Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 112
Filtrar
1.
Int J Mol Sci ; 22(24)2021 Dec 09.
Artigo em Inglês | MEDLINE | ID: mdl-34948054

RESUMO

The brain insulin metabolism alteration has been addressed as a pathophysiological factor underlying Alzheimer's disease (AD). Insulin can be beneficial in AD, but its macro-polypeptide nature negatively influences the chances of reaching the brain. The intranasal (IN) administration of therapeutics in AD suggests improved brain-targeting. Solid lipid nanoparticles (SLNs) and poly(lactic-co-glycolic acid) nanoparticles (PLGA NPs) are promising carriers to deliver the IN-administered insulin to the brain due to the enhancement of the drug permeability, which can even be improved by chitosan-coating. In the present study, uncoated and chitosan-coated insulin-loaded SLNs and PLGA NPs were formulated and characterized. The obtained NPs showed desirable physicochemical properties supporting IN applicability. The in vitro investigations revealed increased mucoadhesion, nasal diffusion, and drug release rate of both insulin-loaded nanocarriers over native insulin with the superiority of chitosan-coated SLNs. Cell-line studies on human nasal epithelial and brain endothelial cells proved the safety IN applicability of nanoparticles. Insulin-loaded nanoparticles showed improved insulin permeability through the nasal mucosa, which was promoted by chitosan-coating. However, native insulin exceeded the blood-brain barrier (BBB) permeation compared with nanoparticulate formulations. Encapsulating insulin into chitosan-coated NPs can be beneficial for ensuring structural stability, enhancing nasal absorption, followed by sustained drug release.


Assuntos
Encéfalo/citologia , Quitosana/química , Insulina/farmacologia , Nariz/citologia , Encéfalo/metabolismo , Linhagem Celular , Liberação Controlada de Fármacos , Células Endoteliais/química , Células Endoteliais/citologia , Insulina/química , Lipossomos/química , Nanopartículas/química , Nariz/química , Tamanho da Partícula , Ácido Poliglicólico/química
2.
Toxicol Lett ; 353: 27-33, 2021 Dec 15.
Artigo em Inglês | MEDLINE | ID: mdl-34627954

RESUMO

Trimellitic anhydride (TMA) is a chemical agent classified as a low molecular weight (LMW) agent causing occupational rhinitis (OR) or asthma. Although TMA is recognized as a respiratory sensitizer, the direct and non-immunologic effects of TMA remain unclear. Air- liquid interface (ALI) cultured human nasal epithelial cells (HNECs) derived from control subjects were treated with TMA, followed by measurement of the transepithelial electrical resistance (TEER), paracellular permeability of fluorescein isothiocyanate (FITC)-dextran and immunofluorescence of tight junction proteins claudin-1 and zonula occludens-1 (ZO-1). The cytotoxicity of TMA was evaluated by lactate dehydrogenase (LDH) assay. TMA at concentrations of 2 and 4 mg/mL significantly reduced the TEER within 10 min (p = 0.0177 on 2 mg/mL; p < 0.0001 on 4 mg/mL). The paracellular permeability of FITC-dextran was significantly increased upon challenge with 4 mg/mL TMA for 3 h (p = 0.0088) and 6 h (p = 0.0004). TMA treatment induced a reduction in the fluorescence intensity of claudin-1 and ZO-1 in a dose-dependent manner. LDH assay revealed 4 mg/mL TMA induced cytotoxicity only after 6 h incubation, while 1 or 2 mg/mL TMA caused no cytotoxicity. Our results suggest that TMA has a potential to penetrate the epithelial barrier by disrupting claudin-1 and ZO-1, indicating an important role for sensitization and OR development.


Assuntos
Células Epiteliais/efeitos dos fármacos , Anidridos Ftálicos/toxicidade , Adulto , Sobrevivência Celular/efeitos dos fármacos , Claudina-1/genética , Claudina-1/metabolismo , Dextranos , Relação Dose-Resposta a Droga , Células Epiteliais/metabolismo , Feminino , Fluoresceína-5-Isotiocianato/análogos & derivados , Técnica Direta de Fluorescência para Anticorpo , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Pessoa de Meia-Idade , Nariz/citologia , Permeabilidade , Anidridos Ftálicos/administração & dosagem , Proteína da Zônula de Oclusão-1/genética , Proteína da Zônula de Oclusão-1/metabolismo
3.
Int J Mol Sci ; 22(19)2021 Sep 25.
Artigo em Inglês | MEDLINE | ID: mdl-34638663

RESUMO

Decellularization has emerged as a potential solution for tracheal replacement. As a fully decellularized graft failed to achieve its purposes, the de-epithelialization partial decellularization protocol appeared to be a promising approach for fabricating scaffolds with preserved mechanical properties and few immune rejection responses after transplantation. Nevertheless, a lack of appropriate concurrent epithelialization treatment can lead to luminal stenosis of the transplant and impede its eventual success. To improve re-epithelialization, autologous nasal epithelial cell sheets generated by our cell sheet engineering platform were utilized in this study under an in vivo rabbit model. The newly created cell sheets have an intact and transplantable appearance, with their specific characteristics of airway epithelial origin being highly expressed upon histological and immunohistochemical analysis. Subsequently, those cell sheets were incorporated with a partially decellularized tracheal graft for autograft transplantation under tracheal partial resection models. The preliminary results two months post operation demonstrated that the transplanted patches appeared to be wholly integrated into the host trachea with adequate healing of the luminal surface, which was confirmed via endoscopic and histologic evaluations. The satisfactory result of this hybrid scaffold protocol could serve as a potential solution for tracheal reconstructions in the future.


Assuntos
Células Epiteliais/citologia , Nariz/citologia , Traqueia/citologia , Animais , Estudos Transversais , Matriz Extracelular/fisiologia , Masculino , Coelhos , Procedimentos de Cirurgia Plástica/métodos , Engenharia Tecidual/métodos , Alicerces Teciduais/química , Transplante Autólogo/métodos
4.
mSphere ; 6(3)2021 05 12.
Artigo em Inglês | MEDLINE | ID: mdl-33980679

RESUMO

Human respiratory syncytial virus (HRSV) is the leading cause of bronchiolitis in infants. Two subgroups of HRSV (A and B) routinely cocirculate. Most research has been performed with HRSV-A strains because these are easier to culture than HRSV-B strains. In this study, we aimed to compare the replicative fitness and HRSV-induced innate cytokine responses of HRSV-A and HRSV-B strains in disease-relevant cell culture models. We used two recombinant (r) clinical isolate-based HRSV strains (A11 and B05) and one recombinant laboratory-adapted HRSV strain (A2) to infect commercially available nasal, bronchial, and small-airway cultures. Epithelial cells from all anatomical locations were susceptible to HRSV infection despite the induction of a dominant type III interferon response. Subgroup A viruses disseminated and replicated faster than the subgroup B virus. Additionally, we studied HRSV infection and innate responses in airway organoids (AOs) cultured at air-liquid interface (ALI). Results were similar to the commercially obtained bronchial cells. In summary, we show that HRSV replicates well in cells from both the upper and the lower airways, with a slight replicative advantage for subgroup A viruses. Lastly, we showed that AOs cultured at ALI are a valuable model for studying HRSV ex vivo and that they can be used in the future to study factors that influence HRSV disease severity.IMPORTANCE Human respiratory syncytial virus (HRSV) is the major cause of bronchiolitis and pneumonia in young infants and causes almost 200,000 deaths per year. Currently, there is no vaccine or treatment available, only a prophylactic monoclonal antibody (palivizumab). An important question in HRSV pathogenesis research is why only a fraction (1 to 3%) of infants develop severe disease. Model systems comprising disease-relevant HRSV isolates and accurate and reproducible cell culture models are indispensable to study infection, replication, and innate immune responses. Here, we used differentiated AOs cultured at ALI to model the human airways. Subgroup A viruses replicated better than subgroup B viruses, which we speculate fits with epidemiological findings that subgroup A viruses cause more severe disease in infants. By using AOs cultured at ALI, we present a highly relevant, robust, and reproducible model that allows for future studies into what drives severe HRSV disease.


Assuntos
Brônquios/virologia , Nariz/virologia , Organoides/microbiologia , Vírus Sincicial Respiratório Humano/genética , Vírus Sincicial Respiratório Humano/fisiologia , Replicação Viral , Brônquios/citologia , Citocinas/análise , Citocinas/imunologia , Células Epiteliais/virologia , Humanos , Imunidade Inata , Técnicas In Vitro , Cinética , Nariz/citologia , Vírus Sincicial Respiratório Humano/classificação , Vírus Sincicial Respiratório Humano/imunologia
5.
mSphere ; 6(2)2021 03 17.
Artigo em Inglês | MEDLINE | ID: mdl-33731473

RESUMO

Staphylococcus aureus is an opportunistic pathogen that colonizes the anterior nares of 30 to 50% of the population. Colonization is most often asymptomatic; however, self-inoculation can give rise to potentially fatal infections of the deeper tissues and blood. Like all bacteria, S. aureus can sense and respond to environmental cues and modify gene expression to adapt to specific environmental conditions. The transition of S. aureus from the nares to the deeper tissues and blood is accompanied by changes in environmental conditions, such as nutrient availability, pH, and temperature. In this study, we perform transcriptomics and proteomics on S. aureus cultures growing at three physiologically relevant temperatures, 34°C (nares), 37°C (body), and 40°C (pyrexia), to determine if small scale, biologically meaningful alterations in temperature impact S. aureus gene expression. Results show that small but definite temperature changes elicit a large-scale restructuring of the S. aureus transcriptome and proteome in a manner that, most often, inversely correlates with increasing temperature. We also provide evidence that a large majority of these changes are modulated at the posttranscriptional level, possibly by sRNA regulatory elements. Phenotypic analyses were also performed to demonstrate that these changes have physiological relevance. Finally, we investigate the impact of temperature-dependent alterations in gene expression on S. aureus pathogenesis and demonstrate decreased intracellular invasion of S. aureus grown at 34°C. Collectively, our results demonstrate that small but biologically meaningful alterations in temperature influence S. aureus gene expression, a process that is likely a major contributor to the transition from a commensal to pathogen.IMPORTANCE Enteric bacterial pathogens, like Escherichia coli, are known to experience large temperature differences as they are transmitted through the fecal oral route. This change in temperature has been demonstrated to influence bacterial gene expression and facilitate infection. Staphylococcus aureus is a human-associated pathogen that can live as a commensal on the skin and nares or cause invasive infections of the deeper tissues and blood. Factors influencing S. aureus nasal colonization are not fully understood; however, individuals colonized with S. aureus are at increased risk of invasive infections through self-inoculation. The transition of S. aureus from the nose (colonization) to the body (infection) is accompanied by a modest but definite temperature increase, from 34°C to 37°C. In this study, we investigate whether these host-associated small temperature changes can influence S. aureus gene expression. Results show widespread changes in the bacterial transcriptome and proteome at three physiologically relevant temperatures (34°C, 37°C, and 40°C).


Assuntos
Proteínas de Bactérias/análise , Regulação Bacteriana da Expressão Gênica , Proteoma , Staphylococcus aureus/genética , Temperatura , Transcriptoma , Células Cultivadas , Células Epiteliais/microbiologia , Humanos , Nariz/citologia , Infecções Estafilocócicas/microbiologia , Staphylococcus aureus/química , Staphylococcus aureus/metabolismo , Fatores de Virulência/genética
6.
J Med Virol ; 93(6): 3484-3495, 2021 06.
Artigo em Inglês | MEDLINE | ID: mdl-33247612

RESUMO

The effects of the clinically used protease inhibitor nafamostat on influenza virus replication have not been well studied. Primary human tracheal (HTE) and nasal (HNE) epithelial cells were pretreated with nafamostat and infected with the 2009 pandemic [A/Sendai-H/108/2009/(H1N1) pdm09] or seasonal [A/New York/55/2004(H3N2)] influenza virus. Pretreatment with nafamostat reduced the titers of the pandemic and seasonal influenza viruses and the secretion of inflammatory cytokines, including interleukin-6 and tumor necrosis factor-α, in the supernatants of the cells infected with the pandemic influenza virus. HTE and HNE cells exhibited mRNA and/or protein expression of transmembrane protease serine 2 (TMPRSS2), TMPRSS4, and TMPRSS11D. Pretreatment with nafamostat reduced cleavage of the precursor protein HA0 of the pandemic influenza virus into subunit HA1 in HTE cells and reduced the number of acidic endosomes in HTE and HNE cells where influenza virus RNA enters the cytoplasm. Additionally, nafamostat (30 mg/kg/day, intraperitoneal administration) reduced the levels of the pandemic influenza virus [A/Hyogo/YS/2011 (H1N1) pdm09] in mouse lung washes. These findings suggest that nafamostat may inhibit influenza virus replication in human airway epithelial cells and mouse lungs and reduce infection-induced airway inflammation by modulating cytokine production.


Assuntos
Benzamidinas/farmacologia , Benzamidinas/uso terapêutico , Células Epiteliais/efeitos dos fármacos , Guanidinas/farmacologia , Guanidinas/uso terapêutico , Pulmão/efeitos dos fármacos , Inibidores de Serina Proteinase/farmacologia , Inibidores de Serina Proteinase/uso terapêutico , Replicação Viral/efeitos dos fármacos , Animais , Células Cultivadas , Citocinas/análise , Citocinas/imunologia , Células Epiteliais/virologia , Feminino , Humanos , Pulmão/virologia , Masculino , Camundongos , Camundongos Endogâmicos BALB C , Nariz/citologia , Traqueia/citologia
7.
Biomolecules ; 10(8)2020 08 14.
Artigo em Inglês | MEDLINE | ID: mdl-32823934

RESUMO

Background: Cilia are actin based cellular protrusions conserved from algae to complex multicellular organisms like Homo sapiens. Respiratory motile cilia line epithelial cells of the tracheobronchial tree, beat in a synchronous, metachronal wave, moving inhaled pollutants and pathogens cephalad. Their role in both congenital disorders like primary ciliary dyskinesia (PCD) to acquired disorders like chronic obstructive pulmonary disease (COPD) continues to evolve. In this current body of work we outline a protocol optimized to reciliate human nasal epithelial cells and mouse tracheal cells in vitro. Using this protocol, we knocked down known cilia genes, as well as use a small molecule inhibitor of Notch, N-[N-(3,5-Difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl Ester (DAPT), to assess the effect of these on ciliogenesis in order to show the validity of our protocol. Methods: Tracheas were harvested from wild-type, adult C57B6 mice, pronase digested and sloughed off epithelial cells grown to confluence in stationary culture on rat-tail collagen coated wells. Upon reaching confluence, collagen was digested and cells placed suspension culture protocol to reciliate the cells. Using this suspension culture protocol, we employed siRNA gene knockdown to assay gene functions required for airway ciliogenesis. Knock down of Dynein axonemal heavy chain 5 (Dnah5), a ciliary structural protein, was confirmed using immunostaining. Mouse tracheal cells were treated in suspension with varying doses of DAPT, an inhibitor of Notch, with the purpose of evaluating its effect and dose response on ciliogenesis. The optimum dose was then used on reciliating human nasal epithelial cells. Results: siRNA knockdown of Foxj1 prevented ciliation, consistent with its role as a master regulator of motile cilia. Knockdown of Dnai1 and Dnah5 resulted in immotile cilia, and Cand1 knockdown, a centrosome protein known to regulate centrosome amplification, inhibited airway ciliogenesis. Dnah5 knockdown was confirmed with significantly decreased immunostaining of cilia for this protein. Inhibiting Notch signaling by inhibiting gamma secretase with DAPT enhanced the percentage of ciliation, and resulted in longer cilia that beat with higher frequency in both mouse and human airway epithelia. Conclusions: Modifying existing reciliation protocols to suit both human nasal epithelial and mouse tracheal tissue, we have shown that knockdown of known cilia-related genes have the expected effects. Additionally, we have demonstrated the optimal dosage for significantly improving reciliation of airway epithelia using DAPT. Given that cilia length and function are significantly compromised in COPD, these findings open up interesting avenues for further exploration.


Assuntos
Cílios/metabolismo , Dipeptídeos/farmacologia , Nariz/citologia , Traqueia/citologia , Animais , Dineínas do Axonema/genética , Técnicas de Cultura de Células , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Cílios/efeitos dos fármacos , Cílios/genética , Relação Dose-Resposta a Droga , Células Epiteliais/citologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Fatores de Transcrição Forkhead/genética , Técnicas de Silenciamento de Genes , Humanos , Camundongos , Camundongos Endogâmicos C57BL , Nariz/efeitos dos fármacos , Traqueia/efeitos dos fármacos , Traqueia/metabolismo , Fatores de Transcrição/genética
8.
mBio ; 11(3)2020 06 02.
Artigo em Inglês | MEDLINE | ID: mdl-32487760

RESUMO

Mucormycosis, caused by Rhizopus species, is a life-threatening fungal infection that occurs in patients immunocompromised by diabetic ketoacidosis (DKA), cytotoxic chemotherapy, immunosuppressive therapy, hematologic malignancies, or severe trauma. Inhaled Rhizopus spores cause pulmonary infections in patients with hematologic malignancies, while patients with DKA are much more prone to rhinoorbital/cerebral mucormycosis. Here, we show that Rhizopus delemar interacts with glucose-regulated protein 78 (GRP78) on nasal epithelial cells via its spore coat protein CotH3 to invade and damage the nasal epithelial cells. Expression of the two proteins is significantly enhanced by high glucose, iron, and ketone body levels (hallmark features of DKA), potentially leading to frequently lethal rhinoorbital/cerebral mucormycosis. In contrast, R. delemar CotH7 recognizes integrin ß1 as a receptor on alveolar epithelial cells, causing the activation of epidermal growth factor receptor (EGFR) and leading to host cell invasion. Anti-integrin ß1 antibodies inhibit R. delemar invasion of alveolar epithelial cells and protect mice from pulmonary mucormycosis. Our results show that R. delemar interacts with different mammalian receptors depending on the host cell type. Susceptibility of patients with DKA primarily to rhinoorbital/cerebral disease can be explained by host factors typically present in DKA and known to upregulate CotH3 and nasal GRP78, thereby trapping the fungal cells within the rhinoorbital milieu, leading to subsequent invasion and damage. Our studies highlight that mucormycosis pathogenesis can potentially be overcome by the development of novel customized therapies targeting niche-specific host receptors or their respective fungal ligands.IMPORTANCE Mucormycosis caused by Rhizopus species is a fungal infection with often fatal prognosis. Inhalation of spores is the major route of entry, with nasal and alveolar epithelial cells among the first cells that encounter the fungi. In patients with hematologic malignancies or those undergoing cytotoxic chemotherapy, Rhizopus causes pulmonary infections. On the other hand, DKA patients predominantly suffer from rhinoorbital/cerebral mucormycosis. The reason for such disparity in disease types by the same fungus is not known. Here, we show that the unique susceptibility of DKA subjects to rhinoorbital/cerebral mucormycosis is likely due to specific interaction between nasal epithelial cell GRP78 and fungal CotH3, the expression of which increases in the presence of host factors present in DKA. In contrast, pulmonary mucormycosis is initiated via interaction of inhaled spores expressing CotH7 with integrin ß1 receptor, which activates EGFR to induce fungal invasion of host cells. These results introduce a plausible explanation for disparate disease manifestations in DKA versus those in hematologic malignancy patients and provide a foundation for development of therapeutic interventions against these lethal forms of mucormycosis.


Assuntos
Células Epiteliais/microbiologia , Proteínas de Choque Térmico/genética , Interações Hospedeiro-Patógeno , Infecções Fúngicas Invasivas/microbiologia , Mucormicose/microbiologia , Receptores de Vitronectina/genética , Rhizopus/patogenicidade , Células A549 , Células Epiteliais Alveolares/microbiologia , Células Epiteliais Alveolares/patologia , Animais , Linhagem Celular , Cetoacidose Diabética/complicações , Cetoacidose Diabética/microbiologia , Chaperona BiP do Retículo Endoplasmático , Células Epiteliais/patologia , Receptores ErbB/genética , Feminino , Humanos , Masculino , Camundongos , Camundongos Endogâmicos ICR , Nariz/citologia , Virulência
9.
Am J Physiol Lung Cell Mol Physiol ; 318(6): L1158-L1164, 2020 06 01.
Artigo em Inglês | MEDLINE | ID: mdl-32267720

RESUMO

Shifts in cellular metabolic phenotypes have the potential to cause disease-driving processes in respiratory disease. The respiratory epithelium is particularly susceptible to metabolic shifts in disease, but our understanding of these processes is limited by the incompatibility of the technology required to measure metabolism in real-time with the cell culture platforms used to generate differentiated respiratory epithelial cell types. Thus, to date, our understanding of respiratory epithelial metabolism has been restricted to that of basal epithelial cells in submerged culture, or via indirect end point metabolomics readouts in lung tissue. Here we present a novel methodology using the widely available Seahorse Analyzer platform to monitor real-time changes in the cellular metabolism of fully differentiated primary human airway epithelial cells grown at air-liquid interface (ALI). We show increased glycolytic, but not mitochondrial, ATP production rates in response to physiologically relevant increases in glucose availability. We also show that pharmacological inhibition of lactate dehydrogenase is able to reduce glucose-induced shifts toward aerobic glycolysis. This method is timely given the recent advances in our understanding of new respiratory epithelial subtypes that can only be observed in vitro through culture at ALI and will open new avenues to measure real-time metabolic changes in healthy and diseased respiratory epithelium, and in turn the potential for the development of novel therapeutics targeting metabolic-driven disease phenotypes.


Assuntos
Ar , Diferenciação Celular , Sistemas Computacionais , Metabolismo Energético , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Nariz/citologia , Ácidos/metabolismo , Diferenciação Celular/efeitos dos fármacos , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Glucose/farmacologia , Humanos , L-Lactato Desidrogenase/antagonistas & inibidores , L-Lactato Desidrogenase/metabolismo , Metabolômica
10.
PLoS One ; 15(3): e0228229, 2020.
Artigo em Inglês | MEDLINE | ID: mdl-32214336

RESUMO

The culture of differentiated human airway epithelial cells allows the study of pathogen-host interactions and innate immune responses in a physiologically relevant in vitro model. As the use of primary cell culture has gained popularity the availability of the reagents needed to generate these cultures has increased. In this study we assessed two different media, Promocell and PneumaCult, during the differentiation and maintenance of well-differentiated primary nasal epithelial cell cultures (WD-PNECs). We compared and contrasted the consequences of these media on WD-PNEC morphological and physiological characteristics and their responses to respiratory syncytial virus (RSV) infection. We found that cultures generated using PneumaCult resulted in greater total numbers of smaller, tightly packed, pseudostratified cells. However, cultures from both media resulted in similar proportions of ciliated and goblet cells. There were no differences in RSV growth kinetics, although more ciliated cells were infected in the PneumaCult cultures. There was also significantly more IL-29/IFNλ1 secreted from PneumaCult compared to Promocell cultures following infection. In conclusion, the type of medium used for the differentiation of primary human airway epithelial cells may impact experimental results.


Assuntos
Diferenciação Celular , Meios de Cultura/química , Células Epiteliais/citologia , Células Epiteliais/virologia , Nariz/citologia , Cultura Primária de Células/métodos , Vírus Sinciciais Respiratórios/fisiologia , Linhagem Celular , Criança , Células Caliciformes/citologia , Humanos
11.
Mol Pharmacol ; 96(4): 515-525, 2019 10.
Artigo em Inglês | MEDLINE | ID: mdl-31427400

RESUMO

ORKAMBI, a combination of the corrector, lumacaftor, and the potentiator, ivacaftor, partially rescues the defective processing and anion channel activity conferred by the major cystic fibrosis-causing mutation, F508del, in in vitro studies. Clinically, the improvement in lung function after ORKAMBI treatment is modest and variable, prompting the search for complementary interventions. As our previous work identified a positive effect of arginine-dependent nitric oxide signaling on residual F508del-Cftr function in murine intestinal epithelium, we were prompted to determine whether strategies aimed at increasing arginine would enhance F508del-cystic fibrosis transmembrane conductance regulator (CFTR) channel activity in patient-derived airway epithelia. Now, we show that the addition of arginine together with inhibition of intracellular arginase activity increased cytosolic nitric oxide and enhanced the rescue effect of ORKAMBI on F508del-CFTR-mediated chloride conductance at the cell surface of patient-derived bronchial and nasal epithelial cultures. Interestingly, arginine addition plus arginase inhibition also enhanced ORKAMBI-mediated increases in ciliary beat frequency and mucociliary movement, two in vitro CF phenotypes that are downstream of the channel defect. This work suggests that strategies to manipulate the arginine-nitric oxide pathway in combination with CFTR modulators may lead to improved clinical outcomes. SIGNIFICANCE STATEMENT: These proof-of-concept studies highlight the potential to boost the response to cystic fibrosis (CF) transmembrane conductance regulator (CFTR) modulators, lumacaftor and ivacaftor, in patient-derived airway tissues expressing the major CF-causing mutant, F508del-CFTR, by enhancing other regulatory pathways. In this case, we observed enhancement of pharmacologically rescued F508del-CFTR by arginine-dependent, nitric oxide signaling through inhibition of endogenous arginase activity.


Assuntos
Aminofenóis/farmacologia , Aminopiridinas/farmacologia , Arginase/antagonistas & inibidores , Arginina/metabolismo , Benzodioxóis/farmacologia , Fibrose Cística/metabolismo , Óxido Nítrico/metabolismo , Quinolonas/farmacologia , Animais , Brônquios/citologia , Brônquios/efeitos dos fármacos , Brônquios/metabolismo , Células Cultivadas , Fibrose Cística/tratamento farmacológico , Fibrose Cística/genética , Regulador de Condutância Transmembrana em Fibrose Cística/genética , Citosol/metabolismo , Combinação de Medicamentos , Humanos , Mucosa Intestinal/metabolismo , Camundongos , Mutação , Nariz/citologia , Nariz/efeitos dos fármacos
12.
Indian J Pathol Microbiol ; 62(3): 451-453, 2019.
Artigo em Inglês | MEDLINE | ID: mdl-31361238

RESUMO

Rhabdomyosarcoma (RMS) is a tumor arising from primitive mesenchymal cell with tendency for myogenesis. WHO classification categorizes this entity as embryonal, alveolar, spindle cell/sclerosing, and pleomorphic subtypes removing botryoid as a separate entity. The alveolar variant has worse prognosis and the cytological features of this entity are similar to embryonal type with little variations. This case report describes the cytohistological features of alveolar RMSfrom a 9-year-old child with nonparameningeal location.


Assuntos
Nariz/patologia , Rabdomiossarcoma Alveolar/diagnóstico , Biópsia , Biópsia por Agulha Fina , Criança , Face/patologia , Técnicas Histológicas , Humanos , Masculino , Nariz/citologia , Rabdomiossarcoma Alveolar/terapia
13.
Clin Exp Pharmacol Physiol ; 46(9): 821-827, 2019 09.
Artigo em Inglês | MEDLINE | ID: mdl-31211861

RESUMO

Allergic rhinitis (AR) is a type of respiratory disease closely associated with chronic inflammation. Esculetin is a natural coumarin derivative and has been reported to possess anti-allergic and anti-inflammatory effects. However, the roles of esculetin in AR have not been studied. In this study, we aimed to examine the effect of esculetin on AR using an in vitro model. The human nasal epithelial cells (HNEpC) were stimulated by histamine for 24 hours with or without the pretreatment of esculetin. The mRNA levels and production of inflammatory cytokines including IL-6 and IL-8, as well as mucin 5AC (MUC5AC) were measured using qRT-PCR and ELISA, respectively. The results showed that esculetin suppressed histamine-induced expression and secretion of IL-6, IL-8, and MUC5AC in HNEpCs. Furthermore, we examined the effect of esculetin on NF-κB pathway by detecting the expression levels of NF-κB p65, p-p65 and IκBα using western blot analysis. Esculetin treatment suppressed the histamine-induced p-p65 expression and p-IκBα degradation. Inhibiting NF-κB pathway suppressed histamine-induced production of IL-6, IL-8, and MUC5AC in HNEpCs. These findings suggested that esculetin suppressed histamine-induced production of inflammatory cytokines and mucin in HNEpCs, which were partly mediated by the inhibition of NF-κB pathway.


Assuntos
Citocinas/metabolismo , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Regulação da Expressão Gênica/efeitos dos fármacos , Histamina/farmacologia , Nariz/citologia , Umbeliferonas/farmacologia , Linhagem Celular , Humanos , Inflamação/metabolismo , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Mucina-5AC/metabolismo , NF-kappa B/metabolismo
14.
Med Eng Phys ; 68: 85-93, 2019 06.
Artigo em Inglês | MEDLINE | ID: mdl-31005567

RESUMO

The ideal simulator for Endoscopic Sinus and Skull Base Surgery (ESSS) training must be supported by a physical model and provide repetitive behavior in a controlled environment. Development of realistic tissue models is a key part of ESSS virtual reality (VR)-based surgical simulation. Considerable research has been conducted to address haptic or force feedback and propose a phenomenological tissue fracture model for sino-nasal tissue during surgical tool indentation. Mechanical properties of specific sino-nasal regions of the sheep head have been studied in various indentation and relaxation experiments. Tool insertion at different indentation rates into coronal orbital floor (COF) tissue is modeled as a sequence of three events: deformation, fracture, and cutting. The behavior in the deformation phase can be characterized using a non-linear, rate-dependent modified Kelvin-Voigt model. A non-linear model for tissue behavior prior to the fracture point is presented. The overall model shows a non-positive dependency of maximum force on tool indentation rate, which indicates faster tool insertion velocity decreases the maximum final fracture force. The tissue cutting phase has been modeled to characterize the force necessary to slice through the COF. The proposed model in this study can help develop VR-based ESSS base simulators in otolaryngology and ophthalmology surgeries. Such simulators are useful in preoperative planning, accurate surgical simulation, intelligent robotic assistance, and treatment applications.


Assuntos
Endoscopia/efeitos adversos , Fenômenos Mecânicos , Modelos Biológicos , Nariz/cirurgia , Base do Crânio/cirurgia , Animais , Fenômenos Biomecânicos , Nariz/citologia , Ovinos , Base do Crânio/citologia
15.
Int J Mol Sci ; 20(6)2019 Mar 19.
Artigo em Inglês | MEDLINE | ID: mdl-30893823

RESUMO

Monoclonal antibodies (mAbs) are promising therapies to treat airway chronic inflammatory disease (asthma or nasal polyps). To date, no study has specifically assessed, in vitro, the potential function of neonatal Fc receptor (FcRn) in IgG transcytosis through the human nasal airway epithelium. The objective of this study was to report the in vitro expression and function of FcRn in nasal human epithelium. FcRn expression was studied in an air⁻liquid interface (ALI) primary culture model of human nasal epithelial cells (HNEC) from polyps. FcRn expression was characterized by quantitative RT-PCR, western blot, and immunolabeling. The ability of HNECs to support mAb transcytosis via FcRn was assessed by transcytosis assay. This study demonstrates the expression of FcRn mRNA and protein in HNEC. We report a high expression of FcRn in the cytosol of ciliated, mucus, and basal cells by immunohistochemistry with a higher level of FcRn proteins in differentiated HNEC. We also proved in vitro transepithelial delivery of an IgG1 therapeutic mAb with a dose⁻response curve. This is the first time that FcRn expression and mAb transcytosis has been shown in a model of human nasal respiratory epithelium in vitro. This study is a prerequisite for FcRn-dependent nasal administration of mAbs.


Assuntos
Anticorpos Monoclonais/metabolismo , Sistemas de Liberação de Medicamentos , Células Epiteliais/metabolismo , Antígenos de Histocompatibilidade Classe I/metabolismo , Nariz/citologia , Receptores Fc/metabolismo , Transcitose , Diferenciação Celular , Células HEK293 , Humanos , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
17.
Int J Mol Sci ; 19(12)2018 Nov 26.
Artigo em Inglês | MEDLINE | ID: mdl-30486295

RESUMO

The effects of the isoflavone daidzein on the ciliary beat distance (CBD, which is a parameter assessing the amplitude of ciliary beating) and the ciliary beat frequency (CBF) were examined in ciliated human nasal epithelial cells (cHNECs) in primary culture. Daidzein decreased [Cl-]i and enhanced CBD in cHNECs. The CBD increase that was stimulated by daidzein was mimicked by Cl--free NO3- solution and bumetanide (an inhibitor of Na⁺/K⁺/2Cl- cotransport), both of which decreased [Cl-]i. Moreover, the CBD increase was inhibited by 5-Nitro-2-(3-phenylpropylamino)benzoic acid (NPPB, a Cl- channel blocker), which increased [Cl-]i. CBF was also decreased by NPPB. The rate of [Cl-]i decrease evoked by Cl--free NO3- solution was enhanced by daidzein. These results suggest that daidzein activates Cl- channels in cHNECs. Moreover, daidzein enhanced the microbead transport driven by beating cilia in the cell sheet of cHNECs, suggesting that an increase in CBD enhances ciliary transport. An [Cl-]i decrease enhanced CBD, but not CBF, in cHNECs at 37 °C, although it enhanced both at 25 °C. Intracellular Cl- affects both CBD and CBF in a temperature-dependent manner. In conclusion, daidzein, which activates Cl- channels to decrease [Cl-]i, stimulated CBD increase in cHNECs at 37 °C. CBD is a crucial factor that can increase ciliary transport in the airways under physiological conditions.


Assuntos
Cloretos/metabolismo , Cílios/metabolismo , Células Epiteliais/metabolismo , Isoflavonas/farmacologia , Nariz/citologia , Bumetanida/farmacologia , Cálcio/farmacologia , Células Cultivadas , Cílios/efeitos dos fármacos , AMP Cíclico/farmacologia , Células Epiteliais/efeitos dos fármacos , Humanos , Látex/química , Microesferas , Movimento
18.
Epigenomics ; 10(8): 1131-1145, 2018 08.
Artigo em Inglês | MEDLINE | ID: mdl-30052057

RESUMO

AIM: To assess whether DNA methylation levels account for the noninherited phenotypic variations observed among cystic fibrosis (CF) patients. PATIENTS & METHODS: Using the 450 K BeadChip, we profiled DNA methylation in nasal epithelial cells collected from 32 CF patients and 16 controls. RESULTS: We detected substantial DNA methylation differences up to 55% (median ß change 0.13; IQR: 0.15-0.11) between CF patients and controls. DNA methylation levels differed between mild and severe CF patients and correlated with lung function at 50 CpG sites. CONCLUSION: In CF samples, dynamic changes of DNA methylation occurred in genes responsible for the integrity of the epithelium and the inflammatory and immune responses, were prominent in transcriptionally active genomic regions and were over-represented in enhancers active in lung tissues. ( Clinicaltrials.gov NCT02884622).


Assuntos
Fibrose Cística/genética , Metilação de DNA , Adulto , Ilhas de CpG , Células Epiteliais/metabolismo , Feminino , Expressão Gênica , Humanos , Masculino , Nariz/citologia
19.
Sci Rep ; 8(1): 9688, 2018 06 26.
Artigo em Inglês | MEDLINE | ID: mdl-29946071

RESUMO

The respiratory epithelium is a barrier against pathogens and allergens and a target for therapy in respiratory allergy, asthma and chronic obstructive pulmonary disease (COPD). We investigated barrier-damaging factors and protective factors by real-time measurement of respiratory cell barrier integrity. Barrier integrity to cigarette smoke extract (CSE), house dust mite (HDM) extract, interferon-γ (IFN-γ) or human rhinovirus (HRV) infection alone or in combination was assessed. Corticosteroids, lipopolysaccharide (LPS), and nasal mucus proteins were tested for their ability to prevent loss of barrier integrity. Real-time impedance-based measurement revealed different patterns of CSE-, HDM-, IFN-γ- and HRV-induced damage. When per se non-damaging concentrations of harmful factors were combined, a synergetic effect was observed only for CSE and HDM. Betamethasone prevented the damaging effect of HRV and CSE, but not damage caused by HDM or IFN-γ. Real-time impedance-based measurement of respiratory epithelial barrier function is useful to study factors, which are harmful or protective. The identification of a synergetic damaging effect of CSE and HDM as well as the finding that Betamethasone protects against HRV- and CSE-induced damage may be important for asthma and COPD.


Assuntos
Betametasona/farmacologia , Mucosa Respiratória/citologia , Rhinovirus/patogenicidade , Fumaça/efeitos adversos , Adulto , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Feminino , Humanos , Interferon gama/metabolismo , Masculino , Pessoa de Meia-Idade , Nariz/citologia , Rhinovirus/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Adulto Jovem
20.
Biomaterials ; 171: 219-229, 2018 07.
Artigo em Inglês | MEDLINE | ID: mdl-29705655

RESUMO

Despite the various reported approaches to generate osteochondral composites by combination of different cell types and materials, engineering of templates with the capacity to autonomously and orderly develop into cartilage-bone bi-layered structures remains an open challenge. Here, we hypothesized that the embedding of cells inducible to endochondral ossification (i.e. bone marrow derived mesenchymal stromal cells, BMSCs) and of cells capable of robust and stable chondrogenesis (i.e. nasal chondrocytes, NCs) adjacent to each other in bi-layered hydrogels would develop directly in vivo into osteochondral tissues. Poly(ethylene glycol) (PEG) hydrogels were functionalized with TGFß3 or BMP-2, enzymatically polymerized encapsulating human BMSCs, combined with a hydrogel layer containing human NCs and ectopically implanted in nude mice without pre-culture. The BMSC-loaded layers reproducibly underwent endochondral ossification and generated ossicles containing bone and marrow. The NC-loaded layers formed cartilage tissues, which (under the influence of BMP-2 but not of TGFß3 from the neighbouring layer) remained phenotypically stable. The proposed strategy, resulting in orderly connected osteochondral composites, should be further assessed for the repair of osteoarticular defects and will be useful to model developmental processes leading to cartilage-bone interfaces.


Assuntos
Hidrogéis/farmacologia , Osteogênese/efeitos dos fármacos , Engenharia Tecidual/métodos , Adulto , Proteína Morfogenética Óssea 2/farmacologia , Condrócitos/efeitos dos fármacos , Condrócitos/metabolismo , Condrogênese/efeitos dos fármacos , Feminino , Humanos , Cartilagem Hialina/efeitos dos fármacos , Cartilagem Hialina/fisiologia , Masculino , Células-Tronco Mesenquimais/citologia , Células-Tronco Mesenquimais/efeitos dos fármacos , Células-Tronco Mesenquimais/metabolismo , Pessoa de Meia-Idade , Nariz/citologia , Polietilenoglicóis/farmacologia , Implantação de Prótese , Fator de Crescimento Transformador beta3/farmacologia
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA