Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Hypertension ; 80(2): 385-402, 2023 02.
Artículo en Inglés | MEDLINE | ID: mdl-36440576

RESUMEN

BACKGROUND: Aortic aneurysm (AA) is a "silent killer" human disease with no effective treatment. Although the therapeutic potential of various pharmacological agents have been evaluated, there are no reports of ß-arrestin-biased AT1R (angiotensin-II type-1 receptor) agonist (TRV027) used to prevent the progression of AA. METHODS: We tested the hypothesis that TRV027 infusion in AngII (angiotensin II)-induced mouse model of AA prevents AA. High-fat-diet-fed ApoE (apolipoprotein E gene)-null mice were infused with AngII to induce AA and co-infused with TRV027 and a clinically used AT1R blocker Olmesartan to prevent AA. Aortas explanted from different ligand infusion groups were compared with assess different grades of AA or lack of AA. RESULTS: AngII produced AA in ≈67% male mice with significant mortality associated with AA rupture. We observed ≈13% mortality due to aortic arch dissection without aneurysm in male mice. AngII-induced AA and mortality was prevented by co-infusion of TRV027 or Olmesartan, but through different mechanisms. In TRV027 co-infused mice aortic wall thickness, elastin content, new DNA, and protein synthesis were higher than untreated and Olmesartan co-infused mice. Co-infusion with both TRV027 and Olmesartan prevented endoplasmic reticulum stress, fibrosis, and vasomotor hyper responsiveness. CONCLUSIONS: TRV027-engaged AT1R prevented AA and associated mortality by distinct molecular mechanisms compared with the AT1R blocker, Olmesartan. Developing novel ß-arrestin-biased AT1R ligands may yield promising drugs to combat AA.


Asunto(s)
Aneurisma de la Aorta , Animales , Femenino , Masculino , Ratones , Angiotensina II/farmacología , Aorta/metabolismo , beta-Arrestinas , Ratones Noqueados , Receptor de Angiotensina Tipo 1/metabolismo
2.
Drug Deliv ; 29(1): 2206-2216, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-35815732

RESUMEN

Cancer is a leading cause of death worldwide and affects society in terms of the number of lives lost. Current cancer treatments are based on conventional chemotherapy which is nonspecific in targeting cancer. Therefore, intensive efforts are underway to better target cancer-specific cells while minimizing the side effects on healthy tissues by using LDL particles as active drug delivery vehicles. The goal is to encapsulate anticancer agents thiosemicarbazone metal-ligand complexes into LDL particles to increase the cytotoxic effect of the agent by internalization through LDL receptors into MCF7, A549, and C42 cancer cell lines as segregate models for biological evaluations targeting tubulin. Zeta potential data of LDL-particles encapsulated anticancer agents showed an acceptable diameter range between 66-91 nm and uniform particle morphology. The results showed cell proliferation reduction in all tested cell lines. The IC50 values of LDL encapsulated thiosemicarbazone metal-ligand complexes treated with MCF7, A549, and C42 ranged between 1.18-6.61 µM, 1.17-9.66 µM, and 1.01-6.62 µM, respectively. Western blot analysis showed a potent decrease in tubulin expression when the cell lines were treated with LDL particles encapsulated with thiosemicarbazone metal-ligand complexes as anticancer agents. In conclusion, the data provide strong evidence that LDL particles are used as an active drug delivery strategy for cancer therapy.


Asunto(s)
Antineoplásicos , Complejos de Coordinación , Neoplasias de la Próstata , Tiosemicarbazonas , Antineoplásicos/farmacología , Complejos de Coordinación/farmacología , Humanos , Ligandos , Lipoproteínas LDL , Pulmón , Masculino , Tiosemicarbazonas/farmacología , Tubulina (Proteína)
3.
Front Immunol ; 13: 853009, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35359954

RESUMEN

Overt and subclinical maternal infections in pregnancy can have multiple and significant pathological consequences for the developing fetus, leading to acute perinatal complications and/or chronic disease throughout postnatal life. In this context, the current concept of pregnancy as a state of systemic immunosuppression seems oversimplified and outdated. Undoubtedly, in pregnancy the maternal immune system undergoes complex changes to establish and maintain tolerance to the fetus while still protecting from pathogens. In addition to downregulated maternal immunity, hormonal changes, and mechanical adaptation (e.g., restricted lung expansion) make the pregnant woman more susceptible to respiratory pathogens, such as influenza virus, respiratory syncytial virus (RSV), and severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). Depending on the infectious agent and timing of the infection during gestation, fetal pathology can range from mild to severe, and even fatal. Influenza is associated with a higher risk of morbidity and mortality in pregnant women than in the general population, and, especially during the third trimester of pregnancy, mothers are at increased risk of hospitalization for acute cardiopulmonary illness, while their babies show higher risk of complications such as prematurity, respiratory and neurological illness, congenital anomalies, and admission to neonatal intensive care. RSV exposure in utero is associated with selective immune deficit, remodeling of cholinergic innervation in the developing respiratory tract, and abnormal airway smooth muscle contractility, which may predispose to postnatal airway inflammation and hyperreactivity, as well as development of chronic airway dysfunction in childhood. Although there is still limited evidence supporting the occurrence of vertical transmission of SARS-CoV-2, the high prevalence of prematurity among pregnant women infected by SARS-CoV-2 suggests this virus may alter immune responses at the maternal-fetal interface, affecting both the mother and her fetus. This review aims at summarizing the current evidence about the short- and long-term consequences of intrauterine exposure to influenza, RSV, and SARS-CoV-2 in terms of neonatal and pediatric outcomes.


Asunto(s)
COVID-19 , Gripe Humana , Complicaciones Infecciosas del Embarazo , Niño , Femenino , Humanos , Recién Nacido , Transmisión Vertical de Enfermedad Infecciosa , Embarazo , Virus Sincitiales Respiratorios , SARS-CoV-2
4.
PLoS One ; 17(3): e0265094, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35263387

RESUMEN

We have shown that respiratory syncytial virus (RSV) can spread hematogenously from infected airways of a pregnant woman to the developing fetal lungs in utero. This study sought to measure RSV replication, cytopathic effects, and protein expression in human lung organoids (HLOs) reproducing architecture and transcriptional profiles of human fetal lungs during the 1st trimester of gestation. HLOs derived from human pluripotent stem cells were microinjected after 50 or 100 days in culture with medium or recombinant RSV-A2 expressing the red fluorescent protein gene (rrRSV). Infection was monitored by fluorescent microscopy and PCR. Immunohistochemistry and proteomic analysis were performed. RSV infected HLOs in a dose- and time-dependent manner. RSV-infected HLOs increased expression of CC10 (Club cells), but had sparse FOXJ1 (ciliated cells). Disruption of F-actin cytoskeleton was consistent with proteomic data showing a significant increase in Rho GTPases proteins. RSV upregulated the transient receptor potential vanilloid 1 (TRPV1) channel and, while ß2 adrenergic receptor (ß2AR) expression was decreased overall, its phosphorylated form increased. Our data suggest that prenatal RSV infection produces profound changes in fetal lungs' architecture and expression profiles and maybe an essential precursor of chronic airway dysfunction. expression profiles, and possibly be an important precursor of chronic airway dysfunction.


Asunto(s)
Neumonía Viral , Infecciones por Virus Sincitial Respiratorio , Virus Sincitial Respiratorio Humano , Femenino , Humanos , Pulmón/metabolismo , Organoides/metabolismo , Embarazo , Proteómica , Virus Sincitial Respiratorio Humano/fisiología
5.
Sci Signal ; 14(685)2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-34074703

RESUMEN

Pharmacologic agonism of the ß2-adrenergic receptor (ß2AR) induces bronchodilation by activating the enzyme adenylyl cyclase to generate cyclic adenosine monophosphate (cAMP). ß2AR agonists are generally the most effective strategy to relieve acute airway obstruction in asthmatic patients, but they are much less effective when airway obstruction in young patients is triggered by infection with respiratory syncytial virus (RSV). Here, we investigated the effects of RSV infection on the abundance and function of ß2AR in primary human airway smooth muscle cells (HASMCs) derived from pediatric lung tissue. We showed that RSV infection of HASMCs resulted in proteolytic cleavage of ß2AR mediated by the proteasome. RSV infection also resulted in ß2AR ligand-independent activation of adenylyl cyclase, leading to reduced cAMP synthesis compared to that in uninfected control cells. Last, RSV infection caused stronger airway smooth muscle cell contraction in vitro due to increased cytosolic Ca2+ concentrations. Thus, our results suggest that RSV infection simultaneously induces loss of functional ß2ARs and activation of multiple pathways favoring airway obstruction in young patients, with the net effect of counteracting ß2AR agonist-induced bronchodilation. These findings not only provide a potential mechanism for the reported lack of clinical efficacy of ß2AR agonists for treating virus-induced wheezing but also open the path to developing more precise therapeutic strategies.


Asunto(s)
Asma , Virus Sincitiales Respiratorios , Niño , AMP Cíclico , Humanos , Pulmón , Miocitos del Músculo Liso
6.
Am J Physiol Lung Cell Mol Physiol ; 321(1): L189-L203, 2021 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-34010080

RESUMEN

The airway epithelium's ability to repair itself after injury, known as epithelial restitution, is an essential mechanism enabling the respiratory tract's normal functions. Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infections worldwide. We sought to determine whether RSV delays the airway epithelium wound repair process both in vitro and in vivo. We found that RSV infection attenuated epithelial cell migration, a step in wound repair, promoted stress fiber formation, and mediated assembly of large focal adhesions. Inhibition of Rho-associated kinase, a master regulator of actin function, reversed these effects. There was increased RhoA and phospho-myosin light chain 2 following RSV infection. In vivo, mice were intraperitoneally inoculated with naphthalene to induce lung injury, followed by RSV infection. RSV infection delayed reepithelialization. There were increased concentrations of phospho-myosin light chain 2 in day 7 naphthalene + RSV animals, which normalized by day 14. This study suggests a key mechanism by which RSV infection delays wound healing.

7.
Am J Physiol Lung Cell Mol Physiol ; 320(6): L1074-L1084, 2021 06 01.
Artículo en Inglés | MEDLINE | ID: mdl-33787326

RESUMEN

The transient receptor potential vanilloid 1 (TRPV1) channel is expressed in human bronchial epithelium (HBE), where it transduces Ca2+ in response to airborne irritants. TRPV1 activation results in bronchoconstriction, cough, and mucus production, and may therefore contribute to the pathophysiology of obstructive airway disease. Since children with asthma face the greatest risk of developing virus-induced airway obstruction, we hypothesized that changes in TRPV1 expression, localization, and function in the airway epithelium may play a role in bronchiolitis and asthma in childhood. We sought to measure TRPV1 protein expression, localization, and function in HBE cells from children with versus without asthma, both at baseline and after RSV infection. We determined changes in TRPV1 protein expression, subcellular localization, and function both at baseline and after RSV infection in primary HBE cells from normal children and children with asthma. Basal TRPV1 protein expression was higher in HBE from children with versus without asthma and primarily localized to plasma membranes (PMs). During RSV infection, TRPV1 protein increased more in the PM of asthmatic HBE as compared with nonasthmatic cells. TRPV1-mediated increase in intracellular Ca2+ was greater in RSV-infected asthmatic cells, but this increase was attenuated when extracellular Ca2+ was removed. Nerve growth factor (NGF) recapitulated the effect of RSV on TRPV1 activation in HBE cells. Our data suggest that children with asthma have intrinsically hyperreactive airways due in part to higher TRPV1-mediated Ca2+ influx across epithelial membranes, and this abnormality is further exacerbated by NGF overexpression during RSV infection driving additional Ca2+ from intracellular stores.


Asunto(s)
Asma/virología , Calcio/metabolismo , Transporte Iónico/fisiología , Canales Catiónicos TRPV/metabolismo , Asma/metabolismo , Broncoconstricción/fisiología , Niño , Preescolar , Células Epiteliales/metabolismo , Epitelio/metabolismo , Humanos , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/virología , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico
8.
Am J Physiol Lung Cell Mol Physiol ; 319(3): L481-L496, 2020 09 01.
Artículo en Inglés | MEDLINE | ID: mdl-32640839

RESUMEN

Respiratory syncytial virus (RSV) is the leading cause of lower respiratory tract infections in children worldwide. While most develop a mild, self-limiting illness, some develop severe acute lower respiratory infection and persistent airway disease. Exposure to ambient particulate matter has been linked to asthma, bronchitis, and viral infection in multiple epidemiological studies. We hypothesized that coexposure to nanoparticles worsens RSV-induced airway epithelial barrier dysfunction. Bronchial epithelial cells were incubated with titanium dioxide nanoparticles (TiO2-NP) or a combination of TiO2-NP and RSV. Structure and function of epithelial cell barrier were analyzed. Viral titer and the role of reactive oxygen species (ROS) generation were evaluated. In vivo, mice were intranasally incubated with TiO2-NP, RSV, or a combination. Lungs and bronchoalveolar lavage (BAL) fluid were harvested for analysis of airway inflammation and apical junctional complex (AJC) disruption. RSV-induced AJC disruption was amplified by TiO2-NP. Nanoparticle exposure increased viral infection in epithelial cells. TiO2-NP induced generation of ROS, and pretreatment with antioxidant, N-acetylcysteine, reversed said barrier dysfunction. In vivo, RSV-induced injury and AJC disruption were augmented in the lungs of mice given TiO2-NP. Airway inflammation was exacerbated, as evidenced by increased white blood cell infiltration into the BAL, along with exaggeration of peribronchial inflammation and AJC disruption. These data demonstrate that TiO2-NP exposure exacerbates RSV-induced AJC dysfunction and increases inflammation by mechanisms involving generation of ROS. Further studies are required to determine whether NP exposure plays a role in the health disparities of asthma and other lung diseases, and why some children experience more severe airway disease with RSV infection.


Asunto(s)
Células Epiteliales/efectos de los fármacos , Infecciones por Virus Sincitial Respiratorio/tratamiento farmacológico , Virus Sincitiales Respiratorios/patogenicidad , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Titanio/farmacología , Animales , Asma/tratamiento farmacológico , Asma/etiología , Bronquios/efectos de los fármacos , Bronquios/virología , Líquido del Lavado Bronquioalveolar/citología , Células Epiteliales/virología , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Pulmón/efectos de los fármacos , Pulmón/virología , Ratones , Virus Sincitiales Respiratorios/efectos de los fármacos
9.
Pediatr Res ; 87(2): 420-426, 2020 01.
Artículo en Inglés | MEDLINE | ID: mdl-31698410

RESUMEN

New information is emerging concerning the influence of environmental factors (e.g., viruses, pollutants, nutrients) on fetal lung development and the prenatal modulation of cellular and molecular effectors essential to the control of airway function, which may shed new light into the pathogenesis of chronic obstructive pulmonary disease in childhood. In particular, recent studies have shown that nanosize biological and inorganic particles (e.g., respiratory viruses and pollutants) are able to spread hematogenously across the placenta from mother to offspring and interfere with lung development during critical "windows of opportunity". Furthermore, the nutritional balance of maternal diet during pregnancy can affect postnatal lung structure and function. Adverse prenatal environmental conditions can predispose to increased airway reactivity by inducing aberrant cholinergic innervation of the respiratory tract, enhanced contractility of the airway smooth muscle, and impaired innate immunity. Such changes can persist long after birth and might provide a plausible explanation to the development of chronic airway dysfunction in children, even in the absence of atopic predisposition. Insight into maternal-fetal interactions will contribute to a better understanding of the pathogenesis of highly prevalent diseases like bronchiolitis and asthma, and may lead to more precise preventative and therapeutic strategies, or new indications for existing ones.


Asunto(s)
Dieta/efectos adversos , Contaminantes Ambientales/efectos adversos , Transmisión Vertical de Enfermedad Infecciosa , Enfermedades Pulmonares/etiología , Pulmón/crecimiento & desarrollo , Intercambio Materno-Fetal , Efectos Tardíos de la Exposición Prenatal , Infecciones del Sistema Respiratorio/transmisión , Virosis/transmisión , Animales , Femenino , Humanos , Pulmón/efectos de los fármacos , Pulmón/virología , Enfermedades Pulmonares/fisiopatología , Enfermedades Pulmonares/virología , Exposición Materna/efectos adversos , Fenómenos Fisiologicos Nutricionales Maternos , Estado Nutricional , Valor Nutritivo , Embarazo , Infecciones del Sistema Respiratorio/fisiopatología , Infecciones del Sistema Respiratorio/virología , Medición de Riesgo , Factores de Riesgo , Virosis/fisiopatología , Virosis/virología
10.
PLoS One ; 14(12): e0225767, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-31790466

RESUMEN

BACKGROUND: Respiratory syncytial virus (RSV) is capable of transient viremia and extrapulmonary dissemination. Recently, this virus has been identified in fetal cord blood, suggesting the possibility of in utero acquisition in humans. Here, we assess permissivity and kinetics of RSV replication in primary human placental cells, examine their potential to transfer this infection to neighboring cells, and measure the inflammatory response evoked by the virus. METHODS AND FINDINGS: Human placental villus tissue was collected immediately upon delivery and processed for isolation of placental cytotrophoblast, fibroblast, and macrophage (Hofbauer) cells. Isolated cells were infected with a recombinant RSV-A2 strain (rrRSV) expressing red fluorescent protein (RFP) and analyzed by fluorescence microscopy, Western blot, and quantitative PCR (qPCR). Based on RFP expression, rrRSV exhibited differential tropism for the three major placental cell types. Placental fibroblasts and Hofbauer cells were permissive and supported productive rrRSV replication. While infected cytotrophoblast cells expressed viral glycoprotein (G protein), only limited RSV replication was detected. Importantly, qPCR and fluorescence-focused unit assay revealed that the viral progeny remains trapped within infected Hofbauer cells for up to 30 days, with no release into surrounding media. Yet, Hofbauer cells passed the infection onto overlaid naïve 16HBE cells, suggesting contact-dependent trans-infection. Lastly, a significant increase in proinflammatory cytokines, particularly IL-6, TNF-alpha, and IFN-gamma was measured in the supernatant of infected Hofbauer cells by multiplex cytokine assay and conventional ELISA. CONCLUSIONS: This study demonstrates that RSV can replicate in human placenta, exhibits differential tropism for distinct placental cell types, can be stored and transferred to neighboring cells by Hofbauer cells, and elicits an inflammatory response. It also supports the hypothesis that this respiratory virus can be vertically transferred to the fetus and potentially affect its development and the outcome of pregnancies.


Asunto(s)
Placenta/patología , Placenta/virología , Virus Sincitial Respiratorio Humano/fisiología , Tropismo , Citocinas/metabolismo , Femenino , Humanos , Mediadores de Inflamación/metabolismo , Cinética , Embarazo , Infecciones por Virus Sincitial Respiratorio/patología , Infecciones por Virus Sincitial Respiratorio/virología
11.
Am J Physiol Lung Cell Mol Physiol ; 316(2): L358-L368, 2019 02 01.
Artículo en Inglés | MEDLINE | ID: mdl-30489157

RESUMEN

Respiratory syncytial virus (RSV) is a major cause of hospitalization for infants and young children worldwide. RSV is known to infect epithelial cells and increase the permeability of model airway epithelial monolayers in vitro. We hypothesized that RSV infection also induces airway barrier dysfunction in vivo. C57BL/6 mice were intranasally inoculated with RSV, and on day 4 post-inoculation were examined for viral replication, lung inflammation, and barrier integrity as well as the structure and molecular composition of epithelial junctions. In parallel, primary mouse tracheal epithelial cells (mTEC) were cultured for in vitro studies. RSV-infected mice lost weight and showed significant peribronchial inflammation compared with noninfected controls and UV-inactivated RSV-inoculated animals. RSV infection increased the permeability of the airway epithelial barrier and altered the molecular composition of epithelial tight junctions. The observed RSV-induced barrier disruption was accompanied by decreased expression of several tight-junction proteins and accumulation of cleaved extracellular fragments of E-cadherin in bronchoalveolar lavage and mTEC supernatants. Similarly, in vitro RSV infection of mTEC monolayers resulted in enhanced permeability and disruption of tight-junction structure. Furthermore, incubation of mTEC monolayers with a recombinant fragment of E-cadherin caused tight-junction disassembly. Taken together, these data indicate that RSV infection leads to airway barrier dysfunction in vivo, mediated by either decreased expression or cleavage of junctional proteins. Our observations provide further insights into the pathophysiology of RSV infection and provide a rationale for development of barrier-protecting agents to alleviate the pathogenic effects of RSV infection.


Asunto(s)
Células Epiteliales/virología , Infecciones por Virus Sincitial Respiratorio/virología , Virus Sincitial Respiratorio Humano/patogenicidad , Uniones Estrechas/virología , Animales , Modelos Animales de Enfermedad , Células Epiteliales/metabolismo , Ratones Endogámicos C57BL , Sistema Respiratorio/metabolismo , Uniones Estrechas/metabolismo
12.
Pediatr Res ; 83(5): 1049-1056, 2018 05.
Artículo en Inglés | MEDLINE | ID: mdl-29329282

RESUMEN

BackgroundDespite decades that have passed since its discovery, accurate biomarkers of respiratory syncytial virus (RSV) disease activity and effective therapeutic strategies are still lacking. The high-mobility group box type 1 (HMGB1) protein has been proposed as a possible link between RSV and immune system, but only limited information is currently available to support this hypothesis.MethodsExpression of HMGB1 gene and protein was analyzed by quantitative PCR, enzyme-linked immunosorbent assay (ELISA), western blot, immunocytochemistry, and confocal microscopy in immortalized and primary human bronchial epithelial cells, as well as in rat pup lungs. The role of HMGB1 in RSV infection was explored using glycyrrhizin, a selective HMGB1 inhibitor.ResultsRSV infection strongly induced HMGB1 expression both in vitro and in vivo. Glycyrrhizin dose-dependently inhibited HMGB1 upregulation in both RSV-infected immortalized and primary human bronchial epithelial cells, and this effect was associated with significant reduction of viral replication.ConclusionOur data suggest that HMGB1 expression increases during RSV replication. This seems to have a critical pathogenic role as its selective inhibition virtually modified the infection. These observations provide further insight into the pathophysiology of RSV infection and uncover a potential biomarker and therapeutic target for the most common respiratory infection of infancy.


Asunto(s)
Proteína HMGB1/metabolismo , Infecciones por Virus Sincitial Respiratorio/metabolismo , Virus Sincitiales Respiratorios/metabolismo , Animales , Biomarcadores/metabolismo , Bronquios/metabolismo , Bronquiolitis/virología , Línea Celular , Células Epiteliales/metabolismo , Perfilación de la Expresión Génica , Humanos , Sistema Inmunológico , Pulmón/metabolismo , Ratas , Ratas Endogámicas F344 , Infecciones del Sistema Respiratorio/metabolismo , Infecciones del Sistema Respiratorio/virología , Regulación hacia Arriba , Replicación Viral
14.
Apoptosis ; 22(12): 1532-1542, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28918507

RESUMEN

The muscle regulatory transcription factor MyoD is a master regulator of skeletal myoblast differentiation. We have previously reported that MyoD is also necessary for the elevated expression of the pro-apoptotic Bcl2 family member PUMA, and the ensuing apoptosis, that occurs in a subset of myoblasts induced to differentiate. Herein, we report the identification of a functional MyoD binding site within the extended PUMA promoter. In silico analysis of the murine PUMA extended promoter revealed three potential MyoD binding sites within 2 kb of the transcription start site. Expression from a luciferase reporter construct containing this 2 kb fragment was enhanced by activation of MyoD in both myoblasts and fibroblasts and diminished by silencing of MyoD in myoblasts. Experiments utilizing truncated versions of this promoter region revealed that the potential binding site at position - 857 was necessary for expression. Chromatin immunoprecipitation (ChIP) analysis confirmed binding of MyoD to the DNA region encompassing position - 857. The increase in MyoD binding to the PUMA promoter as a consequence of culture in differentiation media (DM) was comparable to the increase in MyoD binding at the myogenin promoter and was diminished in myoblasts silenced for MyoD expression. Finally, ChIP analysis using an antibody specific for the transcription factor p53 demonstrated that, in myoblasts silenced for MyoD expression, p53 binding to the PUMA promoter was diminished in response to culture in DM. These data indicate that MyoD plays a direct role in regulating PUMA expression and reveal functional consequences of MyoD expression on p53 mediated transcription of PUMA.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Regulación de la Expresión Génica/efectos de los fármacos , Proteína MioD/metabolismo , Mioblastos/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Proteínas Supresoras de Tumor/genética , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Sitios de Unión , Diferenciación Celular , Línea Celular , Medios de Cultivo , Fibroblastos/citología , Fibroblastos/metabolismo , Silenciador del Gen , Ratones , MicroARNs/genética , Proteína MioD/genética , Mioblastos/citología , Miogenina/genética , Miogenina/metabolismo , Regiones Promotoras Genéticas , Unión Proteica , Proteínas Supresoras de Tumor/metabolismo
15.
PLoS One ; 12(7): e0181876, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28759570

RESUMEN

Airway epithelium forms a barrier to the outside world and has a crucial role in susceptibility to viral infections. Cyclic adenosine monophosphate (cAMP) is an important second messenger acting via two intracellular signaling molecules: protein kinase A (PKA) and the guanidine nucleotide exchange factor, Epac. We sought to investigate effects of increased cAMP level on the disruption of model airway epithelial barrier caused by RSV infection and the molecular mechanisms underlying cAMP actions. Human bronchial epithelial cells were infected with RSV-A2 and treated with either cAMP releasing agent, forskolin, or cAMP analogs. Structure and functions of the Apical Junctional Complex (AJC) were evaluated by measuring transepithelial electrical resistance and permeability to FITC-dextran, and determining localization of AJC proteins by confocal microscopy. Increased intracellular cAMP level significantly attenuated RSV-induced disassembly of AJC. These barrier-protective effects of cAMP were due to the activation of PKA signaling and did not involve Epac activity. Increased cAMP level reduced RSV-induced reorganization of the actin cytoskeleton, including apical accumulation of an essential actin-binding protein, cortactin, and inhibited expression of the RSV F protein. These barrier-protective and antiviral-function of cAMP signaling were evident even when cAMP level was increased after the onset of RSV infection. Taken together, our study demonstrates that cAMP/PKA signaling attenuated RSV-induced disruption of structure and functions of the model airway epithelial barrier by mechanisms involving the stabilization of epithelial junctions and inhibition of viral biogenesis. Improving our understanding of the mechanisms involved in RSV-induced epithelial dysfunction and viral pathogenesis will help to develop novel anti-viral therapeutic approaches.


Asunto(s)
Proteínas Quinasas Dependientes de AMP Cíclico/metabolismo , AMP Cíclico/metabolismo , Epitelio/virología , Virus Sincitial Respiratorio Humano , Infecciones del Sistema Respiratorio/virología , Citoesqueleto de Actina/metabolismo , Bronquios/citología , Colforsina/farmacología , Dextranos/química , Células Epiteliales/metabolismo , Células Epiteliales/virología , Epitelio/patología , Fluoresceína-5-Isotiocianato/análogos & derivados , Factores de Intercambio de Guanina Nucleótido/metabolismo , Humanos , Uniones Intercelulares/metabolismo , Proteínas de Microfilamentos/metabolismo , Microscopía Confocal , Permeabilidad , Transducción de Señal/efectos de los fármacos , Resultado del Tratamiento
16.
Curr Opin Virol ; 24: 70-78, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-28500974

RESUMEN

Respiratory syncytial virus (RSV) is the most common respiratory pathogen in infants and young children worldwide. Lower respiratory tract infection due to RSV is one of the most common causes of hospitalization for infants, especially those born premature or with chronic lung or heart disease. Furthermore, RSV infection is an important cause of morbidity in adults, particularly in the elderly and immunocompromised individuals. The acute phase of this infection is often followed by episodes of wheezing that recur for months or years and usually lead to a physician diagnosis of asthma. RSV was discovered more than 50 years ago, and despite extensive research to identify pharmacological therapies, the most effective management of this infection remains supportive care. The trial of a formalin-inactivated RSV vaccine in the 1960s resulted in priming the severe illness upon natural infection. Currently, Palivizumab is the only available option for RSV prophylaxis, and because of restricted clinical benefits and high costs, it has been limited to a group of high-risk infants. There are several ongoing trials in preclinical, Phase-I, Phase-II, or Phase-III clinical stages for RSV vaccine development based on various strategies. Here we review the existing available prophylactic options, the current stages of RSV vaccine clinical trials, different strategies, and major hurdles in the development of an effective RSV vaccine.


Asunto(s)
Profilaxis Pre-Exposición , Infecciones por Virus Sincitial Respiratorio/prevención & control , Vacunas contra Virus Sincitial Respiratorio , Virus Sincitial Respiratorio Humano/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Antivirales/uso terapéutico , Ensayos Clínicos como Asunto , Hospitalización , Humanos , Inmunogenicidad Vacunal , Lactante , Ratones , Palivizumab/uso terapéutico , Infecciones por Virus Sincitial Respiratorio/diagnóstico , Infecciones por Virus Sincitial Respiratorio/virología , Vacunas contra Virus Sincitial Respiratorio/inmunología , Virus Sincitial Respiratorio Humano/efectos de los fármacos , Virus Sincitial Respiratorio Humano/fisiología , Factores de Riesgo , Vacunas de Productos Inactivados/inmunología
17.
PLoS One ; 12(2): e0168786, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28178290

RESUMEN

Maternal viral infections can have pathological effects on the developing fetus which last long after birth. Recently, maternal-fetal transmission of respiratory syncytial virus (RSV) was shown to cause postnatal airway hyperreactivity (AHR) during primary early-life reinfection; however, the influence of prenatal exposure to RSV on offspring airway immunity and smooth muscle contractility during recurrent postnatal reinfections remains unknown. Therefore, we sought to determine whether maternal RSV infection impairs specific aspects of cell-mediated offspring immunity during early-life reinfections and the mechanisms leading to AHR. Red fluorescent protein-expressing recombinant RSV (rrRSV) was inoculated into pregnant rat dams at midterm, followed by primary and secondary postnatal rrRSV inoculations of their offspring at early-life time points. Pups and weanlings were tested for specific lower airway leukocyte populations by flow cytometry; serum cytokine/chemokine concentrations by multiplex ELISA and neurotrophins concentrations by standard ELISA; and ex vivo lower airway smooth muscle (ASM) contraction by physiological tissue bath. Pups born to RSV-infected mothers displayed elevated total CD3+ T cells largely lacking CD4+ and CD8+ surface expression after both primary and secondary postnatal rrRSV infection. Cytokine/chemokine analyses revealed reduced IFN-γ, IL-2, IL-12, IL-17A, IL-18, and TNF-α, as well as elevated nerve growth factor (NGF) expression. Prenatal exposure to RSV also increased ASM reactivity and contractility during early-life rrRSV infection compared to non-exposed controls. We conclude that maternal RSV infection can predispose offspring to postnatal lower airways dysfunction by altering immunity development, NGF signaling, and ASM contraction during early-life RSV reinfections.


Asunto(s)
Exposición Materna , Efectos Tardíos de la Exposición Prenatal , Infecciones por Virus Sincitial Respiratorio/inmunología , Infecciones por Virus Sincitial Respiratorio/fisiopatología , Virus Sincitiales Respiratorios/inmunología , Animales , Biomarcadores , Hiperreactividad Bronquial/inmunología , Hiperreactividad Bronquial/metabolismo , Hiperreactividad Bronquial/fisiopatología , Citocinas/metabolismo , Modelos Animales de Enfermedad , Femenino , Inmunofenotipificación , Contracción Muscular , Músculo Liso , Fenotipo , Embarazo , Ratas , Infecciones por Virus Sincitial Respiratorio/virología , Subgrupos de Linfocitos T/inmunología , Subgrupos de Linfocitos T/metabolismo
18.
Translation (Austin) ; 1(1): e24391, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-26824017

RESUMEN

The proapoptotic Bcl-2 family member PUMA is a critical regulator of apoptosis. We have previously shown that PUMA plays a pivotal role in the apoptosis associated with skeletal myoblast differentiation and that a MyoD-dependent mechanism is responsible for the increased expression of PUMA in these cells. Herein, we report that the increased expression of PUMA under these conditions involves regulation at the level of translation. Specifically, we have found that the increase in PUMA protein levels occurs under conditions of decreased total protein synthesis, eIF2-alpha phosphorylation and hypophosphorylation of eIF4E-BP, suggesting that PUMA translation is proceeding via an alternative initiation mechanism. Polyribosome analysis of PUMA mRNA further corroborated this suggestion. A combination of in vitro and ex vivo (cellular) approaches has provided evidence suggesting that PUMA mRNA 5'UTR harbors an Internal Ribosome Entry Site (IRES) element. Using mono- and bi-cistronic reporter constructs, we have delineated an mRNA fragment that allows for cap-independent translation in vitro and ex vivo (in skeletal myoblasts) in response to culture in differentiation media (DM), or in response to treatment with the DNA-damaging agent, etoposide. This mRNA fragment also supports translation in HeLa and 293T cells. Thus, our data has revealed a novel IRES-mediated regulation of PUMA expression in several cell types and in response to several stimuli. These findings contribute to our understanding and potential manipulation of any developmental or therapeutic scenario involving PUMA.

19.
Apoptosis ; 15(1): 71-82, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-19943111

RESUMEN

We have previously reported that the level of MyoD expression correlates with the level of apoptosis that occurs in a subpopulation of skeletal myoblasts induced to differentiate by serum withdrawal. Herein we document that MyoD expression contributes to the level of apoptosis in myoblasts and fibroblasts in response to a variety of apoptotic stimuli. Specifically, re-expression of MyoD in skeletal myoblasts rendered defective for both differentiation and apoptosis by the expression of oncogenic Ras restores their ability to undergo both differentiation and apoptosis in response to serum withdrawal. Further, using a fibroblast cell line expressing an estrogen receptor:MyoD fusion protein, we have determined that addition of estrogen sensitizes these fibroblasts to apoptosis induced by serum withdrawal, or by treatment with etoposide or thapsigargin. RNAi mediated silencing of MyoD in either 23A2 or C2C12 myoblasts renders these cells resistant to apoptosis induced by serum withdrawal, or by treatment with etoposide or thapsigargin. Finally, MyoD mediated regulation of the apoptotic response to these various stimuli, in both myoblasts and fibroblasts, correlates with the level of induction of the pro-apoptotic Bcl2 family member PUMA.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/genética , Apoptosis , Estrógenos/metabolismo , Expresión Génica , Proteína MioD/metabolismo , Mioblastos Esqueléticos/citología , Proteínas Supresoras de Tumor/genética , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Línea Celular , Etopósido/farmacología , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/metabolismo , Expresión Génica/efectos de los fármacos , Ratones , Proteína MioD/genética , Mioblastos Esqueléticos/efectos de los fármacos , Mioblastos Esqueléticos/metabolismo , Tapsigargina/farmacología , Proteínas Supresoras de Tumor/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...