
5.
PLoS One ; 11(9): e0162244, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27643599

RESUMO

BACKGROUND: Innate immune responses are fine-tuned by small noncoding RNA molecules termed microRNAs (miRs) that modify gene expression in response to the environment. During acute infections, miRs can be secreted in extracellular vesicles (EV) to facilitate cell-to-cell genetic communication. The purpose of this study was to characterize the baseline population of miRs secreted in EVs in the airways of young children (airway secretory microRNAome) and examine the changes during rhinovirus (RV) infection, the most common cause of asthma exacerbations and the most important early risk factor for the development of asthma beyond childhood. METHODS: Nasal airway secretions were obtained from children (≤3 yrs. old) during PCR-confirmed RV infections (n = 10) and age-matched controls (n = 10). Nasal EVs were isolated with polymer-based precipitation and global miR profiles generated using NanoString microarrays. We validated our in vivo airway secretory miR data in an in vitro airway epithelium model using apical secretions from primary human bronchial epithelial cells (HBEC) differentiated at air-liquid interface (ALI). Bioinformatics tools were used to determine the unified (nasal and bronchial) signature airway secretory miRNAome and changes during RV infection in children. RESULTS: Multiscale analysis identified four signature miRs comprising the baseline airway secretory miRNAome: hsa-miR-630, hsa-miR-302d-3p, hsa- miR-320e, hsa-miR-612. We identified hsa-miR-155 as the main change in the baseline miRNAome during RV infection in young children. We investigated the potential biological relevance of the airway secretion of hsa-mir-155 using in silico models derived from gene datasets of experimental in vivo human RV infection. These analyses confirmed that hsa-miR-155 targetome is an overrepresented pathway in the upper airways of individuals infected with RV. CONCLUSIONS: Comparative analysis of the airway secretory microRNAome in children indicates that RV infection is associated with airway secretion of EVs containing miR-155, which is predicted in silico to regulate antiviral immunity. Further characterization of the airway secretory microRNAome during health and disease may lead to completely new strategies to treat and monitor respiratory conditions in all ages.


Assuntos
Regulação da Expressão Gênica , MicroRNAs/genética , Infecções por Picornaviridae/genética , Infecções Respiratórias/genética , Rhinovirus/fisiologia , Pré-Escolar , Genômica , Humanos , Lactente , Regulação para Cima
6.
Pulm Pharmacol Ther ; 38: 17-26, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-27133900

RESUMO

Overproduction of secretory mucins contributes to morbidity/mortality in inflammatory lung diseases. Inflammatory mediators directly increase expression of mucin genes, but few drugs have been shown to directly repress mucin gene expression. IL-1ß upregulates the MUC5AC mucin gene in part via the transcription factors NFκB while the glucocorticoid Dexamethasone (Dex) transcriptionally represses MUC5AC expression by Dex-activated GR binding to two GRE cis-sites in the MUC5AC promoter in lung epithelial cells. VBP compounds (ReveraGen BioPharma) maintain anti-inflammatory activity through inhibition of NFκB but exhibit reduced GRE-mediated transcriptional properties associated with adverse side-effects and thus have potential to minimize harmful side effects of long-term steroid therapy in inflammatory lung diseases. We investigated VBP15 efficacy as an anti-mucin agent in two types of airway epithelial cells and analyzed the transcription factor activity and promoter binding associated with VBP15-induced MUC5AC repression. VBP15 reduced MUC5AC mRNA abundance in a dose- and time-dependent manner similar to Dex in the presence or absence of IL-1ß in A549 and differentiated human bronchial epithelial cells. Repression was abrogated in the presence of RU486, demonstrating a requirement for GR in the VBP15-induced repression of MUC5AC. Inhibition of NFκB activity resulted in reduced baseline expression of MUC5AC indicating that constitutive activity maintains MUC5AC production. Chromatin immunoprecipitation analysis demonstrated lack of GR and of p65 (NFκB) binding to composite GRE domains in the MUC5AC promoter following VBP15 exposure of cells, in contrast to Dex. These data demonstrate that VBP15 is a novel anti-mucin agent that mediates the reduction of MUC5AC gene expression differently than the classical glucocorticoid, Dex.


Assuntos
Anti-Inflamatórios/farmacologia , Dexametasona/farmacologia , Mucina-5AC/genética , Pregnadienodiois/farmacologia , Células A549 , Anti-Inflamatórios/administração & dosagem , Brônquios/citologia , Brônquios/efeitos dos fármacos , Linhagem Celular , Dexametasona/administração & dosagem , Relação Dose-Resposta a Droga , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Glucocorticoides/administração & dosagem , Glucocorticoides/farmacologia , Humanos , Mediadores da Inflamação/metabolismo , Interleucina-1beta/metabolismo , Mucinas/antagonistas & inibidores , Mucinas/metabolismo , Pregnadienodiois/administração & dosagem , RNA Mensageiro/metabolismo , Fatores de Tempo
7.
PLoS One ; 11(4): e0152865, 2016.
Artigo em Inglês | MEDLINE | ID: mdl-27078692

RESUMO

BACKGROUND: Chronic Otitis Media (COM) is characterized by middle ear effusion (MEE) and conductive hearing loss. MEE reflect mucus hypersecretion, but global proteomic profiling of the mucosal components are limited. OBJECTIVE: This study aimed at characterizing the proteome of MEEs from children with COM with the goal of elucidating important innate immune responses. METHOD: MEEs were collected from children (n = 49) with COM undergoing myringotomy. Mass spectrometry was employed for proteomic profiling in nine samples. Independent samples were further analyzed by cytokine multiplex assay, immunoblotting, neutrophil elastase activity, next generation DNA sequencing, and/or immunofluorescence analysis. RESULTS: 109 unique and common proteins were identified by MS. A majority were innate immune molecules, along with typically intracellular proteins such as histones and actin. 19.5% percent of all mapped peptide counts were from proteins known to be released by neutrophils. Immunofluorescence and immunoblotting demonstrated the presence of neutrophil extracellular traps (NETs) in every MEE, along with MUC5B colocalization. DNA found in effusions revealed unfragmented DNA of human origin. CONCLUSION: Proteomic analysis of MEEs revealed a predominantly neutrophilic innate mucosal response in which MUC5B is associated with NET DNA. NETs are a primary macromolecular constituent of human COM middle ear effusions.


Assuntos
Armadilhas Extracelulares/imunologia , Imunidade Inata , Neutrófilos/citologia , Otite Média com Derrame/imunologia , Otite Média com Derrame/metabolismo , Proteômica , Pré-Escolar , Doença Crônica , Armadilhas Extracelulares/metabolismo , Feminino , Humanos , Lactente , Recém-Nascido , Masculino
8.
Pediatr Neonatol ; 57(1): 27-34, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26117550

RESUMO

BACKGROUND: Human metapneumovirus (HMPV) is a recently discovered respiratory pathogen of the family Paramyxoviridae, the same family as that of respiratory syncytial virus (RSV). Premature children are at high risk of severe RSV infections, however, it is unclear whether HMPV infection is more severe in hospitalized children with a history of severe prematurity. METHODS: We conducted a retrospective analysis of the clinical respiratory presentation of all polymerase chain reaction-confirmed HMPV infections in preschool-age children (≤5 years) with and without history of severe prematurity (<32 weeks gestation). Respiratory distress scores were developed to examine the clinical severity of HMPV infections. Demographic and clinical variables were obtained from reviewing electronic medical records. RESULTS: A total of 571 preschool children were identified using polymerase chain reaction-confirmed viral respiratory tract infection during the study period. HMPV was identified as a causative organism in 63 cases (11%). Fifty-eight (n = 58) preschool-age children with HMPV infection were included in this study after excluding those with significant comorbidities. Our data demonstrated that 32.7% of children admitted with HMPV had a history of severe prematurity. Preschool children with a history of prematurity had more severe HMPV disease as illustrated by longer hospitalizations, new or increased need for supplemental O2, and higher severity scores independently of age, ethnicity, and history of asthma. CONCLUSION: Our study suggests that HMPV infection causes significant disease burden among preschool children with a history of prematurity leading to severe respiratory infections and increasing health care resource utilization due to prolonged hospitalizations.


Assuntos
Metapneumovirus , Infecções por Paramyxoviridae/complicações , Nascimento Prematuro , Infecções Respiratórias/etiologia , Criança Hospitalizada , Pré-Escolar , Estudos Transversais , Feminino , Humanos , Lactente , Recém-Nascido , Masculino , Nascimento Prematuro/epidemiologia , Infecções por Vírus Respiratório Sincicial/complicações , Estudos Retrospectivos
9.
JAMA Otolaryngol Head Neck Surg ; 141(11): 997-1005, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-26512766

RESUMO

IMPORTANCE: Chronic otitis media with effusion is characterized by middle ear secretion of mucin glycoproteins, predominantly MUC5B; MUC5AC, the other secretory mucin studied frequently, has also been identified in the middle ear. Emerging evidence suggests a dichotomous role for these mucins in innate immune responses. We hypothesized that MUC5AC is an acute responder and MUC5B is expressed at later time points, reflecting a chronic situation. OBJECTIVE: To determine middle ear regulation of MUC5B and MUC5AC following in vitro bacterial and cytokine exposure. DESIGN, SETTING, AND SAMPLES: An in vitro cell-based model of mucin gene regulation was conducted in a basic science laboratory at a tertiary pediatric hospital. The study was conducted from July 1, 2014, to June 30, 2015; data analysis was performed in July 2015. INTERVENTIONS: Nontypeable Haemophilus influenzae (NTHi) lysates were generated and used to stimulate mouse middle ear epithelial cells (mMEECs) for 2 hours during 3 weeks. MAIN OUTCOMES AND MEASURES: Real-time quantitative polymerase chain reaction, luciferase assays, Western blot assay, and immunofluorescence techniques were performed to determine Muc5ac and Muc5b expression over time, Cxcl2 chemokine response, and nuclear factor-κB activation. Luciferase reporter assays were performed to evaluate specific promoter responses after NTHi exposure. RESULTS: Nontypeable H influenzae lysates (200 µg/mL) drove differential mucin gene activation, with Muc5ac being induced up to 2.04 fold at 24 hours and 2.79 fold at 96 hours (P < .05) and Muc5b being induced only at more long-term points: 1.61 fold at 96 hours, 1.41 fold at 1 week, and 1.53 fold at 3 weeks (P < .05). Although NTHi lysates induced robust, early nuclear factor-κB nuclear translocation with nuclear factor-κB-dependent induction of Cxlc2 expression, the lysates had minimal to no effect on Muc5ac and Muc5b promoter activity. However, in contrast to NTHi lysates, CXCL2 induced significant transcription of both Muc5b and Muc5ac as early as 24 hours. CONCLUSIONS AND RELEVANCE: Nontypeable H influenzae lysates activate differential mucin gene activation in mMEECs. Although Muc5ac is an early response mucin gene, Muc5b appears to react as a chronic response mucin.


Assuntos
Orelha Média/microbiologia , Células Epiteliais/metabolismo , Haemophilus influenzae/fisiologia , Mucina-5AC/genética , Mucina-5B/genética , Animais , Western Blotting , Células Cultivadas , Quimiocina CXCL2/biossíntese , Orelha Média/citologia , Células Epiteliais/patologia , Regulação da Expressão Gênica , Infecções por Haemophilus/metabolismo , Camundongos , Mucina-5AC/metabolismo , Mucina-5B/metabolismo , NF-kappa B/metabolismo , Reação em Cadeia da Polimerase em Tempo Real , Fatores de Tempo , Ativação Transcricional
10.
Pediatr Res ; 78(4): 389-94, 2015 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-26086642

RESUMO

BACKGROUND: It is unknown why human metapneumovirus (HMPV) and respiratory syncytial virus (RSV) cause severe respiratory infection in children, particularly in premature infants. Our aim was to investigate if there are defective airway antiviral responses to these viruses in young children with history of prematurity. METHODS: Nasal airway secretions were collected from 140 children ≤ 3 y old without detectable virus (n = 80) or with PCR-confirmed HMPV or RSV infection (n = 60). Nasal protein levels of IFNγ, CCL5/RANTES, IL-10, IL-4, and IL-17 were determined using a multiplex magnetic bead immunoassay. RESULTS: Full-term children with HMPV and RSV infection had increased levels of nasal airway IFNγ, CCL5, and IL-10 along with an elevation in Th1 (IFNγ)/Th2 (IL-4) ratios, which is expected during antiviral responses. In contrast, HMPV-infected premature children (< 32 wk gestation) did not exhibit increased Th1/Th2 ratios or elevated nasal airway secretion of IFNγ, CCL5, and IL-10 relative to uninfected controls. CONCLUSION: Our study is the first to demonstrate that premature infants have defective IFNγ, CCL5/RANTES, and IL-10 airway responses during HMPV infection and provides novel insights about the potential reason why HMPV causes severe respiratory disease in children with history of prematurity.


Assuntos
Recém-Nascido Prematuro , Interferon gama/imunologia , Pulmão/imunologia , Metapneumovirus/imunologia , Infecções por Paramyxoviridae/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Vírus Sinciciais Respiratórios/imunologia , Quimiocina CCL5/imunologia , Quimiocina CCL5/metabolismo , Pré-Escolar , Estudos Transversais , DNA Viral/genética , Feminino , Idade Gestacional , Interações Hospedeiro-Patógeno , Humanos , Lactente , Interferon gama/metabolismo , Interleucina-10/imunologia , Interleucina-10/metabolismo , Interleucina-4/imunologia , Interleucina-4/metabolismo , Pulmão/metabolismo , Pulmão/virologia , Masculino , Metapneumovirus/genética , Metapneumovirus/isolamento & purificação , Infecções por Paramyxoviridae/diagnóstico , Infecções por Paramyxoviridae/metabolismo , Infecções por Paramyxoviridae/virologia , Estudos Prospectivos , Infecções por Vírus Respiratório Sincicial/diagnóstico , Infecções por Vírus Respiratório Sincicial/metabolismo , Infecções por Vírus Respiratório Sincicial/virologia , Vírus Sinciciais Respiratórios/genética , Vírus Sinciciais Respiratórios/isolamento & purificação , Células Th1/imunologia , Células Th1/metabolismo , Células Th1/virologia , Células Th17/imunologia , Células Th17/metabolismo , Células Th17/virologia , Células Th2/imunologia , Células Th2/metabolismo , Células Th2/virologia , Regulação para Cima
11.
Pediatr Allergy Immunol ; 26(2): 145-52, 2015 Mar.
Artigo em Inglês | MEDLINE | ID: mdl-25640734

RESUMO

BACKGROUND: Rhinovirus (RV) has been linked to the pathogenesis of asthma. Prematurity is a risk factor for severe RV infection in early life, but is unknown if RV elicits enhanced pro-asthmatic airway cytokine responses in premature infants. This study investigated whether young children born severely premature (<32 wks gestation) exhibit airway secretion of Th2 and Th17 cytokines during natural RV infections and whether RV-induced Th2-Th17 responses are linked to more respiratory morbidity in premature children during the first 2 yrs of life. METHODS: We measured Th2 and Th17 nasal airway cytokines in a retrospective cohort of young children aged 0-2 yrs with PCR-confirmed RV infection or non-detectable virus. Protein levels of IL-4, IL-13, TSLP, and IL-17 were determined with multiplex immunoassays. Demographic and clinical variables were obtained by electronic medical record (EMR) review. RESULTS: The study comprised 214 children born full term (n = 108), preterm (n = 44) or severely premature (n = 62). Natural RV infection in severely premature children was associated with elevated airway secretion of Th2 (IL-4 and IL-13) and Th17 (IL-17) cytokines, particularly in subjects with history of bronchopulmonary dysplasia. Severely premature children with high RV-induced airway IL-4 had recurrent respiratory hospitalizations (median 3.65 hosp/yr; IQR 2.8-4.8) and were more likely to have at least one pediatric intensive care unit admission during the first 2 yrs of life (OR 8.72; 95% CI 1.3-58.7; p = 0.02). CONCLUSIONS: Severely premature children have increased airway secretion of Th2 and Th17 cytokines during RV infections, which is associated with more respiratory morbidity in the first 2 yrs of life.


Assuntos
Resfriado Comum/imunologia , Citocinas/imunologia , Lactente Extremamente Prematuro/imunologia , Sistema Respiratório/imunologia , Sistema Respiratório/virologia , Asma/imunologia , Asma/virologia , Displasia Broncopulmonar/imunologia , Displasia Broncopulmonar/virologia , Estudos de Coortes , Resfriado Comum/complicações , Citocinas/biossíntese , Feminino , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Masculino , Reação em Cadeia da Polimerase Multiplex , Estudos Retrospectivos , Rhinovirus
12.
Am J Respir Cell Mol Biol ; 53(1): 22-32, 2015 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-25692303

RESUMO

Alterations in epithelial secretions and mucociliary clearance contribute to chronic bacterial infection in cystic fibrosis (CF) lung disease, but whether CF lungs are unchanged in the absence of infection remains controversial. A proteomic comparison of airway secretions from subjects with CF and control subjects shows alterations in key biological processes, including immune response and proteolytic activity, but it is unclear if these are due to mutant CF transmembrane conductance regulator (CFTR) and/or chronic infection. We hypothesized that the CF lung apical secretome is altered under constitutive conditions in the absence of inflammatory cells and pathogens. To test this, we performed quantitative proteomics of in vitro apical secretions from air-liquid interface cultures of three life-extended CF (ΔF508/ΔF508) and three non-CF human bronchial epithelial cells after labeling of CF cells by stable isotope labeling with amino acids in cell culture. Mass spectrometry analysis identified and quantitated 666 proteins across samples, of which 70 exhibited differential enrichment or depletion in CF secretions (±1.5-fold change; P < 0.05). The key molecular functions were innate immunity (24%), cytoskeleton/extracellular matrix organization (24%), and protease/antiprotease activity (17%). Oxidative proteins and classical complement pathway proteins that are altered in CF secretions in vivo were not altered in vitro. Specific differentially increased proteins-MUC5AC and MUC5B mucins, fibronectin, and matrix metalloproteinase-9-were validated by antibody-based assays. Overall, the in vitro CF secretome data are indicative of a constitutive airway epithelium with altered innate immunity, suggesting that downstream consequences of mutant CFTR set the stage for chronic inflammation and infection in CF airways.


Assuntos
Brônquios/metabolismo , Fibrose Cística/metabolismo , Proteoma/metabolismo , Proteômica , Mucosa Respiratória/metabolismo , Brônquios/patologia , Linhagem Celular , Doença Crônica , Fibrose Cística/genética , Fibrose Cística/patologia , Humanos , Inflamação/genética , Inflamação/metabolismo , Inflamação/patologia , Proteoma/genética , Mucosa Respiratória/patologia
13.
Am J Respir Cell Mol Biol ; 52(5): 535-42, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25412193

RESUMO

Hyperplasia/hypertrophy of submucosal glands contributes to mucus overproduction in chronic diseases of the upper and lower respiratory tracts, especially in adult and pediatric chronic rhinosinusitis. Mechanisms that lead to glandular hyperplasia/hypertrophy are markedly understudied, reflecting a lack of in vitro model systems wherein airway epithelial progenitor cells differentiate into glandular cells. In this study, we developed and compared several in vitro three-dimensional systems using human nasal epithelial basal cells (HNEBCs) cultured by different methods on two types of extracellular matrices. We demonstrate that HNEBCs cultured on Matrigel (Corning, Tewksbury, MA) form glandular acini-like structures, whereas HNEBCs embedded in a collagen type I matrix form a network of tubules. Fibroblast-conditioned medium increases tubule formation in collagen type I. In contrast, HNEBCs cocultured with fibroblasts self-aggregate into organotypic structures with tubules and acini. These observations provide morphological evidence that HNEBCs are pluripotent and retain the capacity to differentiate into structures resembling specific structural components of submucosal glands depending on the extracellular matrices and culture conditions. The resultant models should prove useful in targeting cross-talk between epithelial cells and fibroblasts to decipher molecular mechanisms and specific signals responsible for the development of glandular hyperplasia/hypertrophy, which in turn may lead to new therapeutic strategies for chronic rhinosinusitis and other inflammatory respiratory diseases characterized by glandular hyperplasia/hypertrophy.


Assuntos
Células Epiteliais/fisiologia , Glândulas Exócrinas/fisiologia , Mucosa Nasal/fisiologia , Células-Tronco Pluripotentes/fisiologia , Engenharia Tecidual/métodos , Diferenciação Celular , Células Cultivadas , Técnicas de Cocultura , Colágeno/metabolismo , Colágeno Tipo I/metabolismo , Meios de Cultivo Condicionados/metabolismo , Combinação de Medicamentos , Células Epiteliais/metabolismo , Glândulas Exócrinas/citologia , Glândulas Exócrinas/metabolismo , Matriz Extracelular/metabolismo , Fibroblastos/metabolismo , Géis , Humanos , Laminina/metabolismo , Mucosa Nasal/citologia , Mucosa Nasal/metabolismo , Organogênese , Comunicação Parácrina , Células-Tronco Pluripotentes/metabolismo , Proteoglicanas/metabolismo , Nicho de Células-Tronco
14.
Pediatr Res ; 77(2): 356-62, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25420179

RESUMO

BACKGROUND: Chronic rhinosinusitis (CRS) is characterized by mucous overproduction and submucosal gland hyperplasia. The global protein profile of sinonasal secretions in pediatric CRS has not been studied. We hypothesized that MUC5B, a glandular mucin, would be relatively increased in CRS secretions compared to other mucins. METHODS: Secretions were collected at Children's National Health System (Children's National) from CRS patients undergoing sinus surgery and from control patients without CRS undergoing craniofacial procedures. Proteins were extracted, digested to peptides, and analyzed by mass spectometry. Fold change significance was calculated using the QSpec algorithm. Western blot analysis was performed to validate proteomic findings. RESULTS: In total, 294 proteins were identified. Although both MUC5B and MUC5AC were identified in a majority of samples, the relative abundance of MUC5B was found to be significantly higher (P < 0.05). Western blot data validated these findings. Other proteins with the highest significant positive-fold change in CRS samples were BP1 fold-containing family A member 1, chitinase-3-like protein 1, plastin-2, serpin 10, and BP1 fold-containing family B member 1. CONCLUSION: Overall, our data demonstrate an increase of MUC5B abundance in the sinus secretions of pediatric patients with CRS.


Assuntos
Mucina-5B/metabolismo , Mucosa/metabolismo , Seios Paranasais/metabolismo , Rinite/metabolismo , Sinusite/metabolismo , Adolescente , Western Blotting , Criança , Pré-Escolar , Eletroforese em Gel de Poliacrilamida , Ontologia Genética , Humanos , Proteômica
15.
PLoS One ; 9(12): e115398, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-25546419

RESUMO

BACKGROUND: Thymic stromal lymphoproetin (TSLP) is a cytokine secreted by the airway epithelium in response to respiratory viruses and it is known to promote allergic Th2 responses in asthma. This study investigated whether virally-induced secretion of TSLP is directional in nature (apical vs. basolateral) and/or if there are TSLP-mediated effects occurring at both sides of the bronchial epithelial barrier in the asthmatic state. METHODS: Primary human bronchial epithelial cells (HBEC) from control (n = 3) and asthmatic (n = 3) donors were differentiated into polarized respiratory tract epithelium under air-liquid interface (ALI) conditions and treated apically with dsRNA (viral surrogate) or TSLP. Sub-epithelial effects of TSLP were examined in human airway smooth muscle cells (HASMC) from normal (n = 3) and asthmatic (n = 3) donors. Clinical experiments examined nasal airway secretions obtained from asthmatic children during naturally occurring rhinovirus-induced exacerbations (n = 20) vs. non-asthmatic uninfected controls (n = 20). Protein levels of TSLP, CCL11/eotaxin-1, CCL17/TARC, CCL22/MDC, TNF-α and CXCL8 were determined with a multiplex magnetic bead assay. RESULTS: Our data demonstrate that: 1) Asthmatic HBEC exhibit an exaggerated apical, but not basal, secretion of TSLP after dsRNA exposure; 2) TSLP exposure induces unidirectional (apical) secretion of CCL11/eotaxin-1 in asthmatic HBEC and enhanced CCL11/eotaxin-1 secretion in asthmatic HASMC; 3) Rhinovirus-induced asthma exacerbations in children are associated with in vivo airway secretion of TSLP and CCL11/eotaxin-1. CONCLUSIONS: There are virally-induced TSLP-driven secretory immune responses at both sides of the bronchial epithelial barrier characterized by enhanced CCL11/eotaxin-1 secretion in asthmatic airways. These results suggest a new model of TSLP-mediated eosinophilic responses in the asthmatic airway during viral-induced exacerbations.


Assuntos
Asma/metabolismo , Quimiocina CCL11/metabolismo , Citocinas/metabolismo , Mucosa Nasal/metabolismo , RNA de Cadeia Dupla/farmacologia , Adolescente , Asma/virologia , Estudos de Casos e Controles , Linhagem Celular , Quimiocina CCL11/genética , Quimiocina CCL17/genética , Quimiocina CCL17/metabolismo , Quimiocina CCL22/genética , Quimiocina CCL22/metabolismo , Criança , Pré-Escolar , Citocinas/genética , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Mucosa Nasal/efeitos dos fármacos , Mucosa Nasal/virologia , Rhinovirus , Fator de Necrose Tumoral alfa/genética , Fator de Necrose Tumoral alfa/metabolismo , Linfopoietina do Estroma do Timo
17.
Am J Respir Cell Mol Biol ; 51(3): 363-9, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-24669775

RESUMO

We recently proposed that mitotic asynchrony in repairing tissue may underlie chronic inflammation and fibrosis, where immune cell infiltration is secondary to proinflammatory cross-talk among asynchronously repairing adjacent tissues. Building on our previous finding that mitotic asynchrony is associated with proinflammatory/fibrotic cytokine secretion (e.g., transforming growth factor [TGF]-ß1), here we provide evidence supporting cause-and-effect. Under normal conditions, primary airway epithelial basal cell populations undergo mitosis synchronously and do not secrete proinflammatory or profibrotic cytokines. However, when pairs of nonasthmatic cultures were mitotically synchronized at 12 hours off-set and then combined, the mixed cell populations secreted elevated levels of TGF-ß1. This shows that mitotic asynchrony is not only associated with but is also causative of TGF-ß1 secretion. The secreted cytokines and other mediators from asthmatic cells were not the cause of asynchronous regeneration; synchronously mitotic nonasthmatic epithelia exposed to conditioned media from asthmatic cells did not show changes in mitotic synchrony. We also tested if resynchronization of regenerating asthmatic airway epithelia reduces TGF-ß1 secretion and found that pulse-dosed dexamethasone, simvastatin, and aphidicolin were all effective. We therefore propose a new model for chronic inflammatory and fibrotic conditions where an underlying factor is mitotic asynchrony.


Assuntos
Asma/metabolismo , Células Epiteliais/metabolismo , Mitose , Fator de Crescimento Transformador beta1/metabolismo , Afidicolina/administração & dosagem , Brônquios/metabolismo , Brônquios/patologia , Células Cultivadas , Meios de Cultivo Condicionados/química , Dexametasona/administração & dosagem , Epitélio/metabolismo , Fibrose , Humanos , Inflamação , Mucosa Respiratória/metabolismo , Sinvastatina/administração & dosagem , Fatores de Tempo
18.
Am J Physiol Lung Cell Mol Physiol ; 306(8): L797-807, 2014 Apr 15.
Artigo em Inglês | MEDLINE | ID: mdl-24487386

RESUMO

Chronic airway diseases are characterized by inflammation and mucus overproduction. The MUC5AC mucin gene is upregulated by the proinflammatory cytokine interleukin-1 ß (IL-1ß) via activation of cAMP response element-binding protein (CREB) in the NCI-H292 cancer cell line and nuclear factor-κB (NF-κB) in the HBE1 transformed cell line, with each transcription factor binding to a cognate cis site in the proximal or distal region, respectively, of the MUC5AC promoter. We utilized primary differentiated human bronchial epithelial (HBE) and A549 lung adenocarcinoma cells to further investigate the contributions of CREB and NF-κB subunits to the IL-1ß-induced upregulation of MUC5AC. Data show that ligand binding of IL-1ß to the IL-1ß receptor is required to increase MUC5AC mRNA abundance. Chromatin immunoprecipitation analyses show direct binding of CREB to the previously identified cAMP response element site and binding of p65 and p50 subunits to a novel NF-κB site in a mucin-regulatory domain in the proximal promoter and to a previously identified NF-κB site in the distal promoter. P50 binds to both NF-κB sites at 1 h following IL-1ß exposure, but is replaced at 2 h by p65 in A549 cells and by a p50/p65 heterodimer in HBE cells. Thus IL-1ß activates multiple domains in the MUC5AC promoter but exhibits some cell-specific responses, highlighting the complexity of MUC5AC transcriptional regulation. Data show that dexamethasone, a glucocorticoid that transcriptionally represses MUC5AC gene expression under constitutive conditions, also represses IL-1ß-mediated upregulation of MUC5AC gene expression. A further understanding of mechanisms mediating MUC5AC regulation should lead to a honing of therapeutic approaches for the treatment of mucus overproduction in inflammatory lung diseases.


Assuntos
Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/metabolismo , Dexametasona/farmacologia , Regulação da Expressão Gênica , Interleucina-1beta/farmacologia , Neoplasias Pulmonares/genética , Mucina-5AC/genética , NF-kappa B/metabolismo , Adenocarcinoma/tratamento farmacológico , Adenocarcinoma/genética , Adenocarcinoma/metabolismo , Anti-Inflamatórios/farmacologia , Brônquios/efeitos dos fármacos , Brônquios/metabolismo , Células Cultivadas , Imunoprecipitação da Cromatina , Proteína de Ligação ao Elemento de Resposta ao AMP Cíclico/genética , RNA Polimerases Dirigidas por DNA/genética , RNA Polimerases Dirigidas por DNA/metabolismo , Ensaio de Desvio de Mobilidade Eletroforética , Humanos , Neoplasias Pulmonares/tratamento farmacológico , Neoplasias Pulmonares/metabolismo , Mucina-5AC/metabolismo , NF-kappa B/genética , Regiões Promotoras Genéticas/genética , RNA Mensageiro/genética , Reação em Cadeia da Polimerase em Tempo Real , Receptores de Interleucina-1/genética , Receptores de Interleucina-1/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa
19.
Am J Respir Cell Mol Biol ; 50(2): 292-300, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24010916

RESUMO

The polarity of the conducting airway epithelium is responsible for its directional secretion. This is an essential characteristic of lung integrity and function that dictates interactions between the external environment (apical) and subepithelial structures (basolateral). Defining the directional secretomes in the in vitro human bronchial epithelial (HBE) differentiated model could bring valuable insights into lung biology and pulmonary diseases. Normal primary HBE cells (n = 3) were differentiated into respiratory tract epithelium. Apical and basolateral secretions (24 h) were processed for proteome profiling and pathway analysis. A total of 243 proteins were identified in secretions from all HBE cultures combined. Of these, 51% were classified as secreted proteins, including true secreted proteins (36%) and exosomal proteins (15%). Close examination revealed consistent secretion of 69 apical proteins and 13 basolateral proteins and differential secretion of 25 proteins across all donors. Expression of Annexin A4 in apical secretions and Desmoglein-2 in basolateral secretions was validated using Western blot or ELISA in triplicate independent experiments. To the best of our knowledge, this is the first study defining apical and basolateral secretomes in the in vitro differentiated HBE model. The data demonstrate that epithelial polarity directs protein secretion with different patterns of biological processes to the apical and basolateral surfaces that are consistent with normal bronchial epithelium homeostatic functions. Applying this in vitro directional secretome model to lung diseases may elucidate their molecular pathophysiology and help define potential therapeutic targets.


Assuntos
Polaridade Celular/fisiologia , Células Epiteliais/metabolismo , Pulmão/metabolismo , Mucosa Respiratória/metabolismo , Asma/metabolismo , Diferenciação Celular/fisiologia , Células Cultivadas , Desmogleína 2/metabolismo , Células Epiteliais/citologia , Humanos
20.
PLoS One ; 8(5): e63871, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-23667681

RESUMO

Asthma is a chronic inflammatory condition of the lower respiratory tract associated with airway hyperreactivity and mucus obstruction in which a majority of cases are due to an allergic response to environmental allergens. Glucocorticoids such as prednisone have been standard treatment for many inflammatory diseases for the past 60 years. However, despite their effectiveness, long-term treatment is often limited by adverse side effects believed to be caused by glucocorticoid receptor-mediated gene transcription. This has led to the pursuit of compounds that retain the anti-inflammatory properties yet lack the adverse side effects associated with traditional glucocorticoids. We have developed a novel series of steroidal analogues (VBP compounds) that have been previously shown to maintain anti-inflammatory properties such as NFκB-inhibition without inducing glucocorticoid receptor-mediated gene transcription. This study was undertaken to determine the effectiveness of the lead compound, VBP15, in a mouse model of allergic lung inflammation. We show that VBP15 is as effective as the traditional glucocorticoid, prednisolone, at reducing three major hallmarks of lung inflammation--NFκB activity, leukocyte degranulation, and pro-inflammatory cytokine release from human bronchial epithelial cells obtained from patients with asthma. Moreover, we found that VBP15 is capable of reducing inflammation of the lung in vivo to an extent similar to that of prednisone. We found that prednisolone--but not VBP15 shortens the tibia in mice upon a 5 week treatment regimen suggesting effective dissociation of side effects from efficacy. These findings suggest that VBP15 may represent a potent and safer alternative to traditional glucocorticoids in the treatment of asthma and other inflammatory diseases.


Assuntos
Glucocorticoides/uso terapêutico , Hipersensibilidade/complicações , Hipersensibilidade/tratamento farmacológico , Pneumonia/complicações , Pneumonia/tratamento farmacológico , Pregnadienodiois/uso terapêutico , Animais , Asma/complicações , Asma/metabolismo , Asma/patologia , Degranulação Celular/efeitos dos fármacos , Movimento Celular/efeitos dos fármacos , Citocinas/metabolismo , Modelos Animais de Doenças , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Glucocorticoides/química , Glucocorticoides/farmacologia , Humanos , Leucócitos/efeitos dos fármacos , Leucócitos/fisiologia , Pulmão/efeitos dos fármacos , Pulmão/patologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , NF-kappa B/metabolismo , Osteogênese/efeitos dos fármacos , Ovalbumina , Pregnadienodiois/química , Pregnadienodiois/farmacologia , Tíbia/efeitos dos fármacos , Tíbia/patologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...