Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 20
Filtrar
1.
J Immunol ; 205(2): 489-501, 2020 07 15.
Artículo en Inglés | MEDLINE | ID: mdl-32540997

RESUMEN

Hypercapnia (HC), elevation of the partial pressure of CO2 in blood and tissues, is a risk factor for mortality in patients with severe acute and chronic lung diseases. We previously showed that HC inhibits multiple macrophage and neutrophil antimicrobial functions and increases the mortality of bacterial pneumonia in mice. In this study, we show that normoxic HC increases viral replication, lung injury, and mortality in mice infected with influenza A virus (IAV). Elevated CO2 increased IAV replication and inhibited antiviral gene and protein expression in macrophages in vivo and in vitro. HC potentiated IAV-induced activation of Akt, whereas specific pharmacologic inhibition or short hairpin RNA knockdown of Akt1 in alveolar macrophages blocked HC's effects on IAV growth and the macrophage antiviral response. Our findings suggest that targeting Akt1 or the downstream pathways through which elevated CO2 signals could enhance macrophage antiviral host defense and improve clinical outcomes in hypercapnic patients with advanced lung disease.


Asunto(s)
Hipercapnia/inmunología , Virus de la Influenza A/fisiología , Gripe Humana/inmunología , Pulmón/patología , Macrófagos/inmunología , Proteína Oncogénica v-akt/metabolismo , Infecciones por Orthomyxoviridae/inmunología , Animales , Células Cultivadas , Regulación de la Expresión Génica , Humanos , Inmunidad Celular , Terapia de Inmunosupresión , Pulmón/virología , Activación de Macrófagos , Ratones , Ratones Endogámicos C57BL , Transducción de Señal , Replicación Viral
2.
FASEB J ; 32(7): 3614-3622, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29405096

RESUMEN

Hypercapnia, elevated levels of CO2 in the blood, is a known marker for poor clinical prognosis and is associated with increased mortality in patients hospitalized with both bacterial and viral pneumonias. Although studies have established a connection between elevated CO2 levels and poor pneumonia outcomes, a mechanistic basis of this association has not yet been established. We previously reported that hypercapnia inhibits expression of key NF-κB-regulated, innate immune cytokines, TNF-α, and IL-6, in LPS-stimulated macrophages in vitro and in mice during Pseudomonas pneumonia. The transcription factor heat shock factor 1 (HSF1) is important in maintaining proteostasis during stress and has been shown to negatively regulate NF-κB activity. In this study, we tested the hypothesis that HSF1 activation in response to hypercapnia results in attenuated NF-κB-regulated gene expression. We found that hypercapnia induced the protein expression and nuclear accumulation of HSF1 in primary murine alveolar macrophages and in an alveolar macrophage cell line (MH-S). In MH-S cells treated with short interfering RNA targeting Hsf1, LPS-induced IL-6 and TNF-α release were elevated during exposure to hypercapnia. Pseudomonas-infected Hsf1+/+ (wild-type) mice, maintained in a hypercapnic environment, showed lower levels of IL-6 and TNF-α in bronchoalveolar lavage fluid and IL-1ß in lung tissue than did infected mice maintained in room air. In contrast, infected Hsf1+/- mice exposed to either hypercapnia or room air had similarly elevated levels of those cytokines. These results suggest that hypercapnia-mediated inhibition of NF-κB cytokine production is dependent on HSF1 expression and/or activation.-Lu, Z., Casalino-Matsuda, S. M., Nair, A., Buchbinder, A., Budinger, G. R. S., Sporn, P. H. S., Gates, K. L. A role for heat shock factor 1 in hypercapnia-induced inhibition of inflammatory cytokine expression.


Asunto(s)
Factores de Transcripción del Choque Térmico/metabolismo , Hipercapnia/metabolismo , Interleucinas/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Línea Celular , Células Cultivadas , Factores de Transcripción del Choque Térmico/genética , Interleucinas/genética , Macrófagos/metabolismo , Ratones , Ratones Endogámicos C57BL , FN-kappa B/metabolismo , Factor de Necrosis Tumoral alfa/genética
3.
J Immunol ; 196(2): 655-667, 2016 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-26643480

RESUMEN

Hypercapnia, elevated partial pressure of CO2 in blood and tissue, develops in many patients with chronic severe obstructive pulmonary disease and other advanced lung disorders. Patients with advanced disease frequently develop bacterial lung infections, and hypercapnia is a risk factor for mortality in such individuals. We previously demonstrated that hypercapnia suppresses induction of NF-κB-regulated innate immune response genes required for host defense in human, mouse, and Drosophila cells, and it increases mortality from bacterial infections in both mice and Drosophila. However, the molecular mediators of hypercapnic immune suppression are undefined. In this study, we report a genome-wide RNA interference screen in Drosophila S2* cells stimulated with bacterial peptidoglycan. The screen identified 16 genes with human orthologs whose knockdown reduced hypercapnic suppression of the gene encoding the antimicrobial peptide Diptericin (Dipt), but did not increase Dipt mRNA levels in air. In vivo tests of one of the strongest screen hits, zinc finger homeodomain 2 (Zfh2; mammalian orthologs ZFHX3/ATBF1 and ZFHX4), demonstrate that reducing zfh2 function using a mutation or RNA interference improves survival of flies exposed to elevated CO2 and infected with Staphylococcus aureus. Tissue-specific knockdown of zfh2 in the fat body, the major immune and metabolic organ of the fly, mitigates hypercapnia-induced reductions in Dipt and other antimicrobial peptides and improves resistance of CO2-exposed flies to infection. Zfh2 mutations also partially rescue hypercapnia-induced delays in egg hatching, suggesting that Zfh2's role in mediating responses to hypercapnia extends beyond the immune system. Taken together, to our knowledge, these results identify Zfh2 as the first in vivo mediator of hypercapnic immune suppression.


Asunto(s)
Proteínas de Unión al ADN/inmunología , Proteínas de Drosophila/inmunología , Hipercapnia/inmunología , Infecciones Estafilocócicas/complicaciones , Animales , Western Blotting , Modelos Animales de Enfermedad , Drosophila , Técnicas de Silenciamiento del Gen , Hipercapnia/microbiología , Inmunidad Innata/inmunología , Interferencia de ARN , Infecciones Estafilocócicas/inmunología , Staphylococcus aureus
4.
J Immunol ; 194(11): 5388-96, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25895534

RESUMEN

Hypercapnia, the elevation of CO2 in blood and tissue, commonly develops in patients with advanced lung disease and severe pulmonary infections, and it is associated with high mortality. We previously reported that hypercapnia alters expression of host defense genes, inhibits phagocytosis, and increases the mortality of Pseudomonas pneumonia in mice. However, the effect of hypercapnia on autophagy, a conserved process by which cells sequester and degrade proteins and damaged organelles that also plays a key role in antimicrobial host defense and pathogen clearance, has not previously been examined. In the present study we show that hypercapnia inhibits autophagy induced by starvation, rapamycin, LPS, heat-killed bacteria, and live bacteria in the human macrophage. Inhibition of autophagy by elevated CO2 was not attributable to acidosis. Hypercapnia also reduced macrophage killing of Pseudomonas aeruginosa. Moreover, elevated CO2 induced the expression of Bcl-2 and Bcl-xL, antiapoptotic factors that negatively regulate autophagy by blocking Beclin 1, an essential component of the autophagy initiation complex. Furthermore, small interfering RNA targeting Bcl-2 and Bcl-xL and the small molecule Z36, which blocks Bcl-2 and Bcl-xL binding to Beclin 1, prevented hypercapnic inhibition of autophagy and bacterial killing. These results suggest that targeting the Bcl-2/Bcl-xL-Beclin 1 interaction may hold promise for ameliorating hypercapnia-induced immunosuppression and improving resistance to infection in patients with advanced lung disease and hypercapnia.


Asunto(s)
Autofagia/inmunología , Hipercapnia/inmunología , Macrófagos Alveolares/inmunología , Proteínas Proto-Oncogénicas c-bcl-2/genética , Proteína bcl-X/genética , Acidosis , Animales , Proteínas Reguladoras de la Apoptosis/antagonistas & inhibidores , Autofagia/efectos de los fármacos , Beclina-1 , Dióxido de Carbono/sangre , Dióxido de Carbono/farmacología , Línea Celular , Humanos , Hipercapnia/sangre , Indoles/farmacología , Lipopolisacáridos , Enfermedades Pulmonares/patología , Macrófagos Alveolares/microbiología , Proteínas de la Membrana/antagonistas & inhibidores , Ratones , Fagocitosis/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Proteínas Proto-Oncogénicas c-bcl-2/biosíntesis , Pseudomonas aeruginosa/inmunología , Interferencia de ARN , ARN Interferente Pequeño , Sirolimus/farmacología , Proteína bcl-X/biosíntesis
6.
JCI Insight ; 9(4)2024 Jan 16.
Artículo en Inglés | MEDLINE | ID: mdl-38227369

RESUMEN

Hypercapnia, elevation of the partial pressure of CO2 in blood and tissues, is a risk factor for mortality in patients with severe acute and chronic lung diseases. We previously showed that hypercapnia inhibits multiple macrophage and neutrophil antimicrobial functions and that elevated CO2 increases the mortality of bacterial and viral pneumonia in mice. Here, we show that normoxic hypercapnia downregulates innate immune and antiviral gene programs in alveolar macrophages (AMØs). We also show that zinc finger homeobox 3 (Zfhx3) - a mammalian ortholog of zfh2, which mediates hypercapnic immune suppression in Drosophila - is expressed in mouse and human macrophages. Deletion of Zfhx3 in the myeloid lineage blocked the suppressive effect of hypercapnia on immune gene expression in AMØs and decreased viral replication, inflammatory lung injury, and mortality in hypercapnic mice infected with influenza A virus. To our knowledge, our results establish Zfhx3 as the first known mammalian mediator of CO2 effects on immune gene expression and lay the basis for future studies to identify therapeutic targets to interrupt hypercapnic immunosuppression in patients with advanced lung disease.


Asunto(s)
Virus de la Influenza A , Enfermedades Pulmonares , Animales , Humanos , Ratones , Dióxido de Carbono/farmacología , Drosophila , Proteínas de Homeodominio/genética , Hipercapnia , Pulmón , Macrófagos , Mamíferos
7.
J Biol Chem ; 287(50): 42288-98, 2012 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-23048036

RESUMEN

Cigarette smoke (CigS) exposure is associated with increased bronchial epithelial permeability and impaired barrier function. Primary cultures of normal human bronchial epithelial cells exposed to CigS exhibit decreased E-cadherin expression and reduced transepithelial electrical resistance. These effects were mediated by hyaluronan (HA) because inhibition of its synthesis with 4-methylumbelliferone prevented these effects, and exposure to HA fragments of <70 kDa mimicked these effects. We show that the HA receptor layilin is expressed apically in human airway epithelium and that cells infected with lentivirus expressing layilin siRNAs were protected against increased permeability triggered by both CigS and HA. We identified RhoA/Rho-associated protein kinase (ROCK) as the signaling effectors downstream layilin. We conclude that HA fragments generated by CigS bind to layilin and signal through Rho/ROCK to inhibit the E-cadherin gene and protein expression, leading to a loss of epithelial cell-cell contact. These studies suggest that HA functions as a master switch protecting or disrupting the epithelial barrier in its high versus low molecular weight form and that its depolymerization is a first and necessary step triggering the inflammatory response to CigS.


Asunto(s)
Cadherinas/biosíntesis , Regulación de la Expresión Génica , Ácido Hialurónico/biosíntesis , Lectinas Tipo C/metabolismo , Mucosa Respiratoria/metabolismo , Transducción de Señal , Fumar/efectos adversos , Cadherinas/genética , Células Cultivadas , Impedancia Eléctrica , Femenino , Humanos , Ácido Hialurónico/genética , Himecromona/análogos & derivados , Himecromona/farmacología , Inflamación/inducido químicamente , Inflamación/genética , Inflamación/metabolismo , Inflamación/patología , Lectinas Tipo C/genética , Lentivirus , Masculino , Permeabilidad/efectos de los fármacos , Mucosa Respiratoria/patología , Fumar/genética , Fumar/metabolismo , Transducción Genética , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo
8.
Front Immunol ; 14: 1251120, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37901225

RESUMEN

Patients with chronic lung disease, obesity, and other co-morbid conditions are at increased risk of severe illness and death when infected with severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Hypercapnia, the elevation of CO2 in blood and tissue, commonly occurs in patients with severe acute and chronic lung disease, including those with pulmonary infections, and is also associated with high mortality risk. We previously reported that hypercapnia increases viral replication and mortality of influenza A virus infection in mice. We have also shown that culture in elevated CO2 upregulates expression of cholesterol synthesis genes in primary human bronchial epithelial cells. Interestingly, factors that increase the cholesterol content of lipid rafts and lipid droplets, platforms for viral entry and assembly, enhance SARS-CoV-2 infection. In the current study, we investigated the effects of hypercapnia on ACE2 expression and entry of SARS-CoV-2 pseudovirus (p-SARS-CoV-2) into airway epithelial cells. We found that hypercapnia increased ACE2 expression and p-SARS-CoV-2 uptake by airway epithelium in mice, and in cultured VERO and human bronchial epithelial cells. Hypercapnia also increased total cellular and lipid raft-associated cholesterol in epithelial cells. Moreover, reducing cholesterol synthesis with inhibitors of sterol regulatory element binding protein 2 (SREBP2) or statins, and depletion of cellular cholesterol, each blocked the hypercapnia-induced increases in ACE2 expression and p-SARS-CoV-2 entry into epithelial cells. Cigarette smoke extract (CSE) also increased ACE2 expression, p-SARS-CoV-2 entry and cholesterol accumulation in epithelial cells, an effect not additive to that of hypercapnia, but also inhibited by statins. These findings reveal a mechanism that may account, in part, for poor clinical outcomes of SARS-CoV-2 infection in patients with advanced lung disease and hypercapnia, and in those who smoke cigarettes. Further, our results suggest the possibility that cholesterol-lowering therapies may be of particular benefit in patients with hypercapnia when exposed to or infected with SARS-CoV-2.


Asunto(s)
Enzima Convertidora de Angiotensina 2 , COVID-19 , Inhibidores de Hidroximetilglutaril-CoA Reductasas , Hipercapnia , Animales , Humanos , Ratones , Enzima Convertidora de Angiotensina 2/genética , Enzima Convertidora de Angiotensina 2/metabolismo , Dióxido de Carbono/metabolismo , Colesterol/metabolismo , COVID-19/metabolismo , Células Epiteliales/metabolismo , Hipercapnia/metabolismo , Pulmón/metabolismo , Peptidil-Dipeptidasa A/metabolismo , SARS-CoV-2/metabolismo
9.
bioRxiv ; 2023 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-36909510

RESUMEN

Hypercapnia, elevation of the partial pressure of CO 2 in blood and tissues, is a risk factor for mortality in patients with severe acute and chronic lung diseases. We previously showed that hypercapnia inhibits multiple macrophage and neutrophil antimicrobial functions, and that elevated CO 2 increases the mortality of bacterial and viral pneumonia in mice. Here, we show that normoxic hypercapnia downregulates innate immune and antiviral gene programs in alveolar macrophages (AMØs). We also show that zinc finger homeobox 3 (Zfhx3), mammalian ortholog of zfh2, which mediates hypercapnic immune suppression in Drosophila , is expressed in mouse and human MØs. Deletion of Zfhx3 in the myeloid lineage blocked the suppressive effect of hypercapnia on immune gene expression in AMØs and decreased viral replication, inflammatory lung injury and mortality in hypercapnic mice infected with influenza A virus. Our results establish Zfhx3 as the first known mammalian mediator of CO 2 effects on immune gene expression and lay the basis for future studies to identify therapeutic targets to interrupt hypercapnic immunosuppression in patients with advanced lung diseases.

10.
JCI Insight ; 8(4)2023 02 22.
Artículo en Inglés | MEDLINE | ID: mdl-36626234

RESUMEN

Persistent symptoms and radiographic abnormalities suggestive of failed lung repair are among the most common symptoms in patients with COVID-19 after hospital discharge. In mechanically ventilated patients with acute respiratory distress syndrome (ARDS) secondary to SARS-CoV-2 pneumonia, low tidal volumes to reduce ventilator-induced lung injury necessarily elevate blood CO2 levels, often leading to hypercapnia. The role of hypercapnia on lung repair after injury is not completely understood. Here - using a mouse model of hypercapnia exposure, cell lineage tracing, spatial transcriptomics, and 3D cultures - we show that hypercapnia limits ß-catenin signaling in alveolar type II (AT2) cells, leading to their reduced proliferative capacity. Hypercapnia alters expression of major Wnts in PDGFRα+ fibroblasts from those maintaining AT2 progenitor activity toward those that antagonize ß-catenin signaling, thereby limiting progenitor function. Constitutive activation of ß-catenin signaling in AT2 cells or treatment of organoid cultures with recombinant WNT3A protein bypasses the inhibitory effects of hypercapnia. Inhibition of AT2 proliferation in patients with hypercapnia may contribute to impaired lung repair after injury, preventing sealing of the epithelial barrier and increasing lung flooding, ventilator dependency, and mortality.


Asunto(s)
Hipercapnia , Vía de Señalización Wnt , Ratones , beta Catenina/metabolismo , Proliferación Celular , COVID-19/complicaciones , Hipercapnia/metabolismo , Animales
11.
J Biol Chem ; 285(34): 26126-34, 2010 Aug 20.
Artículo en Inglés | MEDLINE | ID: mdl-20554532

RESUMEN

Hyaluronidase 2 (Hyal2) is a hyaluronan (HA)-degrading enzyme found intracellularly or/and anchored to the plasma membrane through glycosylphosphatidylinositol (GPI). Normal human bronchial epithelial cells (NHBE) grown at the air-liquid interphase (ALI), treated with PI-specific phospholipase C (PI-PLC), exhibited increased Hyal activity in secretions and decreased protein and activity on the apical membrane, confirming that GPI-anchored Hyal2 is expressed in NHBE cells and it remains active in its soluble form. We have reported that HA degradation was mediated by reactive oxygen species (ROS) in human airways. Here we show that ROS increase Hyal2 expression and activity in NHBE cells and that the p38MAPK signaling pathway is involved in this effect. Hyal2 induction was confirmed by using small interfering RNA (siRNA) expressing lentivirus. These in vitro findings correlated in vivo with smokers, where increased Hyal2 immunoreactivity in the epithelium was associated with augmented levels of HA and the appearance of low molecular mass HA species in bronchial secretions. In summary, this work provides evidence that ROS induce Hyal2, suggesting that Hyal2 is likely responsible for the sustained HA fragmentation in the airway lumen observed in inflammatory conditions associated with oxidative stress.


Asunto(s)
Antígenos de Neoplasias/metabolismo , Histona Acetiltransferasas/metabolismo , Ácido Hialurónico/metabolismo , Hialuronoglucosaminidasa/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Mucosa Respiratoria/metabolismo , Células Cultivadas , Glicosilfosfatidilinositoles , Humanos , Inflamación , Sistema de Señalización de MAP Quinasas , Estrés Oxidativo , Mucosa Respiratoria/citología
12.
Am J Physiol Lung Cell Mol Physiol ; 300(2): L204-15, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21097527

RESUMEN

Cigarette smoke represents a major risk factor for the development of chronic obstructive pulmonary disease (COPD), a respiratory condition associated with airflow obstruction, mucus hypersecretion, chronic inflammation, and upregulation of inflammatory mediators such as the monocyte chemotactic protein-1 (MCP-1). MCP-1 through its receptor CCR2 induces chemotaxis and activates (44/42)MAPK, a kinase known to play a key role in mucin regulation in bronchial epithelium. In the present study we used differentiated primary cultures of normal human bronchial epithelial (NHBE) cells to test whether MCP-1 through its receptor CCR2 induces mucin upregulation. We have provided evidence that NHBE cells release MCP-1 to the epithelial surface and express the CCR2B isoform of the receptor mainly at the apical pole. In addition, we found that MCP-1 has a novel function in airway epithelium, increasing the two major airway mucins MUC5AC and MUC5B, an effect mediated, at least in part, by a cascade of events initiated by interaction of its receptor CCR2B with G(q) subunits in caveolae, followed by PLCß, PKC, and (44/42)MAPK activation. We also have shown that MCP-1 is able to induce its own expression using the same receptor but through a different pathway that involves RhoA GTPase. Furthermore, we found that a single exposure to MCP-1 is enough to induce MCP-1 secretion and sustained mucin upregulation up to 7 days after initial exposure, an effect mediated by CCR2B as confirmed using short hairpin RNA. These results agree with our data in smoker's airway epithelium, where CCR2B is present in MUC5AC- and MUC5B-expressing cells and augmented MCP-1 expression is associated with increased MUC5AC and MUC5B immunolabeling, suggesting that the mechanisms described in primary cell cultures in the present study are operative in vivo. Therefore, therapeutic approaches targeting MCP-1/CCR2B may be useful in preventing not only influx of inflammatory cells to the airways but also mucus hypersecretion and goblet cell hyperplasia.


Asunto(s)
Quimiocina CCL2/metabolismo , Mucina 5AC/metabolismo , Mucina 5B/metabolismo , Receptores CCR2/metabolismo , Mucosa Respiratoria/metabolismo , Adulto , Bronquios/efectos de los fármacos , Bronquios/metabolismo , Caveolas/metabolismo , Caveolina 1/metabolismo , Células Cultivadas , Quimiocina CCL2/genética , Quimiocina CCL2/farmacología , Subunidades alfa de la Proteína de Unión al GTP Gq-G11/metabolismo , Técnicas de Silenciamiento del Gen , Humanos , Sistema de Señalización de MAP Quinasas/efectos de los fármacos , Persona de Mediana Edad , Modelos Biológicos , Enfermedad Pulmonar Obstructiva Crónica/etiología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , ARN Mensajero/genética , ARN Mensajero/metabolismo , Receptores CCR2/antagonistas & inhibidores , Receptores CCR2/genética , Mucosa Respiratoria/efectos de los fármacos , Transducción de Señal/efectos de los fármacos , Fumar/efectos adversos , Fumar/metabolismo , Adulto Joven , Proteína de Unión al GTP rhoA/metabolismo
13.
Interface Focus ; 11(2): 20200039, 2021 Apr 06.
Artículo en Inglés | MEDLINE | ID: mdl-33633835

RESUMEN

Hypercapnia, the elevation of CO2 in blood and tissues, commonly occurs in severe acute and chronic respiratory diseases and is associated with increased risk of death. Recent studies have shown that hypercapnia inhibits expression of select innate immune genes and suppresses host defence against bacterial and viral pneumonia in mice. In the current study, we evaluated the effect of culture under conditions of hypercapnia (20% CO2) versus normocapnia (5% CO2), both with normoxia, on global gene transcription in human THP-1 and mouse RAW 264.7 macrophages stimulated with lipopolysaccharide (LPS). We found that hypercapnia selectively downregulated transcription of LPS-induced genes associated with innate immunity, antiviral response, type I interferon signalling, cytokine signalling and other inflammatory pathways in both human and mouse macrophages. Simultaneously, hypercapnia increased expression of LPS-downregulated genes associated with mitosis, DNA replication and DNA repair. These CO2-induced changes in macrophage gene expression help explain hypercapnic suppression of antibacterial and antiviral host defence in mice and reveal a mechanism that may underlie, at least in part, the high mortality of patients with severe lung disease and hypercapnia.

14.
Sci Adv ; 6(33): eabb7238, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32851183

RESUMEN

Cigarette smoking, the leading cause of chronic obstructive pulmonary disease (COPD), has been implicated as a risk factor for severe disease in patients infected with the severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2). Here we show that mice with lung epithelial cell-specific loss of function of Miz1, which we identified as a negative regulator of nuclear factor κB (NF-κB) signaling, spontaneously develop progressive age-related changes resembling COPD. Furthermore, loss of Miz1 up-regulates the expression of Ace2, the receptor for SARS-CoV-2. Concomitant partial loss of NF-κB/RelA prevented the development of COPD-like phenotype in Miz1-deficient mice. Miz1 protein levels are reduced in the lungs from patients with COPD, and in the lungs of mice exposed to chronic cigarette smoke. Our data suggest that Miz1 down-regulation-induced sustained activation of NF-κB-dependent inflammation in the lung epithelium is sufficient to induce progressive lung and airway destruction that recapitulates features of COPD, with implications for COVID-19.


Asunto(s)
Células Epiteliales/metabolismo , Factores de Transcripción de Tipo Kruppel/metabolismo , Pulmón/metabolismo , Peptidil-Dipeptidasa A/metabolismo , Fenotipo , Proteínas Inhibidoras de STAT Activados/genética , Enfermedad Pulmonar Obstructiva Crónica/genética , Ubiquitina-Proteína Ligasas/genética , Regulación hacia Arriba/genética , Enzima Convertidora de Angiotensina 2 , Animales , Betacoronavirus , COVID-19 , Infecciones por Coronavirus/metabolismo , Infecciones por Coronavirus/virología , Técnicas de Inactivación de Genes , Humanos , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Pandemias , Neumonía Viral/metabolismo , Neumonía Viral/virología , Proteínas Inhibidoras de STAT Activados/metabolismo , Enfermedad Pulmonar Obstructiva Crónica/etiología , Enfermedad Pulmonar Obstructiva Crónica/metabolismo , SARS-CoV-2 , Transducción de Señal/genética , Fumar/efectos adversos , Factor de Transcripción ReIA/metabolismo , Ubiquitina-Proteína Ligasas/metabolismo
15.
Am J Respir Cell Mol Biol ; 40(3): 277-85, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18757307

RESUMEN

Mucus hypersecretion with elevated MUC5B mucin production is a pathologic feature in many airway diseases associated with oxidative stress. In the present work, we evaluated MUC5B expression in airways and in primary cultures of normal human bronchial epithelial (NHBE) cells, as well as the mechanisms involved in its regulation. We found that oxidative stress generated by cigarette smoke or reactive oxygen species (ROS) induces MUC5B up-regulation in airway epithelium from smokers and in NHBE cells, respectively. We have previously shown that ROS-induced MUC5AC expression in NHBE cells is dependent on hyaluronan depolymerization and epidermal growth factor receptor (EGFR)/mitogen-activated protein kinase (MAPK) activation. Since hyaluronan fragments can activate MAPK through the hyaluronan receptor CD44, and CD44 heterodimerizes with EGFR, we tested whether ROS and/or hyaluronan fragments induce MUC5B mRNA and protein expression through CD44/EGFR. We found that ROS promotes CD44/EGFR interaction, EGFR/MAPK activation, and MUC5B up-regulation that are prevented by blocking CD44 and/or EGFR. These results were mimicked by hyaluronan fragments. In summary, our results show that oxidative stress in vivo (cigarette smoke) or in vitro (ROS) induces MUC5B up-regulation. This ROS-induced MUC5B expression requires CD44 as well as EGFR and MAPK activation. In addition, we also provide evidence that hyaluronan fragments are sufficient to induce CD44/EGFR interaction and downstream signaling that results in MUC5B up-regulation, suggesting that hyaluronan depolymerization during inflammatory responses could be directly involved in the induction of mucus hypersecretion.


Asunto(s)
Regulación de la Expresión Génica , Receptores de Hialuranos/metabolismo , Ácido Hialurónico/metabolismo , Mucina 5B/metabolismo , Estrés Oxidativo , Fragmentos de Péptidos/metabolismo , Mucosa Respiratoria/fisiología , Adulto , Animales , Células Cultivadas , Activación Enzimática , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Epiteliales/fisiología , Receptores ErbB/genética , Receptores ErbB/metabolismo , Humanos , Receptores de Hialuranos/genética , Ácido Hialurónico/genética , Persona de Mediana Edad , Proteína Quinasa 1 Activada por Mitógenos/genética , Proteína Quinasa 1 Activada por Mitógenos/metabolismo , Proteína Quinasa 3 Activada por Mitógenos/genética , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Mucina 5B/genética , Fragmentos de Péptidos/genética , Especies Reactivas de Oxígeno/metabolismo , Mucosa Respiratoria/citología , Mucosa Respiratoria/efectos de los fármacos , Transducción de Señal/fisiología , Humo/efectos adversos , Fumar , Nicotiana/efectos adversos , Xantina/metabolismo , Xantina Oxidasa/metabolismo , Adulto Joven
16.
Sci Rep ; 9(1): 18251, 2019 12 03.
Artículo en Inglés | MEDLINE | ID: mdl-31796806

RESUMEN

Carbon dioxide (CO2) is sensed by cells and can trigger signals to modify gene expression in different tissues leading to changes in organismal functions. Despite accumulating evidence that several pathways in various organisms are responsive to CO2 elevation (hypercapnia), it has yet to be elucidated how hypercapnia activates genes and signaling pathways, or whether they interact, are integrated, or are conserved across species. Here, we performed a large-scale transcriptomic study to explore the interaction/integration/conservation of hypercapnia-induced genomic responses in mammals (mice and humans) as well as invertebrates (Caenorhabditis elegans and Drosophila melanogaster). We found that hypercapnia activated genes that regulate Wnt signaling in mouse lungs and skeletal muscles in vivo and in several cell lines of different tissue origin. Hypercapnia-responsive Wnt pathway homologues were similarly observed in secondary analysis of available transcriptomic datasets of hypercapnia in a human bronchial cell line, flies and nematodes. Our data suggest the evolutionarily conserved role of high CO2 in regulating Wnt pathway genes.


Asunto(s)
Caenorhabditis elegans/metabolismo , Dióxido de Carbono/farmacología , Drosophila melanogaster/metabolismo , Vía de Señalización Wnt/efectos de los fármacos , Animales , Bronquios/citología , Bronquios/metabolismo , Caenorhabditis elegans/efectos de los fármacos , Línea Celular , Drosophila melanogaster/efectos de los fármacos , Perfilación de la Expresión Génica , Humanos , Hipercapnia/metabolismo , Masculino , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa , Análisis de Matrices Tisulares
17.
Am J Respir Cell Mol Biol ; 39(3): 289-95, 2008 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-18390475

RESUMEN

Hyaluronan (HA) is present at the apical surface of airway epithelium as a high-molecular-weight polymer. Since HA depolymerization initiates a cascade of events that results in kinin generation and growth factor processing, in the present work we used primary cultures of human bronchial epithelial (HBE) cells grown at the air-liquid interface (ALI) to assess hyaluronidase (Hyal) activity by HA zymography, gene expression by quantitative real-time PCR, and localization by confocal microscopy. Because TNF-alpha and IL-1beta induce Hyals in other cells, we tested their effects on Hyals expression and activity. We found that Hyal-like activity is present in the apical and basolateral secretions from HBE cells where Hyals 1, 2, and 3 are expressed, and that IL-1beta acts synergistically with TNF-alpha to increase gene expression and activity. Confocal microscopy showed that Hyals 1, 2, and 3 were localized intracellularly, while Hyal2 was also expressed at the apical pole associated with the plasma membrane, and in a soluble form on the apical secretions. Tissue sections from normal individuals and from individuals with asthma showed a Hyal distribution pattern similar to that observed on nontreated HBE cells or exposed to cytokines, respectively. In addition, increased expression and activity were observed in tracheal sections and in bronchoalveolar lavage (BAL) obtained from subjects with asthma when compared with normal lung donors and healthy volunteers. Our observations indicate that Hyal 1, 2, and 3 are expressed in airway epithelium and may operate in a coordinated fashion to depolymerize HA during inflammation associated with up-regulation of TNF-alpha and IL-1beta, such as allergen-induced asthmatic responses.


Asunto(s)
Citocinas/fisiología , Hialuronoglucosaminidasa/biosíntesis , Mediadores de Inflamación/fisiología , Mucosa Respiratoria/enzimología , Alérgenos/administración & dosificación , Alérgenos/inmunología , Asma/enzimología , Líquido del Lavado Bronquioalveolar , Células Cultivadas , Técnica del Anticuerpo Fluorescente , Expresión Génica , Humanos , Ácido Hialurónico/biosíntesis , Reacción en Cadena de la Polimerasa , Tráquea/citología , Tráquea/enzimología
18.
J Appl Physiol (1985) ; 105(1): 54-7, 2008 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-18467553

RESUMEN

Cigarette smoking is associated with attenuated endothelium-dependent vasodilation (endothelial dysfunction) in the systemic circulation, including the airway circulation. We wished to determine whether an inhaled corticosteroid could restore endothelial function in the airway of lung-healthy current smokers, ex-smokers, and nonsmokers. We measured baseline airway blood flow (Qaw) and Qaw reactivity to inhaled albuterol as an index of endothelium-dependent vasodilation and to sublingual nitroglycerin as an index of endothelium-independent vasodilation in lung-healthy current smokers, ex-smokers, and nonsmokers. Current smokers were then treated with inhaled fluticasone for 3 wk, and all measurements were repeated after fluticasone treatment and after a subsequent 3-wk fluticasone washout period. Baseline mean Qaw and endothelium-independent Qaw reactivity were similar in the three groups. Mean endothelium-dependent Qaw reactivity was 49.5% in nonsmokers, 42.7% in ex-smokers, and 10.8% in current smokers (P < 0.05 vs. nonsmokers). In current smokers, mean baseline Qaw was unchanged after fluticasone treatment, but endothelium-dependent Qaw reactivity significantly increased to 34.9%. Qaw reactivity was again blunted after fluticasone washout. Endothelial dysfunction, as assessed by vascular reactivity, can be corrected with an inhaled corticosteroid in the airway of lung-healthy current smokers. This proof of concept can serve as the basis for future clinical investigations on the effect of glucocorticoids on endothelial function in smokers.


Asunto(s)
Androstadienos/farmacología , Broncodilatadores/farmacología , Endotelio/efectos de los fármacos , Glucocorticoides/farmacología , Fumar/fisiopatología , Administración por Inhalación , Adolescente , Adulto , Albuterol/administración & dosificación , Albuterol/farmacología , Androstadienos/administración & dosificación , Broncodilatadores/administración & dosificación , Femenino , Fluticasona , Volumen Espiratorio Forzado/efectos de los fármacos , Volumen Espiratorio Forzado/fisiología , Glucocorticoides/administración & dosificación , Humanos , Masculino , Persona de Mediana Edad , Relajación Muscular/efectos de los fármacos , Músculo Liso Vascular/efectos de los fármacos , Nitroglicerina/farmacología , Flujo Sanguíneo Regional/efectos de los fármacos , Pruebas de Función Respiratoria , Sistema Respiratorio/irrigación sanguínea , Espirometría , Vasodilatadores/farmacología
19.
Sci Rep ; 8(1): 13508, 2018 09 10.
Artículo en Inglés | MEDLINE | ID: mdl-30202079

RESUMEN

Hypercapnia, the elevation of CO2 in blood and tissues, commonly occurs in severe acute and chronic respiratory diseases, and is associated with increased risk of mortality. Recent studies have shown that hypercapnia adversely affects innate immunity, host defense, lung edema clearance and cell proliferation. Airway epithelial dysfunction is a feature of advanced lung disease, but the effect of hypercapnia on airway epithelium is unknown. Thus, in the current study we examined the effect of normoxic hypercapnia (20% CO2 for 24 h) vs normocapnia (5% CO2), on global gene expression in differentiated normal human airway epithelial cells. Gene expression was assessed on Affymetrix microarrays, and subjected to gene ontology analysis for biological process and cluster-network representation. We found that hypercapnia downregulated the expression of 183 genes and upregulated 126. Among these, major gene clusters linked to immune responses and nucleosome assembly were largely downregulated, while lipid metabolism genes were largely upregulated. The overwhelming majority of these genes were not previously known to be regulated by CO2. These changes in gene expression indicate the potential for hypercapnia to impact bronchial epithelial cell function in ways that may contribute to poor clinical outcomes in patients with severe acute or advanced chronic lung diseases.


Asunto(s)
Dióxido de Carbono/toxicidad , Regulación de la Expresión Génica/efectos de los fármacos , Hipercapnia/complicaciones , Enfermedades Pulmonares/patología , Mucosa Respiratoria/efectos de los fármacos , Bronquios/citología , Bronquios/efectos de los fármacos , Bronquios/inmunología , Bronquios/patología , Dióxido de Carbono/sangre , Diferenciación Celular , Células Cultivadas , Enfermedad Crónica , Células Epiteliales/efectos de los fármacos , Células Epiteliales/inmunología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Perfilación de la Expresión Génica , Humanos , Hipercapnia/sangre , Inmunidad Innata/efectos de los fármacos , Inmunidad Innata/genética , Metabolismo de los Lípidos/efectos de los fármacos , Metabolismo de los Lípidos/genética , Enfermedades Pulmonares/etiología , Nucleosomas/efectos de los fármacos , Nucleosomas/metabolismo , Mucosa Respiratoria/citología , Mucosa Respiratoria/inmunología , Mucosa Respiratoria/patología , Sarcoglicanopatías
20.
Am J Respir Cell Mol Biol ; 34(5): 581-91, 2006 May.
Artículo en Inglés | MEDLINE | ID: mdl-16424381

RESUMEN

Mucus overproduction in inflammatory and obstructive airway diseases is associated with goblet cell (GC) metaplasia in airways. Although the mechanisms involved in GC metaplasia and mucus hypersecretion are not completely understood, association with oxidative stress and epidermal growth factor receptor (EGFR) signaling has been reported. To explore the mechanisms involved in oxidative stress-induced GC metaplasia, cultures of differentiated normal human bronchial epithelial cells grown at the air-liquid interface were exposed to reactive oxygen species (ROS) generated by xanthine/xanthine oxidase. EGFR activation and signaling was assessed by measuring EGF and transforming growth factor-alpha release and EGFR and (44/42)MAPK phosphorylation. The GC population was evaluated by confocal microscopy. ROS-induced EGFR activation resulted in GC proliferation and increased MUC5AC gene and protein expression. Signaling was due to pro-EGF processing by tissue kallikrein (TK), which was activated by ROS-induced hyaluronan breakdown. It was inhibited by catalase, a TK inhibitor, and EGF-blocking antibodies. Exposure to recombinant TK mimicked the ROS effects, increasing the expression of MUC5AC and lactoperoxidase. In addition, ROS induced the antiapoptotic factor Bcl-2 in a TK-dependent fashion. In conclusion, ROS-induced GC metaplasia in normal human bronchial epithelial cells is associated with HA depolymerization and EGF processing by TK followed by EGFR signaling, suggesting that increases in TK activity could contribute to GC metaplasia and mucus hypersecretion in diseases such as asthma and chronic bronchitis. The data also suggest that increases in GC population could be sustained by the associated upregulation of Bcl-2 in airway epithelial cells.


Asunto(s)
Factor de Crecimiento Epidérmico/metabolismo , Receptores ErbB/metabolismo , Células Caliciformes/efectos de los fármacos , Células Caliciformes/patología , Oxidantes/farmacología , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/patología , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Células Epiteliales/citología , Células Epiteliales/efectos de los fármacos , Células Caliciformes/citología , Humanos , Ácido Hialurónico/metabolismo , Lactoperoxidasa/metabolismo , Metaplasia , Mucina 5AC , Mucinas/metabolismo , Estrés Oxidativo , Polímeros , Precursores de Proteínas/metabolismo , Transporte de Proteínas , Proteínas Proto-Oncogénicas c-bcl-2/genética , ARN Mensajero/genética , ARN Mensajero/metabolismo , Mucosa Respiratoria/metabolismo , Transducción de Señal/efectos de los fármacos , Calicreínas de Tejido/metabolismo , Xantina Oxidasa/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA