Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 913
Filtrar
1.
Nature ; 626(7998): 392-400, 2024 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-38086420

RESUMO

An ideal vaccine both attenuates virus growth and disease in infected individuals and reduces the spread of infections in the population, thereby generating herd immunity. Although this strategy has proved successful by generating humoral immunity to measles, yellow fever and polio, many respiratory viruses evolve to evade pre-existing antibodies1. One approach for improving the breadth of antiviral immunity against escape variants is through the generation of memory T cells in the respiratory tract, which are positioned to respond rapidly to respiratory virus infections2-6. However, it is unknown whether memory T cells alone can effectively surveil the respiratory tract to the extent that they eliminate or greatly reduce viral transmission following exposure of an individual to infection. Here we use a mouse model of natural parainfluenza virus transmission to quantify the extent to which memory CD8+ T cells resident in the respiratory tract can provide herd immunity by reducing both the susceptibility of acquiring infection and the extent of transmission, even in the absence of virus-specific antibodies. We demonstrate that protection by resident memory CD8+ T cells requires the antiviral cytokine interferon-γ (IFNγ) and leads to altered transcriptional programming of epithelial cells within the respiratory tract. These results suggest that tissue-resident CD8+ T cells in the respiratory tract can have important roles in protecting the host against viral disease and limiting viral spread throughout the population.


Assuntos
Linfócitos T CD8-Positivos , Memória Imunológica , Células T de Memória , Infecções por Paramyxoviridae , Sistema Respiratório , Animais , Camundongos , Linfócitos T CD8-Positivos/imunologia , Modelos Animais de Doenças , Células Epiteliais/imunologia , Células Epiteliais/metabolismo , Imunidade Coletiva/imunologia , Memória Imunológica/imunologia , Interferon gama/imunologia , Células T de Memória/imunologia , Paramyxoviridae/imunologia , Paramyxoviridae/fisiologia , Infecções por Paramyxoviridae/imunologia , Infecções por Paramyxoviridae/prevenção & controle , Infecções por Paramyxoviridae/transmissão , Infecções por Paramyxoviridae/virologia , Sistema Respiratório/citologia , Sistema Respiratório/imunologia , Sistema Respiratório/virologia , Transcrição Gênica , Humanos
2.
J Virol ; 97(10): e0105123, 2023 10 31.
Artigo em Inglês | MEDLINE | ID: mdl-37732787

RESUMO

IMPORTANCE: For many years, measles virus (MeV) was assumed to first enter the host via the apical surface of airway epithelial cells and subsequently spread systemically. We and others reported that MeV has an overwhelming preference for entry at the basolateral surface of airway epithelial cells, which led to a fundamental new understanding of how MeV enters a human host. This unexpected observation using well-differentiated primary cultures of airway epithelia from human donors contradicted previous studies using immortalized cultured cells. Here, we show that appropriate differentiation and cell morphology of primary human airway epithelial cells are critical to recapitulate MeV infection patterns and pathogenesis of the in vivo airways. By simply culturing primary cells in media containing serum or passaging primary cultures, erroneous results quickly emerge. These results have broad implications for data interpretation related to respiratory virus infection, spread, and release from human airway epithelial cells.


Assuntos
Células Cultivadas , Células Epiteliais , Vírus do Sarampo , Sarampo , Sistema Respiratório , Humanos , Células Epiteliais/virologia , Epitélio , Sarampo/virologia , Sistema Respiratório/citologia
3.
Nucleic Acids Res ; 51(8): 3650-3670, 2023 05 08.
Artigo em Inglês | MEDLINE | ID: mdl-36772828

RESUMO

Epithelial mesenchymal plasticity (EMP) is a complex cellular reprogramming event that plays a major role in tissue homeostasis. Recently we observed the unfolded protein response (UPR) triggers EMP through the inositol-requiring protein 1 (IRE1α)-X-box-binding protein 1 spliced (XBP1s) axis, enhancing glucose shunting to protein N glycosylation. To better understand the genomic targets of XBP1s, we identified its genomic targets using Cleavage Under Targets and Release Using Nuclease (CUT&RUN) of a FLAG-epitope tagged XBP1s in RSV infection. CUT&RUN identified 7086 binding sites in chromatin that were enriched in AP-1 motifs and GC-sequences. Of these binding sites, XBP1s peaks mapped to 4827 genes controlling Rho-GTPase signaling, N-linked glycosylation and ER-Golgi transport. Strikingly, XBP1s peaks were within 1 kb of transcription start sites of 2119 promoters. In addition to binding core mesenchymal transcription factors SNAI1 and ZEB1, we observed that hexosamine biosynthetic pathway (HBP) enzymes were induced and contained proximal XBP1s peaks. We demonstrate that IRE1α -XBP1s signaling is necessary and sufficient to activate core enzymes by recruiting elongation-competent phospho-Ser2 CTD modified RNA Pol II. We conclude that the IRE1α-XBP1s pathway coordinately regulates mesenchymal transcription factors and hexosamine biosynthesis in EMP by a mechanism involving recruitment of activated pSer2-Pol II to GC-rich promoters.


Assuntos
Epitélio , Sistema Respiratório , Estresse do Retículo Endoplasmático , Endorribonucleases/metabolismo , Genômica , Hexosaminas , Proteínas Serina-Treonina Quinases/genética , Proteínas Serina-Treonina Quinases/metabolismo , Fatores de Transcrição/genética , Fatores de Transcrição/metabolismo , Resposta a Proteínas não Dobradas , Epitélio/fisiologia , Sistema Respiratório/citologia , Humanos
4.
Cell ; 186(1): 112-130.e20, 2023 01 05.
Artigo em Inglês | MEDLINE | ID: mdl-36580912

RESUMO

How SARS-CoV-2 penetrates the airway barrier of mucus and periciliary mucins to infect nasal epithelium remains unclear. Using primary nasal epithelial organoid cultures, we found that the virus attaches to motile cilia via the ACE2 receptor. SARS-CoV-2 traverses the mucus layer, using motile cilia as tracks to access the cell body. Depleting cilia blocks infection for SARS-CoV-2 and other respiratory viruses. SARS-CoV-2 progeny attach to airway microvilli 24 h post-infection and trigger formation of apically extended and highly branched microvilli that organize viral egress from the microvilli back into the mucus layer, supporting a model of virus dispersion throughout airway tissue via mucociliary transport. Phosphoproteomics and kinase inhibition reveal that microvillar remodeling is regulated by p21-activated kinases (PAK). Importantly, Omicron variants bind with higher affinity to motile cilia and show accelerated viral entry. Our work suggests that motile cilia, microvilli, and mucociliary-dependent mucus flow are critical for efficient virus replication in nasal epithelia.


Assuntos
COVID-19 , Sistema Respiratório , SARS-CoV-2 , Humanos , Cílios/fisiologia , Cílios/virologia , COVID-19/virologia , Sistema Respiratório/citologia , Sistema Respiratório/virologia , SARS-CoV-2/fisiologia , Microvilosidades/fisiologia , Microvilosidades/virologia , Internalização do Vírus , Células Epiteliais/fisiologia , Células Epiteliais/virologia
5.
Bioengineered ; 13(2): 3137-3147, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-35037821

RESUMO

Asthma is a respiratory disease with complex pathogenesis. Sterol-responsive element-binding proteins 2 (SREBP2) was found to bind to promoter sequences of ABCA1 to suppress ABCA1 promoter activity. This study aimed to explore the expression level of SREBP2 and ATP-binding cassette transporter A1 (ABCA1), and their effects on the development of airway smooth muscle cells (ASMCs) in asthma. ASMCs were treated with different concentrations of TGF-ß1 (0, 0.5, 1, 5 and 10 ng/mL). Short hairpin SREBP2 (shSREBP2), SREBP2, shABCA1 or ABCA1 were transfected into ASMCs. Cell viability, proliferation, apoptosis, migration, and the expression of SREBP2, ABCA1 and related pathway proteins were detected by MTT assay, Brdu staining, flow cytometer, Transwell assay, qRT-PCR, and Western blotting, respectively. The results showed that TGF-ß1 increased the viability, proliferation, migration and inhibited apoptosis in ASMCs. Moreover, TGF-ß1 also decreased the expression of ABCA1, cleaved caspase-3, cleaved PARP, E-cadherin, and increased the expression of vimentin, TLR2, p-p65 and NFATc1. SREBP2 knockdown alleviated these TGF-ß1-induced changes. SREBP2 overexpression inhibited ABCA1 expression and apoptosis, and promoted cell migration and the expression of TLR2, p-p65, NFATc1 in ASMCs. ABCA1 overexpression alleviated these SREBP2-induced promoting and inhibition effects. In conclusion, SREBP2 activates TLR2/NF-κB/NFATc1 regulatory network and promotes TGF-ß1-induced cell movement through inhibiting ABCA1 expression.


Assuntos
Miócitos de Músculo Liso , Proteína de Ligação a Elemento Regulador de Esterol 2/fisiologia , Fator de Crescimento Transformador beta1/farmacologia , Transportador 1 de Cassete de Ligação de ATP/genética , Transportador 1 de Cassete de Ligação de ATP/metabolismo , Apoptose/efeitos dos fármacos , Apoptose/genética , Movimento Celular/efeitos dos fármacos , Movimento Celular/genética , Proliferação de Células/efeitos dos fármacos , Proliferação de Células/genética , Sobrevivência Celular/efeitos dos fármacos , Sobrevivência Celular/genética , Células Cultivadas , Humanos , Miócitos de Músculo Liso/efeitos dos fármacos , Miócitos de Músculo Liso/fisiologia , NF-kappa B/genética , NF-kappa B/metabolismo , Fatores de Transcrição NFATC/genética , Fatores de Transcrição NFATC/metabolismo , Mucosa Respiratória/citologia , Mucosa Respiratória/efeitos dos fármacos , Mucosa Respiratória/metabolismo , Sistema Respiratório/citologia , Sistema Respiratório/efeitos dos fármacos , Sistema Respiratório/metabolismo , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Receptor 2 Toll-Like/genética , Receptor 2 Toll-Like/metabolismo
6.
Sci Rep ; 12(1): 427, 2022 01 10.
Artigo em Inglês | MEDLINE | ID: mdl-35013475

RESUMO

Neutrophilic airway inflammation is highly prevalent in racehorses in training, with the term mild to moderate equine asthma (MMEA) being applied to the majority of such cases. Our proposed study is largely derived from the strong association between MMEA in racehorses and their entry into a race training program. The objectives of this study are to characterise the effect of training on the local pulmonary immune system by defining the gene and protein expression of tracheal wash (TW) derived samples from Thoroughbred racehorses prior to and following commencement of race training. Multiomics analysis detected 2138 differentially expressed genes and 260 proteins during the training period. Gene and protein sets were enriched for biological processes related to acute phase response, oxidative stress, haemopoietic processes, as well as to immune response and inflammation. This study demonstrated TW samples to represent a rich source of airway cells, protein and RNA to study airway immunity in the horse and highlighted the benefits of a multiomics methodological approach to studying the dynamics of equine airway immunity. Findings likely reflect the known associations between race-training and both airway inflammation and bleeding, offering further insight into the potential mechanisms which underpin training associated airway inflammation.


Assuntos
Cavalos/imunologia , Condicionamento Físico Animal , Proteoma , Sistema Respiratório/imunologia , Transcriptoma , Animais , Perfilação da Expressão Gênica , Masculino , Sistema Respiratório/citologia
7.
Toxicol Lett ; 356: 100-109, 2022 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-34902520

RESUMO

Lung epithelial cells and fibroblasts play key roles in pulmonary fibrosis and are involved in fibrotic signaling and production of the extracellular matrix (ECM), respectively. Recently, 3D airway models consisting of both cell types have been developed to evaluate the fibrotic responses while facilitating cell-cell crosstalk. This study aimed to evaluate the fibrotic responses in these models using different fibrogenic agents, which are known as key events in adverse outcome pathways of pulmonary fibrosis. We quantified cell injury and several sequential steps in fibrogenesis, including inflammation, the epithelial-mesenchymal transition (EMT), fibroblast activation, and ECM accumulation, using two different 3D airway models, the EpiAirway™-full thickness (Epi/FT) and MucilAir™-human fibroblast (Mucil/HF) models. In the Epi/FT model, fibrogenic agents induced the expression of inflammation and EMT-associated markers, while in the Mucil/HF model, they induced fibroblast activation and ECM accumulation. Using this information, we conducted gene ontology term network analysis. In the Epi/FT model, the terms associated with cell migration and response to stimulus made up a large part of the network. In the Mucil/HF model, the terms associated with ECM organization and cell differentiation and proliferation constituted a great part of the network. Collectively, our data suggest that polyhexamethyleneguanidine phosphate and bleomycin induce different responses in the two 3D airway models. While Epi/FT was associated with inflammatory/EMT-associated responses, Mucil/HF was associated with fibroblast-associated responses. This study will provide an important basis for selecting proper 3D airway models and fibrogenic agents to further research or screen chemicals causing inhalation toxicity.


Assuntos
Técnicas de Cultura de Células em Três Dimensões/métodos , Células Epiteliais/fisiologia , Fibroblastos/fisiologia , Fibrose/induzido quimicamente , Sistema Respiratório/citologia , Antineoplásicos/toxicidade , Biomarcadores , Bleomicina/toxicidade , Citocinas/genética , Citocinas/metabolismo , Relação Dose-Resposta a Droga , Regulação da Expressão Gênica/efeitos dos fármacos , Guanidinas/toxicidade , Humanos , Fator de Crescimento Transformador beta
8.
Bioengineered ; 13(1): 1791-1801, 2022 01.
Artigo em Inglês | MEDLINE | ID: mdl-34699311

RESUMO

Childhood asthma is the most universal chronic disease, with significant cases reported. Despite the current progress in treatment, prognosis remains poor and the existing drugs cause serious side effects. This investigation explored the mechanisms and use of miR-335-5p on childhood asthma therapy. MiR-335-5p and ATG5 expression was analyzed in clinical plasma samples through RT-qPCR. Airway smooth muscle cells (ASMCs) were cultured, and transfected with miR-335-5p mimic, miR-335-5p inhibitor, and pcDNA3.1-ATG5, or co-transfected with miR-335-5p mimic + pcDNA3.1-ATG5. Asthma cell models were constructed through TGF-ß1, and animal models through ovalbumin (OVA). Monocyte-macrophage infiltration in bronchoalveolar lavage fluid (BALF) was determined by May-Grunwald-Giemsa staining, and collagen in lung tissue was assessed via Masson staining. Relationship between miR-335-5p and ATG5 was detected by dual-luciferase assay. Cell proliferation was detected by MTT assay. MiR-335-5p and ATG5 RNA expression was determined by RT-qPCR. Collagen I, collagen III, α-SMA, ATG5, LC3I/II, Beclin-1, and p62 protein expression levels in ASMCs were detected by western blot. MiR-335-5p expression was low, but ATG5 expression was high in childhood asthma. Versus OVA+ mimic NC group, the number of eosinophil and collagen in OVA+ miR-335-5p mimic group were reduced. In contrast to TGF-ß1 + mimic NC group, TGF-ß1 + miR-335-5p mimic group reduced inflammatory, airway fibrosis, and autophagy in ASMCs. ATG5 was miR-335-5p target. Overexpressing ATG5 significantly reversed the inhibitory effects of miR-335-5p on inflammatory response, fibrosis, and autophagy in ASMCs. Overall, the study concludes that MiR-335-5p alleviate inflammatory response, airway fibrosis, and autophagy in childhood asthma through targeted regulation of ATG5.


Assuntos
Asma/genética , Proteína 5 Relacionada à Autofagia/genética , MicroRNAs/genética , Ovalbumina/imunologia , Sistema Respiratório/citologia , Fator de Crescimento Transformador beta/imunologia , Animais , Autofagia , Líquido da Lavagem Broncoalveolar/imunologia , Proliferação de Células , Células Cultivadas , Criança , Modelos Animais de Doenças , Feminino , Humanos , Masculino , Miócitos de Músculo Liso/citologia , Transdução de Sinais
9.
STAR Protoc ; 2(3): 100683, 2021 09 17.
Artigo em Inglês | MEDLINE | ID: mdl-34355203

RESUMO

Airway basal cells play an essential role in the maintenance of the airway epithelium. Here, we provide a detailed directed differentiation protocol to generate ''induced basal cells (iBCs)'' from human pluripotent stem cells. iBCs recapitulate biological and functional properties of airway basal cells including mucociliary differentiation in vitro or in vivo in tracheal xenografts, facilitating the study of inherited and acquired diseases of the airway, as well as potential use in regenerative medicine. For complete details on the use and execution of this protocol, please refer to Hawkins et al. (2021).


Assuntos
Técnicas de Cultura de Células/métodos , Sistema Respiratório/citologia , Engenharia Tecidual/métodos , Diferenciação Celular/fisiologia , Células Cultivadas , Endoderma/citologia , Células Epiteliais/citologia , Epitélio/metabolismo , Humanos , Células-Tronco Pluripotentes Induzidas/citologia , Pulmão/citologia , Organoides/citologia , Células-Tronco Pluripotentes/citologia , Traqueia/citologia
10.
Virol J ; 18(1): 169, 2021 08 17.
Artigo em Inglês | MEDLINE | ID: mdl-34404450

RESUMO

BACKGROUND: Transmission of all known pathogenic orthohantaviruses (family Hantaviridae) usually occurs via inhalation of aerosols contaminated with viral particles derived from infected rodents and organ manifestation of infections is characterized by lung and kidney involvement. Orthohantaviruses found in Eurasia cause hemorrhagic fever with renal syndrome (HFRS) and New World orthohantaviruses cause hantavirus cardiopulmonary syndrome (HCPS). However, cases of infection with Old World orthohantaviruses with severe pulmonary manifestations have also been observed. Therefore, human airway cells may represent initial targets for orthohantavirus infection and may also play a role in the pathogenesis of infections with Eurasian orthohantaviruses. METHODS: We analyzed the permissiveness of primary endothelial cells of the human pulmonary microvasculature and of primary human epithelial cells derived from bronchi, bronchioles and alveoli for Old World orthohantavirus Puumala virus (PUUV) in vitro. In addition, we examined the expression of orthohantaviral receptors in these cell types. To minimize donor-specific effects, cells from two different donors were tested for each cell type. RESULTS: Productive infection with PUUV was observed for endothelial cells of the microvasculature and for the three tested epithelial cell types derived from different sites of the respiratory tract. Interestingly, infection and particle release were also detected in bronchial and bronchiolar epithelial cells although expression of the orthohantaviral receptor integrin ß3 was not detectable in these cell types. In addition, replication kinetics and viral release demonstrate enormous donor-specific variations. CONCLUSIONS: The human respiratory epithelium is among the first targets of orthohantaviral infection and may contribute to virus replication, dissemination and pathogenesis of HFRS-causing orthohantaviruses. Differences in initial pulmonary infection due to donor-specific factors may play a role in the observed broad variance of severity and symptoms of orthohantavirus disease in patients. The absence of detectable levels of integrin αVß3 surface expression on bronchial and small airway epithelial cells indicates an alternate mode of orthohantaviral entry in these cells that is independent from integrin ß3.


Assuntos
Células Endoteliais/virologia , Virus Puumala , Replicação Viral , Febre Hemorrágica com Síndrome Renal , Humanos , Integrinas , Cultura Primária de Células , Virus Puumala/fisiologia , Sistema Respiratório/citologia , Sistema Respiratório/virologia
11.
J Med Virol ; 93(12): 6671-6685, 2021 12.
Artigo em Inglês | MEDLINE | ID: mdl-34324210

RESUMO

Infection by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) causes a wide spectrum of syndromes involving multiple organ systems and is primarily mediated by viral spike (S) glycoprotein through the receptor-binding domain (RBD) and numerous cellular proteins including ACE2, transmembrane serine protease 2 (TMPRSS2), kidney injury molecule-1 (Kim-1), and neuropilin-1 (NRP-1). In this study, we examined the entry tropism of SARS-CoV-2 and SARS-CoV using S protein-based pseudoviruses to infect 22 cell lines and 3 types of primary cells isolated from respiratory, urinary, digestive, reproductive, and immune systems. At least one cell line or type of primary cell from each organ system was infected by both pseudoviruses. Infection by pseudoviruses is effectively blocked by S1, RBD, and ACE2 recombinant proteins, and more weakly by Kim-1 and NRP-1 recombinant proteins. Furthermore, cells with robust SARS-CoV-2 pseudovirus infection had strong expression of either ACE2 or Kim-1 and NRP-1 proteins. ACE2 glycosylation appeared to be critical for the infections of both viruses as there was a positive correlation between infectivity of either SARS-CoV-2 or SARS-CoV pseudovirus with the level of glycosylated ACE2 (gly-ACE2). These results reveal that SARS-CoV-2 cell entry could be mediated by either an ACE2-dependent or -independent mechanism, thus providing a likely molecular basis for its broad tropism for a wide variety of cell types.


Assuntos
Enzima de Conversão de Angiotensina 2/metabolismo , Trato Gastrointestinal/virologia , Genitália/virologia , Receptor Celular 1 do Vírus da Hepatite A/metabolismo , Sistema Imunitário/virologia , Neuropilina-1/metabolismo , Sistema Respiratório/virologia , SARS-CoV-2/fisiologia , Serina Endopeptidases/metabolismo , Internalização do Vírus , Western Blotting , COVID-19/metabolismo , COVID-19/virologia , Linhagem Celular , Células Cultivadas , Imunofluorescência , Trato Gastrointestinal/citologia , Genitália/citologia , Humanos , Sistema Imunitário/citologia , Sistema Respiratório/citologia
12.
Am J Physiol Regul Integr Comp Physiol ; 321(1): R79-R90, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-34105399

RESUMO

Although recognized as an important endocrine organ, little is known about the mechanisms through which adipose tissue can regulate inflammatory responses in distant tissues, such as lung that are affected by obesity. To explore potential mechanisms, male C57BL/6J mice were provided either high-fat diet, low-fat diet, or were provided a high-fat diet then switched to the low-fat diet to promote weight loss. Visceral adipocytes were then cultured in vitro to generate conditioned media (CM) that was used to treat both primary (mouse tracheal epithelial cells; MTECs) and immortalized (mouse-transformed club cells; MTCCs) airway epithelial cells. Adiponectin levels were greatly depressed in the CM from both obese and diet-switched adipocytes relative to mice continually fed the low-fat diet. MTECs from mice with obesity secreted higher baseline levels of inflammatory cytokines than MTECs from lean or diet-switched mice. MTECs treated with obese adipocyte CM increased their secretion of these cytokines compared with MTECs treated with lean CM. Diet-switched CM modestly decreased the production of cytokines compared with obese CM, and these effects were recapitulated when the CM was used to treat MTCCs. Adipose stromal vascular cells from mice with obesity expressed genes consistent with an M1 macrophage phenotype and decreased eosinophil abundance compared with lean stromal vascular fraction, a profile that persisted in the lean diet-switched mice despite substantial weight loss. Soluble factors secreted from obese adipocytes exert a proinflammatory effect on airway epithelial cells, and these alterations are attenuated by diet-induced weight loss, which could have implications for the airway dysfunction related to obese asthma and its mitigation by weight loss.


Assuntos
Adipócitos/fisiologia , Tecido Adiposo/citologia , Células Epiteliais/fisiologia , Inflamação/complicações , Obesidade/induzido quimicamente , Animais , Linhagem Celular , Técnicas de Cocultura , Dieta Hiperlipídica , Humanos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Sistema Respiratório/citologia
13.
Vet Microbiol ; 257: 109067, 2021 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-33862331

RESUMO

Respiratory diseases negatively impact the global goat industry, but are understudied. There is a shortage of established and biological relevant in vitro or ex vivo assays to study caprine respiratory infections. Here, we describe the establishment of an in vitro system based on well-differentiated caprine airway epithelial cell (AEC) cultures grown under air liquid interface conditions as an experimental platform to study caprine respiratory pathogens. The functional differentiation of the AEC cultures was monitored and confirmed by light and immunofluorescence microscopy, scanning electron microscopy and examination of histological sections. We validated the functionality of the platform by studying Influenza D Virus (IDV) infection and Mycoplasma mycoides subsp. capri (Mmc) colonization over 5 days, including monitoring of infectious agents by titration and qPCR as well as colour changing units, respectively. The inoculation of caprine AEC cultures with IDV showed that efficient viral replication takes place, and revealed that IDV has a marked cell tropism for ciliated cells. Furthermore, AEC cultures were successfully infected with Mmc using a multiplicity of infection of 0.1 and colonization was monitored over several days. Altogether, these results demonstrate that our newly-established caprine AEC cultures can be used to investigate host-pathogen interactions of caprine respiratory pathogens.


Assuntos
Técnicas de Cultura de Células/métodos , Técnicas de Cultura de Células/veterinária , Células Epiteliais/microbiologia , Células Epiteliais/virologia , Mucosa Respiratória/microbiologia , Mucosa Respiratória/virologia , Sistema Respiratório/citologia , Animais , Brônquios/citologia , Diferenciação Celular , Células Cultivadas , Cabras , Interações Hospedeiro-Patógeno , Microscopia Eletrônica de Varredura , Mycoplasma/fisiologia , Thogotovirus/fisiologia , Tropismo Viral , Replicação Viral/fisiologia
14.
Virology ; 559: 89-99, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33862336

RESUMO

Influenza D virus (IDV) is a novel type of influenza virus that infects and causes respiratory illness in bovines. Lack of host-specific in vitro model that can recapitulate morphology and physiology of in vivo airway epithelial cells has impeded the study of IDV infection. Here, we established and characterized bovine primary respiratory epithelial cells from nasal turbinate, soft palate, and trachea of the same calf. All three cell types showed characteristics peculiar of epithelial cells, polarized into apical-basolateral membrane, and formed tight junctions. Furthermore, these cells expressed both α-2,3- and α-2,6-linked sialic acids with α-2,3 linkage being more abundant. IDV strains replicated to high titers in these cells, while influenza A and B viruses exhibited moderate to low titers, with influenza C virus replication not detected. These findings suggest that bovine primary airway epithelial cells can be utilized to model infection biology and pathophysiology of IDV and other respiratory pathogens.


Assuntos
Células Epiteliais/virologia , Sistema Respiratório/citologia , Thogotovirus/fisiologia , Replicação Viral , Animais , Bovinos , Contagem de Células , Células Cultivadas , Palato Mole/citologia , Palato Mole/virologia , Sistema Respiratório/virologia , Traqueia/citologia , Traqueia/virologia , Conchas Nasais/citologia , Conchas Nasais/virologia , Virologia/métodos
15.
Am J Physiol Lung Cell Mol Physiol ; 321(1): L1-L5, 2021 07 01.
Artigo em Inglês | MEDLINE | ID: mdl-33909498

RESUMO

Abnormal airway remodeling is a common pathological change seen in chronic respiratory diseases. Altered proliferation and differentiation of airway smooth muscle cells (ASMCs) are the major components of airway remodeling, and the resultant structural abnormalities are difficult to restore. Understanding of airway smooth muscle regulation is urgently needed to identify potential intervention targets. MYOCD (or myocardin) and myocardin-related transcription factors (MRTFs) are key cotranscription factors in muscle growth, which have not been extensively investigated in airway smooth muscle cells. In addition, the RhoA/ROCK signaling pathway is known to play an important role in airway remodeling partly through regulating the proliferation and differentiation of ASMCs, which may be connected with MYOCD/MRTF cotranscription factors [Kumawat et al. (Am J Physiol Lung Cell Mol Physiol 311: L529-L537, 2016); Lagna et al. (J Biol Chem 282: 37244-37255, 2007)]. This review focuses on this newly recognized and potentially important RhoA/ROCK-MYOCD/MRTFs pathway in controlling airway smooth muscle growth and remodeling.


Assuntos
Remodelação das Vias Aéreas , Miócitos de Músculo Liso/citologia , Proteínas Nucleares/metabolismo , Sistema Respiratório/citologia , Transativadores/metabolismo , Quinases Associadas a rho/metabolismo , Proteína rhoA de Ligação ao GTP/metabolismo , Humanos , Miócitos de Músculo Liso/fisiologia , Proteínas Nucleares/genética , Sistema Respiratório/metabolismo , Transativadores/genética , Quinases Associadas a rho/genética , Proteína rhoA de Ligação ao GTP/genética
16.
Nat Microbiol ; 6(7): 899-909, 2021 07.
Artigo em Inglês | MEDLINE | ID: mdl-33907312

RESUMO

SARS-CoV-2 entry requires sequential cleavage of the spike glycoprotein at the S1/S2 and the S2' cleavage sites to mediate membrane fusion. SARS-CoV-2 has a polybasic insertion (PRRAR) at the S1/S2 cleavage site that can be cleaved by furin. Using lentiviral pseudotypes and a cell-culture-adapted SARS-CoV-2 virus with an S1/S2 deletion, we show that the polybasic insertion endows SARS-CoV-2 with a selective advantage in lung cells and primary human airway epithelial cells, but impairs replication in Vero E6, a cell line used for passaging SARS-CoV-2. Using engineered spike variants and live virus competition assays and by measuring growth kinetics, we find that the selective advantage in lung and primary human airway epithelial cells depends on the expression of the cell surface protease TMPRSS2, which enables endosome-independent virus entry by a route that avoids antiviral IFITM proteins. SARS-CoV-2 virus lacking the S1/S2 furin cleavage site was shed to lower titres from infected ferrets and was not transmitted to cohoused sentinel animals, unlike wild-type virus. Analysis of 100,000 SARS-CoV-2 sequences derived from patients and 24 human postmortem tissues showed low frequencies of naturally occurring mutants that harbour deletions at the polybasic site. Taken together, our findings reveal that the furin cleavage site is an important determinant of SARS-CoV-2 transmission.


Assuntos
COVID-19/transmissão , Furina/metabolismo , SARS-CoV-2/fisiologia , Glicoproteína da Espícula de Coronavírus/metabolismo , Animais , COVID-19/virologia , Catepsinas/metabolismo , Chlorocebus aethiops , Endossomos/metabolismo , Células Epiteliais , Furões , Humanos , Evasão da Resposta Imune , Proteínas de Membrana/metabolismo , Proteínas de Ligação a RNA/metabolismo , Sistema Respiratório/citologia , Sistema Respiratório/virologia , Serina Endopeptidases/metabolismo , Glicoproteína da Espícula de Coronavírus/química , Glicoproteína da Espícula de Coronavírus/genética , Células Vero , Empacotamento do Genoma Viral , Internalização do Vírus , Replicação Viral , Eliminação de Partículas Virais
17.
Biomed Pharmacother ; 139: 111583, 2021 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-33901875

RESUMO

TMEM16A is a Ca2+-activated Cl- channel involved in mucus secretion in inflamed airways and proposed as a drug target for diseases associated with mucus hypersecretion including asthma. This study aimed to identify novel inhibitors of TMEM16A-mediated Cl- secretion in airway epithelial cells from a collection of compounds isolated from fungi indigenous in Thailand and examine its potential utility in mitigating airway mucus secretion using Calu-3 cells as a study model. Screening of > 400 fungal metabolites revealed purpactin A isolated from a soil-derived fungus Penicillium aculeatum PSU-RSPG105 as an inhibitor of TMEM16A-mediated Cl- transport with an IC50 value of ~2 µM. A consistent inhibitory effect of purpactin A on TMEM16A were observed regardless of TMEM16A activators or in the presence of an inhibitor of Ca2+/calmodulin-dependent protein kinase II (CaMKII), a negative regulator of TMEM16A. In addition, purpactin A did not affect cell viability, epithelial barrier integrity and activities of membrane transport proteins essential for maintaining airway hydration including CFTR Cl- channels and apical BK K+ channels. Intriguingly, purpactin A prevented a Ca2+-induced mucin release in cytokine-treated airway cells. Taken together, purpactin A represents the first class of TMEM16A inhibitor derived from fungus, which may be beneficial for the treatment of diseases associated with mucus hypersecretion.


Assuntos
Anoctamina-1/antagonistas & inibidores , Células Epiteliais/efeitos dos fármacos , Compostos Heterocíclicos com 3 Anéis/farmacologia , Mucinas/metabolismo , Animais , Anoctamina-1/metabolismo , Anoctamina-1/fisiologia , Cálcio/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , Células Epiteliais/metabolismo , Células Epiteliais/fisiologia , Humanos , Ratos Endogâmicos F344 , Sistema Respiratório/citologia , Talaromyces
18.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Artigo em Inglês | MEDLINE | ID: mdl-33836570

RESUMO

Measles virus (MeV) is highly infectious by the respiratory route and remains an important cause of childhood mortality. However, the process by which MeV infection is efficiently established in the respiratory tract is controversial with suggestions that respiratory epithelial cells are not susceptible to infection from the apical mucosal surface. Therefore, it has been hypothesized that infection is initiated in lung macrophages or dendritic cells and that epithelial infection is subsequently established through the basolateral surface by infected lymphocytes. To better understand the process of respiratory tract initiation of MeV infection, primary differentiated respiratory epithelial cell cultures were established from rhesus macaque tracheal and nasal tissues. Infection of these cultures with MeV from the apical surface was more efficient than from the basolateral surface with shedding of viable MeV-producing multinucleated giant cell (MGC) syncytia from the surface. Despite presence of MGCs and infectious virus in supernatant fluids after apical infection, infected cells were not detected in the adherent epithelial sheet and transepithelial electrical resistance was maintained. After infection from the basolateral surface, epithelial damage and large clusters of MeV-positive cells were observed. Treatment with fusion inhibitory peptides showed that MeV production after apical infection was not dependent on infection of the basolateral surface. These results are consistent with the hypothesis that MeV infection is initiated by apical infection of respiratory epithelial cells with subsequent infection of lymphoid tissue and systemic spread.


Assuntos
Diferenciação Celular , Células Gigantes/metabolismo , Vírus do Sarampo/fisiologia , Sistema Respiratório/metabolismo , Animais , Células Cultivadas , Chlorocebus aethiops , Células Epiteliais/metabolismo , Feminino , Macaca mulatta , Masculino , Sistema Respiratório/citologia , Células Vero
19.
Elife ; 102021 04 09.
Artigo em Inglês | MEDLINE | ID: mdl-33835028

RESUMO

Virus propagation methods generally use transformed cell lines to grow viruses from clinical specimens, which may force viruses to rapidly adapt to cell culture conditions, a process facilitated by high viral mutation rates. Upon propagation in VeroE6 cells, SARS-CoV-2 may mutate or delete the multibasic cleavage site (MBCS) in the spike protein. Previously, we showed that the MBCS facilitates serine protease-mediated entry into human airway cells (Mykytyn et al., 2021). Here, we report that propagating SARS-CoV-2 on the human airway cell line Calu-3 - that expresses serine proteases - prevents cell culture adaptations in the MBCS and directly adjacent to the MBCS (S686G). Similar results were obtained using a human airway organoid-based culture system for SARS-CoV-2 propagation. Thus, in-depth knowledge on the biology of a virus can be used to establish methods to prevent cell culture adaptation.


Assuntos
Células Epiteliais , SARS-CoV-2/fisiologia , Glicoproteína da Espícula de Coronavírus/genética , Cultura de Vírus/métodos , Internalização do Vírus , Animais , Linhagem Celular , Chlorocebus aethiops , Células Epiteliais/citologia , Células Epiteliais/metabolismo , Células Epiteliais/virologia , Humanos , Proteólise , Sistema Respiratório/citologia , Sistema Respiratório/virologia , Serina Proteases/metabolismo
20.
Hum Cell ; 34(3): 785-799, 2021 May.
Artigo em Inglês | MEDLINE | ID: mdl-33683656

RESUMO

Club cells are critical in maintaining airway integrity via, in part, secretion of immunomodulatory Club cell 10 kd protein (CC10) and xenobiotic detoxification. Aryl hydrocarbon receptor (AhR) is important in xenobiotic metabolism, but its role in Club cell function is unclear. To this end, an AhR ligand, 6-formylindolo[3,2-b]carbazole (FICZ, 10 nM) was found to induce, in a ligand and AhR-dependent manner, endoplasmic reticulum stress, phospholipid remodeling, free fatty acid and triglyceride synthesis, leading to perilipin 2-dependent lipid droplet (LD) biogenesis in a Club cell-like cell line, NL20. The increase in LDs was due, in part, to the blockade of adipose triglyceride lipase to LDs, while perilipin 5 facilitated LDs-mitochondria connection, leading to the breakdown of LDs via mitochondrial ß-oxidation and acetyl-coA generation. In FICZ-treated cells, increased CC10 secretion and its intracellular association with LDs were noted. Administration of low (0.28 ng), medium (1.42 ng), and high (7.10 ng) doses of FICZ in C57BL/6 mice significantly enhanced lipopolysaccharide (LPS, 0.1 µg)-induced airway inflammation, mucin secretion, pro-inflammatory cytokines and CC10 in the bronchoalveolar lavage fluids, as compared to those seen in mice receiving LPS alone, suggesting the importance of AhR signaling in controlling the metabolic homeostasis and functions of Club cells.


Assuntos
Células Epiteliais/metabolismo , Gotículas Lipídicas/metabolismo , Receptores de Hidrocarboneto Arílico/fisiologia , Sistema Respiratório/citologia , Animais , Carbazóis/farmacologia , Linhagem Celular , Humanos , Inativação Metabólica , Ligantes , Camundongos Endogâmicos C57BL , Mitocôndrias/metabolismo , Perilipina-1/farmacologia , Transdução de Sinais/fisiologia , Uteroglobina/metabolismo , Xenobióticos/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...