Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 14 de 14
Filtrar
1.
N Engl J Med ; 389(23): 2140-2150, 2023 Dec 07.
Artículo en Inglés | MEDLINE | ID: mdl-38055252

RESUMEN

BACKGROUND: Janus kinase (JAK) inhibitors, including baricitinib, block cytokine signaling and are effective disease-modifying treatments for several autoimmune diseases. Whether baricitinib preserves ß-cell function in type 1 diabetes is unclear. METHODS: In this phase 2, double-blind, randomized, placebo-controlled trial, we assigned patients with type 1 diabetes diagnosed during the previous 100 days to receive baricitinib (4 mg once per day) or matched placebo orally for 48 weeks. The primary outcome was the mean C-peptide level, determined from the area under the concentration-time curve, during a 2-hour mixed-meal tolerance test at week 48. Secondary outcomes included the change from baseline in the glycated hemoglobin level, the daily insulin dose, and measures of glycemic control assessed with the use of continuous glucose monitoring. RESULTS: A total of 91 patients received baricitinib (60 patients) or placebo (31 patients). The median of the mixed-meal-stimulated mean C-peptide level at week 48 was 0.65 nmol per liter per minute (interquartile range, 0.31 to 0.82) in the baricitinib group and 0.43 nmol per liter per minute (interquartile range, 0.13 to 0.63) in the placebo group (P = 0.001). The mean daily insulin dose at 48 weeks was 0.41 U per kilogram of body weight per day (95% confidence interval [CI], 0.35 to 0.48) in the baricitinib group and 0.52 U per kilogram per day (95% CI, 0.44 to 0.60) in the placebo group. The levels of glycated hemoglobin were similar in the two trial groups. However, the mean coefficient of variation of the glucose level at 48 weeks, as measured by continuous glucose monitoring, was 29.6% (95% CI, 27.8 to 31.3) in the baricitinib group and 33.8% (95% CI, 31.5 to 36.2) in the placebo group. The frequency and severity of adverse events were similar in the two trial groups, and no serious adverse events were attributed to baricitinib or placebo. CONCLUSIONS: In patients with type 1 diabetes of recent onset, daily treatment with baricitinib over 48 weeks appeared to preserve ß-cell function as estimated by the mixed-meal-stimulated mean C-peptide level. (Funded by JDRF International and others; BANDIT Australian New Zealand Clinical Trials Registry number, ACTRN12620000239965.).


Asunto(s)
Diabetes Mellitus Tipo 1 , Células Secretoras de Insulina , Inhibidores de las Cinasas Janus , Humanos , Australia , Glucemia/análisis , Automonitorización de la Glucosa Sanguínea , Péptido C/sangre , Diabetes Mellitus Tipo 1/sangre , Diabetes Mellitus Tipo 1/tratamiento farmacológico , Hemoglobina Glucada/análisis , Insulina/uso terapéutico , Inhibidores de las Cinasas Janus/efectos adversos , Inhibidores de las Cinasas Janus/farmacología , Inhibidores de las Cinasas Janus/uso terapéutico , Células Secretoras de Insulina/efectos de los fármacos , Método Doble Ciego
2.
Am J Respir Cell Mol Biol ; 46(1): 6-13, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-21778412

RESUMEN

The clinical manifestations of infection in cystic fibrosis (CF) are restricted to the lung, and involve a limited number of pathogens, suggesting a specific defect in mucosal immunity. We postulated that cystic fibrosis transmembrane conductance regulator (CTFR) mutations could affect the activation of type I interferon signaling in airway epithelial cells, which function in immune surveillance and initiate the recruitment and activation of immune cells. In response to infection with Pseudomonas aeruginosa, Ifnb was induced more than 100-fold in the murine lung, and the phosphorylation of STAT1 was similarly induced by the expected TLR4/TRIF/MD2/TBK1 cascade. The stimulation by P. aeruginosa of CF (IB3) cells and control (C-38) human cell lines similarly resulted in the induction of IFN-ß, but to a significantly lower extent in CF airway cells. The potential consequences of diminished type I IFN signaling were demonstrated in a murine model of P. aeruginosa pneumonia, pretreatment with polyinosinic:polycytidylic acid significantly enhanced bacterial clearance and correlated with increased numbers of mature CD11c(+)/CD86(+) dendritic cells (DCs) in the lung. Using culture supernatants from CF or control cell lines stimulated with P. aeruginosa, we similarly demonstrated the diminished activation of human monocyte-derived DCs by incubation with CF compared with normal epithelial cell culture supernatants, which was dependent on IFN-ß. These observations suggest that dysfunction of the CFTR in airway epithelial cells may contribute to impaired immune surveillance in the CF airway and resultant colonization by P. aeruginosa.


Asunto(s)
Fibrosis Quística/inmunología , Fibrosis Quística/microbiología , Interferón Tipo I/metabolismo , Infecciones por Pseudomonas/inmunología , Pseudomonas aeruginosa/inmunología , Proteínas Adaptadoras del Transporte Vesicular/inmunología , Proteínas Adaptadoras del Transporte Vesicular/metabolismo , Animales , Antígeno B7-2/inmunología , Antígeno B7-2/metabolismo , Antígeno CD11c/inmunología , Antígeno CD11c/metabolismo , Células Cultivadas , Fibrosis Quística/genética , Fibrosis Quística/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/inmunología , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Células Dendríticas/inmunología , Células Dendríticas/metabolismo , Células Epiteliales/patología , Humanos , Interferón Tipo I/genética , Interferón Tipo I/inmunología , Interferón beta/genética , Interferón beta/inmunología , Antígeno 96 de los Linfocitos/inmunología , Antígeno 96 de los Linfocitos/metabolismo , Ratones , Ratones Endogámicos C57BL , Mutación , Fosforilación , Infecciones por Pseudomonas/genética , Infecciones por Pseudomonas/metabolismo , Infecciones por Pseudomonas/patología , Factor de Transcripción STAT1/metabolismo , Transducción de Señal , Receptor Toll-Like 4/inmunología , Receptor Toll-Like 4/metabolismo
3.
J Biol Chem ; 286(41): 35891-35898, 2011 Oct 14.
Artículo en Inglés | MEDLINE | ID: mdl-21878647

RESUMEN

Staphyococcus aureus and especially the epidemic methicillin-resistant S. aureus strains cause severe necrotizing pneumonia. The mechanisms whereby these organisms invade across the mucosal epithelial barrier to initiate invasive infection are not well understood. Protein A (SpA), a highly conserved and abundant surface protein of S. aureus, activates TNF receptor 1 and EGF receptor (EGFR) signaling cascades that can perturb the cytoskeleton. We demonstrate that wild-type S. aureus, but not spa mutants, invade across polarized airway epithelial cell monolayers via the paracellular junctions. SpA stimulated a RhoA/ROCK/MLC cascade, resulting in the contraction of the cytoskeleton. SpA(+) but not SpA(-) mutants stimulated activation of EGFR and along with subsequent calpain activity cleaved the membrane-spanning junctional proteins occludin and E-cadherin, facilitating staphylococcal transmigration through the cell-cell junctions. Treatment of polarized human airway epithelial monolayers with inhibitors of ROCK, EGFR, MAPKs, or calpain prevented staphylococcal penetration through the monolayers. In vivo, blocking calpain activity impeded bacterial invasion into the lung parenchyma. Thus, S. aureus exploits multiple receptors available on the airway mucosal surface to facilitate invasion across epithelial barriers.


Asunto(s)
Neumonía Estafilocócica/metabolismo , Mucosa Respiratoria/metabolismo , Transducción de Señal , Proteína Estafilocócica A/metabolismo , Staphylococcus aureus/metabolismo , Proteínas de Unión al GTP rho/metabolismo , Proteína de Unión al GTP rhoA/metabolismo , Animales , Cadherinas/genética , Cadherinas/metabolismo , Calpaína/antagonistas & inhibidores , Calpaína/genética , Calpaína/metabolismo , Línea Celular , Receptores ErbB/genética , Receptores ErbB/metabolismo , Femenino , Humanos , Uniones Intercelulares/genética , Uniones Intercelulares/metabolismo , Ratones , Mutación , Neumonía Estafilocócica/tratamiento farmacológico , Neumonía Estafilocócica/genética , Inhibidores de Proteasas/farmacología , Inhibidores de Proteínas Quinasas/farmacología , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Mucosa Respiratoria/microbiología , Proteína Estafilocócica A/genética , Staphylococcus aureus/genética , Staphylococcus aureus/patogenicidad , Proteínas de Unión al GTP rho/genética , Quinasas Asociadas a rho/antagonistas & inhibidores , Quinasas Asociadas a rho/genética , Quinasas Asociadas a rho/metabolismo , Proteína de Unión al GTP rhoA/genética
4.
Infect Immun ; 79(5): 1898-904, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21402768

RESUMEN

Staphylococcus aureus causes especially severe pulmonary infection, associated with high morbidity and mortality. In addition to the effects of specific virulence factors, it appears that the intensity of the host proinflammatory response, particularly in the initial stages of infection, contributes substantially to pulmonary damage. We tested the hypothesis that the CD11c(+) leukocytes are important in the host response to pulmonary infection with methicillin-resistant S. aureus (MRSA) USA300. Clodronate-induced depletion of the alveolar macrophage population resulted in increased numbers of dendritic cells (DCs) and CD4(+) cells in bronchoalveolar lavage (BAL) fluid and was associated with significantly increased mortality by 18 h following S. aureus inoculation but had no effect on bacterial load or polymorphonuclear leukocyte (PMN) numbers in the lung. These clodronate-treated mice also had increased expression of interleukin-17A/F (IL-17A/F) and CXCL10 but not of gamma interferon (IFN-γ) or tumor necrosis factor (TNF). Depletion of the dendritic cell population in mice expressing a CD11c-enhanced green fluorescent protein (EGFP)-diphtheria toxin receptor (DTR) transgene was associated with an increased bacterial load in the lung but not increased mortality. Both DCs and airway epithelial cells produced CXCL9, -10, and -11 in response to S. aureus. Pretreatment of mice with an anti-CXCR3 antibody prior to inoculation with MRSA substantially reduced CD4(+) cells and decreased pulmonary inflammation at 18 h postinfection compared to pretreatment with an IgG control. The results of these experiments suggest that CD11c(+) cells, the induction of CXCR3 ligand expression, and subsequent CD4(+) cell recruitment have an important role in the pathogenesis of severe MRSA pulmonary infection.


Asunto(s)
Antígeno CD11c/inmunología , Linfocitos T CD4-Positivos/inmunología , Leucocitos/inmunología , Staphylococcus aureus Resistente a Meticilina/inmunología , Infecciones del Sistema Respiratorio/inmunología , Infecciones Estafilocócicas/inmunología , Animales , Antígeno CD11c/metabolismo , Separación Celular , Citocinas/biosíntesis , Citocinas/inmunología , Ensayo de Inmunoadsorción Enzimática , Femenino , Citometría de Flujo , Leucocitos/metabolismo , Pulmón/inmunología , Pulmón/microbiología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Receptores CXCR3/inmunología , Receptores CXCR3/metabolismo , Infecciones del Sistema Respiratorio/metabolismo , Infecciones del Sistema Respiratorio/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Infecciones Estafilocócicas/microbiología
5.
J Infect Dis ; 201(4): 508-15, 2010 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-20078212

RESUMEN

BACKGROUND: Postinfluenza Staphylococcus aureus pneumonias are increasingly recognized as a major form of life-threatening infections. METHODS: A mouse model of postinfluenza S. aureus pneumonia was developed. Mice were intranasally infected with bacteria alone or bacteria plus virus. Infection was assessed by mouse survival, lung histopathology, bacterial density in the lungs, and cellular response to infection. RESULTS: Mice infected with both influenza virus and S. aureus showed higher mortality, greater lung parenchymal damage, and greater bacterial density at metastatic tissue sites than mice infected with only S. aureus. At 4 h, more polymorphonuclear leukocytes and fewer CD11c(+) cells were found in lung samples from mice infected with virus and bacteria than in those from mice infected with bacteria. alpha-Hemolysin and protein A were maximally expressed 4 h after infection, and Panton-Valentine leukocidin was maximally expressed 72 h after infection, with higher levels of alpha-hemolysin expression in mice infected with bacteria alone. Interferon gamma expression was higher in tissue collected from mice infected with virus plus bacteria than in those from bacteria-infected mice. CONCLUSIONS: The results from this model demonstrate diverse effects caused by antecedent influenza virus infection, which have a profound influence on the morbidity and mortality associated with S. aureus pneumonia.


Asunto(s)
Modelos Animales de Enfermedad , Infecciones por Orthomyxoviridae/microbiología , Neumonía Estafilocócica/virología , Staphylococcus aureus/patogenicidad , Análisis de Varianza , Animales , Toxinas Bacterianas/biosíntesis , Toxinas Bacterianas/genética , Líquido del Lavado Bronquioalveolar/citología , Ensayo de Inmunoadsorción Enzimática , Exotoxinas/biosíntesis , Exotoxinas/genética , Femenino , Citometría de Flujo , Proteínas Hemolisinas/biosíntesis , Proteínas Hemolisinas/genética , Histocitoquímica , Subtipo H1N1 del Virus de la Influenza A/crecimiento & desarrollo , Interferón gamma/biosíntesis , Interferón gamma/genética , Leucocidinas/biosíntesis , Leucocidinas/genética , Pulmón/microbiología , Pulmón/patología , Ratones , Ratones Endogámicos BALB C , Infecciones por Orthomyxoviridae/fisiopatología , Neumonía Estafilocócica/microbiología , Neumonía Estafilocócica/fisiopatología , Proteína Estafilocócica A/biosíntesis , Proteína Estafilocócica A/genética , Staphylococcus aureus/genética , Staphylococcus aureus/metabolismo , Estadísticas no Paramétricas
6.
Semin Oncol ; 31(6 Suppl 13): 5-15, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15717735

RESUMEN

Liposomes, closed vesicular structures consisting of one or more lipid bilayers, have generated a great deal of interest as drug delivery vehicles. In particular, they have been investigated for their ability to improve the delivery of chemotherapeutic agents to tumors, in efforts to increase therapeutic efficacy and decrease toxicity to normal cells. Development of liposomal chemotherapeutic agents has, in the past, been hindered primarily by the rapid uptake of liposomes by the reticuloendothelial system. Numerous strategies that seek to either exploit or avoid this phenomenon have been used. As a result, several liposomal chemotherapeutic agents are now available in the clinic. STEALTH, a novel liposomal system coated with polyethylene glycol, avoids uptake by the reticuloendothelial system, thus improving drug delivery to the tumor while decreasing toxicity. In pegylated liposomal doxorubicin (Doxil/Caelyx [PLD]), this delivery system encapsulates doxorubicin within polyethylene glycol-coated liposomes, leading to promising new applications for a well-established drug. Liposome-encapsulated doxorubicin citrate complex (Myocet [NPLD]), another liposomal delivery system for doxorubicin, lacks the polyethylene glycol coating, resulting in much shorter circulation times than those of PLD. Daunorubicin citrate liposome (DaunoXome [DNX]) contains daunorubicin encapsulated in a smaller liposome of a different lipid composition. It has circulation times between those of PLD and NPLD. This article reviews the advantages of liposomal delivery systems in general and the divergent approaches that have been taken in developing these agents.


Asunto(s)
Antraciclinas/administración & dosificación , Antibióticos Antineoplásicos/administración & dosificación , Antraciclinas/farmacocinética , Antibióticos Antineoplásicos/farmacocinética , Humanos , Liposomas/farmacocinética
7.
Semin Oncol ; 31(6 Suppl 13): 196-205, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15717745

RESUMEN

Nanoscale drug delivery systems including liposomes, polymers, and other nanoparticles provide potential solutions for improved cancer therapeutics. Of these drug delivery systems, liposome-based agents, particularly liposomal anthracyclines, have had the greatest impact in oncology to date. Current liposomal drugs evolved from a number of design strategies for improved biodistribution over free drugs. Reticuloendothelial system-targeted formulations significantly reduce systemic exposure to high peak levels of free drug, but do not facilitate targeting to tumors. Passive or physiologic targeting of drugs to tumors is achievable using long-circulating liposomes, including pure lipid systems as well as surface-modified formulations designed to resist recognition and uptake by reticuloendothelial system cells. The latter, represented by pegylated or STEALTH liposomes, circulate for days as stable constructs and slowly extravasate in neoangiogenic vessels in tumors, providing a degree of passive targeting to tumor tissue. Future liposome therapeutics are building on these validated designs as well as on pharmacologic insights into their mechanisms of delivery. For example, camptothecin analogues, anti-angiogenesis agents, and antisense oligonucleotides each represent rational candidates for delivery in highly stabilized and long-circulating liposomes. For such agents, pegylated liposome delivery offers improved chemical stability of encapsulated drug, enhanced accumulation in tumors, and prolonged drug exposure. True molecular targeting can be achieved using liposomes linked to ligands such as monoclonal antibody fragments directed against cancer-associated antigens. Immunoliposomes combine antibody-mediated tumor recognition with liposomal delivery and, when designed for target cell internalization, provide intracellular drug release. Recent advances in immunoliposome design include rapid selection of phage antibody-derived scFv for targeting, and methods for conjugation of ligands to existing US Food and Drug Administration-approved liposomal drugs such as pegylated liposomal doxorubicin (Doxil/Caelxy [PLD]). An immunoliposome consisting of novel anti-HER2 scFv F5 conjugated to PLD, currently in development, selectively binds to and internalizes in HER2-overexpressing tumor cells. The modular organization of immunoliposome technology enables a combinatorial approach in which a repertoire of monoclonal antibody fragments can be used in conjunction with a series of liposomal drugs to yield a new generation of molecularly targeted agents.


Asunto(s)
Antineoplásicos/administración & dosificación , Inmunoconjugados , Liposomas , Anticuerpos Monoclonales , Humanos , Fragmentos de Inmunoglobulinas , Oligonucleótidos Antisentido , Receptor ErbB-2 , Proteínas Recombinantes
8.
Semin Oncol ; 31(6 Suppl 13): 16-35, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15717736

RESUMEN

Encapsulation of doxorubicin in polyethylene glycol-coated liposomes (Doxil/Caelyx [PLD]), was developed to enhance the safety and efficacy of conventional doxorubicin. The liposomes alter pharmacologic and pharmacokinetic parameters of conventional doxorubicin so that drug delivery to the tumor is enhanced while toxicity normally associated with conventional doxorubicin is decreased. In animals and humans, pharmacokinetic advantages of PLD include an increased area under the plasma concentration-time curve, longer distribution half-life, smaller volume of distribution, and reduced clearance. In preclinical models, PLD produced remission and cure against many cancers including tumors of the breast, lung, ovaries, prostate, colon, bladder, and pancreas, as well as lymphoma, sarcoma, and myeloma. It was also found to be effective as adjuvant therapy. In addition, it was found to cross the blood-brain barrier and induce remission in tumors of the central nervous system. Increased potency over conventional doxorubicin was observed and, in contrast to conventional doxorubicin, PLD was equally effective against low- and high-growth fraction tumors. The combination of PLD with vincristine or trastuzumab resulted in additive effects and possible synergy. PLD appeared to overcome multidrug resistance, possibly as the result of increased intracellular concentrations and an interaction between the liposome and P-glycoprotein function. On the basis of pharmacokinetic and preclinical studies, PLD, either alone or as part of combination therapy, has potential applications to treat a variety of cancers.


Asunto(s)
Antibióticos Antineoplásicos/administración & dosificación , Doxorrubicina/administración & dosificación , Animales , Antibióticos Antineoplásicos/farmacocinética , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales Humanizados , Protocolos de Quimioterapia Combinada Antineoplásica/farmacología , Cetuximab , Doxorrubicina/farmacocinética , Evaluación Preclínica de Medicamentos , Resistencia a Antineoplásicos , Sinergismo Farmacológico , Humanos , Liposomas/farmacocinética , Modelos Animales , Trastuzumab
9.
Semin Oncol ; 31(6 Suppl 13): 161-81, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15717742

RESUMEN

Conventional anthracyclines are active against many tumor types, but cardiotoxicity related to the cumulative dose may limit their use; this is particularly problematic for patients with risk factors for increased toxicity, for those who have received any anthracycline in the past, or for those who are to receive other cardiotoxic agents. Preclinical studies determined that encapsulating conventional anthracyclines in liposomes reduced the incidence and severity of cumulative dose-related cardiomyopathy while preserving antitumor activity. In controlled clinical trials, the risk of cardiotoxicity was significantly lower when nonpegylated liposomal doxorubicin (Myocet [NPLD]) was substituted for conventional doxorubicin, but the risk was not significantly different when NPLD was used in place of conventional epirubicin. Direct comparisons to conventional doxorubicin therapy showed comparable efficacy but significantly lower risk of cardiotoxicity with pegylated liposomal doxorubicin (Doxil/Caelyx [PLD]) therapy. Retrospective and prospective trials have not identified a maximum "cardiac safe" dose of PLD, despite use of cumulative doses exceeding 2,000 mg/m2 in some patients. Liposomal daunorubicin (DaunoXome [DNX]) may be associated with a lower risk of cardiotoxicity than conventional anthracyclines, but comparative trials are not available. With respect to combination chemotherapy, early results of clinical trials suggest that combining trastuzumab or a taxane with NPLD or PLD instead of a conventional anthracycline significantly reduces cardiotoxicity risk without reducing chemotherapeutic efficacy. Further results are eagerly awaited from ongoing controlled trials of cardiac safety with long-term liposomal anthracycline therapy, either alone or in combination with other potentially cardiotoxic agents.


Asunto(s)
Antraciclinas/administración & dosificación , Antraciclinas/efectos adversos , Antibióticos Antineoplásicos/administración & dosificación , Antibióticos Antineoplásicos/efectos adversos , Cardiomiopatías/inducido químicamente , Corazón/efectos de los fármacos , Protocolos de Quimioterapia Combinada Antineoplásica/uso terapéutico , Cardiotónicos/administración & dosificación , Ensayos Clínicos como Asunto , Daunorrubicina/administración & dosificación , Daunorrubicina/efectos adversos , Doxorrubicina/administración & dosificación , Doxorrubicina/efectos adversos , Humanos , Liposomas
10.
mBio ; 2(3): e00016-11, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21586648

RESUMEN

UNLABELLED: The mucosal epithelium is the initial target for respiratory pathogens of all types. While type I interferon (IFN) signaling is traditionally associated with antiviral immunity, we demonstrate that the extracellular bacterial pathogen Streptococcus pneumoniae activates the type I IFN cascade in airway epithelial and dendritic cells. This response is dependent upon the pore-forming toxin pneumolysin. Pneumococcal DNA activates IFN-ß expression through a DAI/STING/TBK1/IRF3 cascade. Tlr4(-/-), Myd88(-/-), Trif(-/-), and Nod2(-/-) mutant mice had no impairment of type I IFN signaling. Induction of type I IFN signaling contributes to the eradication of pneumococcal carriage, as IFN-α/ß receptor null mice had significantly increased nasal colonization with S. pneumoniae compared with that of wild-type mice. These studies suggest that the type I IFN cascade is a central component of the mucosal response to airway bacterial pathogens and is responsive to bacterial pathogen-associated molecular patterns that are capable of accessing intracellular receptors. IMPORTANCE: The bacterium Streptococcus pneumoniae is a leading cause of bacterial pneumonia, leading to upwards of one million deaths a year worldwide and significant economic burden. Although it is known that antibody is critical for efficient phagocytosis, it is not known how this pathogen is sensed by the mucosal epithelium. We demonstrate that this extracellular pathogen activates mucosal signaling typically activated by viral pathogens via the pneumolysin pore to activate intracellular receptors and the type I interferon (IFN) cascade. Mice lacking the receptor to type I IFNs have a reduced ability to clear S. pneumoniae, suggesting that the type I IFN cascade is central to the mucosal clearance of this important pathogen.


Asunto(s)
ADN Bacteriano/inmunología , Interferón Tipo I/inmunología , Interferón Tipo I/metabolismo , Sistema Respiratorio/inmunología , Transducción de Señal , Streptococcus pneumoniae/inmunología , Animales , Portador Sano/inmunología , Portador Sano/microbiología , Células Cultivadas , Células Dendríticas/inmunología , Modelos Animales de Enfermedad , Células Epiteliales/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Mucosa Nasal/microbiología , Infecciones Neumocócicas/inmunología , Infecciones Neumocócicas/microbiología , Sistema Respiratorio/microbiología , Streptococcus pneumoniae/genética
11.
J Clin Invest ; 119(7): 1931-9, 2009 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-19603548

RESUMEN

The activation of type I IFN signaling is a major component of host defense against viral infection, but it is not typically associated with immune responses to extracellular bacterial pathogens. Using mouse and human airway epithelial cells, we have demonstrated that Staphylococcus aureus activates type I IFN signaling, which contributes to its virulence as a respiratory pathogen. This response was dependent on the expression of protein A and, more specifically, the Xr domain, a short sequence-repeat region encoded by DNA that consists of repeated 24-bp sequences that are the basis of an internationally used epidemiological typing scheme. Protein A was endocytosed by airway epithelial cells and subsequently induced IFN-beta expression, JAK-STAT signaling, and IL-6 production. Mice lacking IFN-alpha/beta receptor 1 (IFNAR-deficient mice), which are incapable of responding to type I IFNs, were substantially protected against lethal S. aureus pneumonia compared with wild-type control mice. The profound immunological consequences of IFN-beta signaling, particularly in the lung, may help to explain the conservation of multiple copies of the Xr domain of protein A in S. aureus strains and the importance of protein A as a virulence factor in the pathogenesis of staphylococcal pneumonia.


Asunto(s)
Interferón Tipo I/inmunología , Staphylococcus aureus Resistente a Meticilina/inmunología , Staphylococcus aureus Resistente a Meticilina/patogenicidad , Transducción de Señal , Proteína Estafilocócica A/inmunología , Animales , Humanos , Interferón Tipo I/metabolismo , Ratones , Ratones Endogámicos C57BL , Estructura Terciaria de Proteína , Receptor de Interferón alfa y beta/inmunología , Secuencias Repetitivas de Aminoácido , Factor de Transcripción STAT3/metabolismo , Proteína Estafilocócica A/química
12.
J Immunol ; 180(7): 4986-93, 2008 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-18354224

RESUMEN

The innate immune response to inhaled bacteria, such as the opportunist Pseudomonas aeruginosa, is initiated by TLR2 displayed on the apical surface of airway epithelial cells. Activation of TLR2 is accompanied by an immediate Ca(2+) flux that is both necessary and sufficient to stimulate NF-kappaB and MAPK proinflammatory signaling to recruit and activate polymorphonuclear leukocytes in the airway. In human airway cells, gap junction channels were found to provide a regulated conduit for the movement of Ca(2+) from cell to cell. In response to TLR2 stimulation, by either lipid agonists or P. aeruginosa, gap junctions functioned to transiently amplify proinflammatory signaling by communicating Ca(2+) fluxes from stimulated to adjacent, nonstimulated cells thus increasing epithelial CXCL8 production. P. aeruginosa stimulation also induced tyrosine phosphorylation of connexin 43 and association with c-Src, events linked to the closure of these channels. By 4 h postbacterial stimulation, gap junction communication was decreased indicating an autoregulatory control of the connexins. Thus, gap junction channels comprised of connexin 43 and other connexins in airway cells provide a mechanism to coordinate and regulate the epithelial immune response even in the absence of signals from the immune system.


Asunto(s)
Comunicación Celular , Uniones Comunicantes/metabolismo , Sistema Respiratorio/metabolismo , Receptor Toll-Like 2/metabolismo , Animales , Calcio/metabolismo , Línea Celular , Conexinas/metabolismo , Citocinas/metabolismo , Humanos , Ratones , Fosforilación , Transducción de Señal
13.
J Biol Chem ; 280(4): 2737-44, 2005 Jan 28.
Artículo en Inglés | MEDLINE | ID: mdl-15550386

RESUMEN

Skeletal muscle size is regulated by anabolic (hypertrophic) and catabolic (atrophic) processes. We first characterized molecular markers of both hypertrophy and atrophy and identified a small subset of genes that are inversely regulated in these two settings (e.g. up-regulated by an inducer of hypertrophy, insulin-like growth factor-1 (IGF-1), and down-regulated by a mediator of atrophy, dexamethasone). The genes identified as being inversely regulated by atrophy, as opposed to hypertrophy, include the E3 ubiquitin ligase MAFbx (also known as atrogin-1). We next sought to investigate the mechanism by which IGF-1 inversely regulates these markers, and found that the phosphatidylinositol 3-kinase/Akt/mammalian target of rapamycin (PI3K/Akt/mTOR) pathway, which we had previously characterized as being critical for hypertrophy, is also required to be active in order for IGF-1-mediated transcriptional changes to occur. We had recently demonstrated that the IGF1/PI3K/Akt pathway can block dexamethasone-induced up-regulation of the atrophy-induced ubiquitin ligases MuRF1 and MAFbx by blocking nuclear translocation of a FOXO transcription factor. In the current study we demonstrate that an additional step of IGF1 transcriptional regulation occurs downstream of mTOR, which is independent of FOXO. Thus both the Akt/FOXO and the Akt/mTOR pathways are required for the transcriptional changes induced by IGF-1.


Asunto(s)
Regulación Enzimológica de la Expresión Génica , Factor I del Crecimiento Similar a la Insulina/fisiología , Fosfatidilinositol 3-Quinasas/metabolismo , Proteínas Quinasas/metabolismo , Transporte Activo de Núcleo Celular , Animales , Atrofia , Diferenciación Celular , Línea Celular , Núcleo Celular/metabolismo , Dexametasona/farmacología , Relación Dosis-Respuesta a Droga , Regulación hacia Abajo , Regulación de la Expresión Génica , Glucocorticoides/metabolismo , Immunoblotting , Factor I del Crecimiento Similar a la Insulina/genética , Ratones , Proteínas Musculares/metabolismo , Análisis de Secuencia por Matrices de Oligonucleótidos , Fenotipo , Proteínas Serina-Treonina Quinasas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Proteínas Ligasas SKP Cullina F-box/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR , Factores de Tiempo , Transcripción Genética , Ubiquitina-Proteína Ligasas/metabolismo
14.
Exp Neurol ; 187(2): 388-402, 2004 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15144865

RESUMEN

Vascular endothelial growth factor (VEGF) has been shown to induce angiogenesis when infused continuously into adult rat brain tissue. In addition, VEGF has been shown to enhance permeability in brain vasculature. Adult rats were continuously infused with mouse VEGF into neocortex for up to 7 days. We studied the development of VEGF-induced vasculature in rat neocortex and evaluated the temporal expression of a wide variety of markers for inflammation and vascular leak in relation to the angiogenic response using immunohistochemistry and Western blot analysis. We report here that VEGF-mediated inflammation in brain is characterized by upregulation of ICAM-1 and the chemokine MIP-1alpha, as well as a preferential extravasation of monocytes. VEGF causes a dramatic breakdown of the blood-brain barrier, which is characterized by decreased investment of the vasculature with astroglial endfeet. Perivascular cells, in contrast, increase around the newly formed cerebrovasculature. In addition, breakdown of the blood-brain barrier, leukocyte extravasation, and extracellular matrix deposition occur before vascular proliferation. Furthermore, administration of low doses of VEGF induces permeability and inflammation without appreciable vascular proliferation.


Asunto(s)
Encéfalo/irrigación sanguínea , Encéfalo/efectos de los fármacos , Encefalitis/inducido químicamente , Mediadores de Inflamación/farmacología , Neovascularización Fisiológica/efectos de los fármacos , Factor A de Crecimiento Endotelial Vascular/farmacología , Animales , Astrocitos/efectos de los fármacos , Astrocitos/patología , Barrera Hematoencefálica/efectos de los fármacos , Western Blotting , Encéfalo/patología , Permeabilidad Capilar/efectos de los fármacos , División Celular/efectos de los fármacos , Quimiocinas/genética , Relación Dosis-Respuesta a Droga , Vías de Administración de Medicamentos , Encefalitis/metabolismo , Encefalitis/patología , Hibridación in Situ , Mediadores de Inflamación/administración & dosificación , Molécula 1 de Adhesión Intercelular/metabolismo , Antígeno Ki-67/metabolismo , Masculino , Neocórtex/irrigación sanguínea , Neocórtex/efectos de los fármacos , Neocórtex/patología , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Factor A de Crecimiento Endotelial Vascular/administración & dosificación
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA