Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 44
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Am J Physiol Regul Integr Comp Physiol ; 324(1): R109-R119, 2023 01 01.
Artículo en Inglés | MEDLINE | ID: mdl-36409022

RESUMEN

The fundamental body functions that determine maximal O2 uptake (V̇o2max) have not been studied in Aqp5-/- mice (aquaporin 5, AQP5). We measured V̇o2max to globally assess these functions and then investigated why it was found altered in Aqp5-/- mice. V̇o2max was measured by the Helox technique, which elicits maximal metabolic rate by intense cold exposure of the animals. We found V̇o2max reduced in Aqp5-/- mice by 20%-30% compared with wild-type (WT) mice. As AQP5 has been implicated to act as a membrane channel for respiratory gases, we studied whether this is caused by the known lack of AQP5 in the alveolar epithelial membranes of Aqp5-/- mice. Lung function parameters as well as arterial O2 saturation were normal and identical between Aqp5-/- and WT mice, indicating that AQP5 does not contribute to pulmonary O2 exchange. The cause for the decreased V̇o2max thus might be found in decreased O2 consumption of an intensely O2-consuming peripheral organ such as activated brown adipose tissue (BAT). We found indeed that absence of AQP5 greatly reduces the amount of interscapular BAT formed in response to 4 wk of cold exposure, from 63% in WT to 25% in Aqp5-/- animals. We conclude that lack of AQP5 does not affect pulmonary O2 exchange, but greatly inhibits transformation of white to brown adipose tissue. As under cold exposure, BAT is a major source of the animals' heat production, reduction of BAT likely causes the decrease in V̇o2max under this condition.


Asunto(s)
Tejido Adiposo Pardo , Intercambio Gaseoso Pulmonar , Animales , Ratones , Tejido Adiposo Pardo/metabolismo , Termogénesis/fisiología , Pulmón , Consumo de Oxígeno , Frío
2.
J Immunol ; 192(9): 4453-4464, 2014 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-24688024

RESUMEN

Overwhelming lung inflammation frequently occurs following exposure to both direct infectious and noninfectious agents and is a leading cause of mortality worldwide. In that context, immunomodulatory strategies may be used to limit severity of impending organ damage. We sought to determine whether priming the lung by activating the immune system, or immunological priming, could accelerate resolution of severe lung inflammation. We assessed the importance of alveolar macrophages, regulatory T cells, and their potential interaction during immunological priming. We demonstrate that oropharyngeal delivery of low-dose LPS can immunologically prime the lung to augment alveolar macrophage production of IL-10 and enhance resolution of lung inflammation induced by a lethal dose of LPS or by Pseudomonas bacterial pneumonia. IL-10-deficient mice did not achieve priming and were unable to accelerate lung injury resolution. Depletion of lung macrophages or regulatory T cells during the priming response completely abrogated the positive effect of immunological priming on resolution of lung inflammation and significantly reduced alveolar macrophage IL-10 production. Finally, we demonstrated that oropharyngeal delivery of synthetic CpG-oligonucleotides elicited minimal lung inflammation compared with low-dose LPS but nonetheless primed the lung to accelerate resolution of lung injury following subsequent lethal LPS exposure. Immunological priming is a viable immunomodulatory strategy used to enhance resolution in an experimental acute lung injury model with the potential for therapeutic benefit against a wide array of injurious exposures.


Asunto(s)
Macrófagos Alveolares/inmunología , Neumonía/inmunología , Linfocitos T Reguladores/inmunología , Vacunación/métodos , Animales , Citocinas/biosíntesis , Citometría de Flujo , Interleucina-10/inmunología , Lipopolisacáridos/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Neumonía/prevención & control
3.
Am J Respir Cell Mol Biol ; 52(5): 641-52, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25295995

RESUMEN

Acute respiratory distress syndrome (ARDS) is a common and often fatal inflammatory lung condition without effective targeted therapies. Regulatory T cells (Tregs) resolve lung inflammation, but mechanisms that enhance Tregs to promote resolution of established damage remain unknown. DNA demethylation at the forkhead box protein 3 (Foxp3) locus and other key Treg loci typify the Treg lineage. To test how dynamic DNA demethylation affects lung injury resolution, we administered the DNA methyltransferase inhibitor 5-aza-2'-deoxycytidine (DAC) to wild-type (WT) mice beginning 24 hours after intratracheal LPS-induced lung injury. Mice that received DAC exhibited accelerated resolution of their injury. Lung CD4(+)CD25(hi)Foxp3(+) Tregs from DAC-treated WT mice increased in number and displayed enhanced Foxp3 expression, activation state, suppressive phenotype, and proliferative capacity. Lymphocyte-deficient recombinase activating gene-1-null mice and Treg-depleted (diphtheria toxin-treated Foxp3(DTR)) mice did not resolve their injury in response to DAC. Adoptive transfer of 2 × 10(5) DAC-treated, but not vehicle-treated, exogenous Tregs rescued Treg-deficient mice from ongoing lung inflammation. In addition, in WT mice with influenza-induced lung inflammation, DAC rescue treatment facilitated recovery of their injury and promoted an increase in lung Treg number. Thus, DNA methyltransferase inhibition, at least in part, augments Treg number and function to accelerate repair of experimental lung injury. Epigenetic pathways represent novel manipulable targets for the treatment of ARDS.


Asunto(s)
Lesión Pulmonar Aguda/tratamiento farmacológico , Azacitidina/análogos & derivados , Metilasas de Modificación del ADN/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Pulmón/efectos de los fármacos , Neumonía/tratamiento farmacológico , Linfocitos T Reguladores/efectos de los fármacos , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/enzimología , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/virología , Traslado Adoptivo , Animales , Azacitidina/farmacología , Células Cultivadas , Quimiotaxis de Leucocito , Metilasas de Modificación del ADN/metabolismo , Decitabina , Modelos Animales de Enfermedad , Factores de Transcripción Forkhead/genética , Factores de Transcripción Forkhead/metabolismo , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/metabolismo , Subtipo H1N1 del Virus de la Influenza A , Lipopolisacáridos , Pulmón/enzimología , Pulmón/inmunología , Pulmón/virología , Ratones Endogámicos C57BL , Ratones Noqueados , Fenotipo , Neumonía/inducido químicamente , Neumonía/enzimología , Neumonía/inmunología , Neumonía/virología , Linfocitos T Reguladores/enzimología , Linfocitos T Reguladores/inmunología , Linfocitos T Reguladores/trasplante , Linfocitos T Reguladores/virología , Factores de Tiempo
4.
Am J Physiol Lung Cell Mol Physiol ; 306(8): L709-25, 2014 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-24508730

RESUMEN

Acute respiratory distress syndrome (ARDS) is a devastating disease with distinct pathological stages. Fundamental to ARDS is the acute onset of lung inflammation as a part of the body's immune response to a variety of local and systemic stimuli. In patients surviving the inflammatory and subsequent fibroproliferative stages, transition from injury to resolution and recovery is an active process dependent on a series of highly coordinated events regulated by the immune system. Experimental animal models of acute lung injury (ALI) reproduce key components of the injury and resolution phases of human ARDS and provide a methodology to explore mechanisms and potential new therapies. Macrophages are essential to innate immunity and host defense, playing a featured role in the lung and alveolar space. Key aspects of their biological response, including differentiation, phenotype, function, and cellular interactions, are determined in large part by the presence, severity, and chronicity of local inflammation. Studies support the importance of macrophages to initiate and maintain the inflammatory response, as well as a determinant of resolution of lung inflammation and repair. We will discuss distinct roles for lung macrophages during early inflammatory and late resolution phases of ARDS using experimental animal models. In addition, each section will highlight human studies that relate to the diverse role of macrophages in initiation and resolution of ALI and ARDS.


Asunto(s)
Macrófagos Alveolares/fisiología , Neumonía/inmunología , Lesión Pulmonar Aguda/inmunología , Animales , Modelos Animales de Enfermedad , Humanos , Neumonía/fisiopatología
5.
J Immunol ; 189(5): 2234-45, 2012 Sep 01.
Artículo en Inglés | MEDLINE | ID: mdl-22844117

RESUMEN

Although early events in the pathogenesis of acute lung injury (ALI) have been defined, little is known about the mechanisms mediating resolution. To search for determinants of resolution, we exposed wild type (WT) mice to intratracheal LPS and assessed the response at intervals to day 10, when injury had resolved. Inducible NO synthase (iNOS) was significantly upregulated in the lung at day 4 after LPS. When iNOS-/- mice were exposed to intratracheal LPS, early lung injury was attenuated; however, recovery was markedly impaired compared with WT mice. iNOS-/- mice had increased mortality and sustained increases in markers of lung injury. Adoptive transfer of WT (iNOS+/+) bone marrow-derived monocytes or direct adenoviral gene delivery of iNOS into injured iNOS-/- mice restored resolution of ALI. Irradiated bone marrow chimeras confirmed the protective effects of myeloid-derived iNOS but not of epithelial iNOS. Alveolar macrophages exhibited sustained expression of cosignaling molecule CD86 in iNOS-/- mice compared with WT mice. Ab-mediated blockade of CD86 in iNOS-/- mice improved survival and enhanced resolution of lung inflammation. Our findings show that monocyte-derived iNOS plays a pivotal role in mediating resolution of ALI by modulating lung immune responses, thus facilitating clearance of alveolar inflammation and promoting lung repair.


Asunto(s)
Lesión Pulmonar Aguda/enzimología , Lesión Pulmonar Aguda/terapia , Monocitos/enzimología , Monocitos/inmunología , Óxido Nítrico Sintasa de Tipo II/uso terapéutico , Lesión Pulmonar Aguda/inmunología , Animales , Antígeno B7-2/biosíntesis , Línea Celular , Línea Celular Transformada , Modelos Animales de Enfermedad , Mediadores de Inflamación/metabolismo , Mediadores de Inflamación/uso terapéutico , Macrófagos Peritoneales/enzimología , Macrófagos Peritoneales/inmunología , Masculino , Ratones , Ratones Congénicos , Ratones Endogámicos C57BL , Ratones Noqueados , Monocitos/patología , Óxido Nítrico Sintasa de Tipo II/deficiencia
6.
Am J Respir Cell Mol Biol ; 48(1): 35-43, 2013 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-23002097

RESUMEN

Acute lung injury (ALI) causes significant morbidity and mortality. Fibroproliferation in ALI results in worse outcomes, but the mechanisms governing fibroproliferation remain poorly understood. Regulatory T cells (Tregs) are important in lung injury resolution. Their role in fibroproliferation is unknown. We sought to identify the role of Tregs in ALI fibroproliferation, using a murine model of lung injury. Wild-type (WT) and lymphocyte-deficient Rag-1(-/-) mice received intratracheal LPS. Fibroproliferation was characterized by histology and the measurement of lung collagen. Lung fibrocytes were measured by flow cytometry. To dissect the role of Tregs in fibroproliferation, Rag-1(-/-) mice received CD4(+)CD25(+) (Tregs) or CD4(+)CD25(-) Tcells (non-Tregs) at the time of LPS injury. To define the role of the chemokine (C-X-C motif) ligand 12 (CXCL12)-CXCR4 pathway in ALI fibroproliferation, Rag-1(-/-) mice were treated with the CXCR4 antagonist AMD3100 to block fibrocyte recruitment. WT and Rag-1(-/-) mice demonstrated significant collagen deposition on Day 3 after LPS. WT mice exhibited the clearance of collagen, but Rag-1(-/-) mice developed persistent fibrosis. This fibrosis was mediated by the sustained epithelial expression of CXCL12 (or stromal cell-derived factor 1 [SDF-1]) that led to increased fibrocyte recruitment. The adoptive transfer of Tregs resolved fibroproliferation by decreasing CXCL12 expression and subsequent fibrocyte recruitment. Blockade of the CXCL12-CXCR4 axis with AMD3100 also decreased lung fibrocytes and fibroproliferation. These results indicate a central role for Tregs in the resolution of ALI fibroproliferation by reducing fibrocyte recruitment along the CXCL12-CXCR4 axis. A dissection of the role of Tregs in ALI fibroproliferation may inform the design of new therapeutic tools for patients with ALI.


Asunto(s)
Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/patología , Linfocitos T Reguladores/inmunología , Traslado Adoptivo , Animales , Bencilaminas , Proliferación Celular/efectos de los fármacos , Quimiocina CXCL12/metabolismo , Ciclamas , Fibroblastos/inmunología , Fibroblastos/patología , Compuestos Heterocíclicos/farmacología , Proteínas de Homeodominio/genética , Proteínas de Homeodominio/inmunología , Lipopolisacáridos/toxicidad , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Miofibroblastos/inmunología , Miofibroblastos/patología , Receptores CXCR4/antagonistas & inhibidores , Receptores CXCR4/metabolismo , Transducción de Señal/inmunología , Linfocitos T Reguladores/patología
7.
Am J Respir Cell Mol Biol ; 48(5): 635-46, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23349051

RESUMEN

Acute respiratory distress syndrome (ARDS) causes significant morbidity and mortality. Exacerbating factors increasing the risk of ARDS remain unknown. Supplemental oxygen is often necessary in both mild and severe lung disease. The potential effects of supplemental oxygen may include augmentation of lung inflammation by inhibiting anti-inflammatory pathways in alveolar macrophages. We sought to determine oxygen-derived effects on the anti-inflammatory A2A adenosinergic (ADORA2A) receptor in macrophages, and the role of the ADORA2A receptor in lung injury. Wild-type (WT) and ADORA2A(-/-) mice received intratracheal lipopolysaccharide (IT LPS), followed 12 hours later by continuous exposure to 21% oxygen (control mice) or 60% oxygen for 1 to 3 days. We measured the phenotypic endpoints of lung injury and the alveolar macrophage inflammatory state. We tested an ADORA2A-specific agonist, CGS-21680 hydrochloride, in LPS plus oxygen-exposed WT and ADORA2A(-/-) mice. We determined the specific effects of myeloid ADORA2A, using chimera experiments. Compared with WT mice, ADORA2A(-/-) mice exposed to IT LPS and 60% oxygen demonstrated significantly more histologic lung injury, alveolar neutrophils, and protein. Macrophages from ADORA2A(-/-) mice exposed to LPS plus oxygen expressed higher concentrations of proinflammatory cytokines and cosignaling molecules. CGS-21680 prevented the oxygen-induced augmentation of lung injury after LPS only in WT mice. Chimera experiments demonstrated that the transfer of WT but not ADORA2A(-/-) bone marrow cells into irradiated ADORA2A(-/-) mice reduced lung injury after LPS plus oxygen, demonstrating myeloid ADORA2A protection. ADORA2A is protective against lung injury after LPS and oxygen. Oxygen after LPS increases macrophage activation to augment lung injury by inhibiting the ADORA2A pathway.


Asunto(s)
Lesión Pulmonar Aguda/metabolismo , Macrófagos Alveolares/metabolismo , Oxígeno/toxicidad , Receptor de Adenosina A2A/metabolismo , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/inmunología , Adenosina/análogos & derivados , Adenosina/farmacología , Agonistas del Receptor de Adenosina A2/farmacología , Animales , Líquido del Lavado Bronquioalveolar , Células Cultivadas , Quimiocinas/metabolismo , Técnicas de Inactivación de Genes , Mediadores de Inflamación/fisiología , Lipopolisacáridos/farmacología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Pulmón/patología , Macrófagos Alveolares/efectos de los fármacos , Macrófagos Alveolares/inmunología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Terapia por Inhalación de Oxígeno , Fenetilaminas/farmacología , Receptor de Adenosina A2A/genética
9.
Am J Respir Crit Care Med ; 185(8): 825-34, 2012 Apr 15.
Artículo en Inglés | MEDLINE | ID: mdl-22312013

RESUMEN

RATIONALE: Acute lung injury (ALI) is a debilitating condition associated with severe skeletal muscle weakness that persists in humans long after lung injury has resolved. The molecular mechanisms underlying this condition are unknown. OBJECTIVES: To identify the muscle-specific molecular mechanisms responsible for muscle wasting in a mouse model of ALI. METHODS: Changes in skeletal muscle weight, fiber size, in vivo contractile performance, and expression of mRNAs and proteins encoding muscle atrophy-associated genes for muscle ring finger-1 (MuRF1) and atrogin1 were measured. Genetic inactivation of MuRF1 or electroporation-mediated transduction of miRNA-based short hairpin RNAs targeting either MuRF1 or atrogin1 were used to identify their role in ALI-associated skeletal muscle wasting. MEASUREMENTS AND MAIN RESULTS: Mice with ALI developed profound muscle atrophy and preferential loss of muscle contractile proteins associated with reduced muscle function in vivo. Although mRNA expression of the muscle-specific ubiquitin ligases, MuRF1 and atrogin1, was increased in ALI mice, only MuRF1 protein levels were up-regulated. Consistent with these changes, suppression of MuRF1 by genetic or biochemical approaches prevented muscle fiber atrophy, whereas suppression of atrogin1 expression was without effect. Despite resolution of lung injury and down-regulation of MuRF1 and atrogin1, force generation in ALI mice remained suppressed. CONCLUSIONS: These data show that MuRF1 is responsible for mediating muscle atrophy that occurs during the period of active lung injury in ALI mice and that, as in humans, skeletal muscle dysfunction persists despite resolution of lung injury.


Asunto(s)
Lesión Pulmonar Aguda/genética , Lesión Pulmonar Aguda/patología , Proteínas Musculares/genética , Atrofia Muscular/genética , Ubiquitina-Proteína Ligasas/genética , Animales , Western Blotting , Líquido del Lavado Bronquioalveolar/química , Modelos Animales de Enfermedad , Regulación hacia Abajo , Regulación de la Expresión Génica , Inmunohistoquímica , Masculino , Ratones , Ratones Endogámicos C57BL , Análisis Multivariante , Fuerza Muscular/fisiología , Músculo Esquelético/patología , Músculo Esquelético/fisiopatología , Atrofia Muscular/patología , Dominios RING Finger/genética , Distribución Aleatoria , Sensibilidad y Especificidad , Proteínas de Motivos Tripartitos
10.
Am J Physiol Lung Cell Mol Physiol ; 303(4): L343-53, 2012 Aug 15.
Artículo en Inglés | MEDLINE | ID: mdl-22683574

RESUMEN

Pulmonary arterial smooth muscle cell (PASMC) migration is a key component of the vascular remodeling that occurs during the development of hypoxic pulmonary hypertension, although the mechanisms governing this phenomenon remain poorly understood. Aquaporin-1 (AQP1), an integral membrane water channel protein, has recently been shown to aid in migration of endothelial cells. Since AQP1 is expressed in certain types of vascular smooth muscle, we hypothesized that AQP1 would be expressed in PASMCs and would be required for migration in response to hypoxia. Using PCR and immunoblot techniques, we determined the expression of AQPs in pulmonary vascular smooth muscle and the effect of hypoxia on AQP levels, and we examined the role of AQP1 in hypoxia-induced migration in rat PASMCs using Transwell filter assays. Moreover, since the cytoplasmic tail of AQP1 contains a putative calcium binding site and an increase in intracellular calcium concentration ([Ca(2+)](i)) is a hallmark of hypoxic exposure in PASMCs, we also determined whether the responses were Ca(2+) dependent. Results were compared with those obtained in aortic smooth muscle cells (AoSMCs). We found that although AQP1 was abundant in both PASMCs and AoSMCs, hypoxia selectively increased AQP1 protein levels, [Ca(2+)](i), and migration in PASMCs. Blockade of Ca(2+) entry through voltage-dependent Ca(2+) or nonselective cation channels prevented the hypoxia-induced increase in PASMC [Ca(2+)](i), AQP1 levels, and migration. Silencing AQP1 via siRNA also prevented hypoxia-induced migration of PASMCs. Our results suggest that hypoxia induces a PASMC-specific increase in [Ca(2+)](i) that results in increased AQP1 protein levels and cell migration.


Asunto(s)
Acuaporina 1/genética , Calcio/metabolismo , Movimiento Celular , Miocitos del Músculo Liso/metabolismo , Miocitos del Músculo Liso/patología , Arteria Pulmonar/patología , Regulación hacia Arriba/genética , Animales , Aorta/patología , Acuaporina 1/metabolismo , Hipoxia de la Célula , Proliferación Celular , Espacio Intracelular/metabolismo , Masculino , Músculo Liso Vascular/patología , Ratas , Ratas Wistar
11.
Am J Respir Cell Mol Biol ; 45(1): 120-6, 2011 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-20870893

RESUMEN

Epithelial cells have the ability to regulate paracellular permeability dynamically in response to extracellular stimuli. With every respiratory effort, airway epithelial cells are exposed to both physiologic as well as pathologic stimuli, and regulation of the epithelial barrier in response to these stimuli is crucial to respiratory function. We report that increased membrane septin-2 localization mediates decreases in paracellular permeability by altering cortical actin arrangement in human airway epithelial cells. This phenomenon occurs in response to both physiologic levels of shear stress and a pathologic stimulus, particular matter exposure. The resulting changes in barrier function in response to septin-2 redistribution have a significant impact on the ability of the apical ligand, epidermal growth factor, to interact with its receptor, epidermal growth factor receptor, which is segregated to the basolateral side in airway epithelial cells. This suggests that the dynamic regulation of the epithelial barrier function is essential in regulating signaling responses to extracellular stimuli. These findings indicate that septin-2 plays a fundamental role in regulating barrier function by altering cortical actin expression.


Asunto(s)
Membrana Celular/metabolismo , Células Epiteliales/metabolismo , Mucosa Respiratoria/metabolismo , Actinas/metabolismo , Animales , Línea Celular , Membrana Celular/patología , Perros , Factor de Crecimiento Epidérmico/metabolismo , Células Epiteliales/patología , Regulación de la Expresión Génica , Humanos , Transporte de Proteínas , Mucosa Respiratoria/patología , Septinas , Transducción de Señal
12.
Exp Lung Res ; 37(10): 575-84, 2011 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-22044313

RESUMEN

To test the hypothesis that interleukin-6 (IL-6) contributes to the development of ventilator-associated lung injury (VALI), IL-6-deficient (IL6(-/-)) and wild-type control (WT) mice received intratracheal hydrochloric acid followed by randomization to mechanical ventilation (MV + IT HCl) or spontaneous ventilation (IT HCl). After 4 hours, injury was assessed by estimation of lung lavage protein concentration and total and differential cell counts, wet/dry lung weight ratio, pulmonary cell death, histologic inflammation score (LIS), and parenchymal myeloperoxidase (MPO) concentration. Vascular endothelial growth factor (VEGF) concentration was measured in lung lavage and homogenate, as IL-6 and stretch both regulate expression of this potent mediator of permeability. MV-induced increases in alveolar barrier dysfunction and lavage VEGF were attenuated in IL6(-/-) mice as compared with WT controls, whereas tissue VEGF concentration increased. The effects of IL-6 deletion on alveolar permeability and VEGF concentration were inflammation independent, as parenchymal MPO concentration, LIS, and lavage total and differential cell counts did not differ between WT and IL6(-/-) mice following MV + IT HCl. These data support a role for IL-6 in promoting VALI in this two-hit model. Strategies to interfere with IL-6 expression or signaling may represent important therapeutic targets to limit the injurious effects of MV in inflamed lungs.


Asunto(s)
Permeabilidad Capilar/fisiología , Interleucina-6/metabolismo , Lesión Pulmonar/metabolismo , Lesión Pulmonar/patología , Pulmón/metabolismo , Animales , Líquido del Lavado Bronquioalveolar , Inflamación/metabolismo , Inflamación/patología , Interleucina-6/deficiencia , Interleucina-6/genética , Pulmón/patología , Lesión Pulmonar/etiología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Distribución Aleatoria , Respiración Artificial/métodos , Factor A de Crecimiento Endotelial Vascular/metabolismo , Ventiladores Mecánicos
13.
Proc Natl Acad Sci U S A ; 105(9): 3345-50, 2008 Mar 04.
Artículo en Inglés | MEDLINE | ID: mdl-18305162

RESUMEN

As the interface with the outside world, the airway epithelial barrier is critical to lung defense. Because of respiratory efforts, the airways are exposed to shear stress; however, little is known regarding the effects of shear on epithelial function. We report that low-level shear stress enhances epithelial barrier function, an effect that requires serial activation of the transient receptor potential vanilloid (TRPV) 4 and L-type voltage-gated calcium channel (VGCC) and an increase in intracellular calcium. These changes lead to a selective decrease in aquaporin-5 (AQP5) abundance because of protein internalization and degradation. To determine whether AQP5 plays a role in mediating the shear effects on paracellular permeability, we overexpressed hAQP5 in 16HBE cells, an airway epithelial cell line without endogenous AQP5. We found that AQP5 expression was needed for shear-induced barrier enhancement. These findings have direct relevance to the regulation of epithelial barrier function, membrane permeability, and water homeostasis in the respiratory epithelia.


Asunto(s)
Acuaporina 5/fisiología , Permeabilidad de la Membrana Celular , Mucosa Respiratoria/fisiología , Canales Catiónicos TRPV/metabolismo , Animales , Transporte Biológico , Calcio , Canales de Calcio Tipo L/metabolismo , Humanos , Ratones , Ratones Noqueados , Estrés Mecánico , Agua
14.
Physiol Genomics ; 41(2): 109-19, 2010 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-20028937

RESUMEN

In animal models of acute lung injury (ALI), gene expression studies have focused on the acute phase of illness, with little emphasis on resolution. In this study, the acute phase of intratracheal lipopolysaccharide (IT LPS)-induced lung injury was similar in wild-type (WT) and recombinase-activating gene-1-deficient (Rag-1(-/-)) lymphocyte-deficient mice, but resolution was impaired and resolution-phase lung gene expression remained different from baseline only in Rag-1(-/-) mice. By focusing on groups of genes involved in similar biological processes (gene ontologies) pertinent to inflammation and the immune response, we identified 102 genes at days 4 and 10 after IT LPS with significantly different expression between WT and Rag-1(-/-) mice. After adoptive transfer of isolated CD4+CD25+Foxp3+ regulatory T cells (Tregs) to Rag-1(-/-) mice at the time of IT LPS, resolution was similar to that in WT mice. Of the 102 genes distinctly changed in either WT or Rag-1(-/-) mice from our 7 gene ontologies, 19 genes reverted from the Rag-1(-/-) to the WT pattern of expression after adoptive transfer of Tregs, implicating those 19 genes in Treg-mediated resolution of ALI.


Asunto(s)
Lesión Pulmonar Aguda/genética , Perfilación de la Expresión Génica , Pulmón/inmunología , Linfocitos T Reguladores/inmunología , Cicatrización de Heridas/genética , Lesión Pulmonar Aguda/inducido químicamente , Lesión Pulmonar Aguda/inmunología , Lesión Pulmonar Aguda/patología , Traslado Adoptivo , Animales , Líquido del Lavado Bronquioalveolar/inmunología , Análisis por Conglomerados , Modelos Animales de Enfermedad , Perfilación de la Expresión Génica/métodos , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Genes RAG-1 , Lipopolisacáridos , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Análisis de Secuencia por Matrices de Oligonucleótidos , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Linfocitos T Reguladores/trasplante , Factores de Tiempo
15.
Am J Physiol Lung Cell Mol Physiol ; 298(3): L371-81, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20034961

RESUMEN

Despite the associated morbidity and mortality, underlying mechanisms leading to the development of acute lung injury (ALI) remain incompletely understood. Frequently, ALI develops in the hospital, coinciding with institution of various therapies, including the use of supplemental oxygen. Although pathological evidence of hyperoxia-induced ALI in humans has yet to be proven, animal studies involving high oxygen concentration reproducibly induce ALI. The potentially injurious role of lower and presumably safer oxygen concentrations has not been well characterized in any species. We hypothesized that in the setting of a preexisting insult to the lung, the addition of moderate-range oxygen can augment lung injury. Our model of low-dose intratracheal LPS (IT LPS) followed by 60% oxygen caused a significant increase in ALI compared with LPS or oxygen alone with increased alveolar neutrophils, histological injury, and epithelial barrier permeability. In the LPS plus oxygen group, regulatory T cell number was reduced, and macrophage activation markers were increased, compared with LPS alone. Antibody-mediated depletion of neutrophils significantly abrogated the observed lung injury for all measured factors. The enhanced presence of alveolar neutrophils in the setting of LPS and oxygen is due, at least in part, to elevated chemokine gradients signaling neutrophils to the alveolar space. We believe these results strongly support an effect of lower concentrations of oxygen to augment the severity of a mild preexisting lung injury and warrants further investigation in both animals and humans.


Asunto(s)
Lesión Pulmonar/patología , Oxígeno/farmacología , Animales , Líquido del Lavado Bronquioalveolar/citología , Permeabilidad de la Membrana Celular/efectos de los fármacos , Citocinas/metabolismo , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Mediadores de Inflamación/metabolismo , Recuento de Leucocitos , Procedimientos de Reducción del Leucocitos , Lipopolisacáridos/farmacología , Ratones , Ratones Endogámicos C57BL , Neutrófilos/efectos de los fármacos , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/metabolismo
16.
Kidney Int ; 76(7): 717-29, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19625990

RESUMEN

T lymphocytes modulate early ischemia-reperfusion injury in the kidney; however, their role during repair is unknown. We studied the role of TCRbeta(+)CD4(+)CD25(+)Foxp3(+) regulatory T cells (Tregs), known to blunt immune responses, in repair after ischemia-reperfusion injury to the kidney. Using a murine model of ischemic acute kidney injury we found that there was a significant trafficking of Tregs into the kidneys after 3 and 10 days. Post-ischemic kidneys had increased numbers of TCRbeta(+)CD4(+) and TCRbeta(+)CD8(+) T cells with enhanced pro-inflammatory cytokine production. Treg depletion starting 1 day after ischemic injury using anti-CD25 antibodies increased renal tubular damage, reduced tubular proliferation at both time points, enhanced infiltrating T lymphocyte cytokine production at 3 days and TNF-alpha generation by TCRbeta(+)CD4(+) T cells at 10 days. In separate mice, infusion of CD4(+)CD25(+) Tregs 1 day after initial injury reduced INF-gamma production by TCRbeta(+)CD4(+) T cells at 3 days, improved repair and reduced cytokine generation at 10 days. Treg manipulation had minimal effect on neutrophil and macrophage infiltration; Treg depletion worsened mortality and serum creatinine, while Treg infusion had a late beneficial effect on serum creatinine in bilateral ischemia. Our study demonstrates that Tregs infiltrate ischemic-reperfused kidneys during the healing process promoting repair, likely through modulation of pro-inflammatory cytokine production of other T cell subsets. Treg targeting could be a novel therapeutic approach to enhance recovery from ischemic acute kidney injury.


Asunto(s)
Regeneración/fisiología , Daño por Reperfusión/inmunología , Linfocitos T Reguladores/inmunología , Animales , Movimiento Celular/inmunología , Citocinas/biosíntesis , Citocinas/inmunología , Factores de Transcripción Forkhead , Ratones , Linfocitos T/inmunología , Factores de Tiempo
17.
Cell Mol Biol Lett ; 13(2): 195-211, 2008.
Artículo en Inglés | MEDLINE | ID: mdl-17965968

RESUMEN

Altered tissue water homeostasis may contribute to edema formation during various stresses including bacterial infection. We observed induction of aquaporin-1 (AQP1) during Staphylococcus aureus infection of cultured cells indicating a potential mechanism underlying altered water homeostasis during infection. To investigate mechanisms of AQP1 induction, we examined the effects of the S. aureus alpha-hemolysin on AQP1 abundance in Balb/c fibroblasts. Fibroblasts incubated with 30 microg/ml hemolysin exhibited a 5-10 fold increase in AQP1 protein within 4-6 hours of exposure. The use of multiple signaling cascade inhibitors failed to affect hemolysin-mediated accumulation of AQP1. However, immunoprecipitation revealed an initial accumulation of ubiquitinated AQP1 followed by a decrease to baseline levels after 4 hours. Immunofluorescence indicated that following hemolysin exposure, AQP1 was no longer on the plasma membrane, but was found in a population of submembrane vacuoles. AQP1 redistribution was further indicated by surface biotinylation experiments suggesting diminished AQP1 abundance on the plasma membrane as well as redistribution out of lipid raft fractions. Live cell confocal microscopy revealed that the pattern of cell volume change observed following hemolysin exposure was altered in cells in which AQP1 was silenced. We conclude that alpha-toxin alters proteasomal processing and leads to intracellular accumulation of AQP1, which may likely contribute to disrupted cell volume homeostasis in infection.


Asunto(s)
Acuaporina 1/metabolismo , Toxinas Bacterianas/farmacología , Proteínas Hemolisinas/farmacología , Necrosis/patología , Animales , Acuaporina 1/genética , Tamaño de la Célula/efectos de los fármacos , Endocitosis/efectos de los fármacos , Fibroblastos/citología , Fibroblastos/efectos de los fármacos , Fibroblastos/enzimología , Fibroblastos/microbiología , Regulación de la Expresión Génica/efectos de los fármacos , Microdominios de Membrana/efectos de los fármacos , Ratones , Ratones Endogámicos BALB C , Proteínas Quinasas Activadas por Mitógenos/metabolismo , Inhibidores de Proteasoma , Staphylococcus aureus , Ubiquitinación/efectos de los fármacos
18.
Trends Endocrinol Metab ; 13(8): 355-60, 2002 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-12217493

RESUMEN

Recent discovery of a family of water-specific membrane channel proteins, the aquaporins, has provided new insights into the molecular basis of membrane water permeability. Eleven mammalian aquaporins have been identified to date, with homolog present across the spectrum of life, including bacteria, yeast and plants. The distribution of the mammalian aquaporins predicts their participation in a range of pathophysiological events. Empirical evidence of a physiological role for aquaporins is emerging from studies in both mice and humans, and suggests that aquaporins are likely to play significant roles in human pathophysiology.


Asunto(s)
Acuaporinas , Enfermedad , Animales , Acuaporinas/fisiología , Evolución Biológica , Glándulas Exocrinas , Ojo , Oftalmopatías , Humanos , Riñón , Enfermedades Renales , Pulmón , Enfermedades Pulmonares , Ratones
19.
Vaccine ; 33(36): 4495-504, 2015 Aug 26.
Artículo en Inglés | MEDLINE | ID: mdl-26196325

RESUMEN

Live Attenuated Influenza Vaccine (LAIV) strains are associated with cold adapted, temperature sensitive and attenuated phenotypes that have been studied in non-human or immortalized cell cultures as well as in animal models. Using a primary, differentiated human nasal epithelial cell (hNEC) culture system we compared the replication kinetics, levels of cell-associated viral proteins and virus particle release during infection with LAIV or the corresponding wild type (WT) influenza viruses. At both 33 °C and 37 °C, seasonal influenza virus and an antigenically matched LAIV replicated to similar titers in MDCK cells but seasonal influenza virus replicated to higher titers than LAIV in hNEC cultures, suggesting a greater restriction of LAIV replication in hNEC cultures. Despite the disparity in infectious virus production, the supernatants from H1N1 and LAIV infected hNEC cultures had equivalent amounts of viral proteins and hemagglutination titers, suggesting the formation of non-infectious virus particles by LAIV in hNEC cultures.


Asunto(s)
Células Epiteliales/virología , Virus de la Influenza A/fisiología , Vacunas contra la Influenza , Mucosa Nasal/virología , Replicación Viral , Animales , Antígenos Virales/análisis , Células Cultivadas , Medios de Cultivo/química , Perros , Humanos , Mucosa Nasal/citología , Temperatura , Vacunas Atenuadas , Carga Viral
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA