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.
Antibiotics (Basel) ; 12(8)2023 Aug 19.
Artículo en Inglés | MEDLINE | ID: mdl-37627757

RESUMEN

Francisella tularensis subspecies tularensis is a category-A biothreat agent that can cause lethal tularemia. Ceftobiprole medocaril is being explored as a medical countermeasure for the treatment of pneumonic tularemia. The efficacy of ceftobiprole medocaril against inhalational tularemia was evaluated in the Fischer 344 rat model of infection. The dose was expected to be effective against F. tularensis isolates with ceftobiprole minimum inhibitory concentrations ≤0.5 µg/mL. Animals treated with ceftobiprole medocaril exhibited a 92% survival rate 31 days post-challenge, identical to the survival of levofloxacin-treated rats. By comparison, rats receiving placebo experienced 100% mortality. Terminally collected blood, liver, lung, and spleen samples confirmed disseminated F. tularensis infections in most animals that died prior to completing treatments (placebo animals and a rat treated with ceftobiprole medocaril), although levels of bacteria detected in the placebo samples were significantly elevated compared to the ceftobiprole-medocaril-treated group geometric mean. Furthermore, no evidence of infection was detected in any rat that completed ceftobiprole medocaril or levofloxacin treatment and survived to the end of the post-treatment observation period. Overall, survival rates, body weights, and bacterial burdens consistently demonstrated that treatment with ceftobiprole medocaril is efficacious against otherwise fatal cases of pneumonic tularemia in the rat model.

2.
Sci Rep ; 13(1): 4047, 2023 03 10.
Artículo en Inglés | MEDLINE | ID: mdl-36899021

RESUMEN

Melioidosis is an endemic disease in numerous tropical regions. Additionally, the bacterium that causes melioidosis, Burkholderia pseudomallei, has potential to be used as a biological weapon. Therefore, development of effective and affordable medical countermeasures to serve regions affected by the disease and to have medical countermeasures available in the event of a bioterrorism attack remains critical. The current study evaluated the efficacy of eight distinct acute phase ceftazidime treatment regimens administered therapeutically in the murine model. At the conclusion of the treatment period, survival rates were significantly greater in several of the treated groups when compared to the control group. Pharmacokinetics of a single dose of ceftazidime were examined at 150 mg/kg, 300 mg/kg, and 600 mg/kg and were compared to an intravenous clinical dose administered at 2000 mg every eight hours. The clinical dose has an estimated 100% fT > 4*MIC which exceeded the highest murine dose of 300 mg/kg every six hours at 87.2% fT > 4*MIC. Based upon survival at the end of the treatment regimen and supplemented by pharmacokinetic modeling, a daily dose of 1200 mg/kg of ceftazidime, administered every 6 h at 300 mg/kg, provides protection in the acute phase of inhalation melioidosis in the murine model.


Asunto(s)
Burkholderia pseudomallei , Melioidosis , Animales , Ratones , Ceftazidima/farmacología , Melioidosis/microbiología , Modelos Animales de Enfermedad , Aerosoles/farmacología , Antibacterianos/farmacología
3.
Artículo en Inglés | MEDLINE | ID: mdl-29133571

RESUMEN

The recommended management of inhalational anthrax, a high-priority bioterrorist threat, includes antibiotics and antitoxins. Obiltoxaximab, a chimeric monoclonal antibody against anthrax protective antigen (PA), is licensed under the U.S. Food and Drug Administration's (FDA's) Animal Rule for the treatment of inhalational anthrax. Because of spore latency, disease reemergence after treatment cessation is a concern, and there is a need to understand the development of endogenous protective immune responses following antitoxin-containing anthrax treatment regimens. Here, acquired protective immunity was examined in New Zealand White (NZW) rabbits challenged with a targeted lethal dose of Bacillus anthracis spores and treated with antibiotics, obiltoxaximab, or a combination of both. Survivors of the primary challenge were rechallenged 9 months later and monitored for survival. Survival rates after primary and rechallenge for controls and animals treated with obiltoxaximab, levofloxacin, or a combination of both were 0, 65, 100, and 95%, and 0, 100, 95, and 89%, respectively. All surviving immune animals had circulating antibodies to PA and serum toxin-neutralizing titers prior to rechallenge. Following rechallenge, systemic bacteremia and toxemia were not detected in most animals, and the levels of circulating anti-PA IgG titers increased starting at 5 days postrechallenge. We conclude that treatment with obiltoxaximab, alone or combined with antibiotics, significantly improves the survival of rabbits that received a lethal inhalation B. anthracis spore challenge dose and does not interfere with the development of immunity. Survivors of primary challenge are protected against reexposure, have rare incidents of systemic bacteremia and toxemia, and have evidence of an anamnestic response.


Asunto(s)
Carbunco , Antibacterianos , Anticuerpos Monoclonales , Antitoxinas , Bacillus anthracis , Levofloxacino , Infecciones del Sistema Respiratorio , Esporas Bacterianas , Animales , Femenino , Masculino , Conejos , Carbunco/inmunología , Carbunco/microbiología , Carbunco/mortalidad , Carbunco/prevención & control , Antibacterianos/farmacología , Anticuerpos Antibacterianos/biosíntesis , Anticuerpos Monoclonales/farmacología , Antígenos Bacterianos/sangre , Antígenos Bacterianos/inmunología , Antitoxinas/farmacología , Bacillus anthracis/efectos de los fármacos , Bacillus anthracis/inmunología , Bacillus anthracis/patogenicidad , Toxinas Bacterianas/antagonistas & inhibidores , Toxinas Bacterianas/sangre , Toxinas Bacterianas/inmunología , Quimioterapia Combinada , Inmunización Pasiva/métodos , Inmunoglobulina G/biosíntesis , Memoria Inmunológica/efectos de los fármacos , Levofloxacino/farmacología , Distribución Aleatoria , Infecciones del Sistema Respiratorio/inmunología , Infecciones del Sistema Respiratorio/microbiología , Infecciones del Sistema Respiratorio/mortalidad , Infecciones del Sistema Respiratorio/prevención & control , Esporas Bacterianas/efectos de los fármacos , Esporas Bacterianas/inmunología , Esporas Bacterianas/patogenicidad , Análisis de Supervivencia
4.
Artículo en Inglés | MEDLINE | ID: mdl-28784679

RESUMEN

The fluorocycline TP-271 was evaluated in mouse and nonhuman primate (NHP) models of inhalational anthrax. BALB/c mice were exposed by nose-only aerosol to Bacillus anthracis Ames spores at a level of 18 to 88 lethal doses sufficient to kill 50% of exposed individuals (LD50). When 21 days of once-daily dosing was initiated at 24 h postchallenge (the postexposure prophylaxis [PEP] study), the rates of survival for the groups treated with TP-271 at 3, 6, 12, and 18 mg/kg of body weight were 90%, 95%, 95%, and 84%, respectively. When 21 days of dosing was initiated at 48 h postchallenge (the treatment [Tx] study), the rates of survival for the groups treated with TP-271 at 6, 12, and 18 mg/kg TP-271 were 100%, 91%, and 81%, respectively. No deaths of TP-271-treated mice occurred during the 39-day posttreatment observation period. In the NHP model, cynomolgus macaques received an average dose of 197 LD50 of B. anthracis Ames spore equivalents using a head-only inhalation exposure chamber, and once-daily treatment of 1 mg/kg TP-271 lasting for 14 or 21 days was initiated within 3 h of detection of protective antigen (PA) in the blood. No (0/8) animals in the vehicle control-treated group survived, whereas all 8 infected macaques treated for 21 days and 4 of 6 macaques in the 14-day treatment group survived to the end of the study (56 days postchallenge). All survivors developed toxin-neutralizing and anti-PA IgG antibodies, indicating an immunologic response. On the basis of the results obtained with the mouse and NHP models, TP-271 shows promise as a countermeasure for the treatment of inhalational anthrax.


Asunto(s)
Carbunco/tratamiento farmacológico , Antibacterianos/uso terapéutico , Bacillus anthracis/efectos de los fármacos , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Tetraciclinas/uso terapéutico , Animales , Carbunco/microbiología , Carbunco/mortalidad , Anticuerpos Antibacterianos/sangre , Anticuerpos Antibacterianos/inmunología , Anticuerpos Neutralizantes/sangre , Anticuerpos Neutralizantes/inmunología , Antígenos Bacterianos/inmunología , Bacillus anthracis/inmunología , Toxinas Bacterianas/inmunología , Modelos Animales de Enfermedad , Femenino , Inmunoglobulina G/sangre , Inmunoglobulina G/inmunología , Macaca fascicularis , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Profilaxis Posexposición/métodos , Infecciones del Sistema Respiratorio/microbiología , Infecciones del Sistema Respiratorio/mortalidad , Esporas Bacterianas , Tasa de Supervivencia , Tetraciclinas/farmacocinética
5.
Artículo en Inglés | MEDLINE | ID: mdl-28559261

RESUMEN

TP-271 is a novel, fully synthetic fluorocycline in development for complicated bacterial respiratory infections. TP-271 was active in vitro against a panel of 29 Francisella tularensis isolates, showing MICs against 50% and 90% of isolates of 0.25 and 0.5 µg/ml, respectively. In a mouse model of inhalational tularemia, animals were exposed by aerosol to 91 to 283 50% lethal doses (LD50)/mouse of F. tularensis SCHU S4. Following 21 days of once-daily intraperitoneal dosing with TP-271 at 3, 6, 12, and 18 mg/kg of body weight/day, initiating at 24 h postchallenge, survival was 80%, 100%, 100%, and 100%, respectively. When treatment was initiated at 72 h postchallenge, survival was 89%, 100%, 100%, and 100% in the 3-, 6-, 12-, and 18-mg/kg/day TP-271 groups, respectively. No mice treated with the vehicle control survived. Surviving mice treated with TP-271 showed little to no relapse during 14 days posttreatment. In a nonhuman primate model of inhalational tularemia, cynomolgus macaques received an average aerosol exposure of 1,144 CFU of F. tularensis SCHU S4. Once-daily intravenous infusion with 1 or 3 mg/kg TP-271, or vehicle control, for 21 days was initiated within 6 h of confirmed fever. All animals treated with TP-271 survived to the end of the study, with no relapse during 14 days after the last treatment, whereas no vehicle control-treated animals survived. The protection and low relapse afforded by TP-271 treatment in these studies support continued investigation of TP-271 for use in the event of aerosolized exposure to F. tularensis.


Asunto(s)
Antibacterianos/uso terapéutico , Francisella tularensis/efectos de los fármacos , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Tetraciclinas/uso terapéutico , Tularemia/tratamiento farmacológico , Animales , Modelos Animales de Enfermedad , Femenino , Macaca fascicularis , Masculino , Ratones , Ratones Endogámicos BALB C , Pruebas de Sensibilidad Microbiana , Infecciones del Sistema Respiratorio/microbiología , Tularemia/microbiología
6.
Antimicrob Agents Chemother ; 60(10): 5796-805, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27431219

RESUMEN

The Centers for Disease Control and Prevention recommend adjunctive antitoxins when systemic anthrax is suspected. Obiltoxaximab, a monoclonal antibody against protective antigen (PA), is approved for treatment of inhalational anthrax in combination with antibiotics and for prophylaxis when alternative therapies are not available. The impact of toxin neutralization with obiltoxaximab during pre- and postexposure prophylaxis was explored, and efficacy results that supported the prophylaxis indication are presented here. New Zealand White rabbits and cynomolgus macaques received obiltoxaximab as a single intramuscular or intravenous dose of 2 to 16 mg/kg of body weight at various times relative to Bacillus anthracis aerosol spore challenge. The primary endpoint was survival, and effect of treatment timing was explored. In rabbits, obiltoxaximab administration 9 h postchallenge singly or combined with a 5-day levofloxacin regimen protected 89% to 100% of animals compared to 33% with levofloxacin monotherapy. In cynomolgus macaques, a single intramuscular dose of 16 mg/kg obiltoxaximab led to 100% survival when given 1 to 3 days preexposure and 83% to 100% survival when given 18 to 24 h postexposure and prior to systemic bacteremia onset. Obiltoxaximab administration after bacteremia onset resulted in lower (25% to 50%) survival rates reflective of treatment setting. Prophylactic administration of obiltoxaximab before spore challenge or to spore-challenged animals before systemic bacterial dissemination is efficacious in promoting survival, ameliorating toxemia, and inhibiting bacterial spread to the periphery.


Asunto(s)
Carbunco/mortalidad , Carbunco/prevención & control , Anticuerpos Monoclonales/farmacología , Antitoxinas/farmacología , Bacillus anthracis/patogenicidad , Infecciones del Sistema Respiratorio/mortalidad , Infecciones del Sistema Respiratorio/prevención & control , Animales , Carbunco/tratamiento farmacológico , Antibacterianos/administración & dosificación , Antibacterianos/farmacocinética , Antibacterianos/farmacología , Anticuerpos Monoclonales/administración & dosificación , Anticuerpos Monoclonales/farmacocinética , Antitoxinas/administración & dosificación , Bacillus anthracis/efectos de los fármacos , Bacteriemia/tratamiento farmacológico , Bacteriemia/microbiología , Modelos Animales de Enfermedad , Femenino , Inyecciones Intramusculares , Inyecciones Intravenosas , Macaca fascicularis , Masculino , Profilaxis Posexposición , Profilaxis Pre-Exposición , Conejos , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Tasa de Supervivencia
7.
Antimicrob Agents Chemother ; 60(10): 5787-95, 2016 10.
Artículo en Inglés | MEDLINE | ID: mdl-27431222

RESUMEN

Inhalational anthrax has high mortality even with antibiotic treatment, and antitoxins are now recommended as an adjunct to standard antimicrobial regimens. The efficacy of obiltoxaximab, a monoclonal antibody against anthrax protective antigen (PA), was examined in multiple studies conducted in two animal models of inhalational anthrax. A single intravenous bolus of 1 to 32 mg/kg of body weight obiltoxaximab or placebo was administered to New Zealand White rabbits (two studies) and cynomolgus macaques (4 studies) at disease onset (significant body temperature increase or detection of serum PA) following lethal challenge with aerosolized Bacillus anthracis spores. The primary endpoint was survival. The relationship between efficacy and disease severity, defined by pretreatment bacteremia and toxemia levels, was explored. In rabbits, single doses of 1 to 16 mg/kg obiltoxaximab led to 17 to 93% survival. In two studies, survival following 16 mg/kg obiltoxaximab was 93% and 62% compared to 0% and 0% for placebo (P = 0.0010 and P = 0.0013, respectively). Across four macaque studies, survival was 6.3% to 78.6% following 4 to 32 mg/kg obiltoxaximab. In two macaque studies, 16 mg/kg obiltoxaximab reduced toxemia and led to survival rates of 31%, 35%, and 47% versus 0%, 0%, and 6.3% with placebo (P = 0.0085, P = 0.0053, P = 0.0068). Pretreatment bacteremia and toxemia levels inversely correlated with survival. Overall, obiltoxaximab monotherapy neutralized PA and increased survival across the range of disease severity, indicating clinical benefit of toxin neutralization with obiltoxaximab in both early and late stages of inhalational anthrax.


Asunto(s)
Carbunco/tratamiento farmacológico , Antibacterianos/farmacología , Anticuerpos Monoclonales/farmacología , Antitoxinas/farmacología , Infecciones del Sistema Respiratorio/tratamiento farmacológico , Animales , Carbunco/etiología , Carbunco/mortalidad , Antibacterianos/farmacocinética , Anticuerpos Monoclonales/farmacocinética , Femenino , Macaca fascicularis , Masculino , Conejos , Infecciones del Sistema Respiratorio/etiología , Infecciones del Sistema Respiratorio/mortalidad , Tasa de Supervivencia , Resultado del Tratamiento
8.
Clin Vaccine Immunol ; 19(11): 1765-75, 2012 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-22956657

RESUMEN

Appropriate animal models are required to test medical countermeasures to bioterrorist threats. To that end, we characterized a nonhuman primate (NHP) inhalational anthrax therapeutic model for use in testing anthrax therapeutic medical countermeasures according to the U.S. Food and Drug Administration Animal Rule. A clinical profile was recorded for each NHP exposed to a lethal dose of Bacillus anthracis Ames spores. Specific diagnostic parameters were detected relatively early in disease progression, i.e., by blood culture (∼37 h postchallenge) and the presence of circulating protective antigen (PA) detected by electrochemiluminescence (ECL) ∼38 h postchallenge, whereas nonspecific clinical signs of disease, i.e., changes in body temperature, hematologic parameters (ca. 52 to 66 h), and clinical observations, were delayed. To determine whether the presentation of antigenemia (PA in the blood) was an appropriate trigger for therapeutic intervention, a monoclonal antibody specific for PA was administered to 12 additional animals after the circulating levels of PA were detected by ECL. Seventy-five percent of the monoclonal antibody-treated animals survived compared to 17% of the untreated controls, suggesting that intervention at the onset of antigenemia is an appropriate treatment trigger for this model. Moreover, the onset of antigenemia correlated with bacteremia, and NHPs were treated in a therapeutic manner. Interestingly, brain lesions were observed by histopathology in the treated nonsurviving animals, whereas this observation was absent from 90% of the nonsurviving untreated animals. Our results support the use of the cynomolgus macaque as an appropriate therapeutic animal model for assessing the efficacy of medical countermeasures developed against anthrax when administered after a confirmation of infection.


Asunto(s)
Carbunco/patología , Carbunco/terapia , Modelos Animales de Enfermedad , Enfermedades de los Primates/patología , Enfermedades de los Primates/terapia , Infecciones del Sistema Respiratorio/patología , Infecciones del Sistema Respiratorio/terapia , Animales , Carbunco/diagnóstico , Anticuerpos Antibacterianos/administración & dosificación , Anticuerpos Monoclonales/administración & dosificación , Antígenos Bacterianos/sangre , Toxinas Bacterianas/sangre , Biomarcadores/sangre , Encéfalo/patología , Femenino , Adhesión a Directriz , Macaca fascicularis , Masculino , Enfermedades de los Primates/diagnóstico , Infecciones del Sistema Respiratorio/diagnóstico , Análisis de Supervivencia , Factores de Tiempo , Estados Unidos , United States Food and Drug Administration
9.
Clin Vaccine Immunol ; 19(9): 1517-25, 2012 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-22837095

RESUMEN

The development of an appropriate animal therapeutic model is essential to assess the potential efficacy of therapeutics for use in the event of a Bacillus anthracis exposure. We conducted a natural history study that showed New Zealand White rabbits exhibited a significant increase in body temperature (SIBT), changes in hematologic parameters, and increases in C-reactive protein and succumbed to disease with an average time to death of approximately 73 h following aerosol challenge with B. anthracis Ames spores. The SIBT was used as a trigger to treat with a fully human monoclonal antibody directed at protective antigen (PA). Ninety percent (9/10) of the treated rabbits survived the lethal inhalational challenge of B. anthracis. Further characterization investigated the protective window of opportunity for anti-PA antibody administration up to 12 h post-onset of SIBT. Eighty-three percent (5/6) of the rabbits treated at SIBT and 100% (6/6) of those treated at 6 h after SIBT survived challenge. Only 67% (4/6) of the rabbits treated at 12 h after SIBT survived. The increase in body temperature corresponded with both bacteremia and antigenemia (PA in the blood), indicating that SIBT is a suitable trigger to initiate treatment in a therapeutic model of inhalational anthrax.


Asunto(s)
Carbunco/patología , Carbunco/terapia , Bacillus anthracis/patogenicidad , Biomarcadores , Fiebre/diagnóstico , Animales , Anticuerpos Antibacterianos/administración & dosificación , Anticuerpos Monoclonales/administración & dosificación , Antígenos Bacterianos , Toxinas Bacterianas/antagonistas & inhibidores , Células Sanguíneas/fisiología , Proteína C-Reactiva/análisis , Modelos Animales de Enfermedad , Femenino , Inmunoterapia/métodos , Masculino , Conejos , Análisis de Supervivencia , Factores de Tiempo
10.
Sci Rep ; 2: 495, 2012.
Artículo en Inglés | MEDLINE | ID: mdl-22773944

RESUMEN

To characterize the clinical presentation and pathophysiology of inhalational brucellosis, Balb/c mice were challenged with Brucella melitensis 16M in a nose-only aerosol exposure chamber. A low dose of 1000 cfu/animal of B. melitensis resulted in 45% of mice with tissue burdens eight weeks post-challenge. The natural history of brucellosis in mice challenged by higher aerosol doses was examined by serial euthanizing mice over an eight week period. Higher challenge doses of 1.00E+05 and 5.00E+05 cfu resulted in positive blood cultures 14 days post-challenge and bacterial burdens were observed in the lung, liver and/or spleens 14 days post-challenge. In addition, the progression of brucellosis was similar between mice challenged by the intranasal and aerosol routes. The results from this study support the use of the Balb/c aerosol nose-only brucellosis mouse model for the evaluation of therapeutics against inhalational brucellosis.


Asunto(s)
Brucella melitensis , Brucelosis/microbiología , Brucelosis/patología , Administración por Inhalación , Administración Intranasal , Animales , Temperatura Corporal , Brucella melitensis/patogenicidad , Modelos Animales de Enfermedad , Femenino , Pruebas Hematológicas , Hígado/microbiología , Hígado/patología , Pulmón/microbiología , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Bazo/microbiología , Bazo/patología
11.
Infect Immun ; 80(1): 298-310, 2012 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-22064715

RESUMEN

The objective of this study was to characterize the rhesus macaque (RM) as a model for inhalational brucellosis in support of the U.S. Food and Drug Administration's (FDA) Animal Rule. The pathophysiology of chronic Brucella melitensis aerosol infection was monitored in two phases that each occurred over an 8-week time period; dose escalation (8 RMs; targeted doses of 5.0E+03, 5.0E+04, or 5.0E+05 CFU/animal or the unchallenged control) and natural history (12 RMs; targeted dose of 2.50E+05 CFU/animal or the unchallenged control). RMs given an aerosol challenge with B. melitensis developed undulating fevers (6/6 phase I; 8/9 phase II), positive enriched blood cultures (5/10; phase II), and bacterial burdens in tissues starting 14 to 21 days postchallenge (6/6 phase I; 10/10 phase II). In addition, 80% (8/10; phase II) of infected RMs seroconverted 14 to 21 days postchallenge. RMs developed elevations in certain liver enzymes and had an increased inflammatory response by 3 weeks postchallenge as shown by increases in C-reactive protein (6/8) and neopterin (4/8), which correlated with the onset of a fever. As early as 14 days postchallenge, positive liver biopsy specimens were detected (2/8), and ultrasound imaging showed the development of splenomegaly. Finally, histopathologic examination found lesions attributed to Brucella infection in the liver, kidney, lung, and/or spleen of all animals. The disease progression observed with the RMs in this study is analogous to human brucellosis pathophysiology. Thus, the results from this study support the use of the RM as an animal model for inhalational brucellosis to evaluate the efficacy of novel vaccines and therapeutics against B. melitensis.


Asunto(s)
Brucella melitensis/patogenicidad , Brucelosis/patología , Brucelosis/fisiopatología , Exposición por Inhalación , Enfermedades de los Primates/patología , Enfermedades de los Primates/fisiopatología , Estructuras Animales/microbiología , Estructuras Animales/patología , Animales , Carga Bacteriana , Proteína C-Reactiva/análisis , Modelos Animales de Enfermedad , Enzimas/sangre , Femenino , Fiebre/microbiología , Histocitoquímica , Hígado/enzimología , Hígado/patología , Pruebas de Función Hepática , Macaca mulatta , Masculino , Esplenomegalia/diagnóstico , Factores de Tiempo , Estados Unidos , United States Food and Drug Administration
12.
Am J Respir Cell Mol Biol ; 41(2): 226-36, 2009 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-19131640

RESUMEN

The etiology of acute lung injury is complex and associated with numerous, chemically diverse precipitating factors. During acute lung injury in mice, one key event is epithelial cell injury that leads to reduced surfactant biosynthesis. We have previously reported that transgenic mice that express transforming growth factor alpha (TGFA) in the lung were protected during nickel-induced lung injury. Here, we find that the mechanism by which TGFA imparts protection includes maintenance of surfactant-associated protein B (SFTPB) transcript levels and epidermal growth factor receptor-dependent signaling in distal pulmonary epithelial cells. This protection is complex and not accompanied by a diminution in inflammatory mediator transcripts or additional stimulation of antioxidant transcripts. In mouse lung epithelial (MLE-15) cells, microarray analysis demonstrated that nickel increased transcripts of genes enriched in MTF1, E2F-1, and AP-2 transcription factor-binding sites and decreased transcripts of genes enriched in AP-1-binding sites. Nickel also increased Jun transcript and DNA-binding activity, but decreased SFTPB transcript. Expression of SFTPB under the control of a doxycycline-sensitive promoter increased survival during nickel-induced injury as compared with control mice. Together, these findings support the idea that maintenance of SFTPB expression is critical to survival during acute lung injury.


Asunto(s)
Lesión Pulmonar Aguda/inducido químicamente , Níquel/toxicidad , Proteína B Asociada a Surfactante Pulmonar/metabolismo , Administración por Inhalación , Aerosoles , Animales , Células Cultivadas , Células Epiteliales/citología , Células Epiteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Ratones , Ratones Transgénicos , Regiones Promotoras Genéticas , Proteínas Proto-Oncogénicas c-jun/genética , Proteínas Proto-Oncogénicas c-jun/metabolismo , Proteína B Asociada a Surfactante Pulmonar/genética , Mucosa Respiratoria/citología , Tasa de Supervivencia , Factor de Transcripción AP-1/genética , Factor de Transcripción AP-1/metabolismo , Factor de Crecimiento Transformador alfa/genética , Factor de Crecimiento Transformador alfa/metabolismo
13.
J Immunol ; 180(12): 7847-58, 2008 Jun 15.
Artículo en Inglés | MEDLINE | ID: mdl-18523248

RESUMEN

The pulmonary innate immune system responds to various airborne microbes. Although its specificity is broad and based on the recognition of pathogen-associated molecular patterns, it is uniquely regulated to limit inflammation and thereby prevent damage to the gas-exchanging alveoli. Macrophages, critical cell determinants of this system, recognize microbes through pattern recognition receptors such as TLRs, which typically mediate proinflammatory responses. The lung collectin, surfactant protein A (SP-A), has emerged as an important innate immune determinant that regulates microbe-macrophage interactions in this environment. In this study, we report the basal and SP-A-induced transcriptional and posttranslational regulation of TLR2 and TLR4 expression during the differentiation of primary human monocytes into macrophages. Despite SP-A's ability to up-regulate TLR2 expression on human macrophages, it dampens TLR2 and TLR4 signaling in these cells. SP-A decreases the phosphorylation of IkappaBalpha, a key regulator of NF-kappaB activity, and nuclear translocation of p65 which result in diminished TNF-alpha secretion in response to TLR ligands. SP-A also reduces the phosphorylation of TLR signaling proteins upstream of NF-kappaB, including members of the MAPK family. Finally, we report for the first time that SP-A decreases the phosphorylation of Akt, a major cell regulator of NF-kappaB and potentially MAPKs. These data identify a critical role for SP-A in modulating the lung inflammatory response by regulating macrophage TLR activity.


Asunto(s)
Macrófagos Alveolares/inmunología , Macrófagos Alveolares/metabolismo , Proteína A Asociada a Surfactante Pulmonar/fisiología , Receptores Toll-Like/biosíntesis , Receptores Toll-Like/genética , Adulto , Diferenciación Celular/inmunología , Membrana Celular/inmunología , Membrana Celular/metabolismo , Células Cultivadas , Humanos , Líquido Intracelular/inmunología , Líquido Intracelular/metabolismo , Macrófagos Alveolares/citología , Modelos Inmunológicos , Monocitos/citología , Monocitos/inmunología , Monocitos/metabolismo , Unión Proteica/inmunología , Proteína A Asociada a Surfactante Pulmonar/biosíntesis , Proteína A Asociada a Surfactante Pulmonar/genética , ARN Mensajero/antagonistas & inhibidores , ARN Mensajero/biosíntesis , Receptor Toll-Like 2/antagonistas & inhibidores , Receptor Toll-Like 2/biosíntesis , Receptor Toll-Like 2/metabolismo , Receptor Toll-Like 2/fisiología , Receptor Toll-Like 4/biosíntesis , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 4/fisiología , Receptores Toll-Like/metabolismo , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Factor de Necrosis Tumoral alfa/metabolismo , Regulación hacia Arriba/inmunología
14.
Am J Respir Cell Mol Biol ; 34(1): 73-82, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16166738

RESUMEN

Often fatal, acute lung injury has a complicated etiology. Previous studies from our laboratory in mice have demonstrated that survival during acute lung injury is a complex trait governed by multiple loci. We also found that the increase in metallothionein (MT) is one of the greatest noted in transcriptome-wide analyses of gene expression. To assess the role of MT in nickel-induced acute lung injury, the survival of Mt-transgenic, Mt1/2(+/+), and Mt1/2(-/-) mice was compared. Pulmonary inflammation and global gene expression were compared in Mt1/2(+/+) and Mt1/2(-/-) mice. Gene-targeted Mt1/2(-/-) mice were more susceptible than Mt1/2(+/+) mice to nickel-induced inflammation, surfactant-associated protein B transcript loss, and lethality. Similarly, Mt-transgenic mice exhibited increased survival. MAPPFinder analyses also noted significant decreases in genes involved in protein processing (e.g., ubiquitination, folding), which were greater in Mt1/2(-/-) mice as compared with Mt1/2(+/+) mice early in the progression of acute lung injury, possibly due to a zinc-mediated transcript destabilization. In contrast, transcript levels of genes associated with the inflammatory response, extracellular matrix regulation, and coagulation/fibrinolysis were increased more in Mt1/2(-/-) mice as compared with Mt1/2(+/+) mice late in the development of acute lung injury. Thus, MT ultimately improves survival in the progression of acute lung injury in mice. Transcriptome-wide analysis suggests that this survival may be mediated through changes in the destabilization of transcripts associated with protein processing, the subsequent augmentation of transcripts controlling inflammation, extracellular matrix regulation, coagulation/fibrinolysis, and disruption of surfactant homeostasis.


Asunto(s)
Metalotioneína/metabolismo , Síndrome de Dificultad Respiratoria/genética , Síndrome de Dificultad Respiratoria/fisiopatología , Animales , Progresión de la Enfermedad , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Metalotioneína/genética , Ratones , Ratones Endogámicos , Ratones Noqueados , Níquel/toxicidad , Análisis de Secuencia por Matrices de Oligonucleótidos , Síndrome de Dificultad Respiratoria/inducido químicamente , Síndrome de Dificultad Respiratoria/metabolismo , Tasa de Supervivencia
15.
Am J Respir Crit Care Med ; 172(11): 1399-411, 2005 Dec 01.
Artículo en Inglés | MEDLINE | ID: mdl-16100012

RESUMEN

RATIONALE: Acute lung injury can occur from multiple causes, resulting in high mortality. The pathophysiology of nickel-induced acute lung injury in mice is remarkably complex, and the molecular mechanisms are uncertain. OBJECTIVES: To integrate molecular pathways and investigate the role of transforming growth factor beta (TGF-beta) in acute lung injury in mice. METHODS: cDNA microarray analyses were used to identify lung gene expression changes after nickel exposure. MAPPFinder analysis of the microarray data was used to determine significantly altered molecular pathways. TGF-beta1 protein in bronchoalveolar lavage fluid, as well as the effect of inhibition of TGF-beta, was assessed in nickel-exposed mice. The effect of TGF-beta on surfactant-associated protein B (Sftpb) promoter activity was measured in mouse lung epithelial cells. MEASUREMENTS AND MAIN RESULTS: Genes that decreased the most after nickel exposure play important roles in lung fluid absorption or surfactant and phospholipid synthesis, and genes that increased the most were involved in TGF-beta signaling. MAPPFinder analysis further established TGF-beta signaling to be significantly altered. TGF-beta-inducible genes involved in the regulation of extracellular matrix function and fibrinolysis were significantly increased after nickel exposure, and TGF-beta1 protein was also increased in the lavage fluid. Pharmacologic inhibition of TGF-beta attenuated nickel-induced protein in bronchoalveolar lavage. In addition, treatment with TGF-beta1 dose-dependently repressed Sftpb promoter activity in vitro, and a novel TGF-beta-responsive region in the Sftpb promoter was identified. CONCLUSIONS: These data suggest that TGF-beta acts as a central mediator of acute lung injury through the alteration of several different molecular pathways.


Asunto(s)
Expresión Génica , Proteína B Asociada a Surfactante Pulmonar/genética , ARN/genética , Síndrome de Dificultad Respiratoria/genética , Factor de Crecimiento Transformador beta/metabolismo , Animales , Líquido del Lavado Bronquioalveolar/química , Células Cultivadas , Modelos Animales de Enfermedad , Ensayo de Inmunoadsorción Enzimática , Femenino , Fibrinólisis/efectos de los fármacos , Fibrinólisis/genética , Ratones , Níquel/toxicidad , Análisis de Secuencia por Matrices de Oligonucleótidos , Proteína B Asociada a Surfactante Pulmonar/metabolismo , Síndrome de Dificultad Respiratoria/tratamiento farmacológico , Síndrome de Dificultad Respiratoria/metabolismo , Mucosa Respiratoria/efectos de los fármacos , Mucosa Respiratoria/metabolismo , Mucosa Respiratoria/patología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Factor de Crecimiento Transformador beta/antagonistas & inhibidores , Factor de Crecimiento Transformador beta/uso terapéutico , Factor de Crecimiento Transformador beta1
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...