Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 41
Filtrar
1.
PLoS Pathog ; 17(4): e1009529, 2021 04.
Artigo em Inglês | MEDLINE | ID: mdl-33909707

RESUMO

The human respiratory syncytial virus (RSV) is a major cause of severe lower respiratory tract infections in infants, possibly due to the properties of the immature neonatal pulmonary immune system. Using the newborn lamb, a classical model of human lung development and a translational model of RSV infection, we aimed to explore the role of cell-mediated immunity in RSV disease during early life. Remarkably, in healthy conditions, the developing T cell compartment of the neonatal lung showed major differences to that seen in the mature adult lung. The most striking observation being a high baseline frequency of bronchoalveolar IL-4-producing CD4+ and CD8+ T cells, which declined progressively over developmental age. RSV infection exacerbated this pro-type 2 environment in the bronchoalveolar space, rather than inducing a type 2 response per se. Moreover, regulatory T cell suppressive functions occurred very early to dampen this pro-type 2 environment, rather than shutting them down afterwards, while γδ T cells dropped and failed to produce IL-17. Importantly, RSV disease severity was related to the magnitude of those unconventional bronchoalveolar T cell responses. These findings provide novel insights in the mechanisms of RSV immunopathogenesis in early life, and constitute a major step for the understanding of RSV disease severity.


Assuntos
Pulmão/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Infecções Respiratórias/imunologia , Linfócitos T/patologia , Animais , Animais Recém-Nascidos , Diferenciação Celular/imunologia , Células Cultivadas , Pré-Escolar , Modelos Animais de Doenças , Progressão da Doença , Humanos , Pulmão/crescimento & desenvolvimento , Pulmão/patologia , Pulmão/virologia , Infecções por Vírus Respiratório Sincicial/congênito , Infecções por Vírus Respiratório Sincicial/patologia , Infecções Respiratórias/congênito , Infecções Respiratórias/patologia , Ovinos/crescimento & desenvolvimento , Ovinos/imunologia , Linfócitos T/imunologia , Linfócitos T/fisiologia
2.
PLoS Pathog ; 17(7): e1009789, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-34320038

RESUMO

Lung-resident (LR) mesenchymal stem and stromal cells (MSCs) are key elements of the alveolar niche and fundamental regulators of homeostasis and regeneration. We interrogated their function during virus-induced lung injury using the highly prevalent respiratory syncytial virus (RSV) which causes severe outcomes in infants. We applied complementary approaches with primary pediatric LR-MSCs and a state-of-the-art model of human RSV infection in lamb. Remarkably, RSV-infection of pediatric LR-MSCs led to a robust activation, characterized by a strong antiviral and pro-inflammatory phenotype combined with mediators related to T cell function. In line with this, following in vivo infection, RSV invades and activates LR-MSCs, resulting in the expansion of the pulmonary MSC pool. Moreover, the global transcriptional response of LR-MSCs appears to follow RSV disease, switching from an early antiviral signature to repair mechanisms including differentiation, tissue remodeling, and angiogenesis. These findings demonstrate the involvement of LR-MSCs during virus-mediated acute lung injury and may have therapeutic implications.


Assuntos
Lesão Pulmonar Aguda/imunologia , Lesão Pulmonar Aguda/virologia , Pulmão/imunologia , Células-Tronco Mesenquimais/imunologia , Infecções por Vírus Respiratório Sincicial/imunologia , Animais , Humanos , Pulmão/citologia , Pulmão/metabolismo , Células-Tronco Mesenquimais/metabolismo , Infecções por Vírus Respiratório Sincicial/metabolismo , Vírus Sincicial Respiratório Humano/imunologia , Ovinos
3.
Am J Physiol Lung Cell Mol Physiol ; 318(4): L813-L830, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32073879

RESUMO

Our understanding of mesenchymal cell subsets and their function in human lung affected by aging and in certain disease settings remains poorly described. We use a combination of flow cytometry, prospective cell-sorting strategies, confocal imaging, and modeling of microvessel formation using advanced microfluidic chip technology to characterize mesenchymal cell subtypes in human postnatal and adult lung. Tissue was obtained from patients undergoing elective surgery for congenital pulmonary airway malformations (CPAM) and other airway abnormalities including chronic obstructive pulmonary disease (COPD). In microscopically normal postnatal human lung, there was a fivefold higher mesenchymal compared with epithelial (EpCAM+) fraction, which diminished with age. The mesenchymal fraction composed of CD90+ and CD90+CD73+ cells was enriched in CXCL12 and platelet-derived growth factor receptor-α (PDGFRα) and located in close proximity to EpCAM+ cells in the alveolar region. Surprisingly, alveolar organoids generated from EpCAM+ cells supported by CD90+ subset were immature and displayed dysplastic features. In congenital lung lesions, cystic air spaces and dysplastic alveolar regions were marked with an underlying thick interstitium composed of CD90+ and CD90+PDGFRα+ cells. In postnatal lung, a subset of CD90+ cells coexpresses the pericyte marker CD146 and supports self-assembly of perfusable microvessels. CD90+CD146+ cells from COPD patients fail to support microvessel formation due to fibrinolysis. Targeting the plasmin-plasminogen system during microvessel self-assembly prevented fibrin gel degradation, but microvessels were narrower and excessive contraction blocked perfusion. These data provide important new information regarding the immunophenotypic identity of key mesenchymal lineages and their change in a diverse setting of congenital lung lesions and COPD.


Assuntos
Imunomodulação/imunologia , Células-Tronco Mesenquimais/metabolismo , Antígenos Thy-1/imunologia , Antígenos Thy-1/metabolismo , Adolescente , Biomarcadores/metabolismo , Antígeno CD146/imunologia , Antígeno CD146/metabolismo , Separação Celular/métodos , Criança , Pré-Escolar , Molécula de Adesão da Célula Epitelial/imunologia , Molécula de Adesão da Célula Epitelial/metabolismo , Feminino , Humanos , Fatores Imunológicos/imunologia , Fatores Imunológicos/metabolismo , Lactente , Recém-Nascido , Masculino , Células-Tronco Mesenquimais/imunologia , Microvasos/imunologia , Microvasos/metabolismo , Pericitos/imunologia , Pericitos/metabolismo , Estudos Prospectivos
4.
J Virol ; 92(24)2018 12 15.
Artigo em Inglês | MEDLINE | ID: mdl-30282716

RESUMO

The mosquito-borne Japanese encephalitis virus (JEV) causes severe central nervous system diseases and cycles between Culex mosquitoes and different vertebrates. For JEV and some other flaviviruses, oronasal transmission is described, but the mode of infection is unknown. Using nasal mucosal tissue explants and primary porcine nasal epithelial cells (NEC) at the air-liquid interface (ALI) and macrophages as ex vivo and in vitro models, we determined that the nasal epithelium could represent the route of entry and exit for JEV in pigs. Porcine NEC at the ALI exposed to with JEV resulted in apical and basolateral virus shedding and release of monocyte recruiting chemokines, indicating infection and replication in macrophages. Moreover, macrophages stimulated by alarmins, including interleukin-25, interleukin-33, and thymic stromal lymphopoietin, were more permissive to the JEV infection. Altogether, our data are important to understand the mechanism of non-vector-borne direct transmission of Japanese encephalitis virus in pigs.IMPORTANCE JEV, a main cause of severe viral encephalitis in humans, has a complex ecology composed of a mosquito-waterbird cycle and a cycle involving pigs, which amplifies virus transmission to mosquitoes, leading to increased human cases. JEV can be transmitted between pigs by contact in the absence of arthropod vectors. Moreover, virus or viral RNA is found in oronasal secretions and the nasal epithelium. Using nasal mucosa tissue explants and three-dimensional porcine nasal epithelial cells cultures and macrophages as ex vivo and in vitro models, we determined that the nasal epithelium could be a route of entry as well as exit for the virus. Infection of nasal epithelial cells resulted in apical and basolateral virus shedding and release of monocyte recruiting chemokines and therefore infection and replication in macrophages, which is favored by epithelial-cell-derived cytokines. The results are relevant to understand the mechanism of non-vector-borne direct transmission of JEV.


Assuntos
Vírus da Encefalite Japonesa (Espécie)/fisiologia , Encefalite Japonesa/veterinária , Mucosa Nasal/virologia , Doenças dos Suínos/virologia , Animais , Células Cultivadas , Quimiocinas/metabolismo , Vírus da Encefalite Japonesa (Espécie)/imunologia , Encefalite Japonesa/imunologia , Encefalite Japonesa/virologia , Células Epiteliais/citologia , Mosquitos Vetores/virologia , Mucosa Nasal/citologia , Mucosa Nasal/imunologia , Suínos , Doenças dos Suínos/imunologia , Internalização do Vírus , Replicação Viral , Eliminação de Partículas Virais
5.
Am J Physiol Lung Cell Mol Physiol ; 315(6): L921-L932, 2018 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-30211653

RESUMO

Differentiation of primary alveolar type II epithelial cells (AEC II) to AEC type I in culture is a major barrier in the study of the alveolar epithelium in vitro. The establishment of an AEC II cell line derived from induced pluripotent stem cells (iPSC) represents a novel opportunity to study alveolar epithelial cell biology, for instance, in the context of lung injury, fibrosis, and repair. In the present study, we generated long-lasting AEC II from iPSC (LL-iPSC-AEC II). LL-iPSC-AEC II displayed morphological characteristics of AEC II, including growth in a cobblestone monolayer, the presence of lamellar bodies, and microvilli, as shown by electron microscopy. Also, LL-iPSC-AEC II expressed AEC type II proteins, such as cytokeratin, surfactant protein C, and LysoTracker DND 26 (a marker for lamellar bodies). Furthermore, the LL-iPSC-AEC II exhibited functional properties of AEC II by an increase of transepithelial electrical resistance over time, secretion of inflammatory mediators in biologically relevant quantities (IL-6 and IL-8), and efficient in vitro alveolar epithelial wound repair. Consistent with the AEC II phenotype, the cell line showed the ability to uptake and release surfactant protein B, to secrete phospholipids, and to differentiate into AEC type I. In summary, we established a long-lasting, but finite AEC type II cell line derived from iPSC as a novel cellular model to study alveolar epithelial cell biology in lung health and disease.


Assuntos
Células Epiteliais Alveolares/citologia , Células-Tronco Pluripotentes Induzidas/citologia , Diferenciação Celular/fisiologia , Linhagem Celular , Células HEK293 , Humanos , Lesão Pulmonar/patologia , Fenótipo , Alvéolos Pulmonares/citologia , Mucosa Respiratória/citologia
6.
Artigo em Inglês | MEDLINE | ID: mdl-30181371

RESUMO

The virus family Flaviviridae encompasses several viruses, including (re)emerging viruses which cause widespread morbidity and mortality throughout the world. Members of this virus family are positive-strand RNA viruses and replicate their genome in close association with reorganized intracellular host cell membrane compartments. This evolutionarily conserved strategy facilitates efficient viral genome replication and contributes to evasion from host cell cytosolic defense mechanisms. We have previously described the identification of a small-compound inhibitor, K22, which exerts a potent antiviral activity against a broad range of coronaviruses by targeting membrane-bound viral RNA replication. To analyze the antiviral spectrum of this inhibitor, we assessed the inhibitory potential of K22 against several members of the Flaviviridae family, including the reemerging Zika virus (ZIKV). We show that ZIKV is strongly affected by K22. Time-of-addition experiments revealed that K22 acts during a postentry phase of the ZIKV life cycle, and combination regimens of K22 together with ribavirin (RBV) or interferon alpha (IFN-α) further increased the extent of viral inhibition. Ultrastructural electron microscopy studies revealed severe alterations of ZIKV-induced intracellular replication compartments upon infection of K22-treated cells. Importantly, the antiviral activity of K22 was demonstrated against several other members of the Flaviviridae family. It is tempting to speculate that K22 exerts its broad antiviral activity against several positive-strand RNA viruses via a similar mechanism and thereby represents an attractive candidate for development as a panviral inhibitor.


Assuntos
Antivirais/farmacologia , Membrana Celular/efeitos dos fármacos , Infecções por Flaviviridae/tratamento farmacológico , Flaviviridae/efeitos dos fármacos , Aedes , Animais , Linhagem Celular , Membrana Celular/virologia , Chlorocebus aethiops , Infecções por Flaviviridae/virologia , Humanos , Interferon-alfa/farmacologia , RNA Viral/genética , Ribavirina/farmacologia , Células Vero , Replicação Viral/efeitos dos fármacos
7.
J Infect Dis ; 215(6): 902-906, 2017 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-28453839

RESUMO

The World Health Organization (WHO) published 2 alcohol-based formulations to be used in healthcare settings and for outbreak-associated infections, but inactivation efficacies of these products have not been determined against (re-)emerging viruses. In this study, we evaluated the virucidal activity of these WHO products in a comparative analysis. Zika virus (ZIKV), Ebola virus (EBOV), severe acute respiratory syndrome coronavirus (SARS-CoV), and Middle East respiratory syndrome coronavirus (MERS-CoV) as (re-)emerging viral pathogens and other enveloped viruses could be efficiently inactivated by both WHO formulations, implicating their use in healthcare systems and viral outbreak situations.


Assuntos
Antissepsia/métodos , Ebolavirus/efeitos dos fármacos , Higiene das Mãos/normas , Coronavírus da Síndrome Respiratória do Oriente Médio/efeitos dos fármacos , Coronavírus Relacionado à Síndrome Respiratória Aguda Grave/efeitos dos fármacos , Zika virus/efeitos dos fármacos , Infecções por Coronavirus/prevenção & controle , Doença pelo Vírus Ebola/prevenção & controle , Humanos , Guias de Prática Clínica como Assunto , Análise de Regressão , República da Coreia , Síndrome Respiratória Aguda Grave/prevenção & controle , Virulência , Organização Mundial da Saúde , Infecção por Zika virus/prevenção & controle
8.
Respir Res ; 18(1): 215, 2017 12 28.
Artigo em Inglês | MEDLINE | ID: mdl-29282053

RESUMO

BACKGROUND: In vitro systems of primary cystic fibrosis (CF) airway epithelial cells are an important tool to study molecular and functional features of the native respiratory epithelium. However, undifferentiated CF airway cell cultures grown under submerged conditions do not appropriately represent the physiological situation. A more advanced CF cell culture system based on airway epithelial cells grown at the air-liquid interface (ALI) recapitulates most of the in vivo-like properties but requires the use of invasive sampling methods. In this study, we describe a detailed characterization of fully differentiated primary CF airway epithelial cells obtained by non-invasive nasal brushing of pediatric patients. METHODS: Differentiated cell cultures were evaluated with immunolabelling of markers for ciliated, mucus-secreting and basal cells, and tight junction and CFTR proteins. Epithelial morphology and ultrastructure was examined by histology and transmission electron microscopy. Ciliary beat frequency was investigated by a video-microscopy approach and trans-epithelial electrical resistance was assessed with an epithelial Volt-Ohm meter system. Finally, epithelial permeability was analysed by using a cell layer integrity test and baseline cytokine levels where measured by an enzyme-linked immunosorbent assay. RESULTS: Pediatric CF nasal cultures grown at the ALI showed a differentiation into a pseudostratified epithelium with a mucociliary phenotype. Also, immunofluorescence analysis revealed the presence of ciliated, mucus-secreting and basal cells and tight junctions. CFTR protein expression was observed in CF (F508del/F508del) and healthy cultures and baseline interleukin (IL)-8 and IL-6 release were similar in control and CF ALI cultures. The ciliary beat frequency was 9.67 Hz and the differentiated pediatric CF epithelium was found to be functionally tight. CONCLUSION: In summary, primary pediatric CF nasal epithelial cell cultures grown at the ALI showed full differentiation into ciliated, mucus-producing and basal cells, which adequately reflect the in vivo properties of the human respiratory epithelium.


Assuntos
Fibrose Cística/patologia , Microvilosidades/patologia , Mucosa Nasal/patologia , Mucosa Respiratória/patologia , Adolescente , Células Cultivadas , Criança , Pré-Escolar , Fibrose Cística/metabolismo , Regulador de Condutância Transmembrana em Fibrose Cística/biossíntese , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Feminino , Humanos , Masculino , Microvilosidades/metabolismo , Mucosa Nasal/metabolismo , Mucosa Respiratória/metabolismo
9.
Eur Respir J ; 47(2): 520-30, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26585423

RESUMO

Vitamin D has immunomodulatory properties in the defence against pathogens. Its insufficiency is a widespread feature of cystic fibrosis (CF) patients, which are repeatedly suffering from rhinovirus (RV)-induced pulmonary exacerbations.To investigate whether vitamin D has antiviral activity, primary bronchial epithelial cells from CF children were pre-treated with vitamin D and infected with RV16. Antiviral and anti-inflammatory activity of vitamin D was assessed. RV and LL-37 levels were measured in bronchoalveolar lavage (BAL) of CF children infected with RV.Vitamin D reduced RV16 load in a dose-dependent manner in CF cells (10(-7 )M, p<0.01). The antiviral response mediated by interferons remained unchanged by vitamin D in CF cells. Vitamin D did not exert anti-inflammatory properties in RV-infected CF cells. Vitamin D increased the expression of the antimicrobial peptide LL-37 up to 17.4-fold (p<0.05). Addition of exogenous LL-37 decreased viral replication by 4.4-fold in CF cells (p<0.05). An inverse correlation between viral load and LL-37 levels in CF BAL (r=-0.48, p<0.05) was observed.RV replication in primary CF bronchial cells was reduced by vitamin D through the induction of LL-37. Clinical studies are needed to determine the importance of an adequate control of vitamin D for prevention of virus-induced pulmonary CF exacerbations.


Assuntos
Catelicidinas/efeitos dos fármacos , Colecalciferol/farmacologia , Fibrose Cística/metabolismo , Células Epiteliais/efeitos dos fármacos , Rhinovirus/efeitos dos fármacos , Replicação Viral/efeitos dos fármacos , Vitaminas/farmacologia , Adolescente , Peptídeos Catiônicos Antimicrobianos , Brônquios/citologia , Líquido da Lavagem Broncoalveolar/química , Líquido da Lavagem Broncoalveolar/virologia , Estudos de Casos e Controles , Catelicidinas/metabolismo , Criança , Pré-Escolar , Fibrose Cística/virologia , Células Epiteliais/virologia , Feminino , Humanos , Lactente , Masculino , Reação em Cadeia da Polimerase em Tempo Real , Carga Viral , Vitamina D/farmacologia
10.
Respirology ; 21(2): 304-12, 2016 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-26611536

RESUMO

BACKGROUND AND OBJECTIVE: Rhinoviruses (RV) replicate in both upper and lower airway epithelial cells. We evaluated the possibility of using nasal epithelial cells (NEC) as surrogate of bronchial epithelial cells (BEC) for RV pathogenesis cell culture studies. METHODS: We used primary paired NEC and BEC cultures established from healthy subjects and compared the replication of RV belonging to the major (RV16) and minor (RV1B) group, and the cellular antiviral and proinflammatory cytokine responses towards these viruses. We related antiviral and pro-inflammatory responses of NEC isolated from CF and COPD patients with those of BEC. RESULTS: RV16 replication and major group surface receptor (ICAM-1) expression were higher in healthy NEC compared with BEC (P < 0.05); RV1B replication and minor group surface receptor (LDLR) expression were similar. Healthy NEC and BEC produced similar levels of IFN-ß and IFN-λ2/3 upon RV infection or after simulation with poly(IC). IL-8 production was similar between healthy NEC and BEC. IL-6 release at baseline (P < 0.01) and upon infection with RV16 (P < 0.05) and poly(IC) stimulation (P < 0.05) was higher in NEC. RV1B viral load in NEC was related to RV1B viral load in BEC (r = 0.49, P = 0.01). There was a good correlation of IFN levels between NEC and BEC (r = 0.66, P = 0.0004 after RV1B infection). IL-8 production in NEC was related to IL-8 production in BEC (r = 0.48, P = 0.02 after RV1B infection). CONCLUSION: NEC are a suitable alternative cellular system to BEC to study the pathophysiology of RV infections and particularly to investigate IFN responses induced by RV infection.


Assuntos
Fibrose Cística/imunologia , Células Epiteliais/imunologia , Interleucina-6/imunologia , Interleucina-8/imunologia , Infecções por Picornaviridae/imunologia , Doença Pulmonar Obstrutiva Crônica/imunologia , Sistema Respiratório/imunologia , Rhinovirus/imunologia , Adolescente , Idoso , Células Cultivadas , Criança , Pré-Escolar , Feminino , Humanos , Imunidade Inata/imunologia , Molécula 1 de Adesão Intercelular/imunologia , Masculino , Pessoa de Meia-Idade
11.
Rev Med Suisse ; 12(536): 1794-1798, 2016 Oct 26.
Artigo em Francês | MEDLINE | ID: mdl-28692229

RESUMO

Zika virus has recently emerged as new teratogenic agent. Research is drastically increasing to allow a better comprehension of the role of this emerging virus in the induction of fetal cerebral anomalies. Several epidemiological and microbiological aspects may explain a higher virulence of the current strain. Various animal models have been developed and confirm the materno-fetal transmission as well as the induction of cerebral development's disorders. In parallel, various companies are working to develop a vaccine and antiviral medicine against Zika virus. Despite promising results, the road remains long until a vaccine or antiviral medicines are available.


Le virus Zika est récemment apparu comme un nouvel agent tératogène. La recherche s'accélère dans le but de mieux comprendre le rôle de ce virus émergent dans la formation de malformations cérébrales chez le fœtus. Plusieurs aspects épidémiologiques et microbiologiques contribuent à la nouvelle virulence observée chez la souche épidémique actuelle. Divers modèles d'infection chez la souris et les primates ont été développés confirmant la transmission transplacentaire du virus et l'induction de troubles du développement cérébral. En parallèle, plusieurs compagnies travaillent au développement d'un vaccin et d'antiviraux contre le virus Zika. Malgré des résultats prometteurs, la route est encore longue avant la mise sur le marché de vaccins ou d'antiviraux pouvant être administrés à la femme enceinte.


Assuntos
Antivirais/administração & dosagem , Vacinas Virais/administração & dosagem , Infecção por Zika virus/prevenção & controle , Animais , Anormalidades Congênitas/prevenção & controle , Anormalidades Congênitas/virologia , Modelos Animais de Doenças , Desenho de Fármacos , Feminino , Humanos , Gravidez , Complicações Infecciosas na Gravidez/prevenção & controle , Complicações Infecciosas na Gravidez/virologia , Infecção por Zika virus/complicações , Infecção por Zika virus/epidemiologia
13.
Eur Respir J ; 45(2): 428-39, 2015 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-25359346

RESUMO

Virus-associated pulmonary exacerbations, often associated with rhinoviruses (RVs), contribute to cystic fibrosis (CF) morbidity. Currently, there are only a few therapeutic options to treat virus-induced CF pulmonary exacerbations. The macrolide antibiotic azithromycin has antiviral properties in human bronchial epithelial cells. We investigated the potential of azithromycin to induce antiviral mechanisms in CF bronchial epithelial cells. Primary bronchial epithelial cells from CF and control children were infected with RV after azithromycin pre-treatment. Viral RNA, interferon (IFN), IFN-stimulated gene and pattern recognition receptor expression were measured by real-time quantitative PCR. Live virus shedding was assessed by assaying the 50% tissue culture infective dose. Pro-inflammatory cytokine and IFN-ß production were evaluated by ELISA. Cell death was investigated by flow cytometry. RV replication was increased in CF compared with control cells. Azithromycin reduced RV replication seven-fold in CF cells without inducing cell death. Furthermore, azithromycin increased RV-induced pattern recognition receptor, IFN and IFN-stimulated gene mRNA levels. While stimulating antiviral responses, azithromycin did not prevent virus-induced pro-inflammatory responses. Azithromycin pre-treatment reduces RV replication in CF bronchial epithelial cells, possibly through the amplification of the antiviral response mediated by the IFN pathway. Clinical studies are needed to elucidate the potential of azithromycin in the management and prevention of RV-induced CF pulmonary exacerbations.


Assuntos
Antivirais/farmacologia , Azitromicina/farmacologia , Brônquios/citologia , Fibrose Cística/fisiopatologia , Células Epiteliais/efeitos dos fármacos , Adolescente , Estudos de Casos e Controles , Sobrevivência Celular/efeitos dos fármacos , Células Cultivadas , Criança , Pré-Escolar , Células Epiteliais/citologia , Feminino , Humanos , Lactente , Interferons/metabolismo , Masculino , Infecções por Picornaviridae/tratamento farmacológico , Rhinovirus
14.
Exp Lung Res ; 40(7): 327-32, 2014 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-25058379

RESUMO

PURPOSE: Primary nasal epithelial cells are used for diagnostic purposes in clinical routine and have been shown to be good surrogate models for bronchial epithelial cells in studies of airway inflammation and remodeling. We aimed at comparing different instruments allowing isolation of nasal epithelial cells. METHODS: Primary airway epithelial cell cultures were established using cells acquired from the inferior surface of the middle turbinate of both nostrils. Three different instruments to isolate nasal cells were used: homemade cytology brush, nasal swab, and curette. Cell count, viability, time until a confluent cell layer was reached, and success rate in establishing cell cultures were evaluated. A standard numeric pain intensity scale was used to assess the acceptability of each instrument. RESULTS: Sixty healthy adults (median with interquartile range [IQR] age of 31 [26-37] years) participated in the study. Higher number of cells (×10(5) cells/ml) was obtained using brushes (9.8 [5.9-33.5]) compared to swabs (2.4 [1.5-3.9], p < 0.0001) and curettes (5.5 [4.4-6.9], p < 0.01). Cell viability was similar between groups. Cells obtained by brushes had the fastest growth rate, and the success rate in establishing primary cell cultures was highest with brushes (90% vs. 65% for swabs and 70% for curettes). Pain was highest with curettes (VAS score 4.0 [3.0-5.0] out of 10). The epithelial phenotype of the cultures was confirmed through cytokeratin and E-cadherin staining. CONCLUSIONS: All three types of instruments allow collection and growth of human nasal epithelial cells with good acceptability to study participants. The most efficient instrument is the nasal brush.


Assuntos
Técnicas de Cultura de Células/métodos , Células Epiteliais/citologia , Nariz/citologia , Adulto , Brônquios/citologia , Caderinas/metabolismo , Contagem de Células/métodos , Sobrevivência Celular/fisiologia , Células Epiteliais/metabolismo , Feminino , Humanos , Queratinas/metabolismo , Masculino , Mucosa Nasal/metabolismo
15.
Thorax ; 67(6): 517-25, 2012 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-22213737

RESUMO

BACKGROUND: Rhinoviruses are important triggers of pulmonary exacerbations and possible contributors to long-term respiratory morbidity in cystic fibrosis (CF), but mechanisms leading to rhinovirus-induced CF exacerbations are poorly understood. It is hypothesised that there is a deficient innate immune response of the airway epithelium towards rhinovirus infection in CF. METHODS: Early innate immune responses towards rhinoviruses (RV-16, major-type and RV-1B, minor-type) in CF and non-CF bronchial epithelial cell lines and primary nasal and bronchial epithelial cells from patients with CF (n=13) and healthy controls (n=24) were studied. RESULTS: Rhinovirus RNA expression and virus release into supernatants was increased more than tenfold in CF cells compared with controls. CF cells expressed up to 1000 times less interferon (IFN) type I (ß) and type III (λ) mRNA and produced less than half of IFN-ß and IFN-λ protein compared with controls. In contrast, interleukin 8 production was not impaired, indicating a selective deficiency in the innate antiviral defence system. Deficient IFN production was paralleled by lower expression of IFN-stimulated genes including myxovirus resistance A, 2',5'-oligoadenylate synthetase, viperin and nitric oxide synthase 2. Addition of exogenous type I and III IFNs, particularly IFN-ß, restored antiviral pathways and virus control in CF cells, underscoring the crucial role of these molecules. CONCLUSIONS: This study describes a novel mechanism to explain the increased susceptibility of patients with CF to rhinovirus infections. A profound impairment of the antiviral early innate response in CF airway epithelial cells was identified, suggesting a potential use of IFNs in the treatment of rhinovirus-induced CF exacerbations.


Assuntos
Antivirais/farmacologia , Brônquios/imunologia , Fibrose Cística/imunologia , Interferon beta/imunologia , Interferons/deficiência , Interleucinas/imunologia , Infecções por Picornaviridae/imunologia , Mucosa Respiratória/imunologia , Mucosa Respiratória/virologia , Rhinovirus/imunologia , Doença Aguda , Antivirais/imunologia , Brônquios/citologia , Brônquios/virologia , Células Cultivadas , Fibrose Cística/tratamento farmacológico , Fibrose Cística/virologia , Suscetibilidade a Doenças , Células Epiteliais/imunologia , Células Epiteliais/virologia , Humanos , Imunidade Inata/efeitos dos fármacos , Interferon Tipo I/imunologia , Interferon beta/deficiência , Interferon beta/farmacologia , Interferons/imunologia , Interleucinas/deficiência , Infecções por Picornaviridae/tratamento farmacológico , Infecções por Picornaviridae/metabolismo , Infecções por Picornaviridae/virologia , Podócitos/imunologia , Rhinovirus/efeitos dos fármacos , Resultado do Tratamento
16.
STAR Protoc ; 3(2): 101291, 2022 06 17.
Artigo em Inglês | MEDLINE | ID: mdl-35463475

RESUMO

We present a protocol to generate an advanced ex vivo model of human placenta. We use a vibrating tissue slicer to obtain precision-cut slices representative of the entire thickness of human placenta. This approach delivers standardized cultures with a preserved microstructure and cellular composition comparable to the native tissue. We applied this system to study SARS-CoV-2 infection at the maternal-fetal interface. Moreover, this system can be used to investigate the basic functions of the human placenta in health and disease. For complete details on the use and execution of this protocol, please refer to Fahmi et al. (2021).


Assuntos
COVID-19 , SARS-CoV-2 , Feminino , Humanos , Placenta , Gravidez
17.
STAR Protoc ; 3(4): 101688, 2022 12 16.
Artigo em Inglês | MEDLINE | ID: mdl-36116074

RESUMO

Here, we present a protocol to analyze the T cell profiles of the neonatal ovine lung during respiratory syncytial virus (RSV) infection. The protocol delivers standardized multiparameter flow cytometry (FCM) analysis of CD4+, CD8+, regulatory, and γδ T cells isolated from lung, lymph nodes, and bronchoalveolar lavages (BALs). We detail the preparation of RSV and transtracheal inoculation of newborn lambs. We then describe tissue isolation and preparation of cell suspensions, followed by FCM acquisition to identify different T cell subsets. For complete details on the use and execution of this protocol, please refer to Démoulins et al. (2021).


Assuntos
Infecções por Vírus Respiratório Sincicial , Animais , Ovinos , Infecções por Vírus Respiratório Sincicial/patologia , Citometria de Fluxo , Vírus Sinciciais Respiratórios , Pulmão/patologia , Subpopulações de Linfócitos T
18.
Cell Rep ; 39(4): 110736, 2022 04 26.
Artigo em Inglês | MEDLINE | ID: mdl-35476995

RESUMO

The deacetylase HDAC6 has tandem catalytic domains and a zinc finger domain (ZnF) binding ubiquitin (Ub). While the catalytic domain has an antiviral effect, the ZnF facilitates influenza A virus (IAV) infection and cellular stress responses. By recruiting Ub via the ZnF, HDAC6 promotes the formation of aggresomes and stress granules (SGs), dynamic structures associated with pathologies such as neurodegeneration. IAV subverts the aggresome/HDAC6 pathway to facilitate capsid uncoating during early infection. To target this pathway, we generate designed ankyrin repeat proteins (DARPins) binding the ZnF; one of these prevents interaction with Ub in vitro and in cells. Crystallographic analysis shows that it blocks the ZnF pocket where Ub engages. Conditional expression of this DARPin reversibly impairs infection by IAV and Zika virus; moreover, SGs and aggresomes are downregulated. These results validate the HDAC6 ZnF as an attractive target for drug discovery.


Assuntos
Vírus da Influenza A , Influenza Humana , Infecção por Zika virus , Zika virus , Desacetilase 6 de Histona/metabolismo , Humanos , Vírus da Influenza A/metabolismo , Ubiquitina/metabolismo , Zika virus/metabolismo
19.
Nat Commun ; 13(1): 5929, 2022 10 07.
Artigo em Inglês | MEDLINE | ID: mdl-36207334

RESUMO

Variant of concern (VOC) Omicron-BA.1 has achieved global predominance in early 2022. Therefore, surveillance and comprehensive characterization of Omicron-BA.1 in advanced primary cell culture systems and animal models are urgently needed. Here, we characterize Omicron-BA.1 and recombinant Omicron-BA.1 spike gene mutants in comparison with VOC Delta in well-differentiated primary human nasal and bronchial epithelial cells in vitro, followed by in vivo fitness characterization in hamsters, ferrets and hACE2-expressing mice, and immunized hACE2-mice. We demonstrate a spike-mediated enhancement of early replication of Omicron-BA.1 in nasal epithelial cultures, but limited replication in bronchial epithelial cultures. In hamsters, Delta shows dominance over Omicron-BA.1, and in ferrets Omicron-BA.1 infection is abortive. In hACE2-knock-in mice, Delta and a Delta spike clone also show dominance over Omicron-BA.1 and an Omicron-BA.1 spike clone, respectively. Interestingly, in naïve K18-hACE2 mice, we observe Delta spike-mediated increased replication and pathogenicity and Omicron-BA.1 spike-mediated reduced replication and pathogenicity, suggesting that the spike gene is a major determinant of replication and pathogenicity. Finally, the Omicron-BA.1 spike clone is less well-controlled by mRNA-vaccination in K18-hACE2-mice and becomes more competitive compared to the progenitor and Delta spike clones, suggesting that spike gene-mediated immune evasion is another important factor that led to Omicron-BA.1 dominance.


Assuntos
COVID-19 , SARS-CoV-2 , Animais , Cricetinae , Furões , Humanos , Melfalan , Camundongos , Fenótipo , RNA Mensageiro , SARS-CoV-2/genética , Glicoproteína da Espícula de Coronavírus/genética , gama-Globulinas
20.
Physiol Rep ; 9(18): e14994, 2021 09.
Artigo em Inglês | MEDLINE | ID: mdl-34542243

RESUMO

Nasal epithelial cells (NECs) are among the first cells to be exposed to air pollutants and respiratory viruses. Although it is known that air pollution exposure and rhinovirus infections increase the risk for asthma development independently, it is unclear how these risk factors interact on a cellular level. Therefore, we aimed to investigate how exposure to diesel particulate matter (DPM) modifies the response of primary NECs to rhinovirus (RV) infection in vitro. Exposure of re-differentiated, primary NECs (49 healthy children [0-7 years], 12 adults) to DPM modified the mRNA expression of viral cell-surface receptors, pattern recognition receptors, and pro-inflammatory response (also protein levels). After exposure to DPM, we additionally infected the NECs with RV-1b and RV-16. Viral loads (assessed by titration assays) were significantly higher in DPM-exposed compared with non-exposed NECs. Exposure to DPM prior to RV infection resulted in a significant upregulation of pro-inflammatory cytokines (mRNA and protein level) and ß-defensins mRNA, and significant downregulation of pattern recognition receptors mRNA and CXCL10 (mRNA and protein levels). There was no difference between all outcomes of NECs from children and adults. We can conclude that exposure to DPM prior to RV infection increases viral loads by downregulation of viral defense receptors and upregulation of pro-inflammatory cytokines. Our findings indicate a strong interaction between air pollution and the antiviral response to RV infection in NECs. We provide mechanistic evidence that exposure to air pollution increases susceptibility to RV infection.


Assuntos
Poluentes Atmosféricos/toxicidade , Mucosa Nasal/efeitos dos fármacos , Material Particulado/toxicidade , Infecções por Picornaviridae/imunologia , Emissões de Veículos/toxicidade , Adulto , Células Cultivadas , Quimiocina CXCL10/genética , Quimiocina CXCL10/metabolismo , Criança , Pré-Escolar , Humanos , Lactente , Mucosa Nasal/imunologia , Mucosa Nasal/virologia , Rhinovirus/patogenicidade
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA