Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 15 de 15
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Aging (Albany NY) ; 11(1): 209-229, 2019 01 13.
Artículo en Inglés | MEDLINE | ID: mdl-30636723

RESUMEN

Atp8b1 (ATPase, aminophospholipid transporter, class I, type 8B, member 1) is a cardiolipin transporter in the apical membrane of lung epithelial cells. While the role of Atp8b1 in pneumonia-induced acute lung injury (ALI) has been well studied, its potential role in oxidative stress-induced ALI is poorly understood. We herein show that Atp8b1G308V/G308V mice under hyperoxic conditions display exacerbated cell apoptosis at alveolar epithelium and aberrant proliferation of club cells at bronchiolar epithelium. This hyperoxia-induced ambivalent response in Atp8b1G308V/G308V lungs was followed by patchy distribution of non-uniform interstitial fibrosis at late recovery phase under normoxia. Since this club cell abnormality is commonly observed between Atp8b1G308V/G308V lungs under hyperoxic conditions and IPF lungs, we characterized this mouse fibrosis model focusing on club cells. Intriguingly, subcellular morphological analysis of IPF lungs, using transmission electron microscopy (TEM), revealed that metaplastic bronchiolar epithelial cells in fibrotic lesions and deformed type II alveolar epithelial cells (AECs) in alveoli with mild fibrosis, have common morphological features including cytoplasmic vacuolation and dysmorphic lamellar bodies. In conclusion, the combination of Atp8b1 mutation and hyperoxic insult serves as a novel platform to study unfocused role of club cells in IPF.


Asunto(s)
Adenosina Trifosfatasas/metabolismo , Oxígeno/toxicidad , Proteínas de Transferencia de Fosfolípidos/metabolismo , Fibrosis Pulmonar/etiología , Adenosina Trifosfatasas/genética , Animales , Muerte Celular , Proliferación Celular , Células Epiteliales/fisiología , Regulación de la Expresión Génica/efectos de los fármacos , Ratones , Mutación , Estrés Oxidativo , Proteínas de Transferencia de Fosfolípidos/genética , Alveolos Pulmonares/citología , Uteroglobina/genética , Uteroglobina/metabolismo
2.
PLoS One ; 11(1): e0147652, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26807721

RESUMEN

Apoptosis signal-regulating kinase 1 (ASK1), a member of the MAPK kinase kinase kinase (MAP3K) family, is activated by various stimuli, which include oxidative stress, endoplasmic reticulum (ER) stress, calcium influx, DNA damage-inducing agents and receptor-mediated signaling through tumor necrosis factor receptor (TNFR). Inspiration of a high concentration of oxygen is a palliative therapy which counteracts hypoxemia caused by acute lung injury (ALI)-induced pulmonary edema. However, animal experiments so far have shown that hyperoxia itself could exacerbate ALI through reactive oxygen species (ROS). Our previous data indicates that ASK1 plays a pivotal role in hyperoxia-induced acute lung injury (HALI). However, it is unclear whether or not deletion of ASK1 in vivo protects against HALI. In this study, we investigated whether ASK1 deletion would lead to attenuation of HALI. Our results show that ASK1 deletion in vivo significantly suppresses hyperoxia-induced elevation of inflammatory cytokines (i.e. IL-1ß and TNF-α), cell apoptosis in the lung, and recruitment of immune cells. In summary, the results from the study suggest that deletion of ASK1 in mice significantly inhibits hyperoxic lung injury.


Asunto(s)
Lesión Pulmonar Aguda/genética , Hiperoxia/complicaciones , Pulmón/metabolismo , MAP Quinasa Quinasa Quinasa 5/genética , Lesión Pulmonar Aguda/etiología , Lesión Pulmonar Aguda/patología , Animales , Apoptosis/genética , Epitelio/metabolismo , Epitelio/patología , Hiperoxia/genética , Hiperoxia/patología , Interleucina-1beta/metabolismo , Pulmón/patología , MAP Quinasa Quinasa Quinasa 5/metabolismo , Ratones , Ratones Noqueados , Factor de Necrosis Tumoral alfa/metabolismo
3.
Cell Immunol ; 297(1): 40-5, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-26123077

RESUMEN

Incomplete clearance of apoptotic cells and reactive oxygen species (ROS) release are known to trigger inflammasome activation causing severe inflammation in acute lung injury and various metabolic and autoimmune diseases. Moreover, it has been reported that apoptotic cell clearance and ROS-mediated apoptosis critically depend on mitochondrial uncoupling protein-2 (UCP2). However, the relationship between UCP2 and inflammasome activation has not been studied. This report investigates the role of UCP2 in the expression and activation of NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome in human macrophages. We found that UCP2 overexpression significantly enhanced the expression levels of NLRP3. The NLRP3 expression levels were significantly suppressed in THP1 cells treated with genipin, a UCP2 inhibitor, compared to controls. In addition, genipin altered adenosine triphosphate (ATP)- and hydrogen peroxide (H2O2)-mediated interleukin-1 beta (IL-1ß) secretion and significantly suppressed caspase-1 activity in inflammasome-activated human macrophages. Taken together, our results suggest that genipin modulates NLRP3 inflammasome activation and ATP- or H2O2-mediated IL-1ß release.


Asunto(s)
Proteínas Portadoras/antagonistas & inhibidores , Inflamasomas/efectos de los fármacos , Canales Iónicos/inmunología , Iridoides/farmacología , Proteínas Mitocondriales/inmunología , Apoptosis/inmunología , Proteínas Portadoras/biosíntesis , Proteínas Portadoras/metabolismo , Caspasa 1/inmunología , Células Cultivadas , Activación Enzimática/inmunología , Regulación de la Expresión Génica , Humanos , Inflamasomas/metabolismo , Inflamación/inmunología , Interleucina-1beta/inmunología , Canales Iónicos/antagonistas & inhibidores , Canales Iónicos/biosíntesis , Macrófagos/inmunología , Proteínas Mitocondriales/antagonistas & inhibidores , Proteínas Mitocondriales/biosíntesis , Proteína con Dominio Pirina 3 de la Familia NLR , Especies Reactivas de Oxígeno/inmunología , Proteína Desacopladora 2
4.
Cell Physiol Biochem ; 36(5): 2012-24, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-26202361

RESUMEN

BACKGROUND: Neuregulin (NRG)-1-human epidermal receptor (HER)-2 signaling pathway is a key regulator of IL-1ß-mediated pulmonary inflammation and epithelial permeability. The inflammasome is a newly discovered molecular platform required for caspase-1 activation and maturation of IL-1ß. However, the role of the inflammasome in NRG-1-HER2 signaling-mediated alveolar cell permeability is unknown. METHODS: The inflammasome was activated or inhibited in THP-1 cells; supernatants from these cells were added to A549 cells and human small airway epithelial cells (HSAEC). The protein expression of NRG-1 and phospho-HER2 (pHER2) were measured by Western blot analysis and epithelial permeability was measured using Lucifer yellow dye. RESULTS: Results reveal that alveolar permeability in A549 cells and HSAEC is increased when treated with supernatants of inflammasome-activated THP-1 cells. Alveolar permeability is significantly suppressed when treated with supernatant of inflammasome-inhibited THP-1 cells. Inflammasome-mediated permeability is decreased when A549 cells and HSAEC are pretreated with IL-1ß receptor antagonist (IL-1ßRA). In addition, HER2 kinase inhibitor AG825 or NRG-1 inhibitor TAPI inhibits inflammasome-mediated permeability in A549 cells and HSAEC demonstrating critical roles of IL-1ß, NRG-1, and HER2 in inflammasome-mediated alveolar permeability. CONCLUSION: These findings suggest that inflammasome-induced alveolar cell permeability is mediated by NRG-1/HER2 signaling through IL-1ß regulation.


Asunto(s)
Inflamasomas , Neurregulina-1/metabolismo , Línea Celular Tumoral , Genes erbB-2 , Humanos , Interleucina-1beta/metabolismo , Alveolos Pulmonares
5.
Free Radic Biol Med ; 84: 196-205, 2015 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-25850028

RESUMEN

Suppressor of cytokine signaling-1 (SOCS-1) is a member of the suppressor of cytokine signaling family of proteins and an inhibitor of interleukin-6 (IL-6) signaling. SOCS-1 has been shown to protect cells from cellular damage and apoptosis induced by tumor necrosis factor (TNF), lipopolysaccharide (LPS), and interferon gamma (IL-γ). However, it is not known whether increased SOCS-1 is protective during pulmonary oxidative stress. Therefore, we hypothesized that increased SOCS-1 in the lungs of mice would be protective in the setting of hyperoxic lung injury. We administered SOCS-1 adenovirus (Ad-SOCS-1) intratracheally into the lungs and exposed the mice to 100% O2. Mice infected with GFP adenovirus (Ad-GFP) were used as controls. Mice treated with Ad-SOCS-1 had enhanced survival in 100% oxygen compared to Ad-GFP-administered mice. After 3 days of hyperoxia, Ad-GFP mice were ill and tachypnic and died after 4 days. In contrast, all Ad-SOCS-1-treated mice survived for at least 6 days in hyperoxia and 80% survived beyond 7 days. Ad-SOCS-1 transfection protected mouse lungs from injury as indicated by lower lung wet/dry weight, alveolar-capillary protein leakage, reduced infiltration of inflammatory cells, and lower content of thiobarbituric acid-reactive substances in lung homogenate. Our results also indicated that Ad-SOCS-1 significantly inhibits hyperoxia-induced ASK-1 (apoptosis signal-regulating kinase 1) expression. Taken together, these findings show that increased expression of adenovirus-mediated SOCS-1 in the lungs of mice significantly protects against hyperoxic lung injury.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Hiperoxia/metabolismo , Pulmón/metabolismo , Proteínas Supresoras de la Señalización de Citocinas/genética , Lesión Pulmonar Aguda/prevención & control , Adenoviridae/genética , Animales , Femenino , Expresión Génica , Pulmón/patología , Masculino , Ratones Endogámicos C57BL , Proteína 1 Supresora de la Señalización de Citocinas , Proteínas Supresoras de la Señalización de Citocinas/metabolismo , Transducción Genética
6.
Am J Respir Cell Mol Biol ; 53(3): 422-35, 2015 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-25647402

RESUMEN

Acute lung injury (ALI), which presents as acute respiratory failure, is a major clinical problem that requires aggressive care, and patients who require prolonged oxygen exposure are at risk of developing this disease. Although molecular determinants of ALI have been reported, the molecules involved in disease catabasis associated with oxygen toxicity have not been well studied. It has been reported that lung mucosa is rich in omega-3 fatty acid dicosahexanoic acid (DHA), which has antiinflammatory properties. Aspirin-triggered resolvin D1 (AT-RvD1) is a potent proresolution metabolite of DHA that can curb the inflammatory effects in various acute injuries, yet the effect of AT-RvD1 on hyperoxic acute lung injury (HALI) or in the oxygen toxicity setting in general has not been investigated. The effects of AT-RvD1 on HALI were determined for the first time in 8- to 10-week-old C57BL/6 mice that were exposed to hyperoxia (≥95% O2) for 48 hours. Mice were given AT-RvD1 (100 ng) in saline or a saline vehicle for 24 hours in normoxic (≈21% O2) conditions after hyperoxia. Lung tissue and bronchoalveolar lavage (BAL) fluid were collected for analysis associated with proinflammatory signaling and lung inflammation. AT-RvD1 treatment resulted in reduced oxidative stress, increased glutathione production, and significantly decreased tissue inflammation. AT-RvD1 treatment also significantly reduced the lung wet/dry ratio, protein in BAL fluid, and decreased apoptotic and NF-κB signaling. These results show that AT-RvD1 curbs oxygen-induced lung edema, permeability, inflammation, and apoptosis and is thus an effective therapy for prolonged hyperoxia exposure in this murine model.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Antiinflamatorios no Esteroideos/farmacología , Aspirina/farmacología , Ácidos Docosahexaenoicos/fisiología , Hiperoxia/tratamiento farmacológico , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/metabolismo , Resistencia de las Vías Respiratorias , Animales , Apoptosis , Evaluación Preclínica de Medicamentos , Hiperoxia/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Ratones Endogámicos C57BL , Infiltración Neutrófila , Estrés Oxidativo
8.
Am J Physiol Cell Physiol ; 306(12): C1101-5, 2014 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-24717578

RESUMEN

A significant amount of research has been conducted to examine the pathologic processes and epigenetic mechanisms contributing to peripheral hypertension. However, few studies have been carried out to understand the vascular remodeling behind pulmonary hypertension (PH), including peripheral artery muscularization, medial hypertrophy and neointima formation in proximal arteries, and plexiform lesion formation. Similarly, research examining some of the epigenetic principles that may contribute to this vascular remodeling, such as DNA methylation and histone modification, is minimal. The understanding of these principles may be the key to developing new and more effective treatments for PH. The purpose of this review is to summarize epigenetic research conducted in the field of hypertension that could possibly be used to understand the epigenetics of PH. Possible future therapies that could be pursued using information from these studies include selective histone deacetylase inhibitors and targeted DNA methyltransferases. Both of these could potentially be used to silence proproliferative or antiapoptotic genes that lead to decreased smooth muscle cell proliferation. Epigenetics may provide a glimmer of hope for the eventual improved treatment of this highly morbid and debilitating disease.


Asunto(s)
Metilación de ADN/genética , Epigénesis Genética/genética , Histonas/metabolismo , Hipertensión Pulmonar/genética , Apoptosis/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Inhibidores de Histona Desacetilasas/uso terapéutico , Humanos , Hipertensión Pulmonar/tratamiento farmacológico , Hipertensión Pulmonar/patología , Especies Reactivas de Oxígeno/metabolismo
9.
Am J Physiol Cell Physiol ; 306(11): C999-C1007, 2014 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-24696144

RESUMEN

Hyperoxic acute lung injury (HALI) is characterized by inflammation and epithelial cell death. CLOCK genes are master regulators of circadian rhythm also implicated in inflammation and lung diseases. However, the relationship of CLOCK genes in hyperoxia-induced lung injury has not been studied. This study will determine if HALI alters CLOCK gene expression. To test this, wild-type and NALP3(-/-) mice were exposed to room air or hyperoxia for 24, 48, or 72 h. In addition, mice were exposed to different concentrations of hyperoxia (50, 75, or 100% O2) or room air for 72 h. The mRNA and protein levels of lung CLOCK genes, based on quantitative PCR and Western blot analysis, respectively, and their target genes are significantly elevated in mice exposed to hyperoxia compared with controls. Alterations in CLOCK genes are associated with increased inflammatory markers in bronchoalveolar lavage fluid of hyperoxic mice compared with controls. Histological examination of mice lungs exposed to hyperoxia show increased inflammation and alveolar congestion compared with controls. Our results indicate sequential increase in CLOCK gene expression in lungs of mice exposed to hyperoxia compared with controls. Additionally, data suggest a dose-dependent increase in CLOCK gene expression with increased oxygen concentrations. To validate if the expression changes related to CLOCK genes are indeed associated with inflammation, NALP3(-/-) was introduced to analyze loss of function in inflammation. Western blot analysis showed significant CLOCK gene downregulation in NALP3(-/-) mice compared with wild-type controls. Together, our results demonstrate that hyperoxia-mediated lung inflammation is associated with alterations in CLOCK gene expression.


Asunto(s)
Proteínas CLOCK/biosíntesis , Regulación de la Expresión Génica , Hiperoxia/metabolismo , Lesión Pulmonar/metabolismo , Animales , Proteínas CLOCK/genética , Hiperoxia/genética , Hiperoxia/patología , Lesión Pulmonar/genética , Lesión Pulmonar/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , ARN Mensajero/biosíntesis , ARN Mensajero/genética
11.
Biochem Biophys Res Commun ; 439(3): 407-12, 2013 Sep 27.
Artículo en Inglés | MEDLINE | ID: mdl-23988448

RESUMEN

Inflammasomes are multimeric protein complexes involved in the processing of IL-1ß through Caspase-1 cleavage. NLRP3 is the most widely studied inflammasome, which has been shown to respond to a large number of both endogenous and exogenous stimuli. Although studies have begun to define basic pathways for the activation of inflammasome and have been instrumental in identifying therapeutics for inflammasome related disorders; understanding the inflammasome activation at the molecular level is still incomplete. Recent functional studies indicate that microRNAs (miRs) regulate molecular pathways and can lead to diseased states when hampered or overexpressed. Mechanisms involving the miRNA regulatory network in the activation of inflammasome and IL-1ß processing is yet unknown. This report investigates the involvement of miR-133a-1 in the activation of inflammasome (NLRP3) and IL-1ß production. miR-133a-1 is known to target the mitochondrial uncoupling protein 2 (UCP2). The role of UCP2 in inflammasome activation has remained elusive. To understand the role of miR-133a-1 in regulating inflammasome activation, we either overexpressed or suppressed miR-133a-1 in differentiated THP1 cells that express the NLRP3 inflammasome. Levels of Caspase-1 and IL-1ß were analyzed by Western blot analysis. For the first time, we showed that overexpression of miR-133a-1 increases Caspase-1 p10 and IL-1ß p17 cleavage, concurrently suppressing mitochondrial uncoupling protein 2 (UCP2). Surprisingly, our results demonstrated that miR-133A-1 controls inflammasome activation without affecting the basal expression of the individual inflammasome components NLRP3 and ASC or its immediate downstream targets proIL-1ß and pro-Caspase-1. To confirm the involvement of UCP2 in the regulation of inflammasome activation, Caspase-1 p10 and IL-1ß p17 cleavage in UCP2 of overexpressed and silenced THP1 cells were studied. Suppression of UCP2 by siRNA enhanced the inflammasome activity stimulated by H2O2 and, conversely, overexpression of UCP2 decreased the inflammasome activation. Collectively, these studies suggest that miR-133a-1 suppresses inflammasome activation via the suppression of UCP2.


Asunto(s)
Inflamasomas/inmunología , Interleucina-1beta/inmunología , Canales Iónicos/inmunología , MicroARNs/inmunología , Proteínas Mitocondriales/inmunología , Animales , Proteínas Reguladoras de la Apoptosis , Proteínas Adaptadoras de Señalización CARD , Proteínas Portadoras/genética , Caspasa 1/inmunología , Técnicas de Cultivo de Célula , Línea Celular Tumoral , Proteínas del Citoesqueleto/genética , Regulación hacia Abajo , Silenciador del Gen , Humanos , Canales Iónicos/genética , Ratones , Ratones Noqueados , Proteínas Mitocondriales/genética , Proteína con Dominio Pirina 3 de la Familia NLR , Proteína Desacopladora 2 , Regulación hacia Arriba
12.
Am J Physiol Cell Physiol ; 305(2): C182-9, 2013 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-23636457

RESUMEN

Inspiration of a high concentration of oxygen, a therapy for acute lung injury (ALI), could unexpectedly lead to reactive oxygen species (ROS) production and hyperoxia-induced acute lung injury (HALI). Nucleotide-binding domain and leucine-rich repeat PYD-containing protein 3 (NLRP3) senses the ROS, triggering inflammasome activation and interleukin-1ß (IL-1ß) production and secretion. However, the role of NLRP3 inflammasome in HALI is unclear. The main aim of this study is to determine the effect of NLRP3 gene deletion on inflammatory response and lung epithelial cell death. Wild-type (WT) and NLRP3(-/-) mice were exposed to 100% O2 for 48-72 h. Bronchoalveolar lavage fluid and lung tissues were examined for proinflammatory cytokine production and lung inflammation. Hyperoxia-induced lung pathological score was suppressed in NLRP3(-/-) mice compared with WT mice. Hyperoxia-induced recruitment of inflammatory cells and elevation of IL-1ß, TNFα, macrophage inflammatory protein-2, and monocyte chemoattractant protein-1 were attenuated in NLRP3(-/-) mice. NLRP3 deletion decreased lung epithelial cell death and caspase-3 levels and a suppressed NF-κB levels compared with WT controls. Taken together, this research demonstrates for the first time that NLRP3-deficient mice have suppressed inflammatory response and blunted lung epithelial cell apoptosis to HALI.


Asunto(s)
Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/prevención & control , Proteínas Portadoras/metabolismo , Hiperoxia/complicaciones , Oxígeno/efectos adversos , Lesión Pulmonar Aguda/genética , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Proteínas Portadoras/genética , Quimiocina CXCL2/genética , Quimiocina CXCL2/metabolismo , Fragmentación del ADN , Regulación de la Expresión Génica/efectos de los fármacos , Regulación de la Expresión Génica/fisiología , Predisposición Genética a la Enfermedad , Hiperoxia/metabolismo , Inflamasomas/efectos adversos , Inflamasomas/metabolismo , Interleucina-1beta/análisis , Interleucina-6/análisis , Pulmón/metabolismo , Pulmón/patología , Ratones , Ratones Noqueados , Proteína con Dominio Pirina 3 de la Familia NLR , Oxígeno/administración & dosificación , Oxígeno/uso terapéutico , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
13.
PLoS One ; 7(10): e46808, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-23071643

RESUMEN

Pulmonary Arterial Hypertension (PAH) is a progressive devastating disease characterized by excessive proliferation of the Pulmonary Arterial Smooth Muscle Cells (PASMCs). Studies suggest that PAH and cancers share an apoptosis-resistant state featuring excessive cell proliferation. MicroRNA-206 (miR-206) is known to regulate proliferation and is implicated in various types of cancers. However, the role of miR-206 in PAH has not been studied. In this study, it is hypothesized that miR-206 could play a role in the proliferation of PASMCs. In the present study, the expression patterns of miR-206 were investigated in normal and hypertensive mouse PASMCs. The effects of miR-206 in modulating cell proliferation, apoptosis and smooth muscle cell markers in human pulmonary artery smooth muscle cells (hPASMCs) were investigated in vitro. miR-206 expression in mouse PASMCs was correlated with an increase in right ventricular systolic pressure. Reduction of miR-206 levels in hPASMCs causes increased proliferation and reduced apoptosis and these effects were reversed by the overexpression of miR-206. miR-206 over expression also increased the levels of smooth muscle cell differentiation markers α-smooth muscle actin and calponin implicating its importance in the differentiation of SMCs. miR-206 overexpression down regulated Notch-3 expression, which is key a factor in PAH development. These results suggest that miR-206 is a potential regulator of proliferation, apoptosis and differentiation of PASMCs, and that it could be used as a novel treatment strategy in PAH.


Asunto(s)
Diferenciación Celular/genética , Proliferación Celular , MicroARNs/genética , Miocitos del Músculo Liso/metabolismo , Animales , Apoptosis/genética , Western Blotting , Movimiento Celular/genética , Células Cultivadas , Regulación hacia Abajo , Hipertensión Pulmonar Primaria Familiar , Femenino , Perfilación de la Expresión Génica , Humanos , Hipertensión Pulmonar/genética , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/fisiopatología , Hipoxia , Masculino , Ratones , Ratones Endogámicos C57BL , Miocitos del Músculo Liso/citología , Arteria Pulmonar/citología , Receptor Notch3 , Receptores Notch/genética , Receptores Notch/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa
14.
J Cell Physiol ; 227(9): 3310-6, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22169929

RESUMEN

The hallmark of acute lung injury (ALI) is the influx of proinflammatory cytokines into lung tissue and alveolar permeability that ultimately leads to pulmonary edema. However, the mechanisms involved in inflammatory cytokine production and alveolar permeability are unclear. Recent studies suggest that excessive production of ceramide has clinical relevance as a mediator of pulmonary edema and ALI. Our earlier studies indicate that the activation of inflammasome promotes the processing and secretion of proinflammatory cytokines and causes alveolar permeability in ALI. However, the role of ceramide in inflammasome activation and the underlying mechanism in relation to alveolar permeability is not known. We hypothesized that ceramide activates the inflammasome and causes inflammatory cytokine production and alveolar epithelial permeability. To test this hypothesis, we analyzed the lung ceramide levels during hyperoxic ALI in mice. The effect of ceramide on activation of inflammasome and production of inflammatory cytokine was assessed in primary mouse alveolar macrophages and THP-1 cells. Alveolar transepithelial permeability was determined in alveolar epithelial type-II cells (AT-II) and THP-1 co-cultures. Our results reveal that ceramide causes inflammasome activation, induction of caspase-1, IL-1ß cleavage, and release of proinflammatory cytokines. In addition, ceramide further induces alveolar epithelial permeability. Short-hairpin RNA silencing of inflammasome components abrogated ceramide-induced secretion of proinflammatory cytokines in vitro. Inflammasome silencing abolishes ceramide-induced alveolar epithelial permeability in AT-II. Collectively, our results demonstrate for the first time that ceramide-induced secretion of proinflammatory cytokines and alveolar epithelial permeability occurs though inflammasome activation.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Proteínas Portadoras/antagonistas & inhibidores , Ceramidas/administración & dosificación , Citocinas/genética , Inflamación/metabolismo , Macrófagos Alveolares/efectos de los fármacos , Lesión Pulmonar Aguda/inducido químicamente , Animales , Células Cultivadas , Citocinas/metabolismo , Relación Dosis-Respuesta a Droga , Células Epiteliales/efectos de los fármacos , Regulación de la Expresión Génica/efectos de los fármacos , Hiperoxia/inducido químicamente , Inflamación/genética , Macrófagos Alveolares/fisiología , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR , Oxígeno/administración & dosificación , Permeabilidad/efectos de los fármacos , ARN Interferente Pequeño/genética , Migración Transendotelial y Transepitelial/efectos de los fármacos , Migración Transendotelial y Transepitelial/fisiología
15.
J Bacteriol ; 193(20): 5784-92, 2011 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-21840976

RESUMEN

Antiterminator RNA directly modifies the transcription elongation complex so that it terminates less efficiently at intrinsic and factor-dependent terminators. These unusual RNAs were first discovered in bacteriophage HK022, where the nascent transcripts of the phage put sites promote full expression of phage genes during lytic infection. The activity of antiterminator RNA depends on specific structural elements that form as the transcript exits RNA polymerase. To further our understanding of the critical sequence features that permit RNA to serve as a transcriptional antiterminator, we have identified eight antiterminator RNA sequences in bacteriophages or prophages. There is strong sequence conservation among most of the put sequences, but sequence divergence is tolerated if critical structural elements are preserved. The most diverged antiterminator RNA is found in bacteriophage HK639. The HK639 putL transcript is an efficient antiterminator, and it has a novel structural feature that is critical for its activity. HK639 also displays a unique pattern of sensitivity to amino acid substitutions in the ß' subunit zinc binding domain of RNA polymerase, adding to existing evidence that this domain interacts specifically with antiterminator RNA.


Asunto(s)
Bacteriófagos/genética , Regulación Viral de la Expresión Génica , ARN Viral/química , ARN Viral/genética , Regiones Terminadoras Genéticas , Bacteriófagos/química , Bacteriófagos/enzimología , Bacteriófagos/metabolismo , Secuencia de Bases , Codón de Terminación , ARN Polimerasas Dirigidas por ADN/genética , Escherichia coli/virología , Datos de Secuencia Molecular , Conformación de Ácido Nucleico , ARN Viral/metabolismo , Transcripción Genética , Proteínas Virales/genética , Proteínas Virales/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA