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.
PLoS One ; 14(2): e0213092, 2019.
Artículo en Inglés | MEDLINE | ID: mdl-30807600

RESUMEN

[This corrects the article DOI: 10.1371/journal.pone.0096681.].

2.
J Cyst Fibros ; 17(4): 454-461, 2018 07.
Artículo en Inglés | MEDLINE | ID: mdl-29241629

RESUMEN

INTRODUCTION: Cystic fibrosis (CF) is a multi-organ disorder characterized by chronic sino-pulmonary infections and inflammation. Many patients with CF suffer from repeated pulmonary exacerbations that are predictors of worsened long-term morbidity and mortality. There are no reliable markers that associate with the onset or progression of an exacerbation or pulmonary deterioration. Previously, we found that the Mirc1/Mir17-92a cluster which is comprised of 6 microRNAs (Mirs) is highly expressed in CF mice and negatively regulates autophagy which in turn improves CF transmembrane conductance regulator (CFTR) function. Therefore, here we sought to examine the expression of individual Mirs within the Mirc1/Mir17-92 cluster in human cells and biological fluids and determine their role as biomarkers of pulmonary exacerbations and response to treatment. METHODS: Mirc1/Mir17-92 cluster expression was measured in human CF and non-CF plasma, blood-derived neutrophils, and sputum samples. Values were correlated with pulmonary function, exacerbations and use of CFTR modulators. RESULTS: Mirc1/Mir17-92 cluster expression was not significantly elevated in CF neutrophils nor plasma when compared to the non-CF cohort. Cluster expression in CF sputum was significantly higher than its expression in plasma. Elevated CF sputum Mirc1/Mir17-92 cluster expression positively correlated with pulmonary exacerbations and negatively correlated with lung function. Patients with CF undergoing treatment with the CFTR modulator Ivacaftor/Lumacaftor did not demonstrate significant change in the expression Mirc1/Mir17-92 cluster after six months of treatment. CONCLUSIONS: Mirc1/Mir17-92 cluster expression is a promising biomarker of respiratory status in patients with CF including pulmonary exacerbation.


Asunto(s)
Aminofenoles/administración & dosificación , Aminopiridinas/administración & dosificación , Benzodioxoles/administración & dosificación , Fibrosis Quística , MicroARNs/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Quinolonas/administración & dosificación , Sistema Respiratorio , Adolescente , Adulto , Biomarcadores/metabolismo , Agonistas de los Canales de Cloruro/administración & dosificación , Correlación de Datos , Fibrosis Quística/tratamiento farmacológico , Fibrosis Quística/genética , Fibrosis Quística/metabolismo , Fibrosis Quística/fisiopatología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Progresión de la Enfermedad , Combinación de Medicamentos , Monitoreo de Drogas/métodos , Femenino , Perfilación de la Expresión Génica , Humanos , Masculino , ARN Largo no Codificante , Pruebas de Función Respiratoria/métodos , Sistema Respiratorio/efectos de los fármacos , Sistema Respiratorio/metabolismo , Sistema Respiratorio/fisiopatología , Esputo/metabolismo
3.
Autophagy ; 12(11): 2026-2037, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27541364

RESUMEN

Cystic fibrosis (CF) is a fatal, genetic disorder that critically affects the lungs and is directly caused by mutations in the CF transmembrane conductance regulator (CFTR) gene, resulting in defective CFTR function. Macroautophagy/autophagy is a highly regulated biological process that provides energy during periods of stress and starvation. Autophagy clears pathogens and dysfunctional protein aggregates within macrophages. However, this process is impaired in CF patients and CF mice, as their macrophages exhibit limited autophagy activity. The study of microRNAs (Mirs), and other noncoding RNAs, continues to offer new therapeutic targets. The objective of this study was to elucidate the role of Mirs in dysregulated autophagy-related genes in CF macrophages, and then target them to restore this host-defense function and improve CFTR channel function. We identified the Mirc1/Mir17-92 cluster as a potential negative regulator of autophagy as CF macrophages exhibit decreased autophagy protein expression and increased cluster expression when compared to wild-type (WT) counterparts. The absence or reduced expression of the cluster increases autophagy protein expression, suggesting the canonical inverse relationship between Mirc1/Mir17-92 and autophagy gene expression. An in silico study for targets of Mirs that comprise the cluster suggested that the majority of the Mirs target autophagy mRNAs. Those targets were validated by luciferase assays. Notably, the ability of macrophages expressing mutant F508del CFTR to transport halide through their membranes is compromised and can be restored by downregulation of these inherently elevated Mirs, via restoration of autophagy. In vivo, downregulation of Mir17 and Mir20a partially restored autophagy expression and hence improved the clearance of Burkholderia cenocepacia. Thus, these data advance our understanding of mechanisms underlying the pathobiology of CF and provide a new therapeutic platform for restoring CFTR function and autophagy in patients with CF.


Asunto(s)
Autofagia/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Fibrosis Quística/genética , Fibrosis Quística/patología , Regulación de la Expresión Génica , Macrófagos/metabolismo , MicroARNs/genética , Regiones no Traducidas 3'/genética , Animales , Antagomirs/farmacología , Autofagia/efectos de los fármacos , Proteínas Relacionadas con la Autofagia/metabolismo , Burkholderia cenocepacia/fisiología , Células Cultivadas , Fibrosis Quística/microbiología , Regulación de la Expresión Génica/efectos de los fármacos , Homocigoto , Pulmón/metabolismo , Pulmón/microbiología , Pulmón/patología , Macrófagos/efectos de los fármacos , Macrófagos/patología , Ratones , Ratones Endogámicos C57BL , MicroARNs/metabolismo , Células 3T3 NIH
4.
Epigenetics ; 11(5): 381-8, 2016 05 03.
Artículo en Inglés | MEDLINE | ID: mdl-26909551

RESUMEN

Autophagy is a biological process characterized by self-digestion and involves induction of autophagosome formation, leading to degradation of autophagic cargo. Aging is associated with the reduction of autophagy activity leading to neurodegenerative disorders, chronic inflammation, and susceptibility to infection; however, the underlying mechanism is unclear. DNA methylation by DNA methyltransferases reduces the expression of corresponding genes. Since macrophages are major players in inflammation and defense against infection we determined the differences in methylation of autophagy genes in macrophages derived from young and aged mice. We found that promoter regions of Atg5 and LC3B are hypermethylated in macrophages from aged mice and this is accompanied by low gene expression. Treatment of aged mice and their derived macrophages with methyltransferase inhibitor (2)-epigallocatechin-3-gallate (EGCG) or specific DNA methyltransferase 2 (DNMT2) siRNA restored the expression of Atg5 and LC3 in vivo and in vitro. Our study builds a foundation for the development of novel therapeutics aimed to improve autophagy in the elderly population and suggests a role for DNMT2 in DNA methylation activities.


Asunto(s)
Envejecimiento/genética , Proteína 5 Relacionada con la Autofagia/genética , ADN (Citosina-5-)-Metiltransferasas/genética , Metilación de ADN/genética , Proteínas Asociadas a Microtúbulos/genética , Envejecimiento/patología , Animales , Autofagosomas/efectos de los fármacos , Autofagia/efectos de los fármacos , Autofagia/genética , Catequina/administración & dosificación , Catequina/análogos & derivados , ADN (Citosina-5-)-Metiltransferasas/antagonistas & inhibidores , Metilación de ADN/efectos de los fármacos , Inhibidores Enzimáticos/administración & dosificación , Humanos , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Ratones , ARN Interferente Pequeño/genética
5.
Sci Rep ; 5: 18479, 2015 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-26686473

RESUMEN

Inflammasomes are multiprotein complexes that include members of the NOD-like receptor family and caspase-1. Caspase-1 is required for the fusion of the Legionella vacuole with lysosomes. Caspase-11, independently of the inflammasome, also promotes phagolysosomal fusion. However, it is unclear how these proteases alter intracellular trafficking. Here, we show that caspase-11 and caspase-1 function in opposing manners to phosphorylate and dephosphorylate cofilin, respectively upon infection with Legionella. Caspase-11 targets cofilin via the RhoA GTPase, whereas caspase-1 engages the Slingshot phosphatase. The absence of either caspase-11 or caspase-1 maintains actin in the polymerized or depolymerized form, respectively and averts the fusion of pathogen-containing vacuoles with lysosomes. Therefore, caspase-11 and caspase-1 converge on the actin machinery with opposing effects to promote vesicular trafficking.


Asunto(s)
Actinas/metabolismo , Caspasa 1/genética , Cofilina 1/genética , Enfermedad de los Legionarios/genética , Fosfoproteínas Fosfatasas/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Actinas/genética , Animales , Caspasa 1/metabolismo , Cofilina 1/metabolismo , Humanos , Inflamasomas/genética , Inflamasomas/metabolismo , Legionella pneumophila/genética , Legionella pneumophila/patogenicidad , Enfermedad de los Legionarios/metabolismo , Enfermedad de los Legionarios/patología , Lisosomas/genética , Lisosomas/metabolismo , Ratones , Ratones Noqueados , Complejos Multiproteicos/genética , Complejos Multiproteicos/metabolismo , Fosfoproteínas Fosfatasas/genética , Receptores de Superficie Celular/genética , Receptores de Superficie Celular/metabolismo , Vacuolas/genética , Vacuolas/metabolismo , Proteínas de Transporte Vesicular/genética , Proteínas de Transporte Vesicular/metabolismo , Proteína de Unión al GTP rhoA/genética
6.
Am J Physiol Lung Cell Mol Physiol ; 308(11): L1136-44, 2015 Jun 01.
Artículo en Inglés | MEDLINE | ID: mdl-25840995

RESUMEN

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


Asunto(s)
Lesión Pulmonar Aguda/virología , Virus de la Influenza A/inmunología , Interleucina-6/fisiología , Infecciones por Orthomyxoviridae/inmunología , Factor de Crecimiento Transformador beta/fisiología , Lesión Pulmonar Aguda/inmunología , Animales , Líquido del Lavado Bronquioalveolar , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Inmunidad Innata , Macrófagos Alveolares/metabolismo , Macrófagos Alveolares/virología , Ratones de la Cepa 129 , Ratones Endogámicos C57BL , Ratones Endogámicos CFTR , Eliminación de Secuencia
7.
PLoS One ; 9(5): e96681, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24798083

RESUMEN

Burkholderia cenocepacia is a virulent pathogen that causes significant morbidity and mortality in patients with cystic fibrosis (CF), survives intracellularly in macrophages, and uniquely causes systemic infections in CF. Autophagy is a physiologic process that involves engulfing non-functional organelles and proteins and delivering them for lysosomal degradation, but also plays a role in eliminating intracellular pathogens, including B. cenocepacia. Autophagy is defective in CF but can be stimulated in murine CF models leading to increased clearance of B. cenocepacia, but little is known about autophagy stimulation in human CF macrophages. IFN-γ activates macrophages and increases antigen presentation while also inducing autophagy in macrophages. We therefore, hypothesized that treatment with IFN-γ would increase autophagy and macrophage activation in patients with CF. Peripheral blood monocyte derived macrophages (MDMs) were obtained from CF and non-CF donors and subsequently infected with B. cenocepacia. Basal serum levels of IFN-γ were similar between CF and non-CF patients, however after B. cenocepacia infection there is deficient IFN-γ production in CF MDMs. IFN-γ treated CF MDMs demonstrate increased co-localization with the autophagy molecule p62, increased autophagosome formation, and increased trafficking to lysosomes compared to untreated CF MDMs. Electron microscopy confirmed IFN-γ promotes double membrane vacuole formation around bacteria in CF MDMs, while only single membrane vacuoles form in untreated CF cells. Bacterial burden is significantly reduced in autophagy stimulated CF MDMs, comparable to non-CF levels. IL-1ß production is decreased in CF MDMs after IFN-γ treatment. Together, these results demonstrate that IFN-γ promotes autophagy-mediated clearance of B. cenocepacia in human CF macrophages.


Asunto(s)
Autofagia , Infecciones por Burkholderia/inmunología , Burkholderia cenocepacia , Fibrosis Quística/inmunología , Interferón gamma/inmunología , Macrófagos/inmunología , Adolescente , Adulto , Azitromicina/química , Niño , Fibrosis Quística/microbiología , Femenino , Humanos , Interleucina-1beta/inmunología , Lisosomas/metabolismo , Macrófagos/microbiología , Masculino , Microscopía Confocal , Monocitos/inmunología , Monocitos/microbiología , Fagosomas , Adulto Joven
8.
Artículo en Inglés | MEDLINE | ID: mdl-23750338

RESUMEN

Legionella pneumophila, the causative agent of Legionnaire's disease, replicates in human alveolar macrophages to establish infection. There is no human-to-human transmission and the main source of infection is L. pneumophila biofilms established in air conditioners, water fountains, and hospital equipments. The biofilm structure provides protection to the organism from disinfectants and antibacterial agents. L. pneumophila infection in humans is characterized by a subtle initial immune response, giving time for the organism to establish infection before the patient succumbs to pneumonia. Planktonic L. pneumophila elicits a strong immune response in murine, but not in human macrophages enabling control of the infection. Interactions between planktonic L. pneumophila and murine or human macrophages have been studied for years, yet the interface between biofilm-derived L. pneumophila and macrophages has not been explored. Here, we demonstrate that biofilm-derived L. pneumophila replicates significantly more in murine macrophages than planktonic bacteria. In contrast to planktonic L. pneumophila, biofilm-derived L. pneumophila lacks flagellin expression, do not activate caspase-1 or -7 and trigger less cell death. In addition, while planktonic L. pneumophila is promptly delivered to lysosomes for degradation, most biofilm-derived bacteria were enclosed in a vacuole that did not fuse with lysosomes in murine macrophages. This study advances our understanding of the innate immune response to biofilm-derived L. pneumophila and closely reproduces the natural mode of infection in human.


Asunto(s)
Biopelículas/crecimiento & desarrollo , Interacciones Huésped-Patógeno , Evasión Inmune , Inmunidad Innata , Legionella pneumophila/inmunología , Macrófagos/inmunología , Macrófagos/microbiología , Animales , Carga Bacteriana , Legionella pneumophila/crecimiento & desarrollo , Legionella pneumophila/aislamiento & purificación , Legionella pneumophila/fisiología , Ratones , Ratones Endogámicos C57BL
9.
Eur J Immunol ; 43(5): 1333-44, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23420491

RESUMEN

Legionella pneumophila (L. pneumophila) is an intracellular bacterium of human alveolar macrophages that causes Legionnaires' disease. In contrast to humans, most inbred mouse strains are restrictive to L. pneumophila replication. We demonstrate that autophagy targets L. pneumophila vacuoles to lysosomes and that this process requires ubiquitination of L. pneumophila vacuoles and the subsequent binding of the autophagic adaptor p62/SQSTM1 to ubiquitinated vacuoles. The L. pneumophila legA9 encodes for an ankyrin-containing protein with unknown role. We show that the legA9 mutant replicate in WT mice and their bone marrow-derived macrophages. This is the first L. pneumophila mutant to be found to replicate in WT bone marrow-derived macrophages other than the Fla mutant. Less legA9 mutant-containing vacuoles acquired ubiquitin labeling and p62/SQSTM1 staining, evading autophagy uptake and avoiding lysosomal fusion. Thus, we describe a bacterial protein that targets the L. pneumophila-containing vacuole for autophagy uptake.


Asunto(s)
Autofagia/inmunología , Proteínas Bacterianas/genética , Legionella pneumophila/genética , Macrófagos/microbiología , Mutación , Vacuolas/inmunología , Proteínas Adaptadoras Transductoras de Señales/genética , Proteínas Adaptadoras Transductoras de Señales/inmunología , Animales , Proteínas Bacterianas/inmunología , Células Cultivadas , Proteínas de Choque Térmico/genética , Proteínas de Choque Térmico/inmunología , Especificidad del Huésped , Interacciones Huésped-Patógeno , Humanos , Evasión Inmune/genética , Legionella pneumophila/inmunología , Lisosomas/inmunología , Lisosomas/metabolismo , Lisosomas/microbiología , Macrófagos/inmunología , Ratones , Fagosomas/inmunología , Fagosomas/metabolismo , Fagosomas/microbiología , Unión Proteica , Proteína Sequestosoma-1 , Ubiquitina/metabolismo , Ubiquitinación , Vacuolas/metabolismo , Vacuolas/microbiología
10.
J Biol Chem ; 288(3): 2049-58, 2013 Jan 18.
Artículo en Inglés | MEDLINE | ID: mdl-23148214

RESUMEN

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


Asunto(s)
Proteínas Adaptadoras Transductoras de Señales/genética , Autofagia/genética , Infecciones por Burkholderia/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Fibrosis Quística/genética , Proteínas de Choque Térmico/genética , Macrófagos/metabolismo , Proteínas Adaptadoras Transductoras de Señales/antagonistas & inhibidores , Proteínas Adaptadoras Transductoras de Señales/metabolismo , Animales , Biomarcadores/metabolismo , Infecciones por Burkholderia/complicaciones , Infecciones por Burkholderia/metabolismo , Infecciones por Burkholderia/microbiología , Burkholderia cenocepacia/fisiología , Fibrosis Quística/complicaciones , Fibrosis Quística/metabolismo , Fibrosis Quística/microbiología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Expresión Génica , Proteínas de Choque Térmico/antagonistas & inhibidores , Proteínas de Choque Térmico/metabolismo , Humanos , Macrófagos/microbiología , Macrófagos/patología , Ratones , Ratones Transgénicos , Viabilidad Microbiana , Proteínas Asociadas a Microtúbulos/metabolismo , Fagosomas/metabolismo , Transporte de Proteínas , ARN Interferente Pequeño/genética , Proteína Sequestosoma-1 , Transfección , Ubiquitina/genética , Ubiquitina/metabolismo
11.
Biochem Biophys Res Commun ; 424(2): 221-7, 2012 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-22728038

RESUMEN

Cystic fibrosis (CF) is accompanied with heightened inflammation worsened by drug resistant Burkholderia cenocepacia. Human CF macrophage responses to B. cenocepacia are poorly characterized and variable in the literature. Therefore, we examined human macrophage responses to the epidemic B. cenocepacia J2315 strain in order to identify novel anti-inflammatory targets. Peripheral blood monocyte derived macrophages were obtained from 23 CF and 27 non-CF donors. Macrophages were infected with B. cenocepacia J2315 and analyzed for cytokines, cytotoxicity, and microscopy. CF macrophages demonstrated significant increases in IL-1ß, IL-10, MCP-1, and IFN-γ production in comparison to non-CF controls. CF patients on prednisone exhibited globally diminished cytokines compared to controls and other CF patients. CF macrophages also displayed increased bacterial burden and cell death. In conclusion, CF macrophages demonstrate exaggerated IL-1ß, IL-10, MCP-1, and IFN-γ production and cell death during B. cenocepacia infection. Treatment with corticosteroids acutely suppressed cytokine responses.


Asunto(s)
Infecciones por Burkholderia/microbiología , Burkholderia cenocepacia , Fibrosis Quística/microbiología , Fibrosis Quística/patología , Macrófagos/inmunología , Adolescente , Corticoesteroides/farmacología , Adulto , Apoptosis , Quimiocina CCL2/metabolismo , Niño , Preescolar , Fibrosis Quística/inmunología , Femenino , Humanos , Lactante , Inflamación/microbiología , Inflamación/patología , Interleucina-10/metabolismo , Interleucina-1beta/metabolismo , Lisosomas/microbiología , Macrófagos/efectos de los fármacos , Macrófagos/ultraestructura , Masculino , Microscopía Electrónica , Persona de Mediana Edad , Adulto Joven
12.
Immunity ; 37(1): 35-47, 2012 Jul 27.
Artículo en Inglés | MEDLINE | ID: mdl-22658523

RESUMEN

Inflammasomes are multiprotein complexes that include members of the NLR (nucleotide-binding domain leucine-rich repeat containing) family and caspase-1. Once bacterial molecules are sensed within the macrophage, the inflammasome is assembled, mediating the activation of caspase-1. Caspase-11 mediates caspase-1 activation in response to lipopolysaccharide and bacterial toxins, and yet its role during bacterial infection is unknown. Here, we demonstrated that caspase-11 was dispensable for caspase-1 activation in response to Legionella, Salmonella, Francisella, and Listeria. We also determined that active mouse caspase-11 was required for restriction of L. pneumophila infection. Similarly, human caspase-4 and caspase-5, homologs of mouse caspase-11, cooperated to restrict L. pneumophila infection in human macrophages. Caspase-11 promoted the fusion of the L. pneumophila vacuole with lysosomes by modulating actin polymerization through cofilin. However, caspase-11 was dispensable for the fusion of lysosomes with phagosomes containing nonpathogenic bacteria, uncovering a fundamental difference in the trafficking of phagosomes according to their cargo.


Asunto(s)
Actinas/metabolismo , Bacterias/inmunología , Caspasas/metabolismo , Lisosomas/metabolismo , Fagosomas/metabolismo , Multimerización de Proteína , Factores Despolimerizantes de la Actina/metabolismo , Animales , Bacterias/crecimiento & desarrollo , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/metabolismo , Caspasa 1/deficiencia , Caspasa 1/genética , Caspasa 1/metabolismo , Caspasas/deficiencia , Caspasas/genética , Caspasas Iniciadoras , Humanos , Macrófagos/inmunología , Macrófagos/metabolismo , Macrófagos/microbiología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fagosomas/microbiología , Fosforilación
13.
Infect Immun ; 80(4): 1390-8, 2012 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-22311922

RESUMEN

The mechanisms by which Streptococcus pneumoniae obtains carbohydrates for growth during airway colonization remain to be elucidated. The low concentration of free carbohydrates in the normal human airway suggests that pneumococci must utilize complex glycan structures for growth. The glycosaminoglycan hyaluronic acid is present on the apical surface of airway epithelial cells. As pneumococci express a hyaluronate lyase (Hyl) that cleaves hyaluronic acid into disaccharides, we hypothesized that during colonization pneumococci utilize the released carbohydrates for growth. Hyaluronic acid supported significant pneumococcal growth in an hyl-dependent manner. A phosphoenolpyruvate-dependent phosphotransferase system (PTS) and an unsaturated glucuronyl hydrolase (Ugl) encoded downstream of hyl are also essential for growth on hyaluronic acid. This genomic arrangement is present in several other organisms, suggesting conservation of the utilization mechanism between species. In vivo experiments support the hypothesis that S. pneumoniae utilizes hyaluronic acid as a carbon source during colonization. We also demonstrate that pneumococci can utilize the hyaluronic acid capsule of other bacterial species for growth, suggesting an alternative carbohydrate source for pneumococcal growth. Together, these data support a novel function for pneumococcal degradation of hyaluronic acid in vivo and provide mechanistic details of growth on this glycosaminoglycan.


Asunto(s)
Ácido Hialurónico/metabolismo , Streptococcus pneumoniae/metabolismo , Animales , Proteínas Bacterianas/genética , Proteínas Bacterianas/metabolismo , Glicósido Hidrolasas/metabolismo , Humanos , Ratones , Fosfotransferasas/metabolismo , Polisacárido Liasas/metabolismo , Streptococcus pneumoniae/enzimología , Streptococcus pneumoniae/crecimiento & desarrollo
14.
Free Radic Biol Med ; 48(9): 1228-35, 2010 May 01.
Artículo en Inglés | MEDLINE | ID: mdl-20156552

RESUMEN

The development of smart anticancer drugs that can selectively kill cancer cells while sparing the surrounding healthy tissues/cells is of paramount importance for safe and effective cancer therapy. We report a novel class of bifunctional compounds based on diarylidenyl piperidone (DAP) conjugated to an N-hydroxypyrroline (NOH; a nitroxide precursor) group. We hypothesized that the DAP would have cytotoxic (anticancer) activity, whereas the NOH moiety would function as a tissue-specific modulator (antioxidant) of cytotoxicity. The study used four DAPs, namely H-4073 and H-4318 without NOH and HO-3867 and HO-4200 with NOH substitution. The goal of the study was to evaluate the proof-of-concept anticancer-versus-antioxidant efficacy of the DAPs using a number of cancerous (breast, colon, head and neck, liver, lung, ovarian, and prostate cancer) and noncancerous (smooth muscle, aortic endothelial, and ovarian surface epithelial) human cell lines. Cytotoxicity was determined using an MTT-based cell viability assay. All four compounds induced significant loss of cell viability in cancer cells, whereas HO-3867 and HO-4200 showed significantly less cytotoxicity in noncancerous cells. EPR measurements showed a metabolic conversion of the N-hydroxylamine function to nitroxide with significantly higher levels of the metabolite and superoxide radical-scavenging (antioxidant) activity in noncancerous cells compared to cancer cells. Western blot analysis showed that the DAP-induced growth arrest and apoptosis in cancer cells were mediated by inhibition of STAT3 phosphorylation at the Tyr705 and Ser727 residues and induction of apoptotic markers of cleaved caspase-3 and PARP. The results suggest that the antioxidant-conjugated DAPs will be useful as safe and effective anticancer agents for cancer therapy.


Asunto(s)
Antineoplásicos/farmacología , Antioxidantes/farmacología , Proliferación Celular/efectos de los fármacos , Piperidonas/farmacología , Transducción de Señal/efectos de los fármacos , Apoptosis/efectos de los fármacos , Western Blotting , Línea Celular Tumoral , Supervivencia Celular/efectos de los fármacos , Espectroscopía de Resonancia por Spin del Electrón , Humanos , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción STAT3/efectos de los fármacos , Factor de Transcripción STAT3/metabolismo
15.
Int J Cancer ; 125(9): 2198-204, 2009 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-19623660

RESUMEN

Signal transducer and activator of transcription 3 (STAT3) is activated in a variety of human cancers, including ovarian cancer. The molecular mechanism by which the STAT3 is activated in cancer cells is poorly understood. We observed that human ovarian xenograft tumors (A2780) in mice were severely hypoxic (pO(2) approximately 2 mmHg). We further observed that hypoxic exposure significantly increased the phosphorylation of STAT3 (pSTAT3) at the Tyr705 residue in A2780 cell line. The pSTAT3 (Tyr705) level was highly dependent on cellular oxygenation levels, with a significant increase at <2% O(2), and without any change in the pSTAT3 (Ser727) or total STAT3 levels. The pSTAT3 (Tyr705) elevation following hypoxic exposure could be reversed within 12 hr after returning the cells to normoxia. The increased level of pSTAT3 was partly mediated by increased levels of reactive oxygen species generation in the hypoxic cancer cells. Conventional chemotherapeutic drugs cisplatin and taxol were far less effective in eliminating the hypoxic ovarian cancer cells suggesting a role for pSTAT3 in cellular resistance to chemotherapy. Inhibition of STAT3 by AG490 followed by treatment with cisplatin or taxol resulted in a significant increase in apoptosis suggesting that hypoxia-induced STAT3 activation is responsible for chemoresistance. The results have important clinical implications for the treatment of hypoxic ovarian tumors using STAT3-specific inhibitors.


Asunto(s)
Hipoxia de la Célula , Neoplasias Ováricas/tratamiento farmacológico , Factor de Transcripción STAT3/fisiología , Línea Celular Tumoral , Proliferación Celular , Resistencia a Antineoplásicos , Femenino , Humanos , Neoplasias Ováricas/metabolismo , Neoplasias Ováricas/patología , Oxígeno/farmacología , Especies Reactivas de Oxígeno/metabolismo , Factor de Transcripción STAT3/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...