Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 73
Filtrar
Más filtros

País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 18(2): 225-235, 2017 02.
Artículo en Inglés | MEDLINE | ID: mdl-28024153

RESUMEN

The mechanisms by which human immunodeficiency virus 1 (HIV-1) avoids immune surveillance by dendritic cells (DCs), and thereby prevents protective adaptive immune responses, remain poorly understood. Here we showed that HIV-1 actively arrested antiviral immune responses by DCs, which contributed to efficient HIV-1 replication in infected individuals. We identified the RNA helicase DDX3 as an HIV-1 sensor that bound abortive HIV-1 RNA after HIV-1 infection and induced DC maturation and type I interferon responses via the signaling adaptor MAVS. Notably, HIV-1 recognition by the C-type lectin receptor DC-SIGN activated the mitotic kinase PLK1, which suppressed signaling downstream of MAVS, thereby interfering with intrinsic host defense during HIV-1 infection. Finally, we showed that PLK1-mediated suppression of DDX3-MAVS signaling was a viral strategy that accelerated HIV-1 replication in infected individuals.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/metabolismo , Células Dendríticas/virología , Infecciones por VIH/inmunología , VIH-1/fisiología , Evasión Inmune , Inmunidad , Macrófagos/virología , Proteínas Adaptadoras Transductoras de Señales/genética , Extractos Celulares , Repeticiones Palindrómicas Cortas Agrupadas y Regularmente Espaciadas/genética , Estudios de Cohortes , ARN Helicasas DEAD-box/metabolismo , Células Dendríticas/inmunología , Regulación Viral de la Expresión Génica , Células HEK293 , Infecciones por VIH/virología , Interacciones Huésped-Patógeno/genética , Humanos , Interferón beta/sangre , Macrófagos/inmunología , Polimorfismo de Nucleótido Simple , ARN Viral/inmunología , ARN Viral/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Transducción de Señal , Carga Viral/genética
2.
Am J Respir Crit Care Med ; 209(4): 374-389, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38016030

RESUMEN

Rationale: Non-cystic fibrosis bronchiectasis (NCFB) may originate in bronchiolar regions of the lung. Accordingly, there is a need to characterize the morphology and molecular characteristics of NCFB bronchioles. Objectives: Test the hypothesis that NCFB exhibits a major component of bronchiolar disease manifest by mucus plugging and ectasia. Methods: Morphologic criteria and region-specific epithelial gene expression, measured histologically and by RNA in situ hybridization and immunohistochemistry, identified proximal and distal bronchioles in excised NCFB lungs. RNA in situ hybridization and immunohistochemistry assessed bronchiolar mucus accumulation and mucin gene expression. CRISPR-Cas9-mediated IL-1R1 knockout in human bronchial epithelial cultures tested IL-1α and IL-1ß contributions to mucin production. Spatial transcriptional profiling characterized NCFB distal bronchiolar gene expression. Measurements and Main Results: Bronchiolar perimeters and lumen areas per section area were increased in proximal, but not distal, bronchioles in NCFB versus control lungs, suggesting proximal bronchiolectasis. In NCFB, mucus plugging was observed in ectatic proximal bronchioles and associated nonectatic distal bronchioles in sections with disease. MUC5AC and MUC5B mucins were upregulated in NCFB proximal bronchioles, whereas MUC5B was selectively upregulated in distal bronchioles. Bronchiolar mucus plugs were populated by IL-1ß-expressing macrophages. NCFB sterile sputum supernatants induced human bronchial epithelial MUC5B and MUC5AC expression that was >80% blocked by IL-1R1 ablation. Spatial transcriptional profiling identified upregulation of genes associated with secretory cells, hypoxia, interleukin pathways, and IL-1ß-producing macrophages in mucus plugs and downregulation of epithelial ciliogenesis genes. Conclusions: NCFB exhibits distinctive proximal and distal bronchiolar disease. Both bronchiolar regions exhibit bronchiolar secretory cell features and mucus plugging but differ in mucin gene regulation and ectasia.


Asunto(s)
Bronquiectasia , Fibrosis Quística , Humanos , Bronquiolos , Dilatación Patológica , Bronquiectasia/genética , Mucinas/metabolismo , Interleucina-1beta , Fibrosis , ARN , Mucina 5AC/genética
3.
J Infect Dis ; 230(1): e43-e47, 2024 Jul 25.
Artículo en Inglés | MEDLINE | ID: mdl-39052703

RESUMEN

Dysbiosis of the vaginal microbiome poses a serious risk for sexual human immunodeficiency virus type 1 (HIV-1) transmission. Prevotella spp are abundant during vaginal dysbiosis and associated with enhanced HIV-1 susceptibility; however, underlying mechanisms remain unclear. Here, we investigated the direct effect of vaginal bacteria on HIV-1 susceptibility of vaginal CD4+ T cells. Notably, pre-exposure to Prevotella timonensis enhanced HIV-1 uptake by vaginal T cells, leading to increased viral fusion and enhanced virus production. Pre-exposure to antiretroviral inhibitors abolished P timonensis-enhanced infection. Our study shows that the vaginal microbiome directly affects mucosal CD4+ T-cell susceptibility, emphasizing importance of vaginal dysbiosis diagnosis and treatment.


Asunto(s)
Linfocitos T CD4-Positivos , Disbiosis , Infecciones por VIH , VIH-1 , Prevotella , Vagina , Humanos , Femenino , Prevotella/aislamiento & purificación , Disbiosis/microbiología , Vagina/microbiología , Vagina/virología , Vagina/inmunología , Linfocitos T CD4-Positivos/inmunología , Infecciones por VIH/microbiología , Infecciones por VIH/inmunología , Infecciones por VIH/virología , Susceptibilidad a Enfermedades , Microbiota , Internalización del Virus
5.
Int J Mol Sci ; 24(5)2023 Mar 05.
Artículo en Inglés | MEDLINE | ID: mdl-36902441

RESUMEN

Cystic fibrosis transmembrane conductance regulator (CFTR) modulators, a new series of therapeutics that correct and potentiate some classes of mutations of the CFTR, have provided a great therapeutic advantage to people with cystic fibrosis (pwCF). The main hindrances of the present CFTR modulators are related to their limitations in reducing chronic lung bacterial infection and inflammation, the main causes of pulmonary tissue damage and progressive respiratory insufficiency, particularly in adults with CF. Here, the most debated issues of the pulmonary bacterial infection and inflammatory processes in pwCF are revisited. Special attention is given to the mechanisms favoring the bacterial infection of pwCF, the progressive adaptation of Pseudomonas aeruginosa and its interplay with Staphylococcus aureus, the cross-talk among bacteria, the bronchial epithelial cells and the phagocytes of the host immune defenses. The most recent findings of the effect of CFTR modulators on bacterial infection and the inflammatory process are also presented to provide critical hints towards the identification of relevant therapeutic targets to overcome the respiratory pathology of pwCF.


Asunto(s)
Fibrosis Quística , Infecciones Estafilocócicas , Adulto , Humanos , Fibrosis Quística/tratamiento farmacológico , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Pulmón/patología , Interacciones Huésped-Patógeno , Pseudomonas aeruginosa/genética
6.
Nicotine Tob Res ; 24(3): 395-399, 2022 02 14.
Artículo en Inglés | MEDLINE | ID: mdl-34519792

RESUMEN

INTRODUCTION: Alveolar macrophages (AMs) are lung-resident immune cells that phagocytose inhaled particles and pathogens, and help coordinate the lung's immune response to infection. Little is known about the impact of chronic e-cigarette use (ie, vaping) on this important pulmonary cell type. Thus, we determined the effect of vaping on AM phenotype and gene expression. AIMS AND METHODS: We recruited never-smokers, smokers, and e-cigarette users (vapers) and performed research bronchoscopies to isolate AMs from bronchoalveolar lavage fluid samples and epithelial cells from bronchial brushings. We then performed morphological analyses and used the Nanostring platform to look for changes in gene expression. RESULTS: AMs obtained from smokers and vapers were phenotypically distinct from those obtained from nonsmokers, and from each other. Immunocytochemistry revealed that vapers AMs had significantly elevated inducible nitric oxide synthase (M1) expression and significantly reduced CD301a (M2) expression compared with nonsmokers or smokers. Vapers' AMs and bronchial epithelia exhibited unique changes in gene expression compared with nonsmokers or smokers. Moreover, vapers' AMs were the most affected of all groups and had 124 genes uniquely downregulated. Gene ontology analysis revealed that vapers and smokers had opposing changes in biological processes. CONCLUSIONS: These data indicate that vaping causes unique changes to AMs and bronchial epithelia compared with nonsmokers and smokers which may impact pulmonary host defense. IMPLICATIONS: These data indicate that normal "healthy" vapers have altered AMs and may be at risk of developing abnormal immune responses to inflammatory stimuli.


Asunto(s)
Sistemas Electrónicos de Liberación de Nicotina , Productos de Tabaco , Vapeo , Expresión Génica , Humanos , Macrófagos Alveolares , Vapeo/efectos adversos
7.
Nature ; 540(7633): 448-452, 2016 12 15.
Artículo en Inglés | MEDLINE | ID: mdl-27919079

RESUMEN

The most prevalent route of HIV-1 infection is across mucosal tissues after sexual contact. Langerhans cells (LCs) belong to the subset of dendritic cells (DCs) that line the mucosal epithelia of vagina and foreskin and have the ability to sense and induce immunity to invading pathogens. Anatomical and functional characteristics make LCs one of the primary targets of HIV-1 infection. Notably, LCs form a protective barrier against HIV-1 infection and transmission. LCs restrict HIV-1 infection through the capture of HIV-1 by the C-type lectin receptor Langerin and subsequent internalization into Birbeck granules. However, the underlying molecular mechanism of HIV-1 restriction in LCs remains unknown. Here we show that human E3-ubiquitin ligase tri-partite-containing motif 5α (TRIM5α) potently restricts HIV-1 infection of LCs but not of subepithelial DC-SIGN+ DCs. HIV-1 restriction by TRIM5α was thus far considered to be reserved to non-human primate TRIM5α orthologues, but our data strongly suggest that human TRIM5α is a cell-specific restriction factor dependent on C-type lectin receptor function. Our findings highlight the importance of HIV-1 binding to Langerin for the routeing of HIV-1 into the human TRIM5α-mediated restriction pathway. TRIM5α mediates the assembly of an autophagy-activating scaffold to Langerin, which targets HIV-1 for autophagic degradation and prevents infection of LCs. By contrast, HIV-1 binding to DC-SIGN+ DCs leads to disassociation of TRIM5α from DC-SIGN, which abrogates TRIM5α restriction. Thus, our data strongly suggest that restriction by human TRIM5α is controlled by C-type-lectin-receptor-dependent uptake of HIV-1, dictating protection or infection of human DC subsets. Therapeutic interventions that incorporate C-type lectin receptors and autophagy-targeting strategies could thus provide cell-mediated resistance to HIV-1 in humans.


Asunto(s)
Antígenos CD/metabolismo , Autofagia , Proteínas Portadoras/metabolismo , VIH-1/fisiología , Células de Langerhans/metabolismo , Células de Langerhans/virología , Lectinas Tipo C/metabolismo , Lectinas de Unión a Manosa/metabolismo , Receptores del VIH/metabolismo , Factores de Restricción Antivirales , Moléculas de Adhesión Celular/metabolismo , Línea Celular , Infecciones por VIH/inmunología , Infecciones por VIH/prevención & control , Infecciones por VIH/transmisión , VIH-1/inmunología , Interacciones Huésped-Patógeno , Humanos , Inmunidad Mucosa , Células de Langerhans/citología , Células de Langerhans/inmunología , Receptores de Superficie Celular/metabolismo , Proteínas de Motivos Tripartitos , Ubiquitina-Proteína Ligasas
8.
Int J Mol Sci ; 22(6)2021 Mar 17.
Artículo en Inglés | MEDLINE | ID: mdl-33802742

RESUMEN

New anti-inflammatory treatments are needed for CF airway disease. Studies have implicated the endoplasmic reticulum stress transducer inositol requiring enzyme 1α (IRE1α) in CF airway inflammation. The activation of IRE1α promotes activation of its cytoplasmic kinase and RNase, resulting in mRNA splicing of X-box binding protein-1 (XBP-1s), a transcription factor required for cytokine production. We tested whether IRE1α kinase and RNase inhibition decreases cytokine production induced by the exposure of primary cultures of homozygous F508del CF human bronchial epithelia (HBE) to supernatant of mucopurulent material (SMM) from CF airways. We evaluated whether IRE1α expression is increased in freshly isolated and native CF HBE, and couples with increased XBP-1s levels. A FRET assay confirmed binding of the IRE1α kinase and RNase inhibitor, KIRA6, to the IRE1α kinase. F508del HBE cultures were exposed to SMM with or without KIRA6, and we evaluated the mRNA levels of XBP-1s, IL-6, and IL-8, and the secretion of IL-6 and IL-8. IRE1α mRNA levels were up-regulated in freshly isolated CF vs. normal HBE and coupled to increased XBP-1s mRNA levels. SMM increased XBP-1s, IL-6, and IL-8 mRNA levels and up-regulated IL-6 and IL-8 secretion, and KIRA6 blunted these responses in a dose-dependent manner. Moreover, a triple combination of CFTR modulators currently used in the clinic had no effect on SMM-increased XBP-1s levels coupled with increased cytokine production in presence or absence of KIRA6. These findings indicate that IRE1α mediates cytokine production in CF airways. Small molecule IRE1α kinase inhibitors that allosterically reduce RNase-dependent XBP-1s may represent a new therapeutic strategy for CF airway inflammation.


Asunto(s)
Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/patología , Endorribonucleasas/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/patología , Pulmón/patología , Terapia Molecular Dirigida , Proteínas Serina-Treonina Quinasas/metabolismo , Células Cultivadas , Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Citocinas/biosíntesis , Endorribonucleasas/genética , Epitelio/efectos de los fármacos , Epitelio/patología , Humanos , Imidazoles/química , Imidazoles/farmacología , Inflamación/genética , Modelos Biológicos , Naftalenos/química , Naftalenos/farmacología , Proteínas Serina-Treonina Quinasas/genética , Pirazinas/química , Pirazinas/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Regulación hacia Arriba/efectos de los fármacos , Regulación hacia Arriba/genética , Proteína 1 de Unión a la X-Box/metabolismo
9.
Am J Respir Crit Care Med ; 200(2): 220-234, 2019 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-30973754

RESUMEN

Rationale: The goal was to connect elements of idiopathic pulmonary fibrosis (IPF) pathogenesis, including chronic endoplasmic reticulum stress in respiratory epithelia associated with injury/inflammation and remodeling, distal airway mucus obstruction and honeycomb cyst formation with accumulation of MUC5B (mucin 5B), and associations between IPF risk and polymorphisms in the MUC5B promoter. Objectives: To test whether the endoplasmic reticulum (ER) stress sensor protein ERN2 (ER-to-nucleus signaling 2) and its downstream effector, the spliced form of XBP1S (X-box-binding protein 1), regulate MUC5B expression and differentially activate the MUC5B promoter variant in respiratory epithelia. Methods: Primary human airway epithelial (HAE) cells, transgenic mouse models, human IPF lung tissues, and cell lines expressing XBP1S and MUC5B promoters were used to explore relationships between the ERN2/XBP1S pathway and MUC5B. An inhibitor of the pathway, KIRA6, and XBP1 CRISPR-Cas9 were used in HAE cells to explore therapeutic potential. Measurements and Main Results: ERN2 regulated MUC5B and MUC5AC mRNAs. Downstream XBP1S selectively promoted MUC5B expression in vitro and in distal murine airway epithelia in vivo. XBP1S bound to the proximal region of the MUC5B promoter and differentially upregulated MUC5B expression in the context of the MUC5B promoter rs35705950 variant. High levels of ERN2 and XBP1S were associated with excessive MUC5B mRNAs in distal airways of human IPF lungs. Cytokine-induced MUC5B expression in HAE cells was inhibited by KIRA6 and XBP1 CRISPR-Cas9. Conclusions: A positive feedback bistable ERN2-XBP1S pathway regulates MUC5B-dominated mucus obstruction in IPF, providing an unfolded protein response-dependent mechanism linking the MUC5B promoter rs35705950 polymorphism with IPF pathogenesis. Inhibiting ERN2-dependent pathways/elements may provide a therapeutic option for IPF.


Asunto(s)
Endorribonucleasas/genética , Fibrosis Pulmonar Idiopática/genética , Proteínas de la Membrana/genética , Proteínas Serina-Treonina Quinasas/genética , Mucosa Respiratoria/metabolismo , Proteína 1 de Unión a la X-Box/genética , Animales , Sistemas CRISPR-Cas , Línea Celular , Estrés del Retículo Endoplásmico/genética , Endorribonucleasas/metabolismo , Regulación de la Expresión Génica , Humanos , Fibrosis Pulmonar Idiopática/metabolismo , Proteínas de la Membrana/metabolismo , Ratones , Ratones Transgénicos , Polimorfismo Genético , Cultivo Primario de Células , Regiones Promotoras Genéticas , Proteínas Serina-Treonina Quinasas/metabolismo , Proteína 1 de Unión a la X-Box/metabolismo
10.
Int J Mol Sci ; 21(21)2020 Oct 24.
Artículo en Inglés | MEDLINE | ID: mdl-33114346

RESUMEN

Hijacking and manipulation of host cell biosynthetic pathways by human enveloped viruses are essential for the viral lifecycle. Flaviviridae members, including hepatitis C, dengue and Zika viruses, extensively manipulate host lipid metabolism, underlining the importance of lipid droplets (LDs) in viral infection. LDs are dynamic cytoplasmic organelles that can act as sequestration platforms for a unique subset of host and viral proteins. Transient recruitment and mobilization of proteins to LDs during viral infection impacts host-cell biological properties, LD functionality and canonical protein functions. Notably, recent studies identified LDs in the nucleus and also identified that LDs are transported extracellularly via an autophagy-mediated mechanism, indicating a novel role for autophagy in Flaviviridae infections. These developments underline an unsuspected diversity and localization of LDs and potential moonlighting functions of LD-associated proteins during infection. This review summarizes recent breakthroughs concerning the LD hijacking activities of hepatitis C, dengue and Zika viruses and potential roles of cytoplasmic, nuclear and extracellular LD-associated viral proteins during infection.


Asunto(s)
Flaviviridae/patogenicidad , Gotas Lipídicas/metabolismo , Proteínas Virales/metabolismo , Animales , Autofagia , Núcleo Celular/metabolismo , Virus del Dengue/metabolismo , Virus del Dengue/patogenicidad , Espacio Extracelular/metabolismo , Flaviviridae/metabolismo , Hepacivirus/metabolismo , Hepacivirus/patogenicidad , Humanos , Gotas Lipídicas/virología , Virus Zika/metabolismo , Virus Zika/patogenicidad
11.
Am J Respir Crit Care Med ; 197(4): 481-491, 2018 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-29099608

RESUMEN

RATIONALE: Cystic fibrosis (CF) airways disease produces a mucoobstructive lung phenotype characterized by airways mucus plugging, epithelial mucous cell metaplasia/hyperplasia, chronic infection, and inflammation. Simultaneous biochemical and functional in vivo studies of mucin synthesis and secretion from CF airways are not available. In vitro translational models may quantitate differential CF versus normal mucin and fluid secretory responses to infectious/inflammatory stimuli. OBJECTIVES: We tested the hypothesis that CF airways exhibit defective epithelial fluid, but not mucin, secretory responses to bacterial/inflammatory host products. METHODS: Well-differentiated primary human bronchial epithelial cultures were exposed to supernatant from mucopurulent material (SMM) from human CF airways as a test of bacterial/inflammatory host product stimulus. Human bronchial epithelia (HBE) with normal CF transmembrane conductance regulator function were compared with ΔF508/ΔF508 CF HBE. MEASUREMENTS AND MAIN RESULTS: Acute (up to 60 min) SMM exposure promoted mucin secretion, but mucins were degraded by the proteolytic enzymes present in SMM. Chronic SMM exposure induced upregulation of mucin synthesis and storage and generated absolute increases in basal and stimulated mucin release in normal and CF cultures. These responses were similar in normal and CF cultures. In contrast, SMM produced a coordinated CF transmembrane conductance regulator-mediated Cl- secretory response in normal HBE, but not in CF HBE. The absence of the fluid secretory response in CF produced quantitatively more dehydrated mucus. CONCLUSIONS: Our study reveals the interplay between regulation of mucin and fluid secretion rates in inflamed versus noninflamed conditions and why a hyperconcentrated mucus is produced in CF airways.


Asunto(s)
Fibrosis Quística/metabolismo , Fluidoterapia , Pulmón/metabolismo , Mucinas/biosíntesis , Mucosa Respiratoria/metabolismo , Técnicas de Cultivo de Célula , Fibrosis Quística/patología , Ensayo de Inmunoadsorción Enzimática , Epitelio/metabolismo , Epitelio/patología , Humanos , Pulmón/patología , Mucinas/metabolismo , Reacción en Cadena de la Polimerasa , Mucosa Respiratoria/patología
12.
Am J Respir Cell Mol Biol ; 59(4): 428-436, 2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-29668297

RESUMEN

The lungs of patients with cystic fibrosis (CF) are characterized by an exaggerated inflammation driven by secretion of IL-8 from bronchial epithelial cells and worsened by Pseudomonas aeruginosa infection. To identify novel antiinflammatory molecular targets, we previously performed a genetic study of 135 genes of the immune response, which identified the c.2534C>T (p.S845L) variant of phospholipase C-ß3 (PLCB3) as being significantly associated with mild progression of pulmonary disease. Silencing PLCB3 revealed that it potentiates the Toll-like receptor's inflammatory signaling cascade originating from CF bronchial epithelial cells. In the present study, we investigated the role of the PLCB3-S845L variant together with two synthetic mutants paradigmatic of impaired catalytic activity or lacking functional activation in CF bronchial epithelial cells. In experiments in which cells were exposed to P. aeruginosa, the supernatant of mucopurulent material from the airways of patients with CF or different agonists revealed that PLCB3-S845L has defects of 1) agonist-induced Ca2+ release from endoplasmic reticulum and rise of Ca2+ concentration, 2) activation of conventional protein kinase C isoform ß, and 3) induction of IL-8 release. These results, besides identifying S845L as a loss-of-function variant, strengthen the importance of targeting PLCB3 to mitigate the CF inflammatory response in bronchial epithelial cells without blunting the immune response.


Asunto(s)
Fibrosis Quística/metabolismo , Fibrosis Quística/patología , Interleucina-8/metabolismo , Fosfolipasa C beta/deficiencia , Pseudomonas aeruginosa/fisiología , Bronquios/patología , Señalización del Calcio , Línea Celular , Simulación por Computador , Humanos , Moco/metabolismo , Mutación/genética , Fosfolipasa C beta/química , Fosfolipasa C beta/genética , Fosfolipasa C beta/metabolismo , Serina/metabolismo , Relación Estructura-Actividad
13.
Eur Respir J ; 52(4)2018 10.
Artículo en Inglés | MEDLINE | ID: mdl-30190268

RESUMEN

The multi-organ disease cystic fibrosis (CF) is caused by mutations in the cystic fibrosis transmembrane regulator gene (CFTR) that lead to diminished transepithelial anion transport. CF lungs are characterised by airway surface liquid (ASL) dehydration, chronic infection/inflammation and neutrophilia. Dysfunctional CFTR may upregulate the epithelial Na+ channel (ENaC), further exacerbating dehydration. We previously demonstrated that short palate lung and nasal epithelial clone 1 (SPLUNC1) negatively regulates ENaC in normal airway epithelia.Here, we used pulmonary tissue samples, sputum and human bronchial epithelial cells (HBECs) to determine whether SPLUNC1 could regulate ENaC in a CF-like environment.We found reduced endogenous SPLUNC1 in CF secretions, and rapid degradation of recombinant SPLUNC1 (rSPLUNC1) by CF secretions. Normal sputum, containing SPLUNC1 and SPLUNC1-derived peptides, inhibited ENaC in both normal and CF HBECs. Conversely, CF sputum activated ENaC, and rSPLUNC1 could not reverse this phenomenon. Additionally, we observed upregulation of ENaC protein levels in human CF bronchi. Unlike SPLUNC1, the novel SPLUNC1-derived peptide SPX-101 resisted protease degradation, bound apically to HBECs, inhibited ENaC and prevented ASL dehydration following extended pre-incubation with CF sputum.Our data indicate that CF mucosal secretions drive ASL hyperabsorption and that protease-resistant peptides, e.g. SPX-101, can reverse this effect to rehydrate CF ASL.


Asunto(s)
Fibrosis Quística/metabolismo , Deshidratación/patología , Células Epiteliales/metabolismo , Glicoproteínas/metabolismo , Fosfoproteínas/metabolismo , Células Cultivadas , Canales Epiteliales de Sodio/metabolismo , Glicoproteínas/genética , Humanos , Transporte Iónico , Pulmón/metabolismo , Fosfoproteínas/genética , Mucosa Respiratoria/metabolismo
14.
Nicotine Tob Res ; 19(2): 222-230, 2017 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-27613917

RESUMEN

INTRODUCTION: In 2013, the National Institutes of Health and the Food and Drug Administration funded a network of 14 Tobacco Centers of Regulatory Science (TCORS) with a mission that included research and training. A cross-TCORS Panel was established to define tobacco regulatory science (TRS) competencies to help harmonize and guide their emerging educational programs. The purpose of this paper is to describe the Panel's work to develop core TRS domains and competencies. METHODS: The Panel developed the list of domains and competencies using a semistructured Delphi method divided into four phases occurring between November 2013 and August 2015. RESULTS: The final proposed list included a total of 51 competencies across six core domains and 28 competencies across five specialized domains. CONCLUSIONS: There is a need for continued discussion to establish the utility of the proposed set of competencies for emerging TRS curricula and to identify the best strategies for incorporating these competencies into TRS training programs. Given the field's broad multidisciplinary nature, further experience is needed to refine the core domains that should be covered in TRS training programs versus knowledge obtained in more specialized programs. IMPLICATIONS: Regulatory science to inform the regulation of tobacco products is an emerging field. The paper provides an initial list of core and specialized domains and competencies to be used in developing curricula for new and emerging training programs aimed at preparing a new cohort of scientists to conduct critical TRS research.


Asunto(s)
Benchmarking , Competencia Clínica , Consejo Directivo/organización & administración , Cese del Hábito de Fumar , Fumar/legislación & jurisprudencia , Industria del Tabaco/legislación & jurisprudencia , Conferencias de Consenso como Asunto , Humanos , National Institutes of Health (U.S.) , Prevención del Hábito de Fumar , Estados Unidos , United States Food and Drug Administration
15.
Int J Mol Sci ; 18(1)2017 Jan 09.
Artículo en Inglés | MEDLINE | ID: mdl-28075361

RESUMEN

Cystic fibrosis (CF) pulmonary disease is characterized by chronic airway infection and inflammation. The infectious and inflamed CF airway environment impacts on the innate defense of airway epithelia and airway macrophages. The CF airway milieu induces an adaptation in these cells characterized by increased basal inflammation and a robust inflammatory response to inflammatory mediators. Recent studies have indicated that these responses depend on activation of the unfolded protein response (UPR). This review discusses the contribution of airway epithelia and airway macrophages to CF airway inflammatory responses and specifically highlights the functional importance of the UPR pathway mediated by IRE1/XBP-1 in these processes. These findings suggest that targeting the IRE1/XBP-1 UPR pathway may be a therapeutic strategy for CF airway disease.


Asunto(s)
Fibrosis Quística/patología , Inflamación/patología , Proteínas Serina-Treonina Quinasas/metabolismo , Sistema Respiratorio/patología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Humanos , Respuesta de Proteína Desplegada
16.
Am J Respir Cell Mol Biol ; 55(5): 645-656, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27281024

RESUMEN

Pseudomonas aeruginosa colonization, prominent inflammation with massive expression of the neutrophil chemokine IL-8, and luminal infiltrates of neutrophils are hallmarks of chronic lung disease in patients with cystic fibrosis (CF). The nociceptive transient receptor potential ankyrin (TRPA) 1 calcium channels have been recently found to be involved in nonneurogenic inflammation. Here, we investigate the role of TRPA1 in CF respiratory inflammatory models in vitro. Expression of TRPA1 was evaluated in CF lung tissue sections and cells by immunohistochemistry and immunofluorescence. Epithelial cell lines (A549, IB3-1, CuFi-1, CFBE41o-) and primary cells from patients with CF were used to: (1) check TRPA1 function modulation, by Fura-2 calcium imaging; (2) down-modulate TRPA1 function and expression, by pharmacological inhibitors (HC-030031 and A-967079) and small interfering RNA silencing; and (3) assess the effect of TRPA1 down-modulation on expression and release of cytokines upon exposure to proinflammatory challenges, by quantitative RT-PCR and 27-protein Bioplex assay. TRPA1 channels are expressed in the CF pseudostratified columnar epithelium facing the bronchial lumina exposed to bacteria, where IL-8 is coexpressed. Inhibition of TRPA1 expression results in a relevant reduction of release of several cytokines, including IL-8 and the proinflammatory cytokines IL-1ß and TNF-α, in CF primary bronchial epithelial cells exposed to P. aeruginosa and to the supernatant of mucopurulent material derived from the chronically infected airways of patients with CF. In conclusion, TRPA1 channels are involved in regulating the extent of airway inflammation driven by CF bronchial epithelial cells.


Asunto(s)
Canales de Calcio/metabolismo , Fibrosis Quística/complicaciones , Pulmón/patología , Proteínas del Tejido Nervioso/metabolismo , Neumonía/complicaciones , Neumonía/patología , Canales de Potencial de Receptor Transitorio/metabolismo , Células A549 , Adulto , Bronquios/patología , Fibrosis Quística/genética , Fibrosis Quística/patología , Citocinas/genética , Células Epiteliales/metabolismo , Células Epiteliales/patología , Femenino , Silenciador del Gen , Humanos , Interleucina-8/genética , Interleucina-8/metabolismo , Masculino , Persona de Mediana Edad , Modelos Biológicos , Proteínas del Tejido Nervioso/antagonistas & inhibidores , Neumonía/genética , Pseudomonas aeruginosa/fisiología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Canal Catiónico TRPA1 , Donantes de Tejidos , Transcripción Genética , Canales de Potencial de Receptor Transitorio/antagonistas & inhibidores , Adulto Joven
17.
Am J Respir Crit Care Med ; 192(12): 1449-61, 2015 Dec 15.
Artículo en Inglés | MEDLINE | ID: mdl-26331676

RESUMEN

RATIONALE: Alveolar macrophages (AMs) play a key role in host defense to inhaled bacterial pathogens, in part by secreting inflammatory mediators. Cystic fibrosis (CF) airways exhibit a persistent, robust inflammatory response that may contribute to the pathophysiology of CF. Recent findings have linked endoplasmic reticulum stress responses mediated by inositol-requiring enzyme 1α-dependent messenger RNA splicing (activation) of X-box-binding protein-1 (XBP-1s) to inflammation in peripheral macrophages. However, the role of XBP-1s in CF AM function is not known. OBJECTIVES: To evaluate inflammatory responses of AMs from chronically infected/inflamed human CF lungs and test whether XBP-1s is required for AM-mediated inflammation. METHODS: Basal and LPS-induced inflammatory responses were evaluated in primary cultures of non-CF versus CF AMs. XBP-1s was measured and its function was evaluated in AMs using 8-formyl-7-hydroxy-4-methylcoumarin (4µ8C), an inhibitor of inositol-requiring enzyme 1α-dependent XBP-1s, and in THP-1 cells stably expressing XBP-1 shRNA, XBP-1s, or a dominant-negative XBP-1. MEASUREMENTS AND MAIN RESULTS: CF AMs exhibited exaggerated basal and LPS-induced production of tumor necrosis factor-α and IL-6, and these responses were coupled to increased levels of XBP-1s. In non-CF and CF AMs, LPS-induced cytokine production was blunted by 4µ8C. A role for XBP-1s in AM inflammatory responses was further established by data from dTHP-1 cells indicating that expression of XBP-1 shRNA reduced XBP-1s levels and LPS-induced inflammatory responses; and LPS-induced inflammation was up-regulated by expression of XBP-1s and inhibited by dominant-negative XBP-1. CONCLUSIONS: These findings suggest that AMs contribute to the robust inflammation of CF airways via an up-regulation of XBP-1s-mediated cytokine production.


Asunto(s)
Fibrosis Quística/genética , Fibrosis Quística/inmunología , Proteínas de Unión al ADN/genética , Proteínas de Unión al ADN/inmunología , Inmunidad Innata/inmunología , Macrófagos Alveolares/inmunología , Factores de Transcripción/genética , Factores de Transcripción/inmunología , Células Cultivadas , Fibrosis Quística/complicaciones , Humanos , Inmunidad Innata/genética , Inflamación/complicaciones , Inflamación/genética , Inflamación/inmunología , Factores de Transcripción del Factor Regulador X , Proteína 1 de Unión a la X-Box
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA