Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Vet Intern Med ; 36(3): 1089-1099, 2022 May.
Artículo en Inglés | MEDLINE | ID: mdl-35484990

RESUMEN

BACKGROUND: New drugs for veterinary patients with acute respiratory distress syndrome (ARDS) are urgently needed. Early or late postinfection treatment of influenza-infected mice with the liponucleotide cytidine diphosphocholine (CDP-choline) resulted in decreased hypoxemia, pulmonary edema, lung dysfunction, and inflammation without altering viral replication. These findings suggested CDP-choline could have benefit as adjunctive treatment for ARDS in veterinary patients (VetARDS). OBJECTIVES: Determine if parenterally administered CDP-choline can attenuate mild VetARDS in dogs with aspiration pneumonia. ANIMALS: Dogs admitted to a veterinary intensive care unit (ICU) for aspiration pneumonia. METHODS: Subjects were enrolled in a randomized, double-blinded, placebo-controlled trial of treatment with vehicle (0.1 mL/kg sterile 0.9% saline, IV; n = 8) or CDP-choline (5 mg/kg in 0.1 mL/kg 0.9% saline, IV; n = 9) q12h over the first 48 hours after ICU admission. RESULTS: No significant differences in signalment or clinical findings were found between placebo- and CDP-choline-treated dogs on admission. All dogs exhibited tachycardia, tachypnea, hypertension, hypoxemia, hypocapnia, lymphopenia, and neutrophilia. CDP-choline administration resulted in rapid, progressive, and clinically relevant increases in oxygenation as determined by pulse oximetry and ratios of arterial oxygen partial pressure (Pa O2 mmHg) to fractional inspired oxygen (% Fi O2 ) and decreases in alveolar-arterial (A-a) gradients that did not occur in placebo (saline)-treated animals. Treatment with CDP-choline was also associated with less platelet consumption over the first 48 hours, but had no detectable detrimental effects. CONCLUSIONS AND CLINICAL IMPORTANCE: Ctyidine diphosphcholine acts rapidly to promote gas exchange in dogs with naturally occurring aspiration pneumonia and is a potential adjunctive treatment in VetARDS patients.


Asunto(s)
Enfermedades de los Perros , Neumonía por Aspiración , Síndrome de Dificultad Respiratoria , Animales , Perros , Citidina , Citidina Difosfato Colina/uso terapéutico , Enfermedades de los Perros/tratamiento farmacológico , Hipoxia/tratamiento farmacológico , Hipoxia/veterinaria , Pulmón , Oxígeno/uso terapéutico , Neumonía por Aspiración/veterinaria , Síndrome de Dificultad Respiratoria/veterinaria , Solución Salina
2.
Virology ; 559: 111-119, 2021 07.
Artículo en Inglés | MEDLINE | ID: mdl-33865074

RESUMEN

Influenza A virus (IAV) infection alters lung epithelial cell metabolism in vitro by promoting a glycolytic shift. We hypothesized that this shift benefits the virus rather than the host and that inhibition of glycolysis would improve infection outcomes. A/WSN/33 IAV-inoculated C57BL/6 mice were treated daily from 1 day post-inoculation (d.p.i.) with 2-deoxy-d-glucose (2-DG) to inhibit glycolysis and with the pyruvate dehydrogenase kinase (PDK) inhibitor dichloroacetate (DCA) to promote flux through the TCA cycle. To block OXPHOS, mice were treated every other day from 1 d.p.i. with the Complex I inhibitor rotenone (ROT). 2-DG significantly decreased nocturnal activity, reduced respiratory exchange ratios, worsened hypoxemia, exacerbated lung dysfunction, and increased humoral inflammation at 6 d.p.i. DCA and ROT treatment normalized oxygenation and airway resistance and attenuated IAV-induced pulmonary edema, histopathology, and nitrotyrosine formation. None of the treatments altered viral replication. These data suggest that a shift to glycolysis is host-protective in influenza.


Asunto(s)
Células Epiteliales/metabolismo , Glucólisis , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Lesión Pulmonar/virología , Pulmón/metabolismo , Animales , Femenino , Pulmón/química , Pulmón/virología , Lesión Pulmonar/prevención & control , Masculino , Ratones , Ratones Endogámicos C57BL , Piruvato Deshidrogenasa Quinasa Acetil-Transferidora/antagonistas & inhibidores , Tirosina/análogos & derivados , Tirosina/análisis , Tirosina/metabolismo , Replicación Viral
4.
Ann Diagn Pathol ; 36: 12-20, 2018 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-29966831

RESUMEN

Spinal cord paralysis is relatively common after surgical repair of thoraco-abdominal aortic aneurysm (TAAA) and its etiology is unknown. The present study was designed to examine the histopathology of the disease and investigate whether miR-155 ablation would reduce spinal cord ischemic damage and delayed hindlimb paralysis induced by aortic cross-clamping (ACC) in our mouse model. The loss of locomotor function in ACC-paralyzed mice correlated with the presence of extensive gray matter damage and central cord edema, with minimal white matter histopathology. qRTPCR and Western blotting showed that the spinal cords of wild-type ACC mice that escaped paralysis showed lower miR-155 expression and higher levels of transcripts encoding Mfsd2a, which is implicated in the maintenance of blood-brain barrier integrity. In situ based testing demonstrated that increased miR-155 detection in neurons was highly correlated with the gray matter damage and the loss of one of its targets, Mfsd2a, could serve as a good biomarker of the endothelial cell damage. In vitro, we demonstrated that miR-155 targeted Mfsd2a in endothelial cells and motoneurons and increased endothelial cell permeability. Finally, miR-155 ablation slowed the progression of central cord edema, and reduced the incidence of paralysis by 40%. In sum, the surgical pathology findings clearly indicated that the epicenter of the ischemic-induced paralysis was the gray matter and that endothelial cell damage correlated to Mfsd2a loss is a good biomarker of the disease. MiR-155 targeting therefore offers new therapeutic opportunity for edema caused by traumatic spinal cord injury and diagnostic pathologists, by using immunohistochemistry, can clarify if this mechanism also is important in other ischemic diseases of the CNS, including stroke.


Asunto(s)
Isquemia/metabolismo , Proteínas de Transporte de Membrana/genética , MicroARNs/genética , Traumatismos de la Médula Espinal/genética , Animales , Modelos Animales de Enfermedad , Inmunohistoquímica/métodos , Isquemia/genética , Ratones Endogámicos C57BL , Ratones Noqueados , MicroARNs/metabolismo , Enfermedades del Sistema Nervioso/genética , Neuronas/metabolismo , Médula Espinal/patología , Traumatismos de la Médula Espinal/fisiopatología , Simportadores , Proteínas Supresoras de Tumor/genética
5.
Am J Physiol Lung Cell Mol Physiol ; 314(1): L83-L92, 2018 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-28982733

RESUMEN

Influenza A viruses are highly contagious respiratory pathogens that are responsible for significant morbidity and mortality worldwide on an annual basis. We have shown previously that influenza infection of mice leads to increased ATP and adenosine accumulation in the airway lumen. Moreover, we demonstrated that A1-adenosine receptor activation contributes significantly to influenza-induced acute respiratory distress syndrome (ARDS). However, we found that development of ARDS in influenza-infected mice does not require catabolism of ATP to adenosine by ecto-5'-nucleotidase (CD73). Hence, we hypothesized that increased adenosine generation in response to infection is mediated by tissue nonspecific alkaline phosphatase (TNAP), which is a low-affinity, high-capacity enzyme that catabolizes nucleotides in a nonspecific manner. In the current study, we found that whole lung and BALF TNAP expression and alkaline phosphatase enzymatic activity increased as early as 2 days postinfection (dpi) of C57BL/6 mice with 10,000 pfu/mouse of influenza A/WSN/33 (H1N1). Treatment at 2 and 4 dpi with a highly specific quinolinyl-benzenesulfonamide TNAP inhibitor (TNAPi) significantly reduced whole lung alkaline phosphatase activity at 6 dpi but did not alter TNAP gene or protein expression. TNAPi treatment attenuated hypoxemia, lung dysfunction, histopathology, and pulmonary edema at 6 dpi without impacting viral replication or BALF adenosine. Treatment also improved epithelial barrier function and attenuated cellular and humoral immune responses to influenza infection. These data indicate that TNAP inhibition can attenuate influenza-induced ARDS by reducing inflammation and fluid accumulation within the lung. They also further emphasize the importance of adenosine generation for development of ARDS in influenza-infected mice.


Asunto(s)
Adenosina Trifosfato/metabolismo , Fosfatasa Alcalina/metabolismo , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Infecciones por Orthomyxoviridae/complicaciones , Edema Pulmonar/etiología , Síndrome de Dificultad Respiratoria/etiología , 5'-Nucleotidasa/metabolismo , Animales , Ratones , Ratones Endogámicos C57BL , Infecciones por Orthomyxoviridae/virología , Edema Pulmonar/metabolismo , Edema Pulmonar/patología , Síndrome de Dificultad Respiratoria/metabolismo , Síndrome de Dificultad Respiratoria/patología , Replicación Viral
6.
Am J Physiol Lung Cell Mol Physiol ; 309(11): L1313-22, 2015 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-26432867

RESUMEN

Extracellular nucleotides and nucleosides are important signaling molecules in the lung. Nucleotide and nucleoside concentrations in alveolar lining fluid are controlled by a complex network of surface ectonucleotidases. Previously, we demonstrated that influenza A/WSN/33 (H1N1) virus resulted in increased levels of the nucleotide ATP and the nucleoside adenosine in bronchoalveolar lavage fluid (BALF) of wild-type (WT) C57BL/6 mice. Influenza-induced acute lung injury (ALI) was highly attenuated in A1-adenosine receptor-knockout mice. Because AMP hydrolysis by the ecto-5'-nucleotidase (CD73) plays a central role in and is rate-limiting for generation of adenosine in the normal lung, we hypothesized that ALI would be attenuated in C57BL/6-congenic CD73-knockout (CD73-KO) mice. Infection-induced hypoxemia, bradycardia, viral replication, and bronchoconstriction were moderately increased in CD73-KO mice relative to WT controls. However, postinfection weight loss, pulmonary edema, and parenchymal dysfunction were not altered. Treatment of WT mice with the CD73 inhibitor 5'-(α,ß-methylene) diphosphate (APCP) also had no effect on infection-induced pulmonary edema but modestly attenuated hypoxemia. BALF from CD73-KO and APCP-treated WT mice contained more IL-6 and CXCL-10/IFN-γ-induced protein 10, less CXCL-1/keratinocyte chemoattractant, and fewer neutrophils than BALF from untreated WT controls. BALF from APCP-treated WT mice also contained fewer alveolar macrophages and more transforming growth factor-ß than BALF from untreated WT mice. These results indicate that CD73 is not necessary for development of ALI following influenza A virus infection and suggest that tissue-nonspecific alkaline phosphatase may be responsible for increased adenosine generation in the infected lung. However, they do suggest that CD73 has a previously unrecognized immunomodulatory role in influenza.


Asunto(s)
5'-Nucleotidasa/metabolismo , Lesión Pulmonar Aguda/enzimología , Lesión Pulmonar Aguda/inmunología , Inmunidad Innata , Gripe Humana/inmunología , Infecciones por Orthomyxoviridae/enzimología , Infecciones por Orthomyxoviridae/inmunología , 5'-Nucleotidasa/genética , Lesión Pulmonar Aguda/complicaciones , Lesión Pulmonar Aguda/virología , Adenosina Trifosfato/análogos & derivados , Adenosina Trifosfato/farmacología , Fosfatasa Alcalina/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/citología , Quimiocinas/metabolismo , Adaptabilidad , Proteínas Ligadas a GPI/genética , Proteínas Ligadas a GPI/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Corazón/efectos de los fármacos , Corazón/fisiopatología , Humanos , Inmunidad Innata/efectos de los fármacos , Subtipo H1N1 del Virus de la Influenza A/inmunología , Recuento de Leucocitos , Pulmón/efectos de los fármacos , Pulmón/fisiopatología , Ratones Endogámicos C57BL , Ratones Noqueados , Neutrófilos/citología , Neutrófilos/efectos de los fármacos , Infecciones por Orthomyxoviridae/complicaciones , Infecciones por Orthomyxoviridae/virología , Edema Pulmonar/etiología , Edema Pulmonar/patología , Edema Pulmonar/fisiopatología , Replicación Viral/efectos de los fármacos
7.
Am J Physiol Lung Cell Mol Physiol ; 308(11): L1136-44, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25840995

RESUMEN

As the eighth leading cause of annual mortality in the USA, influenza A viruses are a major public health concern. In 20% of patients, severe influenza progresses to acute lung injury (ALI). However, pathophysiological mechanisms underlying ALI development are poorly defined. We reported that, unlike wild-type (WT) C57BL/6 controls, influenza A virus-infected mice that are heterozygous for the F508del mutation in the cystic fibrosis transmembrane conductance regulator (HETs) did not develop ALI. This effect was associated with higher IL-6 and alveolar macrophages (AMs) at 6 days postinfection (d.p.i.) in HET bronchoalveolar lavage fluid (BALF). In the present study, we found that HET AMs were an important source of IL-6 at 6 d.p.i. Infection also induced TGF-ß production by HET but not WT mice at 2 d.p.i. TGF-ß neutralization at 2 d.p.i. (TGF-N) significantly reduced BALF IL-6 in HETs at 6 d.p.i. Neither TGF-N nor IL-6 neutralization at 4 d.p.i. (IL-6-N) altered postinfection weight loss or viral replication in either mouse strain. However, both treatments increased influenza A virus-induced hypoxemia, pulmonary edema, and lung dysfunction in HETs to WT levels at 6 d.p.i. TGF-N and IL-6-N did not affect BALF AM and neutrophil numbers but attenuated the CXCL-1/keratinocyte chemokine response in both strains and reduced IFN-γ production in WT mice. Finally, bone marrow transfer experiments showed that HET stromal and myeloid cells are both required for protection from ALI in HETs. These findings indicate that TGF-ß-dependent production of IL-6 by AMs later in infection prevents ALI development in influenza A virus-infected HET mice.


Asunto(s)
Lesión Pulmonar Aguda/virología , Virus de la Influenza A/inmunología , Interleucina-6/fisiología , Infecciones por Orthomyxoviridae/inmunología , Factor de Crecimiento Transformador beta/fisiología , Lesión Pulmonar Aguda/inmunología , Animales , Líquido del Lavado Bronquioalveolar , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Inmunidad Innata , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/virología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos CFTR , Eliminación de Secuencia
8.
J Control Release ; 203: 140-9, 2015 Apr 10.
Artículo en Inglés | MEDLINE | ID: mdl-25687308

RESUMEN

Alveolar type II (ATII) respiratory epithelial cells are essential to normal lung function. They may be also central to the pathogenesis of diseases such as acute lung injury, pulmonary fibrosis, and pulmonary adenocarcinoma. Hence, ATII cells are important therapeutic targets. However, effective ATII cell-specific drug delivery in vivo requires carriers of an appropriate size, which can cross the hydrophobic alveolar surfactant film and polar aqueous layer overlying ATII cells, and be taken up without inducing ATII cell dysfunction, pulmonary inflammation, lung damage, or excessive systemic spread and side-effects. We have developed lipoplexes as a versatile nanoparticle carrier system for drug/RNA delivery. To optimize their pulmonary localization and ATII cell specificity, lipoplexes were conjugated to an antibody directed against the ATII cell-specific antigen surfactant protein-C (SP-C) then administered to C57BL/6 mice via the nares. Intranasally-administered, anti-SP-C-conjugated lipoplexes targeted mouse ATII cells with >70% specificity in vivo, were retained within ATII cells for at least 48h, and did not accumulate at significant levels in other lung cell types or viscera. 48h after treatment with anti-SP-C-conjugated lipoplexes containing the test microRNA miR-486, expression of mature miR-486 was approximately 4-fold higher in ATII cells than whole lung by qRT-PCR, and was undetectable in other viscera. Lipoplexes induced no weight loss, hypoxemia, lung dysfunction, pulmonary edema, or pulmonary inflammation over a 6-day period. These findings indicate that ATII cell-targeted lipoplexes exhibit all the desired characteristics of an effective drug delivery system for the treatment of pulmonary diseases that result primarily from ATII cell dysfunction.


Asunto(s)
Células Epiteliales Alveolares/inmunología , Técnicas de Transferencia de Gen , Inmunoconjugados/inmunología , Liposomas/inmunología , MicroARNs/administración & dosificación , Proteína C Asociada a Surfactante Pulmonar/inmunología , Animales , Línea Celular , Células Cultivadas , Sistemas de Liberación de Medicamentos , Femenino , Humanos , Inmunoconjugados/química , Inmunoconjugados/farmacocinética , Liposomas/química , Liposomas/farmacocinética , Pulmón/inmunología , Ratones Endogámicos C57BL
9.
Am J Physiol Lung Cell Mol Physiol ; 308(7): L628-38, 2015 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-25595651

RESUMEN

Influenza viruses cause acute respiratory disease of great importance to public health. Alveolar type II (ATII) respiratory epithelial cells are central to normal lung function and are a site of influenza A virus replication in the distal lung. However, the consequences of infection for ATII cell function are poorly understood. To determine the impact of influenza infection on ATII cells we used C57BL/6-congenic SP-C(GFP) mice that express green fluorescent protein (GFP) under the control of the surfactant protein-C (SP-C) promoter, which is only active in ATII cells. Most cells isolated from the lungs of uninfected SP-C(GFP) mice were GFP(+) but did not express the alveolar type I (ATI) antigen podoplanin (PODO). ATII cells were also EpCAM(+) and α2,3-linked sialosaccharide(+). Infection with influenza A/WSN/33 virus caused severe hypoxemia and pulmonary edema. This was accompanied by loss of whole lung GFP fluorescence, reduced ATII cell yields, increased ATII cell apoptosis, reduced SP-C gene and protein expression in ATII cell lysates, and increased PODO gene and protein levels. Flow cytometry indicated that infection decreased GFP(+)/PODO(-) cells and increased GFP(-)/PODO(+) and GFP(-)/PODO(-) cells. Very few GFP(+)/PODO(+) cells were detectable. Finally, infection resulted in a significant decline in EpCAM expression by PODO(+) cells, but had limited effects on α2,3-linked sialosaccharides. Our findings indicate that influenza infection results in a progressive differentiation of ATII cells into ATI-like cells, possibly via an SP-C(-)/PODO(-) intermediate, to replace dying or dead ATI cells. However, impaired SP-C synthesis is likely to contribute significantly to reduced lung compliance in infected mice.


Asunto(s)
Células Epiteliales Alveolares/metabolismo , Subtipo H1N1 del Virus de la Influenza A , Infecciones por Orthomyxoviridae/patología , Infecciones del Sistema Respiratorio/patología , Células Epiteliales Alveolares/virología , Animales , Antígenos de Neoplasias/metabolismo , Moléculas de Adhesión Celular/metabolismo , Molécula de Adhesión Celular Epitelial , Péptidos y Proteínas de Señalización Intercelular , Pulmón/patología , Pulmón/virología , Ratones Endogámicos C57BL , Ratones Transgénicos , Infecciones por Orthomyxoviridae/metabolismo , Infecciones por Orthomyxoviridae/virología , Péptidos/metabolismo , Fenotipo , Proteína C Asociada a Surfactante Pulmonar , Infecciones del Sistema Respiratorio/metabolismo , Infecciones del Sistema Respiratorio/virología , Ácidos Siálicos/metabolismo
10.
J Virol ; 88(17): 10214-27, 2014 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-24965449

RESUMEN

UNLABELLED: We have shown that bronchoalveolar epithelial A1-adenosine receptors (A1-AdoR) are activated in influenza A virus-infected mice. Alveolar macrophages and neutrophils also express A1-AdoRs, and we hypothesized that activation of A1-AdoRs on these cells will promote macrophage and neutrophil chemotaxis and activation and thereby play a role in the pathogenesis of influenza virus-induced acute lung injury. Wild-type (WT) C57BL/6 mice, congenic A1-AdoR knockout (A1-KO) mice, and mice that had undergone reciprocal bone marrow transfer were inoculated intranasally with 10,000 PFU/mouse influenza A/WSN/33 (H1N1) virus. Alternatively, WT mice underwent daily treatment with the A1-AdoR antagonist 8-cyclopentyl-1,3-dipropylxanthine (DPCPX) from 1 day prior to inoculation. Infection increased bronchoalveolar lining fluid (BALF) adenosine comparably in WT and A1-KO mice. Infection of WT mice resulted in reduced carotid arterial O2 saturation (hypoxemia), lung pathology, pulmonary edema, reduced lung compliance, increased basal airway resistance, and hyperresponsiveness to methacholine. These effects were absent or significantly attenuated in A1-KO mice. Levels of BALF leukocytes, gamma interferon (IFN-γ), and interleukin 10 (IL-10) were significantly reduced in infected A1-KO mice, but levels of KC, IP-10, and MCP-1 were increased. Reciprocal bone marrow transfer resulted in WT-like lung injury severity, but BALF leukocyte levels increased only in WT and A1-KO mice with WT bone barrow. Hypoxemia, pulmonary edema, and levels of BALF alveolar macrophages, neutrophils, IFN-γ, and IL-10 were reduced in DPCPX-treated WT mice. Levels of viral replication did not differ between mouse strains or treatment groups. These findings indicate that adenosine activation of leukocyte A1-AdoRs plays a significant role in their recruitment to the infected lung and contributes to influenza pathogenesis. A1-AdoR inhibitor therapy may therefore be beneficial in patients with influenza virus-induced lung injury. IMPORTANCE: Because antiviral drugs are of limited efficacy in patients hospitalized for influenza virus-induced respiratory failure, there is an urgent need for new therapeutics that can limit the progression of lung injury and reduce influenza death rates. We show that influenza A virus infection results in increased production of the nucleoside adenosine in the mouse lung and that activation of A1-subtype adenosine receptors by adenosine contributes significantly to both recruitment of innate immune cells to the lung and development of acute lung injury following influenza virus infection. We also show that treatment with an A1-adenosine receptor antagonist reduces the severity of lung injury in influenza virus-infected mice. Our findings indicate that adenosine plays an important and previously unrecognized role in the innate immune response to influenza virus infection and suggest that drugs which can inhibit either generation of adenosine or activation of A1-adenosine receptors may be beneficial in treating influenza patients hospitalized for respiratory failure.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Subtipo H1N1 del Virus de la Influenza A/fisiología , Leucocitos/inmunología , Pulmón/inmunología , Infecciones por Orthomyxoviridae/inmunología , Receptor de Adenosina A1/metabolismo , Lesión Pulmonar Aguda/patología , Traslado Adoptivo , Animales , Movimiento Celular , Modelos Animales de Enfermedad , Subtipo H1N1 del Virus de la Influenza A/inmunología , Leucocitos/fisiología , Pulmón/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Infecciones por Orthomyxoviridae/patología , Receptor de Adenosina A1/deficiencia
11.
J Infect Dis ; 208(5): 780-9, 2013 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-23749967

RESUMEN

BACKGROUND: Seasonal and pandemic influenza are significant public health concerns. Influenza stimulates respiratory epithelial Cl(-) secretion via the cystic fibrosis transmembrane conductance regulator (CFTR). The purpose of this study was to determine the contribution of this effect to influenza pathogenesis in mice with reduced CFTR activity. METHODS: C57BL/6-congenic mice heterozygous for the F508del CFTR mutation (HET) and wild-type (WT) controls were infected intranasally with 10 000 focus-forming units of influenza A/WSN/33 (H1N1) per mouse. Body weight, arterial O2 saturation, and heart rate were monitored daily. Pulmonary edema and lung function parameters were derived from ratios of wet weight to dry weight and the forced-oscillation technique, respectively. Levels of cytokines and chemokines in bronchoalveolar lavage fluid were measured by enzyme-linked immunosorbent assay. RESULTS: Relative to WT mice, influenza virus-infected HET mice showed significantly delayed mortality, which was accompanied by attenuated hypoxemia, cardiopulmonary dysfunction, and pulmonary edema. However, viral replication and weight loss did not differ. The protective HET phenotype was correlated with exaggerated alveolar macrophage and interleukin 6 responses to infection and was abrogated by alveolar macrophage depletion, using clodronate liposomes. CONCLUSIONS: Reduced CFTR expression modulates the innate immune response to influenza and alters disease pathogenesis. CFTR-mediated Cl(-) secretion is therefore an important host determinant of disease, and CFTR inhibition may be of therapeutic benefit in influenza.


Asunto(s)
Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Heterocigoto , Subtipo H1N1 del Virus de la Influenza A/patogenicidad , Infecciones por Orthomyxoviridae/genética , Infecciones por Orthomyxoviridae/patología , Eliminación de Secuencia , Animales , Líquido del Lavado Bronquioalveolar/química , Líquido del Lavado Bronquioalveolar/inmunología , Citocinas/análisis , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Humanos , Pulmón/patología , Pulmón/fisiopatología , Ratones , Ratones Endogámicos C57BL , Mutación , Orthomyxoviridae
12.
Mol Ther Nucleic Acids ; 2: e84, 2013 Apr 16.
Artículo en Inglés | MEDLINE | ID: mdl-23591808

RESUMEN

MicroRNA-29b (miR-29b) expression has been shown to be reduced in non-small-cell lung cancer (NSCLC) tissues. Here, we have identified the oncogene cyclin-dependent protein kinase 6 (CDK6) as a direct target of miR-29b in lung cancer. We hypothesized that in vivo restoration of miR-29b and thus targeting of genes important to tumor initiation and progression may represent an option for lung cancer treatment. We developed a cationic lipoplexes (LPs)-based carrier that efficiently delivered miR-29b both in vitro and in vivo. LPs containing miR-29b (LP-miR-29b) efficiently delivered miR-29b to NSCLC A549 cells, reduced the expression of key targets CDK6, DNMT3B, and myeloid cell leukemia sequence 1 (MCL1), as well as cell growth and clonogenicity of A549 cells. In addition, the IC50 for cisplatin in the miR-29b-treated cells was effectively reduced. In a xenograft murine model, LPs efficiently accumulated at tumor sites. Systemic delivery of LP-miR-29b increased the tumor miR-29b expression by approximately fivefold, downregulated the tumor mRNA expression of CDK6, DNMT3B, and MCL1 by ~57.4, ~40.5, and ~52.4%, respectively, and significantly inhibited tumor growth by ~60% compared with LP-miR-NC (negative control). Our results demonstrate that cationic LPs represent an efficient delivery system that holds great potential in the development of miRNA-based therapeutics for lung cancer treatment.Molecular Therapy-Nucleic Acids (2013) 2, e84; doi:10.1038/mtna.2013.14; published online 16 April 2013.

13.
Influenza Other Respir Viruses ; 7(3): 472-9, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-22862736

RESUMEN

BACKGROUND: Patients with severe seasonal or pandemic influenza pneumonia frequently develop acute respiratory distress syndrome (ARDS). One clinical diagnostic criterion for ARDS is the P(a)O(2):F(i)O(2) ratio, which is an index of alveolar gas exchange. However, effects of H1N1 influenza infection on P(a)O(2):F(i)O(2) ratios and related pathophysiologic readouts of lung function have not been reported in mice. METHODS: To develop a method for determining P(a)O(2):F(i)O(2) ratios, uninfected mice were anesthetized with pentobarbital, diazepam/ketamine, or inhaled isoflurane. Subsequently, they were allowed to breathe spontaneously or were mechanically ventilated. After 15 minutes exposure to room air (F(i)O(2) = 0·21) or 100% O(2) (F(i)O(2) = 1·0), carotid P(a)O(2) was measured. To determine influenza effects on P(a)O(2):F(i)O(2), mice were challenged with 10,000 p.f..u./mouse influenza A/WSN/33. RESULTS: P(a)O(2):F(i)O(2) ratios were abnormally low (≤400 mmHg) in spontaneously breathing mice. Mechanical ventilation with positive end-expiratory pressure was required to obtain P(a)O(2):F(i)O(2) ratios in uninfected mice consistent with normal values in humans (≥600 mmHg). At day 2 following infection P(a)O(2):F(i)O(2) ratios indicated the onset of acute lung injury. By day 6, P(a)O(2):F(i)O(2) ratios were <200 mmHg, indicating progression to ARDS. Impaired gas exchange in influenza-infected mice was accompanied by progressive hemoglobin desaturation, hypercapnia, uncompensated respiratory acidosis, hyperkalemia, and polycythemia. CONCLUSIONS: Influenza infection of mice results in impairment of alveolar gas exchange consistent with rapid development of acute lung injury and progression to ARDS. P(a)O(2):F(i)O(2) ratios may be of utility as clinically relevant and predictive outcome measures in influenza pathogenesis and treatment studies that use mouse models.


Asunto(s)
Lesión Pulmonar Aguda/etiología , Subtipo H1N1 del Virus de la Influenza A/fisiología , Gripe Humana/complicaciones , Alveolos Pulmonares/fisiopatología , Síndrome de Dificultad Respiratoria/etiología , Lesión Pulmonar Aguda/diagnóstico , Lesión Pulmonar Aguda/patología , Lesión Pulmonar Aguda/fisiopatología , Animales , Regulación hacia Abajo , Femenino , Humanos , Gripe Humana/fisiopatología , Gripe Humana/virología , Masculino , Ratones , Ratones Endogámicos BALB C , Alveolos Pulmonares/virología , Intercambio Gaseoso Pulmonar , Síndrome de Dificultad Respiratoria/diagnóstico , Síndrome de Dificultad Respiratoria/patología , Síndrome de Dificultad Respiratoria/fisiopatología
14.
J Biol Chem ; 288(3): 2049-58, 2013 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-23148214

RESUMEN

Cystic fibrosis is the most common inherited lethal disease in Caucasians. It is caused by mutations in the cystic fibrosis transmembrane conductance regulator (CFTR), of which the cftr ΔF508 mutation is the most common. ΔF508 macrophages are intrinsically defective in autophagy because of the sequestration of essential autophagy molecules within unprocessed CFTR aggregates. Defective autophagy allows Burkholderia cenocepacia (B. cepacia) to survive and replicate in ΔF508 macrophages. Infection by B. cepacia poses a great risk to cystic fibrosis patients because it causes accelerated lung inflammation and, in some cases, a lethal necrotizing pneumonia. Autophagy is a cell survival mechanism whereby an autophagosome engulfs non-functional organelles and delivers them to the lysosome for degradation. The ubiquitin binding adaptor protein SQSTM1/p62 is required for the delivery of several ubiquitinated cargos to the autophagosome. In WT macrophages, p62 depletion and overexpression lead to increased and decreased bacterial intracellular survival, respectively. In contrast, depletion of p62 in ΔF508 macrophages results in decreased bacterial survival, whereas overexpression of p62 leads to increased B. cepacia intracellular growth. Interestingly, the depletion of p62 from ΔF508 macrophages results in the release of the autophagy molecule beclin1 (BECN1) from the mutant CFTR aggregates and allows its redistribution and recruitment to the B. cepacia vacuole, mediating the acquisition of the autophagy marker LC3 and bacterial clearance via autophagy. These data demonstrate that p62 differentially dictates the fate of B. cepacia infection in WT and ΔF508 macrophages.


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Autofagia/genética , Infecciones por Burkholderia/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/genética , Proteínas de Choque Térmico/genética , Macrófagos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Biomarcadores/metabolismo , Infecciones por Burkholderia/complicaciones , Infecciones por Burkholderia/metabolismo , Infecciones por Burkholderia/microbiología , Burkholderia cenocepacia/fisiología , Fibrosis Quística/complicaciones , Fibrosis Quística/metabolismo , Fibrosis Quística/microbiología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Expresión Génica , Proteínas de Choque Térmico/antagonistas & inhibidores , Proteínas de Choque Térmico/metabolismo , Humanos , Macrófagos/microbiología , Macrófagos/patología , Ratones , Ratones Transgénicos , Viabilidad Microbiana , Proteínas Asociadas a Microtúbulos/metabolismo , Fagosomas/metabolismo , Transporte de Proteínas , ARN Interferente Pequeño/genética , Proteína Sequestosoma-1 , Transfección , Ubiquitina/genética , Ubiquitina/metabolismo
15.
Am J Physiol Lung Cell Mol Physiol ; 301(1): L99-L109, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-21478252

RESUMEN

Both respiratory syncytial virus (RSV) and influenza A virus induce nucleotide/P2Y purinergic receptor-mediated impairment of alveolar fluid clearance (AFC), which contributes to formation of lung edema. Although genetically dissimilar, both viruses generate double-stranded RNA replication intermediates, which act as Toll-like receptor (TLR)-3 ligands. We hypothesized that double-stranded RNA/TLR-3 signaling underlies nucleotide-mediated inhibition of amiloride-sensitive AFC in both infections. We found that addition of the synthetic double-stranded RNA analog poly-inosinic-cytidylic acid [poly(I:C)] (500 ng/ml) to the AFC instillate resulted in nucleotide/P2Y purinergic receptor-mediated inhibition of amiloride-sensitive AFC in BALB/c mice but had no effect on cystic fibrosis transmembrane regulator (CFTR)-mediated Cl(-) transport. Poly(I:C) also induced acute keratinocyte cytokine-mediated AFC insensitivity to stimulation by the ß-adrenergic agonist terbutaline. Inhibitory effects of poly(I:C) on AFC were absent in TLR-3(-/-) mice and were not replicated by addition to the AFC instillate of ligands for other TLRs except TLR-2. Intranasal poly(I:C) administration (250 µg/mouse) similarly induced nucleotide-dependent AFC inhibition 2-3 days later, together with increased lung water content and neutrophilic inflammation. Intranasal treatment of BALB/c mice with poly(I:C) did not induce airway hyperresponsiveness at day 2 but did result in insensitivity to airway bronchodilation by ß-adrenergic agonists. These findings suggest that viral double-stranded RNA replication intermediates induce nucleotide-mediated impairment of amiloride-sensitive AFC in both infections, together with ß-adrenergic agonist insensitivity. Both of these effects also occur in RSV infection. However, double-stranded RNA replication intermediates do not appear to be sufficient to induce either adenosine-mediated, CFTR-dependent Cl(-) secretion in the lung or severe, lethal hypoxemia, both of which are features of influenza infection.


Asunto(s)
Pulmón/fisiopatología , Pulmón/virología , ARN Bicatenario/farmacología , Infecciones por Virus Sincitial Respiratorio/fisiopatología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitiales Respiratorios/fisiología , Administración Intranasal , Agonistas Adrenérgicos beta/farmacología , Animales , Líquidos Corporales/efectos de los fármacos , Exposición por Inhalación , Ligandos , Pulmón/efectos de los fármacos , Lesión Pulmonar/complicaciones , Lesión Pulmonar/patología , Lesión Pulmonar/fisiopatología , Ratones , Ratones Endogámicos BALB C , Poli I-C/administración & dosificación , Poli I-C/farmacología , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/metabolismo , Receptores Purinérgicos P2Y/metabolismo , Infecciones por Virus Sincitial Respiratorio/complicaciones , Virus Sincitiales Respiratorios/efectos de los fármacos , Factores de Tiempo , Receptor Toll-Like 3/metabolismo
16.
Endocrinology ; 152(2): 483-94, 2011 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-21190962

RESUMEN

Respiratory syncytial virus (RSV) is a common cause of bronchiolitis in infants. Although antiinflammatory in nature, glucocorticoids have been shown to be ineffective in the treatment of RSV-induced bronchiolitis and wheezing. In addition, the effectiveness of glucocorticoids at inhibiting RSV-induced proinflammatory cytokine production in cell culture has been questioned. In this study, we have investigated the effect of RSV infection on glucocorticoid-induced gene activation in lung epithelium-derived cells. We show that RSV infection inhibits dexamethasone induction of three glucocorticoid receptor (GR)-regulated genes (glucocorticoid-inducible leucine zipper, FK506 binding protein, and MAPK phosphatase 1) in A549, BEAS-2B cells, and primary small airway epithelial cells. UV irradiation of the virus prevents this repression, suggesting that viral replication is required. RSV is known to activate the nuclear factor κB (NFκB) pathway, which is mutually antagonistic towards the GR pathway. However, specific inhibition of NFκB had no effect on the repression of GR-induced genes by RSV infection, indicating that RSV repression of GR is independent of NFκB. RSV infection of A549 cells does not alter GR protein levels or GR nuclear translocation but does reduce GR binding to the promoters of the glucocorticoid responsive genes analyzed in this study. Repression of GR by RSV infection may account for the apparent clinical ineffectiveness of glucocorticoids in RSV bronchiolitis therapy. In addition, this data adds to our previously published data suggesting that GR may be a general target for infectious agents. Identifying the mechanisms through which this suppression occurs may lead to the development of novel therapeutics.


Asunto(s)
Receptores de Glucocorticoides/metabolismo , Infecciones por Virus Sincitial Respiratorio/metabolismo , Virus Sincitiales Respiratorios/fisiología , Activación Transcripcional/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/genética , Células Cultivadas , Inmunoprecipitación de Cromatina , Dexametasona/farmacología , Ensayo de Inmunoadsorción Enzimática , Silenciador del Gen , Humanos , Inmunohistoquímica , Interleucina-1beta/farmacología , Interleucina-6/metabolismo , Interleucina-8/metabolismo , Reacción en Cadena de la Polimerasa , ARN Interferente Pequeño/genética , Receptores de Glucocorticoides/genética , Infecciones por Virus Sincitial Respiratorio/virología , Activación Transcripcional/genética
17.
Toxicol Sci ; 116(1): 349-58, 2010 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20363832

RESUMEN

Cadmium is a toxic heavy metal ranked seventh on the Priority List of Hazardous Substances. As a byproduct of smelters, cadmium is a prevalent environmental contaminant. It is also a major component of cigarette smoke, and its inhalation is associated with decreased pulmonary function, lung cancer, and chronic obstructive pulmonary disease. Ion channels, including the cystic fibrosis transmembrane conductance regulator (CFTR), play a central role in maintaining fluid homeostasis and lung functions. CFTR is mostly expressed in epithelial cells, and little is known about the effect of cadmium exposure on lung epithelial cell function. We show that exposure to cadmium decreases the expression of the CFTR protein and subsequent chloride transport in human airway epithelial cells in vitro. Impairment of CFTR protein expression was also observed in vivo in the lung of mice after intranasal instillation of cadmium. We established that the inhibitory effect of cadmium was not a nonspecific effect of heavy metals, as nickel had no effect on CFTR protein levels. Finally, we show that selected antioxidants, including alpha-tocopherol (vitamin E), but not N-acetylcysteine, can prevent the cadmium-induced suppression of CFTR. In summary, we have identified cadmium as a regulator of the CFTR chloride channel present in lung epithelial cells. Future strategies to prevent the deleterious effect of cadmium on epithelial cells and lung functions may benefit from the finding that alpha-tocopherol protects CFTR expression and function.


Asunto(s)
Cadmio/toxicidad , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Expresión Génica/efectos de los fármacos , Tráquea/efectos de los fármacos , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Humanos , Reacción en Cadena de la Polimerasa , Tráquea/citología , Tráquea/metabolismo
18.
Am J Physiol Lung Cell Mol Physiol ; 297(4): L785-93, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19684202

RESUMEN

High tidal volume ventilation is detrimental to alveolar fluid clearance (AFC), but effects of ventilation pressure (P) on AFC are unknown. In anesthetized BALB/c mice ventilated at constant tidal volume (8 ml/kg), mean AFC rate was 12.8% at 6 cmH(2)O P, but increased to 37.3% at 18 cmH(2)O P. AFC rate declined at 22 cmH(2)O P, which also induced lung damage. Increased AFC at 18 cmH(2)O P did not result from elevated plasma catecholamines, hypercapnia, or hypocapnia, but was due to augmented Na(+) and Cl(-) absorption. PKA agonists and beta-agonists stimulated AFC at 10 cmH(2)O P by upregulating amiloride-sensitive Na(+) transport. However, at 18 cmH(2)O P, PKA agonists and beta-agonists reduced AFC. At 15 cmH(2)O P, the AFC rate was intermediate (mean 26.6%), and forskolin and beta-agonists had no effect. Comparable P dependency of AFC and beta-agonist responsiveness was found in C57BL/6 mice. The effect on AFC of increasing P to 18 cmH(2)O was blocked by adenosine deaminase or an A(2b)-adenosine receptor antagonist, and could be mimicked by adenosine in mice ventilated at 10 cmH(2)O P. Modulation of adenosine signaling also resulted in altered responsiveness to beta-agonists. These findings indicate that, in the normal mouse lung, basal AFC rates and responses to beta-agonists are impacted by ventilation pressure in an adenosine-dependent manner.


Asunto(s)
Agonistas de Receptores Adrenérgicos beta 2 , Agonistas Adrenérgicos beta/farmacología , Pulmón/metabolismo , Depuración Mucociliar , Alveolos Pulmonares/metabolismo , Respiración Artificial , Mucosa Respiratoria/metabolismo , Amilorida/farmacología , Animales , Líquido del Lavado Bronquioalveolar , Colforsina/farmacología , Células Epiteliales/metabolismo , Femenino , Pulmón/citología , Pulmón/efectos de los fármacos , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Oxígeno/metabolismo , Alveolos Pulmonares/citología , Alveolos Pulmonares/efectos de los fármacos , Mucosa Respiratoria/citología , Mucosa Respiratoria/efectos de los fármacos
19.
Br J Pharmacol ; 156(1): 189-200, 2009 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19134000

RESUMEN

BACKGROUND AND PURPOSE: Injury to the lung parenchyma is a constitutional feature shared by many lung diseases. The protein, phosphatase and tensin homologue deleted on chromosome Ten (PTEN) is a major suppressor of phosphoinositide-3 kinase/Akt signalling, a vital survival pathway in lung parenchymal cells. Based on this, we hypothesized that PTEN inhibition in vivo would enhance cell tolerance to stress thereby preventing acute lung injury. EXPERIMENTAL APPROACH: We evaluated the ability of a PTEN inhibitor, potassium bisperoxo (1,10-phenanthroline) oxovanadate [bpV(phen)], to prevent acute lung injury induced by oleic acid (OA) in adult C57BL/6 mice. Lung assessments included bronchoalveolar lavage, tissue morphology, immunostaining for markers of cell death, cell identity, phospho-Akt and phospho-ERK levels and oximetry. KEY RESULTS: OA induced acute lung injury in a dose- and time-dependent manner. No injury was observed in the vehicle control or bpV(phen) treatment groups. PTEN inhibition by bpV(phen) increased lung tissue levels of phospho-Akt and ERK and but not focal adhesion kinase. This occurred in conjunction with a statistically significant reduction in protein content, lactate dehydrogenase, as well as tumour necrosis factor-alpha and chemokines in bronchoalveolar lavage fluid when compared with OA treatment alone. The incidence of alveolar lesions, consistent with acute lung injury, and terminal uridine deoxynucleotidyl transferase dUTP nick end labelling (TUNEL)-positive cells was also significantly reduced. Importantly, PTEN suppression maintained pulmonary function. CONCLUSIONS AND IMPLICATIONS: Treatment with bpV(phen) significantly reduced the severity of acute lung injury in mice indicating that additional investigation is warranted to understand the important role that this phosphatase may play in the lung.


Asunto(s)
Lesión Pulmonar Aguda/prevención & control , Compuestos Organometálicos/farmacología , Fosfohidrolasa PTEN/antagonistas & inhibidores , Fenantrolinas/farmacología , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/enzimología , Animales , Líquido del Lavado Bronquioalveolar , Quimiocinas/metabolismo , Relación Dosis-Respuesta a Droga , Quinasas MAP Reguladas por Señal Extracelular/metabolismo , Etiquetado Corte-Fin in Situ , L-Lactato Deshidrogenasa/metabolismo , Pulmón/enzimología , Pulmón/patología , Ratones , Ratones Endogámicos C57BL , Ácido Oléico , Compuestos Organometálicos/administración & dosificación , Compuestos Organometálicos/uso terapéutico , Fenantrolinas/administración & dosificación , Fenantrolinas/uso terapéutico , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal , Factor de Necrosis Tumoral alfa/metabolismo
20.
Am J Respir Cell Mol Biol ; 40(5): 588-600, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-18952569

RESUMEN

We investigated the mechanisms by which respiratory syncytial virus (RSV) infection decreases vectorial Na+ transport across respiratory epithelial cells. Mouse tracheal epithelial (MTE) cells from either BALB/c or C57BL/6 mice and human airway H441 cells were grown on semipermeable supports under an air-liquid interface. Cells were infected with RSV-A2 and mounted in Ussing chambers for measurements of short-circuit currents (I(sc)). Infection with RSV for 24 hours (multiplicity of infection = 1) resulted in positive immunofluorescence for RSV antigen in less than 10% of MTE or H441 cells. In spite of the limited number of cells infected, RSV reduced both basal and amiloride-sensitive I(sc) in both MTE and H441 cells by approximately 50%, without causing a concomitant reduction in transepithelial resistance. Agents that increased intracellular cAMP (forskolin, cpt-CAMP, and IBMX) increased mainly Cl(-) secretion in MTE cells and Na+ absorption in H441 cells. RSV infection for 24 hours blunted both variables. In contrast, ouabain sensitive I(sc), measured across apically permeabilized H441 monolayers, remained unchanged. Western blot analysis of H441 cell lysates demonstrated reductions in alpha- but not gamma-ENaC subunit protein levels at 24 hours after RSV infection. The reduction in amiloride-sensitive I(sc) in H441 cells was prevented by pretreatment with inhibitors of de novo pyrimidine or purine synthesis (A77-1726 and 6-MP, respectively, 50 microM). Our results suggest that infection of both murine and human respiratory epithelial cells with RSV inhibits vectorial Na+ transport via nucleotide release. These findings are consistent with our previous studies showing reduced alveolar fluid clearance after RSV infection of BALB/c mice.


Asunto(s)
Células Epiteliales/metabolismo , Células Epiteliales/virología , Pulmón/citología , Pulmón/virología , Infecciones por Virus Sincitial Respiratorio/metabolismo , Sodio/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Transporte Biológico Activo/efectos de los fármacos , Línea Celular , Colforsina/farmacología , Células Epiteliales/efectos de los fármacos , Canales Epiteliales de Sodio/metabolismo , Técnica del Anticuerpo Fluorescente , Humanos , Activación del Canal Iónico/efectos de los fármacos , Pulmón/metabolismo , Ratones , Subunidades de Proteína/metabolismo , Purinas/biosíntesis , Pirimidinas/biosíntesis , Terbutalina/farmacología , Tráquea/citología , Tráquea/virología , Uridina Trifosfato/farmacología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA