Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 24
Filtrar
1.
Am J Respir Cell Mol Biol ; 66(3): 271-282, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-34807800

RESUMO

Orai1 is a plasma membrane Ca2+ channel that mediates store-operated Ca2+ entry (SOCE) and regulates inflammation. Short palate lung and nasal epithelial clone 1 (SPLUNC1) is an asthma gene modifier that inhibits Orai1 and SOCE via its C-terminal α6 region. SPLUNC1 levels are diminished in asthma patient airways. Thus, we hypothesized that inhaled α6 peptidomimetics could inhibit Orai1 and reduce airway inflammation in a murine asthma model. To evaluate α6-Orai1 interactions, we used fluorescent assays to measure Ca2+ signaling, Förster resonance energy transfer, fluorescent recovery after photobleaching, immunostaining, total internal reflection microscopy, and Western blotting. To test whether α6 peptidomimetics inhibited SOCE and decreased inflammation in vivo, wild-type and SPLUNC1-/- mice were exposed to house dust mite (HDM) extract with or without α6 peptide. We also performed nebulization, jet milling, and scanning electron microscopy to evaluate α6 for inhalation. SPLUNC1-/- mice had an exaggerated response to HDM. In BAL-derived immune cells, Orai1 levels increased after HDM exposure in SPLUNC1-/- but not wild-type mice. Inhaled α6 reduced Orai1 levels in mice regardless of genotype. In HDM-exposed mice, α6 dose-dependently reduced eosinophilia and neutrophilia. In vitro, α6 inhibited SOCE in multiple immune cell types, and α6 could be nebulized or jet milled without loss of function. These data suggest that α6 peptidomimetics may be a novel, effective antiinflammatory therapy for patients with asthma.


Assuntos
Asma , Peptidomiméticos , Animais , Asma/tratamento farmacológico , Cálcio/metabolismo , Glicoproteínas , Humanos , Inflamação , Pulmão/metabolismo , Camundongos , Proteína ORAI1/genética , Proteína ORAI1/metabolismo , Fosfoproteínas
2.
Respir Res ; 21(1): 190, 2020 Jul 17.
Artigo em Inglês | MEDLINE | ID: mdl-32680508

RESUMO

Cystic fibrosis (CF) patients are extremely vulnerable to Burkholderia cepacia complex (Bcc) infections. However, the underlying etiology is poorly understood. We tested the hypothesis that short palate lung and nasal epithelial clone 1 (SPLUNC1)-epithelial sodium channel (ENaC) interactions at the plasma membrane are required to reduce Bcc burden in normal airways. To determine if SPLUNC1 was needed to reduce Bcc burden in the airways, SPLUNC1 knockout mice and their wild-type littermates were infected with B. cenocepacia strain J2315. SPLUNC1 knockout mice had increased bacterial burden in the lungs compared to wild-type littermate mice. SPLUNC1-knockdown primary human bronchial epithelia (HBECs) were incubated with J2315, which resulted in increased bacterial burden compared to non-transduced HBECs. We next determined the interaction of the SPLUNC1-ENaC complex during J2315 infection. SPLUNC1 remained at the apical plasma membrane of normal HBECs but less was present at the apical plasma membrane of CF HBECs. Additionally, SPLUNC1-ßENaC complexes reduced intracellular J2315 burden. Our data indicate that (i) secreted SPLUNC1 is required to reduce J2315 burden in the airways and (ii) its interaction with ENaC prevents cellular invasion of J2315.


Assuntos
Infecções por Burkholderia/microbiologia , Burkholderia cenocepacia/patogenicidade , Fibrose Cística/microbiologia , Células Epiteliais/microbiologia , Canais Epiteliais de Sódio/metabolismo , Glicoproteínas/metabolismo , Pulmão/microbiologia , Fosfoproteínas/metabolismo , Infecções Respiratórias/microbiologia , Adolescente , Adulto , Animais , Carga Bacteriana , Infecções por Burkholderia/genética , Infecções por Burkholderia/metabolismo , Estudos de Casos e Controles , Fibrose Cística/genética , Fibrose Cística/metabolismo , Modelos Animais de Doenças , Células Epiteliais/metabolismo , Canais Epiteliais de Sódio/genética , Feminino , Glicoproteínas/genética , Células HEK293 , Interações Hospedeiro-Patógeno , Humanos , Pulmão/metabolismo , Masculino , Camundongos Endogâmicos C57BL , Camundongos Knockout , Pessoa de Meia-Idade , Fosfoproteínas/genética , Infecções Respiratórias/genética , Infecções Respiratórias/metabolismo , Adulto Jovem
3.
Respir Res ; 20(1): 94, 2019 May 21.
Artigo em Inglês | MEDLINE | ID: mdl-31113421

RESUMO

BACKGROUND: Tobacco smoke exposure impairs the lung's innate immune response, leading to an increased risk of chronic infections. SPLUNC1 is a secreted, multifunctional innate defense protein that has antimicrobial activity against Gram negative organisms. We hypothesize that tobacco smoke-induced SPLUNC1 dysfunction contributes to the observed defect in innate immunity in tobacco smokers and that this dysfunction can be used as a potential biomarker of harm. METHODS: We collected sputum from never-smokers and otherwise healthy smokers. We performed Western blotting to determine SPLUNC1 levels and determined antimicrobial activity against nontypeable Haemophilus influenzae. An in vitro exposure model was utilized to measure the effect of tobacco exposure on human bronchial epithelial culture (HBEC) antimicrobial activity against H. influenzae. The direct effects of cigarette and little cigar smoke exposure on SPLUNC1 function was determined using 24 h growth measurements and LPS binding assays. RESULTS: H. influenzae growth in cigarette smoker's sputum was significantly greater compared to never-smokers sputum over 24 h. HBEC supernatants and lysates contained significantly higher numbers of H. influenzae following chronic cigarette and little cigar smoke exposure compared to air-exposed controls. Furthermore, SPLUNC1's antimicrobial activity and LPS-binding capability against both H. influenzae and P. aeruginosa was attenuated following cigarette and little cigar exposure. CONCLUSIONS: These data suggest that cigarette and little cigar exposure impairs SPLUNC1's antimicrobial ability and that this inhibition may serve as a novel biomarker of harm that can be used to assess the toxicity of commercial tobacco products.


Assuntos
Anti-Infecciosos/farmacologia , Glicoproteínas/farmacologia , Fosfoproteínas/farmacologia , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/microbiologia , Fumar Tabaco/efeitos adversos , Adulto , Idoso , Células Cultivadas , Feminino , Haemophilus influenzae/efeitos dos fármacos , Haemophilus influenzae/fisiologia , Humanos , Masculino , Pessoa de Meia-Idade , Pseudomonas aeruginosa/efeitos dos fármacos , Pseudomonas aeruginosa/fisiologia , Produtos do Tabaco/efeitos adversos , Poluição por Fumaça de Tabaco/efeitos adversos
4.
FASEB J ; 32(5): 2478-2491, 2018 05.
Artigo em Inglês | MEDLINE | ID: mdl-29295861

RESUMO

Cystic fibrosis (CF) is a common genetic disease with significantly increased mortality. CF airways exhibit ion transport abnormalities, including hyperactivity of the epithelial Na+ channel (ENaC). Short-palate lung and nasal epithelial clone 1 (SPLUNC1) is a multifunctional innate defense protein that is secreted into the airway lumen. We have previously demonstrated that SPLUNC1 binds to and inhibits ENaC to maintain fluid homeostasis in airway epithelia and that this process fails in CF airways. Despite this, how SPLUNC1 actually regulates ENaC is unknown. Here, we found that SPLUNC1 caused αγ-ENaC to internalize, whereas SPLUNC1 and ß-ENaC remained at the plasma membrane. Additional studies revealed that SPLUNC1 increased neural precursor cell-expressed developmentally down-regulated protein 4-2-dependent ubiquitination of α- but not ß- or γ-ENaC. We also labeled intracellular ENaC termini with green fluorescent protein and mCherry, and found that extracellular SPLUNC1 altered intracellular ENaC Forster resonance energy transfer. Taken together, our data indicate that SPLUNC1 is an allosteric regulator of ENaC that dissociates αßγ-ENaC to generate a new SPLUNC1-ß-ENaC complex. These data indicate a novel mode for regulating ENaC at the plasma membrane.-Kim, C. S., Ahmad, S., Wu, T., Walton, W. G., Redinbo, M. R., Tarran, R. SPLUNC1 is an allosteric modulator of the epithelial sodium channel.


Assuntos
Membrana Celular/metabolismo , Células Epiteliais/metabolismo , Canais Epiteliais de Sódio/metabolismo , Glicoproteínas/metabolismo , Complexos Multiproteicos/química , Mucosa Nasal/metabolismo , Fosfoproteínas/metabolismo , Regulação Alostérica/fisiologia , Membrana Celular/química , Membrana Celular/genética , Células Epiteliais/química , Canais Epiteliais de Sódio/química , Canais Epiteliais de Sódio/genética , Transferência Ressonante de Energia de Fluorescência , Glicoproteínas/química , Glicoproteínas/genética , Células HEK293 , Humanos , Proteínas Luminescentes , Complexos Multiproteicos/genética , Complexos Multiproteicos/metabolismo , Mucosa Nasal/química , Fosfoproteínas/química , Fosfoproteínas/genética , Proteína Vermelha Fluorescente
5.
FASEB J ; : fj201800345R, 2018 Jun 11.
Artigo em Inglês | MEDLINE | ID: mdl-29890087

RESUMO

Chronic obstructive pulmonary disease (COPD) is a growing cause of morbidity and mortality worldwide. Cigarette smoke (CS) exposure, a major cause of COPD, dysregulates airway epithelial ion transport and diminishes airway surface liquid (ASL) volume. Short palate lung and nasal epithelial clone 1 (SPLUNC1) is secreted into the airway lumen where it maintains airway hydration via interactions with the epithelial Na+ channel (ENaC). Although ASL hydration is dysregulated in CS-exposed/COPD airways, effects of CS on SPLUNC1 have not been elucidated. We hypothesized that CS alters SPLUNC1 activity, therefore contributing to ASL dehydration. CS exposure caused irreversible SPLUNC1 aggregation and prevented SPLUNC1 from internalizing ENaC and maintaining ASL hydration. Proteomic analysis revealed αß-unsaturated aldehyde modifications to SPLUNC1's cysteine residues. Removal of these cysteines prevented SPLUNC1 from regulating ENaC/ASL volume. In contrast, SPX-101, a peptide mimetic of natural SPLUNC1, that internalizes ENaC, but does not contain cysteines was unaffected by CS. SPX-101 increased ASL hydration and attenuated ENaC activity in airway cultures after CS exposure and prolonged survival in a chronic airway disease model. These findings suggest that the CS-induced defects in SPLUNC1 can be circumvented, thus making SPX-101 a novel candidate for the treatment of mucus dehydration in COPD. -Moore, P. J., Reidel, B., Ghosh, A., Sesma, J., Kesimer, M., Tarran, R. Cigarette smoke modifies and inactivates SPLUNC1, leading to airway dehydration.

6.
Am J Respir Cell Mol Biol ; 52(5): 525-34, 2015 May.
Artigo em Inglês | MEDLINE | ID: mdl-25265466

RESUMO

Bactericidal/permeability-increasing protein fold-containing family member A1 (BPIFA1), formerly known as SPLUNC1, is one of the most abundant proteins in respiratory secretions and has been identified with increasing frequency in studies of pulmonary disease. Its expression is largely restricted to the respiratory tract, being highly concentrated in the upper airways and proximal trachea. BPIFA1 is highly responsive to airborne pathogens, allergens, and irritants. BPIFA1 actively participates in host protection through antimicrobial, surfactant, airway surface liquid regulation, and immunomodulatory properties. Its expression is modulated in multiple lung diseases, including cystic fibrosis, chronic obstructive pulmonary disease, respiratory malignancies, and idiopathic pulmonary fibrosis. However, the role of BPIFA1 in pulmonary pathogenesis remains to be elucidated. This review highlights the versatile properties of BPIFA1 in antimicrobial protection and its roles as a sensor of environmental exposure and regulator of immune cell function. A greater understanding of the contribution of BPIFA1 to disease pathogenesis and activity may clarify if BPIFA1 is a biomarker and potential drug target in pulmonary disease.


Assuntos
Glicoproteínas/metabolismo , Pneumopatias/microbiologia , Fosfoproteínas/metabolismo , Mucosa Respiratória/metabolismo , Animais , Regulação da Expressão Gênica , Glicoproteínas/genética , Interações Hospedeiro-Patógeno , Humanos , Imunidade Inata , Mediadores da Inflamação/metabolismo , Pneumopatias/genética , Pneumopatias/imunologia , Pneumopatias/virologia , Fosfoproteínas/genética , Mucosa Respiratória/imunologia , Mucosa Respiratória/microbiologia , Mucosa Respiratória/virologia , Transdução de Sinais
7.
Am J Respir Cell Mol Biol ; 53(5): 607-14, 2015 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-25574903

RESUMO

BPI fold containing family A, member 1 (BPIFA1) and BPIFB1 are putative innate immune molecules expressed in the upper airways. Because of their hypothesized roles in airway defense, these molecules may contribute to lung disease severity in cystic fibrosis (CF). We interrogated BPIFA1/BPIFB1 single-nucleotide polymorphisms in data from an association study of CF modifier genes and found an association of the G allele of rs1078761 with increased lung disease severity (P = 2.71 × 10(-4)). We hypothesized that the G allele of rs1078761 is associated with decreased expression of BPIFA1 and/or BPIFB1. Genome-wide lung gene expression and genotyping data from 1,111 individuals with lung disease, including 51 patients with CF, were tested for associations between genotype and BPIFA1 and BPIFB1 gene expression levels. Findings were validated by quantitative PCR in a subset of 77 individuals. Western blotting was used to measure BPIFA1 and BPIFB1 protein levels in 93 lung and 101 saliva samples. The G allele of rs1078761 was significantly associated with decreased mRNA levels of BPIFA1 (P = 4.08 × 10(-15)) and BPIFB1 (P = 0.0314). These findings were confirmed with quantitative PCR and Western blotting. We conclude that the G allele of rs1078761 may be detrimental to lung function in CF owing to decreased levels of BPIFA1 and BPIFB1.


Assuntos
Autoantígenos/genética , Fibrose Cística/genética , Glicoproteínas/genética , Pulmão/metabolismo , Fosfoproteínas/genética , Polimorfismo de Nucleotídeo Único , Proteínas/genética , Adolescente , Adulto , Alelos , Autoantígenos/imunologia , Estudos de Casos e Controles , Criança , Fibrose Cística/imunologia , Fibrose Cística/patologia , Proteínas de Ligação a Ácido Graxo , Feminino , Regulação da Expressão Gênica , Estudo de Associação Genômica Ampla , Glicoproteínas/imunologia , Humanos , Imunidade Inata , Pulmão/imunologia , Pulmão/patologia , Masculino , Fosfoproteínas/imunologia , Proteínas/imunologia , Locos de Características Quantitativas , RNA Mensageiro/genética , RNA Mensageiro/imunologia , Saliva/química , Índice de Gravidade de Doença , Transdução de Sinais
8.
Heliyon ; 10(8): e29587, 2024 Apr 30.
Artigo em Inglês | MEDLINE | ID: mdl-38660271

RESUMO

Background: Pulmonary arterial hypertension (PAH) represents a substantial global risk to human health. This study aims to identify diagnostic biomarkers for PAH and assess their association with the immune microenvironment through the utilization of sophisticated bioinformatics techniques. Methods: Based on two microarray datasets, differentially expressed genes (DEGs) were detected, and hub genes underwent a sequence of machine learning analyses. After pathways associated with PAH were assessed by gene enrichment analysis, the identified genes were validated using external datasets and confirmed in a monocrotaline (MCT)-induced rat model. In addition, three algorithms were employed to estimate the proportions of various immune cell types, and the link between hub genes and immune cells was substantiated. Results: Using SVM, LASSO, and WGCNA, we identified seven hub genes, including (BPIFA1, HBA2, HBB, LOC441081, PI15, S100A9, and WIF1), of which only BPIFA1 remained stable in the external datasets and was validated in an MCT-induced rat model. Furthermore, the results of the functional enrichment analysis established a link between PAH and both metabolism and the immune system. Correlation assessment showed that BPIFA1 expression in the MCP-counter algorithm was negatively associated with various immune cell types, positively correlated with macrophages in the ssGSEA algorithm, and correlated with M1 and M2 macrophages in the CIBERSORT algorithm. Conclusion: BPIFA1 serves as a modulator of PAH, with the potential to impact the immune microenvironment and disease progression, possibly through its regulatory influence on both M1 and M2 macrophages.

10.
Am J Physiol Lung Cell Mol Physiol ; 305(12): L990-L1001, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24124190

RESUMO

The epithelial sodium channel (ENaC) is responsible for Na(+) and fluid absorption across colon, kidney, and airway epithelia. Short palate lung and nasal epithelial clone 1 (SPLUNC1) is a secreted, innate defense protein and an autocrine inhibitor of ENaC that is highly expressed in airway epithelia. While SPLUNC1 has a bactericidal permeability-increasing protein (BPI)-type structure, its NH2-terminal region lacks structure. Here we found that an 18 amino acid peptide, S18, which corresponded to residues G22-A39 of the SPLUNC1 NH2 terminus inhibited ENaC activity to a similar degree as full-length SPLUNC1 (∼2.5 fold), while SPLUNC1 protein lacking this region was without effect. S18 did not inhibit the structurally related acid-sensing ion channels, indicating specificity for ENaC. However, S18 preferentially bound to the ßENaC subunit in a glycosylation-dependent manner. ENaC hyperactivity is contributory to cystic fibrosis (CF) lung disease. Unlike control, CF human bronchial epithelial cultures (HBECs) where airway surface liquid (ASL) height was abnormally low (4.2 ± 0.6 µm), addition of S18 prevented ENaC-led ASL hyperabsorption and maintained CF ASL height at 7.9 ± 0.6 µm, even in the presence of neutrophil elastase, which is comparable to heights seen in normal HBECs. Our data also indicate that the ENaC inhibitory domain of SPLUNC1 may be cleaved away from the main molecule by neutrophil elastase, suggesting that it may still be active during inflammation or neutrophilia. Furthermore, the robust inhibition of ENaC by the S18 peptide suggests that this peptide may be suitable for treating CF lung disease.


Assuntos
Absorção/fisiologia , Fibrose Cística/metabolismo , Células Epiteliais/metabolismo , Glicoproteínas/metabolismo , Fosfoproteínas/metabolismo , Sódio/metabolismo , Células Cultivadas , Canais Epiteliais de Sódio/metabolismo , Glicoproteínas/genética , Humanos , Transporte de Íons/fisiologia , Elastase de Leucócito/metabolismo , Pulmão/metabolismo , Fosfoproteínas/genética , Mucosa Respiratória/metabolismo
11.
Front Cell Infect Microbiol ; 12: 1035566, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36519134

RESUMO

In vitro culture and differentiation of human-derived airway basal cells under air-liquid interface (ALI) into a pseudostratified mucociliated mucosal barrier has proven to be a powerful preclinical tool to study pathophysiology of respiratory epithelium. As such, identifying differentiation stage-specific biomarkers can help investigators better characterize, standardize, and validate populations of regenerating epithelial cells prior to experimentation. Here, we applied longitudinal transcriptomic analysis and observed that the pattern and the magnitude of OMG, KRT14, STC1, BPIFA1, PLA2G7, TXNIP, S100A7 expression create a unique biosignature that robustly indicates the stage of epithelial cell differentiation. We then validated our findings by quantitative hemi-nested real-time PCR from in vitro cultures sourced from multiple donors. In addition, we demonstrated that at protein-level secretion of BPIFA1 accurately reflects the gene expression profile, with very low quantities present at the time of ALI induction but escalating levels were detectable as the epithelial cells terminally differentiated. Moreover, we observed that increase in BPIFA1 secretion closely correlates with emergence of secretory cells and an anti-inflammatory phenotype as airway epithelial cells undergo mucociliary differentiation under air-liquid interface in vitro.


Assuntos
Células Epiteliais , Mucosa Respiratória , Humanos , Células Cultivadas , Células Epiteliais/metabolismo , Diferenciação Celular , Epitélio , Biomarcadores/metabolismo , Glicoproteínas/genética , Glicoproteínas/metabolismo , Fosfoproteínas/metabolismo
12.
Genes (Basel) ; 14(1)2022 12 21.
Artigo em Inglês | MEDLINE | ID: mdl-36672756

RESUMO

Bactericidal/permeability-increasing protein, a primary factor of the innate immune system of mammals, participates in natural immune protection against invading bacteria. BPIFA1 actively contributes to host defense via multiple mechanisms, such as antibacterial, surfactant, airway surface liquid control, and immunomodulatory activities. However, the evolutionary history and selection forces on the BPIFA1 gene in mammals during adaptive evolution are poorly understood. This study examined the BPIFA1 gene of humans compared with that of other mammalian species to estimate the selective pressure derived by adaptive evolution. To assess whether or not positive selection occurred, we employed several different possibility tests (M1 vs. M2 and M7 vs. M8). The proportions of positively selected sites were significant, with a likelihood log value of 93.63 for the BPIFA1 protein. The Selecton server was used on the same dataset to reconfirm positive selection for specific sites by employing the Mechanistic-Empirical Combination model, thus providing additional evidence supporting the findings of positive selection. There was convincing evidence for positive selection signals in the BPIFA1 genes of mammalian species, which was more significant for selection signs and creating signals. We performed probability tests comparing various models based on dN/dS ratios to recognize specific codons under positive selection pressure. We identified positively selected sites in the LBP-BPI domain of BPIFA1 proteins in the mammalian genome, including a lipid-binding domain with a very high degree of selectivity for DPPC. BPIFA1 activates the upper airway's innate immune system in response to numerous genetic signals in the mammalian genome. These findings highlight evolutionary advancements in immunoregulatory effects that play a significant role in the antibacterial and antiviral defenses of mammalian species.


Assuntos
Glicoproteínas , Fosfoproteínas , Humanos , Animais , Glicoproteínas/genética , Fosfoproteínas/genética , Mamíferos/genética , Mamíferos/metabolismo , Imunidade Inata/genética , Evolução Molecular , Permeabilidade
13.
Physiol Rep ; 10(10): e15306, 2022 05.
Artigo em Inglês | MEDLINE | ID: mdl-35581745

RESUMO

Orai1 is a ubiquitously-expressed plasma membrane Ca2+ channel that is involved in store-operated Ca2+ entry (SOCE): a fundamental biological process that regulates gene expression, the onset of inflammation, secretion, and the contraction of airway smooth muscle (ASM). During SOCE, Ca2+ leaves the endoplasmic reticulum, which then stimulates a second, amplifying wave of Ca2+ influx through Orai1 into the cytoplasm. Short Palate LUng and Nasal epithelial Clone 1 (SPLUNC1; gene name BPIFA1) is a multi-functional, innate defense protein that is highly abundant in the lung. We have previously reported that SPLUNC1 was secreted from epithelia, where it bound to and inhibited Orai1, leading to reduced SOCE and ASM relaxation. However, the underlying mechanism of action is unknown. Here, we probed the SPLUNC1-Orai1 interactions in ASM and HEK293T cells using biochemical and imaging techniques. We observed that SPLUNC1 caused a conformational change in Orai1, as measured using Forster resonance energy transfer (FRET). SPLUNC1 binding also led to Nedd4-2 dependent ubiquitination of Orai1. Moreover, SPLUNC1 internalized Orai1 to lysosomes, leading to Orai1 degradation. Thus, we conclude that SPLUNC1 is an allosteric regulator of Orai1. Our data indicate that SPLUNC1-mediated Orai1 inhibition could be utilized as a therapeutic strategy to reduce SOCE.


Assuntos
Glicoproteínas/metabolismo , Pulmão , Músculo Liso , Fosfoproteínas/metabolismo , Cálcio/metabolismo , Sinalização do Cálcio/fisiologia , Membrana Celular/metabolismo , Células HEK293 , Humanos , Pulmão/metabolismo , Músculo Liso/metabolismo , Proteína ORAI1/genética , Proteína ORAI1/metabolismo
14.
Innate Immun ; 28(2): 67-78, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35201913

RESUMO

Bactericidal/permeability-increasing fold containing family A, member 1 (BPIFA1) is an innate immunity defense protein. Our previous studies proved its antibacterial and antiviral effects, but its role in fungi remains unknown. The study aimed to identify antifungal peptides (AFP) derived from BPIFA1, and three antimicrobial peptides (AMP1-3) were designed. The antifungal effects were proved by growth inhibition assay. AMP3 activity was confirmed by germ tube growth experiment and XTT assay. Its effects on cell wall and membrane of Candida albicans were assessed by tannic acid and Annexin V-FITC/PI double staining, respectively. Additionally, scanning electron microscope (SEM) and transmission electron microscopy (TEM) were used for morphological and ultrastructural observation. The expression of ALS1, EAP1, and SUN41 was tested by qPCR. Ultimately, three AMPs could fight against C. albicans in vitro, and AMP3 was highly effective. It functioned by destroying the integrity of cell wall and normal structure of cell membrane. It also inhibited biofilm formation of C. albicans. In addition, AMP3 down-regulated the expression of ALS1, EAP1, and SUN41, those are known to be involved in virulence of C. albicans. Altogether, the study reported successful development of a novel AFP, which could be used as a new strategy for antifungal therapy.


Assuntos
Antifúngicos , Candida albicans , Antifúngicos/química , Antifúngicos/metabolismo , Antifúngicos/farmacologia , Peptídeos Antimicrobianos , Biofilmes , Candida albicans/metabolismo , Glicoproteínas/metabolismo , Humanos , Testes de Sensibilidade Microbiana , Fosfoproteínas/metabolismo , alfa-Fetoproteínas/metabolismo , alfa-Fetoproteínas/farmacologia
15.
Int J Biol Macromol ; 164: 4022-4031, 2020 Dec 01.
Artigo em Inglês | MEDLINE | ID: mdl-32890564

RESUMO

Bacterial permeability family member A1 (BPIFA1) is one of the most abundant proteins present in normal airway surface liquid (ASL). It is known to be diminished in asthmatic patients' sputum, which causes airway hyperresponsiveness (AHR). What is currently unclear is how environmental factors, such as allergens' impact on BPIFA1's abundance and functions in the context of allergic asthma. House dust mite (HDM) is a predominant domestic source of aeroallergens. The group of proteases found in HDM is thought to cleave multiple cellular protective mechanisms, and therefore foster the development of allergic asthma. Here, we show that BPIFA1 is cleaved by HDM proteases in a time-, dose-, and temperature-dependent manner. We have also shown the main component in HDM that is responsible for BPIFA1's degradation is Der p1. Fragmented BPIFA1 failed to bind E. coli lipopolysaccharide (LPS), and hence elevated TNFα and IL-6 secretion in human whole blood. BPIFA1 degradation is also observed in vivo in bronchoalveolar fluid (BALF) of mice which are intranasally instilled with HDM. These data suggest that proteases associated with environmental allergens such as HDM cleave BPIFA1 and therefore impair its immune modulator function.


Assuntos
Antígenos de Dermatophagoides/metabolismo , Proteínas de Artrópodes/metabolismo , Cisteína Endopeptidases/metabolismo , Glicoproteínas/metabolismo , Imunomodulação , Fosfoproteínas/metabolismo , Alérgenos/imunologia , Animais , Antígenos de Dermatophagoides/imunologia , Proteínas de Artrópodes/imunologia , Cálcio/metabolismo , Sinalização do Cálcio , Linhagem Celular , Cisteína Endopeptidases/imunologia , Inibidores de Cisteína Proteinase/farmacologia , Citocinas/metabolismo , Glicoproteínas/farmacologia , Humanos , Imunomodulação/efeitos dos fármacos , Mediadores da Inflamação/metabolismo , Camundongos , Fosfoproteínas/farmacologia , Proteólise/efeitos dos fármacos , Temperatura
16.
Dis Model Mech ; 12(3)2019 03 21.
Artigo em Inglês | MEDLINE | ID: mdl-30898767

RESUMO

Auditory bulla cavitation defects are a cause of otitis media, but the normal cellular pattern of bulla mesenchyme regression and its failure are not well understood. In mice, neural-crest-derived mesenchyme occupies the bulla from embryonic day 17.5 (E17.5) to postnatal day 11 (P11) and then regresses to form the adult air-filled bulla cavity. We report that bulla mesenchyme is bordered by a single layer of non-ciliated epithelium characterized by interdigitating cells with desmosome cell junctions and a basal lamina, and by Bpifa1 gene expression and laminin staining of the basal lamina. At P11-P12, the mesenchyme shrinks: mesenchyme-associated epithelium shortens, and mesenchymal cells and extracellular matrix collagen fibrils condense, culminating in the formation of cochlea promontory mucosa bordered by compact non-ciliated epithelial cells. FBXO11 is a candidate disease gene in human chronic otitis media with effusion and we report that a bulla cavitation defect initiates the pathogenesis of otitis media in the established mouse model Jeff (Fbxo11Jf/+ ). Persistent mesenchyme in Fbxo11Jf/+ bullae has limited mesenchymal cell condensation, fibrosis and hyperplasia of the mesenchyme-associated epithelium. Subsequent modification forms fibrous adhesions that link the mucosa and the tympanic membrane, and this is accompanied by dystrophic mineralization and accumulation of serous effusion in the bulla cavity. Mouse models of bulla cavitation defects are important because their study in humans is limited to post-mortem samples. This work indicates new diagnostic criteria for this otitis media aetiology in humans, and the prospects of studying the molecular mechanisms of murine bulla cavitation in organ culture.


Assuntos
Orelha Média/metabolismo , Orelha Média/patologia , Proteínas F-Box/metabolismo , Otite Média/patologia , Animais , Animais Recém-Nascidos , Doença Crônica , Modelos Animais de Doenças , Orelha Média/embriologia , Orelha Média/ultraestrutura , Epitélio/embriologia , Epitélio/ultraestrutura , Feminino , Proteína do Locus do Complexo MDS1 e EVI1/metabolismo , Masculino , Mesoderma/embriologia , Mesoderma/ultraestrutura , Camundongos Endogâmicos C57BL , Otite Média/embriologia , Proteínas Proto-Oncogênicas c-bcl-6/metabolismo , Fatores de Transcrição da Família Snail/metabolismo , Fatores de Tempo , Aderências Teciduais/patologia
17.
World Allergy Organ J ; 12(11): 100077, 2019 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-31871532

RESUMO

CLCA1 is a member of the CLCA (calcium-activated chloride channel regulator) family and plays an essential role in goblet cell mucus production from the respiratory tract epithelium. CLCA1 also regulates Ca2+-dependent Cl- transport that involves the channel protein transmembrane protein 16A (TMEM16A) and its accessary molecules. CLCA1 modulates epithelial cell chloride current and participates in the pathogenesis of mucus hypersecretory-associated respiratory and gastrointestinal diseases, including asthma, chronic obstructive pulmonary disease, cystic fibrosis, pneumonia, colon colitis, cystic fibrosis intestinal mucous disease, ulcerative colitis, and gastrointestinal parasitic infection. Most studies have been focused on the expression regulation of CLCA1 in human specimens. Limited studies used the CLCA1-deficient mice and CLCA1 blocking agents and yielded inconsistent conclusions regarding its role in these diseases. CLCA1 not only regulates mucin expression, but also participates in innate immune responses by binding to yet unidentified molecules on inflammatory cells for cytokine and chemokine production. CLCA1 also targets lymphatic endothelial cells and cancer cells by regulating lymphatic cell proliferation and lymphatic sinus growth in the lymphatic organs and controlling cancer cell differentiation, proliferation, and apoptosis, all which depend on the location of the lymphatic vessels, the type of cancers, the presence of Th2 cytokines, and possibly the availability and type of CLCA1-binding proteins. Here we summarize available studies related to these different activities of CLCA1 to assist our understanding of how this secreted modifier of calcium-activated chloride channels (CaCCs) affects mucus production and innate immunity during the pathogenesis of respiratory, gastrointestinal, and malignant diseases.

18.
Artigo em Inglês | MEDLINE | ID: mdl-29296079

RESUMO

Chronic obstructive pulmonary disease (COPD) is characterized by an abnormal inflammatory response in the lungs caused by the inhalation of noxious particles and gases. The airway epithelium has a protective function against these harmful agents by maintaining a physical barrier and by secreting defensive proteins, such as bactericidal/permeability-increasing fold-containing (BPIF) proteins, BPIFA1 and BPIFB1. However, inconsistent data regarding BPIFA1 expression in smokers and COPD patients have been reported to date. Therefore, we investigated the expression of BPIFA1 and BPIFB1 in a large cohort of never-smokers and smokers with and without COPD, both on the messenger RNA (mRNA) level in lung tissue and on the protein level in airway epithelium. Furthermore, we examined the correlation between BPIFA1 and BPIFB1 levels, goblet cell hyperplasia, and lung function measurements. BPIFA1 and BPIFB1 mRNA expressions were significantly increased in stage III-IV COPD patients compared with stage II COPD patients and subjects without COPD. In addition, protein levels in COPD patients were significantly increased in comparison with subjects without COPD. BPIFA1 and BPIFB1 levels were inversely correlated with measurements of airflow limitation and positively correlated with goblet cell hyperplasia. In addition, by the use of immunofluorescence double staining, we demonstrated the expression of BPIFB1 in goblet cells. In conclusion, we show that BPIFA1 and BPIFB1 levels are elevated in COPD patients and correlate with disease severity.


Assuntos
Autoantígenos/metabolismo , Glicoproteínas/metabolismo , Células Caliciformes/metabolismo , Pulmão/metabolismo , Fosfoproteínas/metabolismo , Proteínas/metabolismo , Doença Pulmonar Obstrutiva Crônica/metabolismo , Idoso , Autoantígenos/genética , Biomarcadores/metabolismo , Estudos de Casos e Controles , Proteínas de Ligação a Ácido Graxo , Feminino , Volume Expiratório Forçado , Glicoproteínas/genética , Células Caliciformes/patologia , Humanos , Hiperplasia , Pulmão/patologia , Pulmão/fisiopatologia , Masculino , Pessoa de Meia-Idade , Fosfoproteínas/genética , Valor Preditivo dos Testes , Proteínas/genética , Doença Pulmonar Obstrutiva Crônica/diagnóstico , Doença Pulmonar Obstrutiva Crônica/genética , Doença Pulmonar Obstrutiva Crônica/fisiopatologia , RNA Mensageiro/genética , Índice de Gravidade de Doença , Fumar/efeitos adversos , Fumar/genética , Fumar/metabolismo , Regulação para Cima , Capacidade Vital
19.
Acta Crystallogr F Struct Biol Commun ; 74(Pt 5): 268-276, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29717993

RESUMO

Bacterial permeability-increasing family member A1 (BPIFA1) is an innate immunity factor and one of the most abundantly secreted proteins in the upper airways. BPIFA1 is multifunctional, with antimicrobial, surfactant and lipopolysaccharide-binding activities, as well as established roles in lung hydration. Here, the 2.5 Šresolution crystal structure of BPIFA1 from Mus musculus (mBPIFA1) is presented and compared with those of human BPIFA1 (hBPIFA1) and structural homologs. Structural distinctions between mBPIFA1 and hBPIFA1 suggest potential differences in biological function, including the regulation of a key pulmonary ion channel.


Assuntos
Glicoproteínas/química , Glicoproteínas/genética , Imunidade Inata/genética , Fosfoproteínas/química , Fosfoproteínas/genética , Sequência de Aminoácidos , Animais , Cristalografia por Raios X/métodos , Glicoproteínas/metabolismo , Camundongos , Permeabilidade , Fosfoproteínas/metabolismo , Estrutura Secundária de Proteína , Estrutura Terciária de Proteína
20.
Dis Model Mech ; 9(11): 1405-1417, 2016 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-27660200

RESUMO

Otitis media (OM), or middle ear inflammation, is the most common paediatric disease and leads to significant morbidity. Although understanding of underlying disease mechanisms is hampered by complex pathophysiology it is clear that epithelial abnormalities underpin the disease. There is currently a lack of a well-characterised in vitro model of the middle ear (ME) epithelium that replicates the complex cellular composition of the middle ear. Here, we report the development of a novel in vitro model of mouse middle ear epithelial cells (mMECs) at an air-liquid interface (ALI) that recapitulates the characteristics of the native murine ME epithelium. We demonstrate that mMECs undergo differentiation into the varied cell populations seen within the native middle ear. Proteomic analysis confirmed that the cultures secrete a multitude of innate defence proteins from their apical surface. We showed that the mMECs supported the growth of the otopathogen, nontypeable Haemophilus influenzae (NTHi), suggesting that the model can be successfully utilised to study host-pathogen interactions in the middle ear. Overall, our mMEC culture system can help to better understand the cell biology of the middle ear and improve our understanding of the pathophysiology of OM. The model also has the potential to serve as a platform for validation of treatments designed to reverse aspects of epithelial remodelling that underpin OM development.


Assuntos
Orelha Média/anatomia & histologia , Epitélio/anatomia & histologia , Animais , Biomarcadores/metabolismo , Técnicas de Cultura de Células , Forma Celular , Células Cultivadas , Cílios/metabolismo , Células Epiteliais/metabolismo , Células Epiteliais/ultraestrutura , Epitélio/metabolismo , Epitélio/ultraestrutura , Infecções por Haemophilus/microbiologia , Infecções por Haemophilus/patologia , Haemophilus influenzae/fisiologia , Espectrometria de Massas , Camundongos Endogâmicos C57BL , Modelos Animais , Proteoma/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA