Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 911
Filtrar
Más filtros

Tipo del documento
Intervalo de año de publicación
1.
Cell ; 186(1): 112-130.e20, 2023 01 05.
Artículo en Inglés | MEDLINE | ID: mdl-36580912

RESUMEN

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.


Asunto(s)
COVID-19 , Sistema Respiratorio , SARS-CoV-2 , Humanos , Cilios/fisiología , Cilios/virología , COVID-19/virología , Sistema Respiratorio/citología , Sistema Respiratorio/virología , SARS-CoV-2/fisiología , Microvellosidades/fisiología , Microvellosidades/virología , Internalización del Virus , Células Epiteliales/fisiología , Células Epiteliales/virología
2.
Nature ; 626(7998): 392-400, 2024 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-38086420

RESUMEN

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.


Asunto(s)
Linfocitos T CD8-positivos , Memoria Inmunológica , Células T de Memoria , Infecciones por Paramyxoviridae , Sistema Respiratorio , Animales , Ratones , Linfocitos T CD8-positivos/inmunología , Modelos Animales de Enfermedad , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Inmunidad Colectiva/inmunología , Memoria Inmunológica/inmunología , Interferón gamma/inmunología , Células T de Memoria/inmunología , Paramyxoviridae/inmunología , Paramyxoviridae/fisiología , Infecciones por Paramyxoviridae/inmunología , Infecciones por Paramyxoviridae/prevención & control , Infecciones por Paramyxoviridae/transmisión , Infecciones por Paramyxoviridae/virología , Sistema Respiratorio/citología , Sistema Respiratorio/inmunología , Sistema Respiratorio/virología , Transcripción Genética , Humanos
4.
Nucleic Acids Res ; 51(8): 3650-3670, 2023 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-36772828

RESUMEN

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.


Asunto(s)
Epitelio , Sistema Respiratorio , Estrés del Retículo Endoplásmico , Endorribonucleasas/metabolismo , Genómica , Hexosaminas , Proteínas Serina-Treonina Quinasas/genética , Proteínas Serina-Treonina Quinasas/metabolismo , Factores de Transcripción/genética , Factores de Transcripción/metabolismo , Respuesta de Proteína Desplegada , Epitelio/fisiología , Sistema Respiratorio/citología , Humanos
5.
J Virol ; 97(10): e0105123, 2023 10 31.
Artículo en Inglés | MEDLINE | ID: mdl-37732787

RESUMEN

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.


Asunto(s)
Células Cultivadas , Células Epiteliales , Virus del Sarampión , Sarampión , Sistema Respiratorio , Humanos , Células Epiteliales/virología , Epitelio , Sarampión/virología , Sistema Respiratorio/citología
6.
Proc Natl Acad Sci U S A ; 118(11)2021 03 16.
Artículo en Inglés | MEDLINE | ID: mdl-33836570

RESUMEN

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.


Asunto(s)
Diferenciación Celular , Células Gigantes/metabolismo , Virus del Sarampión/fisiología , Sistema Respiratorio/metabolismo , Animales , Células Cultivadas , Chlorocebus aethiops , Células Epiteliales/metabolismo , Femenino , Macaca mulatta , Masculino , Sistema Respiratorio/citología , Células Vero
7.
Nature ; 539(7630): 570-574, 2016 11 24.
Artículo en Inglés | MEDLINE | ID: mdl-27820945

RESUMEN

Professional phagocytes (such as macrophages) and non-professional phagocytes (such as epithelial cells) clear billions of apoptotic cells and particles on a daily basis. Although professional and non-professional macrophages reside in proximity in most tissues, whether they communicate with each other during cell clearance, and how this might affect inflammation, is not known. Here we show that macrophages, through the release of a soluble growth factor and microvesicles, alter the type of particles engulfed by non-professional phagocytes and influence their inflammatory response. During phagocytosis of apoptotic cells or in response to inflammation-associated cytokines, macrophages released insulin-like growth factor 1 (IGF-1). The binding of IGF-1 to its receptor on non-professional phagocytes redirected their phagocytosis, such that uptake of larger apoptotic cells was reduced whereas engulfment of microvesicles was increased. IGF-1 did not alter engulfment by macrophages. Macrophages also released microvesicles, whose uptake by epithelial cells was enhanced by IGF-1 and led to decreased inflammatory responses by epithelial cells. Consistent with these observations, deletion of IGF-1 receptor in airway epithelial cells led to exacerbated lung inflammation after allergen exposure. These genetic and functional studies reveal that IGF-1- and microvesicle-dependent communication between macrophages and epithelial cells can critically influence the magnitude of tissue inflammation in vivo.


Asunto(s)
Células Epiteliales/citología , Macrófagos/citología , Fagocitos/citología , Fagocitosis , Neumonía , Alérgenos/inmunología , Animales , Apoptosis , Comunicación Celular , Citocinas/metabolismo , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Femenino , Fibroblastos/citología , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Masculino , Ratones , Fagocitos/inmunología , Fagocitos/metabolismo , Neumonía/inmunología , Neumonía/metabolismo , Receptor IGF Tipo 1/deficiencia , Receptor IGF Tipo 1/metabolismo , Sistema Respiratorio/citología , Somatomedinas/metabolismo
8.
Am J Respir Cell Mol Biol ; 64(3): 308-317, 2021 03.
Artículo en Inglés | MEDLINE | ID: mdl-33196316

RESUMEN

The human airway epithelium is essential in homeostasis, and epithelial dysfunction contributes to chronic airway disease. Development of flow-cytometric methods to characterize subsets of airway epithelial cells will enable further dissection of airway epithelial biology. Leveraging single-cell RNA-sequencing data in combination with known cell type-specific markers, we developed panels of antibodies to characterize and isolate the major airway epithelial subsets (basal, ciliated, and secretory cells) from human bronchial epithelial-cell cultures. We also identified molecularly distinct subpopulations of secretory cells and demonstrated cell subset-specific expression of low-abundance transcripts and microRNAs that are challenging to analyze with current single-cell RNA-sequencing methods. These new tools will be valuable for analyzing and separating airway epithelial subsets and interrogating airway epithelial biology.


Asunto(s)
Separación Celular/métodos , Células Epiteliales/citología , Citometría de Flujo/métodos , Sistema Respiratorio/citología , Anticuerpos/metabolismo , Biomarcadores/metabolismo , Humanos
9.
J Biol Chem ; 295(19): 6721-6740, 2020 05 08.
Artículo en Inglés | MEDLINE | ID: mdl-32241907

RESUMEN

Protease-activated receptor 2 (PAR-2) is activated by secreted proteases from immune cells or fungi. PAR-2 is normally expressed basolaterally in differentiated nasal ciliated cells. We hypothesized that epithelial remodeling during diseases characterized by cilial loss and squamous metaplasia may alter PAR-2 polarization. Here, using a fluorescent arrestin assay, we confirmed that the common fungal airway pathogen Aspergillus fumigatus activates heterologously-expressed PAR-2. Endogenous PAR-2 activation in submerged airway RPMI 2650 or NCI-H520 squamous cells increased intracellular calcium levels and granulocyte macrophage-colony-stimulating factor, tumor necrosis factor α, and interleukin (IL)-6 secretion. RPMI 2650 cells cultured at an air-liquid interface (ALI) responded to apically or basolaterally applied PAR-2 agonists. However, well-differentiated primary nasal epithelial ALIs responded only to basolateral PAR-2 stimulation, indicated by calcium elevation, increased cilia beat frequency, and increased fluid and cytokine secretion. We exposed primary cells to disease-related modifiers that alter epithelial morphology, including IL-13, cigarette smoke condensate, and retinoic acid deficiency, at concentrations and times that altered epithelial morphology without causing breakdown of the epithelial barrier to model early disease states. These altered primary cultures responded to both apical and basolateral PAR-2 stimulation. Imaging nasal polyps and control middle turbinate explants, we found that nasal polyps, but not turbinates, exhibit apical calcium responses to PAR-2 stimulation. However, isolated ciliated cells from both polyps and turbinates maintained basolateral PAR-2 polarization, suggesting that the calcium responses originated from nonciliated cells. Altered PAR-2 polarization in disease-remodeled epithelia may enhance apical responses and increase sensitivity to inhaled proteases.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias) , Receptor PAR-2/metabolismo , Sistema Respiratorio/citología , Transducción de Señal , Aspergillus fumigatus/fisiología , Línea Celular , Citocinas/metabolismo , Epitelio/metabolismo , Humanos , Sistema Respiratorio/microbiología
10.
Am J Physiol Lung Cell Mol Physiol ; 321(1): L1-L5, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33909498

RESUMEN

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.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias) , Miocitos del Músculo Liso/citología , Proteínas Nucleares/metabolismo , Sistema Respiratorio/citología , Transactivadores/metabolismo , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Humanos , Miocitos del Músculo Liso/fisiología , Proteínas Nucleares/genética , Sistema Respiratorio/metabolismo , Transactivadores/genética , Quinasas Asociadas a rho/genética , Proteína de Unión al GTP rhoA/genética
11.
Clin Exp Immunol ; 204(1): 134-143, 2021 04.
Artículo en Inglés | MEDLINE | ID: mdl-33423291

RESUMEN

Interferon regulatory factor 5 (IRF5) is a master regulator of macrophage phenotype and a key transcription factor involved in expression of proinflammatory cytokine responses to microbial and viral infection. Here, we show that IRF5 controls cellular and metabolic responses. By integrating ChIP sequencing (ChIP-Seq) and assay for transposase-accessible chromatin using sequencing (ATAC)-seq data sets, we found that IRF5 directly regulates metabolic genes such as hexokinase-2 (Hk2). The interaction of IRF5 and metabolic genes had a functional consequence, as Irf5-/- airway macrophages but not bone marrow-derived macrophages (BMDMs) were characterized by a quiescent metabolic phenotype at baseline and had reduced ability to utilize oxidative phosphorylation after Toll-like receptor (TLR)-3 activation, in comparison to controls, ex vivo. In a murine model of influenza infection, IRF5 deficiency had no effect on viral load in comparison to wild-type controls but controlled metabolic responses to viral infection, as IRF5 deficiency led to reduced expression of Sirt6 and Hk2. Together, our data indicate that IRF5 is a key component of AM metabolic responses following influenza infection and TLR-3 activation.


Asunto(s)
Metabolismo Energético/inmunología , Regulación de la Expresión Génica/inmunología , Factores Reguladores del Interferón/inmunología , Macrófagos/inmunología , Sistema Respiratorio/citología , Animales , Células Cultivadas , Secuenciación de Inmunoprecipitación de Cromatina/métodos , Metabolismo Energético/genética , Femenino , Hexoquinasa/genética , Hexoquinasa/inmunología , Hexoquinasa/metabolismo , Humanos , Factores Reguladores del Interferón/genética , Factores Reguladores del Interferón/metabolismo , Macrófagos/metabolismo , Ratones Endogámicos C57BL , Ratones Noqueados , Sirtuinas/genética , Sirtuinas/inmunología , Sirtuinas/metabolismo , Receptor Toll-Like 3/genética , Receptor Toll-Like 3/inmunología , Receptor Toll-Like 3/metabolismo
12.
Am J Physiol Regul Integr Comp Physiol ; 321(1): R79-R90, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-34105399

RESUMEN

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.


Asunto(s)
Adipocitos/fisiología , Tejido Adiposo/citología , Células Epiteliales/fisiología , Inflamación/complicaciones , Obesidad/inducido químicamente , Animales , Línea Celular , Técnicas de Cocultivo , Dieta Alta en Grasa , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Sistema Respiratorio/citología
13.
J Med Virol ; 93(12): 6671-6685, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34324210

RESUMEN

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.


Asunto(s)
Enzima Convertidora de Angiotensina 2/metabolismo , Tracto Gastrointestinal/virología , Genitales/virología , Receptor Celular 1 del Virus de la Hepatitis A/metabolismo , Sistema Inmunológico/virología , Neuropilina-1/metabolismo , Sistema Respiratorio/virología , SARS-CoV-2/fisiología , Serina Endopeptidasas/metabolismo , Internalización del Virus , Western Blotting , COVID-19/metabolismo , COVID-19/virología , Línea Celular , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Tracto Gastrointestinal/citología , Genitales/citología , Humanos , Sistema Inmunológico/citología , Sistema Respiratorio/citología
14.
FASEB J ; 34(1): 1652-1664, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31914670

RESUMEN

Conditions such as asthma and inflammatory bowel disease are characterized by aberrant smooth muscle contraction. It has proven difficult to develop human cell-based models that mimic acute muscle contraction in 2D in vitro cultures due to the nonphysiological chemical and mechanical properties of lab plastics that do not allow for muscle cell contraction. To enhance the relevance of in vitro models for human disease, we describe how functional 3D smooth muscle tissue that exhibits physiological and pharmacologically relevant acute contraction and relaxation responses can be reproducibly fabricated using a unique microfluidic 3D bioprinting technology. Primary human airway and intestinal smooth muscle cells were printed into rings of muscle tissue at high density and viability. Printed tissues contracted to physiological concentrations of histamine (0.01-100 µM) and relaxed to salbutamol, a pharmacological compound used to relieve asthmatic exacerbations. The addition of TGFß to airway muscle rings induced an increase in unstimulated muscle shortening and a decreased response to salbutamol, a phenomenon which also occurs in chronic lung diseases. Results indicate that the 3D bioprinted smooth muscle is a physiologically relevant in vitro model that can be utilized to study disease pathways and the effects of novel therapeutics on acute contraction and chronic tissue stenosis.


Asunto(s)
Bioimpresión/métodos , Microfluídica/métodos , Músculo Liso/citología , Miocitos del Músculo Liso/citología , Sistema Respiratorio/citología , Albuterol/farmacología , Asma/tratamiento farmacológico , Asma/patología , Células Cultivadas , Humanos , Contracción Muscular/efectos de los fármacos , Contracción Muscular/fisiología , Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/efectos de los fármacos , Impresión Tridimensional , Sistema Respiratorio/efectos de los fármacos , Ingeniería de Tejidos/métodos
15.
Virol J ; 18(1): 169, 2021 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-34404450

RESUMEN

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.


Asunto(s)
Células Endoteliales/virología , Virus Puumala , Replicación Viral , Fiebre Hemorrágica con Síndrome Renal , Humanos , Integrinas , Cultivo Primario de Células , Virus Puumala/fisiología , Sistema Respiratorio/citología , Sistema Respiratorio/virología
16.
Ann Allergy Asthma Immunol ; 126(2): 143-151, 2021 02.
Artículo en Inglés | MEDLINE | ID: mdl-33122124

RESUMEN

OBJECTIVE: To review the latest discoveries regarding the role of tuft cells in the pathogenesis of chronic rhinosinusitis (CRS) with nasal polyposis and asthma. DATA SOURCES: Reviews and primary research manuscripts were identified from PubMed, Google, and bioRxiv using the search words airway epithelium, nasal polyposis, CRS or asthma and chemoreceptor cell, solitary chemosensory cell, brush cell, microvillus cell, and tuft cell. STUDY SELECTIONS: Studies were selected on the basis of novelty and likely relevance to the functions of tuft cells in chronic inflammatory diseases in the upper and lower airways. RESULTS: Tuft cells coordinate a variety of immune responses throughout the body. After the activation of bitter-taste receptors, tuft cells coordinate the secretion of antimicrobial products by adjacent epithelial cells and initiate the calcium-dependent release of acetylcholine resulting in neurogenic inflammation, including mast cell degranulation and plasma extravasation. Tuft cells are also the dominant source of interleukin-25 and a significant source of cysteinyl leukotrienes that play a role in initiating inflammatory processes in the airway. Tuft cells have also been found to seem de novo in the distal airway after a viral infection, implicating these cells in dysplastic remodeling in the distal lung in the pathogenesis of asthma. CONCLUSION: Tuft cells bridge innate and adaptive immunes responses and play an upstream role in initiating type 2 inflammation in the upper and possibly the lower airway. The role of tuft cells in respiratory pathophysiology must be further investigated, because tuft cells are putative high-value therapeutic targets for novel therapeutics in CRS with nasal polyps and asthma.


Asunto(s)
Asma/inmunología , Células Epiteliales/inmunología , Pólipos Nasales/inmunología , Sistema Respiratorio/citología , Rinitis/inmunología , Sinusitis/inmunología , Acetilcolina/inmunología , Animales , Enfermedad Crónica , Eicosanoides/inmunología , Humanos , Interleucina-17/inmunología , Sistema Respiratorio/inmunología
17.
Nature ; 526(7571): 122-5, 2015 Oct 01.
Artículo en Inglés | MEDLINE | ID: mdl-26416728

RESUMEN

Influenza A viruses pose a major public health threat by causing seasonal epidemics and sporadic pandemics. Their epidemiological success relies on airborne transmission from person to person; however, the viral properties governing airborne transmission of influenza A viruses are complex. Influenza A virus infection is mediated via binding of the viral haemagglutinin (HA) to terminally attached α2,3 or α2,6 sialic acids on cell surface glycoproteins. Human influenza A viruses preferentially bind α2,6-linked sialic acids whereas avian influenza A viruses bind α2,3-linked sialic acids on complex glycans on airway epithelial cells. Historically, influenza A viruses with preferential association with α2,3-linked sialic acids have not been transmitted efficiently by the airborne route in ferrets. Here we observe efficient airborne transmission of a 2009 pandemic H1N1 (H1N1pdm) virus (A/California/07/2009) engineered to preferentially bind α2,3-linked sialic acids. Airborne transmission was associated with rapid selection of virus with a change at a single HA site that conferred binding to long-chain α2,6-linked sialic acids, without loss of α2,3-linked sialic acid binding. The transmissible virus emerged in experimentally infected ferrets within 24 hours after infection and was remarkably enriched in the soft palate, where long-chain α2,6-linked sialic acids predominate on the nasopharyngeal surface. Notably, presence of long-chain α2,6-linked sialic acids is conserved in ferret, pig and human soft palate. Using a loss-of-function approach with this one virus, we demonstrate that the ferret soft palate, a tissue not normally sampled in animal models of influenza, rapidly selects for transmissible influenza A viruses with human receptor (α2,6-linked sialic acids) preference.


Asunto(s)
Adaptación Fisiológica , Subtipo H1N1 del Virus de la Influenza A/fisiología , Paladar Blando/metabolismo , Paladar Blando/virología , Receptores Virales/metabolismo , Selección Genética , Adaptación Fisiológica/genética , Animales , Células Epiteliales/metabolismo , Células Epiteliales/virología , Femenino , Hurones/virología , Glicoproteínas Hemaglutininas del Virus de la Influenza/genética , Glicoproteínas Hemaglutininas del Virus de la Influenza/metabolismo , Humanos , Subtipo H1N1 del Virus de la Influenza A/química , Subtipo H1N1 del Virus de la Influenza A/genética , Masculino , Datos de Secuencia Molecular , Infecciones por Orthomyxoviridae/transmisión , Infecciones por Orthomyxoviridae/virología , Paladar Blando/química , Sistema Respiratorio/citología , Sistema Respiratorio/metabolismo , Sistema Respiratorio/virología , Selección Genética/genética , Ácidos Siálicos/química , Ácidos Siálicos/metabolismo , Porcinos/virología
18.
Ecotoxicol Environ Saf ; 210: 111857, 2021 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-33421718

RESUMEN

OBJECTIVES: Benzo(a)pyrene (BaP) is a ubiquitous air pollutants, and BaP exposure leads to a risk of respiratory diseases. The oversecretion of airway mucus and high expression of mucin 5AC (MUC5AC) are associated with common respiratory disorders caused by air pollution. We aimed to investigate the effect of BaP on MUC5AC expression, especially the mechanisms by which BaP induces MUC5AC gene expression. METHODS: The human airway epithelial cell NCI-H292 was used to test the effects of BaP on the expression of MUC5AC in vitro. MUC5AC mRNA and protein expression were assessed with real-time quantitative PCR, immunochemistry, and western blotting. A luciferase assay was conducted to detect the activity of the promoter. The total cellular ROS and mitochondrial ROS were measured by corresponding probes. Small-interfering RNAs were used for gene silencing. AhR-overexpressing cell lines were constructed by transfection with AhR overexpression lentivirus. RESULTS: We found that BaP stimulation upregulated the MUC5AC mRNA and protein levels and activated the ERK pathway. Suppressing ERK with U0126 (an ERK inhibitor) or knocking down ERK with siRNA decreased BaP-induced MUC5AC expression. The luciferase activity transfected with the MUC5AC promoter and cAMP-response element (CRE) was increased after BaP treatment, whereas CREB siRNA suppressed the BaP-induced overexpression of MUC5AC. In addition, BaP increased mitochondrial ROS production, and Mito-TEMP, a mitochondrial ROS inhibitor, inhibited BaP-induced MUC5AC expression and ERK activation. BaP increased the mRNA levels of CYP1A1 and CYP1B1, while Alizarin, a CYP1s inhibitor, suppressed the effects of BaP, including the MUC5AC overexpression, ERK activation and mitochondrial ROS generation. BaP induced the translocation of aryl hydrocarbon receptor (AhR) from the cytoplasm to the nucleus. SiRNA-mediated knockdown or chemical inhibition of AhR decreased the BaP-induced expression of MUC5AC, while the overexpression of AhR significantly enhanced the BaP-induced expression of MUC5AC. ITE, an endogenous AhR ligand, also upregulated the mRNA and protein expression of MUC5AC. Furthermore, resveratrol treatment inhibited the BaP-induced MUC5AC overexpression, AhR translocation, mitochondrial ROS production and ERK pathway activation. CONCLUSION: Here, we highlighted the crucial role of AhR/mitochondrial ROS/ERK pathway activation in BaP-induced MUC5AC overexpression and identified resveratrol as a promising drug to reduce BaP-induced MUC5AC overexpression.


Asunto(s)
Contaminantes Atmosféricos/toxicidad , Benzo(a)pireno/toxicidad , Células Epiteliales/efectos de los fármacos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/metabolismo , Línea Celular , Células Epiteliales/metabolismo , Quinasas MAP Reguladas por Señal Extracelular/genética , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Humanos , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mucina 5AC/genética , Mucina 5AC/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Receptores de Hidrocarburo de Aril/genética , Receptores de Hidrocarburo de Aril/metabolismo , Sistema Respiratorio/citología , Transducción de Señal/efectos de los fármacos
19.
J Biol Chem ; 294(33): 12472-12482, 2019 08 16.
Artículo en Inglés | MEDLINE | ID: mdl-31248983

RESUMEN

Type 2 taste receptors (TAS2R) are G protein-coupled receptors first described in the gustatory system, but have also been shown to have extraoral localizations, including airway smooth muscle (ASM) cells, in which TAS2R have been reported to induce relaxation. TAS2R46 is an unexplored subtype that responds to its highly specific agonist absinthin. Here, we first demonstrate that, unlike other bitter-taste receptor agonists, absinthin alone (1 µm) in ASM cells does not induce Ca2+ signals but reduces histamine-induced cytosolic Ca2+ increases. To investigate this mechanism, we introduced into ASM cells aequorin-based Ca2+ probes targeted to the cytosol, subplasma membrane domain, or the mitochondrial matrix. We show that absinthin reduces cytosolic histamine-induced Ca2+ rises and simultaneously increases Ca2+ influx into mitochondria. We found that this effect is inhibited by the potent human TAS2R46 (hTAS2R46) antagonist 3ß-hydroxydihydrocostunolide and is no longer evident in hTAS2R46-silenced ASM cells, indicating that it is hTAS2R46-dependent. Furthermore, these changes were sensitive to the mitochondrial uncoupler carbonyl cyanide p-(trifluoromethoxy)phenyl-hydrazone (FCCP); the mitochondrial calcium uniporter inhibitor KB-R7943 (carbamimidothioic acid); the cytoskeletal disrupter latrunculin; and an inhibitor of the exchange protein directly activated by cAMP (EPAC), ESI-09. Similarly, the ß2 agonist salbutamol also could induce Ca2+ shuttling from cytoplasm to mitochondria, suggesting that this new mechanism might be generalizable. Moreover, forskolin and an EPAC activator mimicked this effect in HeLa cells. Our findings support the hypothesis that plasma membrane receptors can positively regulate mitochondrial Ca2+ uptake, adding a further facet to the ability of cells to encode complex Ca2+ signals.


Asunto(s)
Señalización del Calcio/efectos de los fármacos , Retículo Endoplásmico/metabolismo , Mitocondrias/metabolismo , Miocitos del Músculo Liso/metabolismo , Receptores Acoplados a Proteínas G/agonistas , Sistema Respiratorio/metabolismo , Sesquiterpenos de Guayano/farmacología , Calcio/metabolismo , Carbonil Cianuro p-Trifluorometoxifenil Hidrazona/farmacología , Línea Celular , Retículo Endoplásmico/genética , Células HeLa , Humanos , Mitocondrias/genética , Miocitos del Músculo Liso/citología , Receptores Acoplados a Proteínas G/genética , Receptores Acoplados a Proteínas G/metabolismo , Sistema Respiratorio/citología , Tiourea/análogos & derivados , Tiourea/farmacología
20.
Am J Physiol Lung Cell Mol Physiol ; 319(5): L843-L847, 2020 11 01.
Artículo en Inglés | MEDLINE | ID: mdl-32996784

RESUMEN

The incidence, severity, and mortality of ongoing coronavirus infectious disease 19 (COVID-19) is greater in men compared with women, but the underlying factors contributing to this sex difference are still being explored. In the current study, using primary isolated human airway smooth muscle (ASM) cells from normal males versus females as a model, we explored the effect of estrogen versus testosterone in modulating the expression of angiotensin converting enzyme 2 (ACE2), a cell entry point for severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Using confocal imaging, we found that ACE2 is expressed in human ASM. Furthermore, Western analysis of ASM cell lysates showed significantly lower ACE2 expression in females compared with males at baseline. In addition, ASM cells exposed to estrogen and testosterone for 24 h showed that testosterone significantly upregulates ACE2 expression in both males and females, whereas estrogen downregulates ACE2, albeit not significant compared with vehicle. These intrinsic and sex steroids induced differences may help explain sex differences in COVID-19.


Asunto(s)
Infecciones por Coronavirus/metabolismo , Peptidil-Dipeptidasa A/biosíntesis , Neumonía Viral/metabolismo , Sistema Respiratorio/metabolismo , Adulto , Anciano , Enzima Convertidora de Angiotensina 2 , COVID-19 , Células Cultivadas , Infecciones por Coronavirus/enzimología , Estrógenos/metabolismo , Estrógenos/farmacología , Femenino , Humanos , Masculino , Persona de Mediana Edad , Miocitos del Músculo Liso/citología , Miocitos del Músculo Liso/efectos de los fármacos , Miocitos del Músculo Liso/enzimología , Miocitos del Músculo Liso/metabolismo , Pandemias , Peptidil-Dipeptidasa A/metabolismo , Neumonía Viral/enzimología , Sistema Respiratorio/citología , Sistema Respiratorio/efectos de los fármacos , Sistema Respiratorio/enzimología , Factores Sexuales , Testosterona/metabolismo , Testosterona/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA