Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 11 de 11
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Thorac Cardiovasc Surg ; 52(6): 338-43, 2004 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-15573274

RESUMEN

BACKGROUND: We investigated the effects of PARS inhibition on intestinal injury in a canine model of cardiopulmonary bypass (CPB). METHODS: Twelve dogs underwent 90 minutes of hypothermic CPB. 6 dogs received 5 mg/kg PJ34, a selective PARP inhibitor during CPB, 6 vehicle-treated animals served as controls. Mesenteric blood flow (MBF) and mesenteric vascular resistance (MVR) were measured before and 60 minutes after weaning from CPB. Endothelium-dependent vasorelaxation to acetylcholine (ACH) and endothelium-independent vasorelaxation to sodium-nitroprusside (SNP) were expressed as percent change of MVR. In addition, mesenteric creatine kinase (CK) and lactate release were determined. RESULTS: Baseline hemodynamics, MBF, response to ACH (- 41 +/- 3 vs. - 55 +/- 6 %) and SNP (- 60 +/- 2 vs. - 56 +/- 4 %) did not differ significantly between the groups. The response to ACH decreased significantly in the control group while it remained unchanged in the PJ34 group (- 29 +/- 5 vs. - 46 +/- 9 %, p < 0.05). The response to SNP did not change. Mesenteric CK release (325 +/- 99 vs. 16 +/- 10 U/l, p < 0.05) and lactate production (0.96 +/- 0.17 vs. 0.4 +/- 0.2 mmol/l, p < 0.05) were significantly lower in the PJ34 group. CONCLUSION: PARP inhibition prevents CPB-induced mesenteric endothelial dysfunction and tissue damage.


Asunto(s)
Puente Cardiopulmonar/efectos adversos , Inhibidores Enzimáticos/farmacología , Arteria Mesentérica Superior/lesiones , Venas Mesentéricas/lesiones , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Poli(ADP-Ribosa) Polimerasas/efectos de los fármacos , Acetilcolina/farmacología , Animales , Creatina Quinasa/metabolismo , Modelos Animales de Enfermedad , Perros , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/lesiones , Ácido Láctico/metabolismo , Arteria Mesentérica Superior/fisiopatología , Venas Mesentéricas/fisiopatología , Nitroprusiato/farmacología , Poli(ADP-Ribosa) Polimerasas/metabolismo , Flujo Sanguíneo Regional/efectos de los fármacos , Factores de Tiempo , Resistencia Vascular/efectos de los fármacos , Vasodilatación/efectos de los fármacos , Vasodilatadores/farmacología
2.
Blood ; 98(9): 2845-52, 2001 Nov 01.
Artículo en Inglés | MEDLINE | ID: mdl-11675359

RESUMEN

Macrophage colony-stimulating factor (M-CSF) is one of several hematologic growth factors capable of regulating the survival, proliferation, and differentiation of macrophages, but its role in modulation of the accumulation and function of alveolar macrophages (AMs) in vivo is not well defined. Osteopetrotic (Op/Op) mice have no detectable M-CSF and show variable tissue-specific reductions in macrophage numbers. It was hypothesized that AMs would be decreased in number and have altered function in Op/Op mice because of the absence of M-CSF. Lung macrophages identified by Mac-3 staining in lung sections were decreased in number in 20-day-old Op/Op mice (P <.001) but not Op/Op mice older than 4 months (P =.68) compared with findings in age-matched littermate controls. The numbers of AMs recovered by bronchoalveolar lavage (BAL) were also reduced in young but not adult Op/Op mice compared with controls. Expression of interleukin-3 (IL-3) was increased in the lungs of Op/Op mice compared with controls as determined by quantification of IL-3 cytokine levels (P =.04), bioactivity (P =.02), and messenger RNA transcript levels. AMs of Op/Op mice spontaneously released higher levels of matrix metalloproteinases (MMPs) than AMs of controls as determined by immunohistochemical staining of AMs and zymographic assessment of BAL fluid and AM lysates. Consistent with an increased release of MMP, Op/Op mice had abnormal elastin deposition and spontaneously developed emphysema in the absence of molecular or cellular evidence of lung inflammation. These data show that the AM deficiency observed in young Op/Op mice is spontaneously corrected with age and is associated with increased lung levels of IL-3, spontaneous MMP expression by AMs, and destruction of lung tissue.


Asunto(s)
Factor Estimulante de Colonias de Macrófagos/fisiología , Macrófagos Alveolares/enzimología , Macrófagos Alveolares/patología , Metaloproteinasas de la Matriz/metabolismo , Osteopetrosis/patología , Factores de Edad , Animales , Enfisema/enzimología , Enfisema/etiología , Enfisema/patología , Interleucina-3/metabolismo , Pulmón/enzimología , Pulmón/metabolismo , Pulmón/patología , Factor Estimulante de Colonias de Macrófagos/deficiencia , Factor Estimulante de Colonias de Macrófagos/farmacología , Macrófagos Alveolares/efectos de los fármacos , Metaloproteinasas de la Matriz/efectos adversos , Ratones , Ratones Mutantes , Osteopetrosis/genética
3.
Am J Physiol Lung Cell Mol Physiol ; 280(3): L503-11, 2001 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-11159034

RESUMEN

Host inflammatory and immune responses limit viral gene expression after administration of replication-deficient adenoviruses to the lung. The current study asks whether inducible nitric oxide synthase (iNOS) expression and peroxynitrite generation accompanied the inflammatory response following intratracheal administration of adenovirus. Pulmonary iNOS mRNA and protein were increased 2, 7, and 14 days following administration of 2 x 10(9) plaque-forming units of recombinant adenovirus (Av1Luc1) to BALB/c mice. Adenovirus infection was associated with a marked increase in nitrotyrosine staining. Intense nitrotyrosine staining was observed in alveolar macrophages, respiratory epithelial cells, conducting airways, and alveolar spaces 2 days postinfection. Two weeks after exposure to adenovirus, nitrotyrosine staining was detected within alveolar macrophages, suggesting adenovirus enhanced the nitration of proteins that were subsequently taken up by alveolar macrophages. Western blot analysis using anti-nitrotyrosine antibody did not demonstrate accumulation of nitrated surfactant protein A (SP-A), although a small fraction of aggregated SP-A comigrated with a nitrotyrosine-positive protein. iNOS expression, peroxynitrite, and nitrotyrosine generation accompany and may contribute to inflammatory responses to adenovirus in the lung.


Asunto(s)
Infecciones por Adenoviridae/metabolismo , Pulmón/metabolismo , Nitratos/metabolismo , Óxido Nítrico Sintasa/biosíntesis , Tirosina/análogos & derivados , Infecciones por Adenoviridae/patología , Animales , Western Blotting , Líquido del Lavado Bronquioalveolar/química , Femenino , Inmunohistoquímica , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Óxido Nítrico Sintasa de Tipo II , Surfactantes Pulmonares/metabolismo , Tirosina/metabolismo
4.
J Virol ; 74(20): 9655-67, 2000 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-11000238

RESUMEN

Adenovirus is a common respiratory pathogen which causes a broad range of distinct clinical syndromes and has recently received attention for its potential for in vivo gene delivery. Although adenovirus respiratory tract infection (ARTI) results in dose-dependent, local inflammation, the pathogenesis of this remains unclear. We hypothesized that alveolar macrophages (AMphi) rapidly internalize adenovirus following in vivo pulmonary administration and then initiate inflammatory signaling within the lung. To evaluate the role of AMphi in the induction of lung inflammation during ARTI in vivo, we directly assessed adenovirus uptake by murine AMphi and correlated uptake with the initiation of proinflammatory gene expression. Stimulation of cytokine (tumor necrosis factor alpha [TNF-alpha], interleukin-6 [IL-6], macrophage inflammatory protein-2 [MIP-2], and MIP-1alpha) expression in the lung was evaluated at the level of mRNA (by reverse transcription-PCR [RT-PCR]) and protein (by enzyme-linked immunosorbent assay) and by identification of cells expressing TNF-alpha and IL-6 mRNA in lung tissues (by in situ hybridization) and isolated lung lavage cells (by RT-PCR). Adenovirus, labeled with the fluorescent dye (Cy3), was rapidly and widely distributed on epithelial surfaces of airways and alveoli and was very rapidly ( approximately 1 min) localized within AMphi. At 30 min after infection AMphi but not airway epithelial or vascular endothelial cells expressed mRNA for TNF-alpha and IL-6, thus identifying AMphi as the cell source of initial cytokine signaling. IL-6, TNF-alpha, MIP-2, and MIP-1alpha levels progressively increased in bronchoalveolar lavage fluid after pulmonary adenovirus infection, and all were significantly elevated at 6 h (P < 0.05). To begin to define the molecular mechanism(s) by which adenovirus initiates the inflammatory signaling in macrophages, TNF-alpha expression from adenovirus-infected RAW264.7 macrophages was evaluated in vitro. TNF-alpha expression was readily detected in adenovirus-infected RAW cell supernatant with kinetics similar to AMphi during in vivo infection. Blockage of virus uptake at specific cellular sites, including internalization (by wortmannin), endosome acidification and/or lysis (by chloroquine) or by Ca(2+) chelation (by BAPTA) completely blocked TNF-alpha expression. In conclusion, results showed that during ARTI, (i) AMphi rapidly internalized adenovirus, (ii) expression of inflammatory mediators was initiated within AMphi and not airway epithelial or other cells, and (iii) the initiation of inflammatory signaling was linked to virion uptake by macrophages occurring at a point after vesicle acidification. These results have implications for our understanding of the role of the AMphi in the initiation of inflammation following adenovirus infection and adenovirus-mediated gene transfer to the lung.


Asunto(s)
Infecciones por Adenoviridae/virología , Adenoviridae/fisiología , Inflamación/etiología , Macrófagos Alveolares/virología , Infecciones del Sistema Respiratorio/virología , Enfermedad Aguda , Animales , Calcio/fisiología , Línea Celular , Citocinas/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Ratones , Ratones Endogámicos BALB C , ARN Mensajero/análisis , Factor de Necrosis Tumoral alfa/fisiología , Virión/aislamiento & purificación
5.
Hum Gene Ther ; 10(3): 341-53, 1999 Feb 10.
Artículo en Inglés | MEDLINE | ID: mdl-10048387

RESUMEN

Cell proliferation is required for transduction by standard retrovirus vectors derived from viruses in the murine leukemia virus (MuLV) group. Since proliferation rates are low in the mature pulmonary epithelium, we tested the hypothesis that the efficiency of retrovirus-mediated transduction of respiratory epithelial cells can be enhanced by stimulation of cell proliferation with recombinant human keratinocyte growth factor (rhKGF). A marked increase in proliferation of bronchiolar and alveolar epithelial cells was observed after intratracheal administration of rhKGF (30 mg/kg) to adult FVB/N mice. Two days after rhKGF or saline treatment, 10(7) AP+ FFU of LAPSN, a recombinant amphotropic retrovirus that expresses human placental alkaline phosphatase (AP), was instilled intratracheally into the mice. Transduction efficiency, measured 2 days after infection, was increased approximately 70-fold by rhKGF pretreatment. However, even after KGF treatment the total numbers of AP-expressing cells were few. Transduction efficiency was similar using either LAPSN packaged by amphotropic host range packaging cells or LAPSN pseudotyped with 10A1 MuLV envelope protein (0.091 +/- 0.006 versus 0.094 +/- 0.028 transduction events/mm2, respectively). Amphotropic vectors use Pit-2 for cell entry, while 10A1 MuLV vectors can use Pit-1 or Pit-2 for cell entry. By in situ hybridization the retroviral receptor Pit-2 (Ram-1) mRNA was expressed only in the pulmonary vasculature, and Pit-1 (Glvr-1) mRNA was expressed at low levels throughout the lung. In vitro studies demonstrated that retrovirus was inactivated by pulmonary surfactant. Stimulating proliferation of the respiratory epithelium increased retroviral transduction in vivo, but the paucity of retroviral receptors and inactivation by surfactant are additional barriers to high-level retroviral gene transfer in the lung.


Asunto(s)
Bronquios/efectos de los fármacos , Factores de Crecimiento de Fibroblastos , Técnicas de Transferencia de Gen , Sustancias de Crecimiento/farmacología , Proteínas de la Membrana , Alveolos Pulmonares/efectos de los fármacos , Transducción Genética , Animales , Proteínas Portadoras/análisis , División Celular/efectos de los fármacos , Células Cultivadas , Proteínas de Unión al ADN/análisis , Relación Dosis-Respuesta a Droga , Epitelio/anatomía & histología , Epitelio/inmunología , Epitelio/metabolismo , Femenino , Factor 10 de Crecimiento de Fibroblastos , Factor 7 de Crecimiento de Fibroblastos , Vectores Genéticos , Humanos , Ratones , Ratones Endogámicos , Proteínas de Transferencia de Fosfolípidos , Fosfolípidos/análisis , Proteínas Recombinantes/farmacología , Retroviridae/genética , Factores de Tiempo , Factor de Transcripción Pit-1 , Factores de Transcripción/análisis
6.
Hum Gene Ther ; 9(14): 2101-9, 1998 Sep 20.
Artículo en Inglés | MEDLINE | ID: mdl-9759936

RESUMEN

Mutation of the granulocyte-macrophage colony-stimulating factor (GM-CSF) gene by homologous recombination causes progressive pulmonary alveolar proteinosis (PAP) in GM-CSF-deficient mice (GM-/-). The present study tested whether adenovirus-mediated expression of GM-CSF alters the progression of PAP in GM-/- mice. Adult mice were pretreated with an anti-T cell receptor (TCR) antibody to block T cell-mediated immune response, followed by intratracheal instillation of deltaE1-E3 replication-deficient adenovirus expressing mouse GM-CSF (Av1mGM). Mice were killed 1, 3, and 5 weeks after treatment to assess lungs for GM-CSF, surfactant protein B (SP-B), alveolar macrophage maturation, and type II cell proliferation. GM-CSF was detected in BAL fluid from GM-/- mice 1 week after Av1mGM treatment, and GM-CSF mRNA was detected by RT-PCR through 5 weeks. Five weeks after Av1mGM treatment, PAP was improved and SP-B decreased as assessed by ELISA and immunostaining. Increased numbers of alveolar macrophages stained with alpha-naphthyl acetate esterase (alpha-NAE) following treatment with Av1mGM. Local expression of GM-CSF with a recombinant adenovirus ameliorated PAP in the GM-/- mice in association with enhanced maturation of alveolar macrophages.


Asunto(s)
Adenoviridae/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/deficiencia , Pulmón/patología , Proteinosis Alveolar Pulmonar/genética , Animales , Modelos Animales de Enfermedad , Expresión Génica/genética , Terapia Genética/métodos , Vectores Genéticos/genética , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Histocitoquímica , Macrófagos Alveolares/enzimología , Ratones , Ratones Noqueados , Naftoles/metabolismo , Antígeno Nuclear de Célula en Proliferación/metabolismo , Proteolípidos/metabolismo , Proteinosis Alveolar Pulmonar/terapia , Surfactantes Pulmonares/metabolismo , Receptores de Antígenos de Linfocitos T/antagonistas & inhibidores , Receptores de Antígenos de Linfocitos T/inmunología
7.
Am J Physiol ; 273(4): L715-25, 1997 10.
Artículo en Inglés | MEDLINE | ID: mdl-9357845

RESUMEN

The human surfactant protein (SP)-C gene promoter was used to direct expression of mouse granulocyte macrophage colony-stimulating factor (GM-CSF; SP-C-GM mice) in lung epithelial cells in GM-CSF-replete (GM+/+) or GM-CSF null mutant (GM-/-) mice. Lung weight and volume were significantly increased in SP-C-GM mice compared with GM+/+ or GM-/- control mice. Immunohistochemical staining demonstrated marked type II cell hyperplasia, and immunofluorescent labeling for proliferating cell nuclear antigen was increased in type II cells of SP-C-GM mice. Abundance of type II cells per mouse lung was increased three- to fourfold in SP-C-GM mice compared with GM+/+ and GM-/- mice. GM-CSF increased bromodeoxyuridine labeling of isolated type II cells in vitro. Type II cells, alveolar macrophages, and endothelial and bronchiolar epithelial cells were stained by antibodies to the GM-CSF receptor alpha-subunit in both GM+/+ mice and GM-CSF gene-targeted mice that are also homozygous for the SP-C-GM transgene. High levels of GM-CSF expression in type II cells of transgenic mice increased lung size and caused type II cell hyperplasia, demonstrating an unexpected role for the molecule in the regulation of type II cell proliferation and differentiation.


Asunto(s)
Factor Estimulante de Colonias de Granulocitos y Macrófagos/fisiología , Pulmón/fisiología , Proteolípidos/fisiología , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Surfactantes Pulmonares/fisiología , Animales , Peso Corporal , Líquido del Lavado Bronquioalveolar/química , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/metabolismo , Células Epiteliales/patología , Factor Estimulante de Colonias de Granulocitos y Macrófagos/biosíntesis , Factor Estimulante de Colonias de Granulocitos y Macrófagos/genética , Humanos , Hiperplasia , Lipopolisacáridos/toxicidad , Pulmón/crecimiento & desarrollo , Pulmón/patología , Ratones , Ratones Noqueados , Ratones Transgénicos , Tamaño de los Órganos , Proteolípidos/biosíntesis , Surfactantes Pulmonares/biosíntesis , Proteínas Recombinantes de Fusión/biosíntesis , Salmonella typhimurium
8.
Hum Gene Ther ; 8(11): 1331-44, 1997 Jul 20.
Artículo en Inglés | MEDLINE | ID: mdl-9295128

RESUMEN

Although replication-deficient adenoviruses efficiently transfer genes into epithelial cells of the lung, host immune responses limit the extent and duration of gene expression. To define further the role of inflammatory responses to first-generation, recombinant, deltaE1, deltaE3 adenovirus in lung pathology and surfactant protein homeostasis, expression of the surfactant proteins SP-A, SP-B, and proSP-C was determined by immunohistochemistry 2, 7, and 14 days following intratracheal administration of 2 x 10(9) pfu of a recombinant adenovirus, Av1Luc1, to BALB/c nu/nu and BALB/c wild-type mice. Two to 7 days after virus administration, an acute inflammatory response was observed in both mouse strains. Respiratory epithelial cells were sloughed, and extracellular accumulation of SP-A and SP-B was detected in the airways. Diminished immunostaining for SP-A and SP-B was noted in type II cells, and SP-A and SP-B mRNA expression was decreased in focal regions of the lungs from both mouse strains. One week after virus administration, immunostaining for proSP-C was markedly increased in cells lining the regenerating alveolar epithelial surfaces. Two weeks after Av1Luc1 treatment of nu/nu mice, immunostaining for SP-A, SP-B, and proSP-C was similar to those patterns observed prior to adenoviral administration. In immunocompetent wild-type mice, however, immunostaining for surfactant proteins was absent in areas associated with chronic lymphocytic infiltration. The recombinant adenoviral vector, Av1Luc1, caused acute inflammatory responses in the respiratory epithelium with disruption of surfactant protein homeostasis in both wild-type and nu/nu mice. Alterations in surfactant homeostasis persisted in wild-type mice. Thus, both acute and thymic-dependent immune responses limit transgene expression and disrupt surfactant protein gene expression and homeostasis. Because surfactant proteins are critical to host defense and to the maintenance of alveolar stability following injury, these findings raise concerns regarding both acute and chronic toxicity of first-generation recombinant adenoviral vectors for gene transfer.


Asunto(s)
Infecciones por Adenoviridae/metabolismo , Adenoviridae , Homeostasis , Pulmón/fisiología , Surfactantes Pulmonares/metabolismo , Infecciones por Adenoviridae/complicaciones , Animales , Vectores Genéticos/efectos adversos , Vectores Genéticos/farmacología , Inflamación/fisiopatología , Pulmón/virología , Ratones , Ratones Endogámicos BALB C , Ratones Desnudos , Proteolípidos/genética , Proteolípidos/metabolismo , Proteína A Asociada a Surfactante Pulmonar , Proteínas Asociadas a Surfactante Pulmonar , Surfactantes Pulmonares/genética , ARN Mensajero/metabolismo
9.
Hum Gene Ther ; 8(8): 935-41, 1997 May 20.
Artículo en Inglés | MEDLINE | ID: mdl-9195216

RESUMEN

Replication-deficient delta E1a-E3 adenovirus mediates efficient gene transfer to the mouse lung; however it induces a host immune response mediated, in part, by T cells. This immune response is associated with loss of transgene expression. Monoclonal antibodies (mAb) against the T cell receptor (TCR) complex can inhibit both CD4+ and CD8+ T cell responses in vivo and are the most potent anti-T cell agents in clinical use. To determine whether such mAbs can be used to prolong adenovirus-mediated transgene expression, the vector Av1Luc1 (delta E1a-E3 recombinant adenovirus encoding the firefly luciferase gene) was administered intratracheally to C57BL/6 mice on day 0. Three days prior to adenovirus administration (day -3), mice were treated with a single i.p. injection of the anti-TCR mAb H57. Controls received phosphate-buffered saline. Animals were sacrificed on days 3, 14, 28, and 56 and lungs were assessed for transgene expression and histopathology. Luciferase activity decreased markedly in the controls by day 14, but was maintained at high levels in animals receiving anti-TCR mAb. A mild, focal, predominantly neutrophilic inflammation was observed in the alveoli of all mice 3 days after virus administration. In PBS-treated controls, this inflammation progressed to a moderate to severe multifocal, perivascular and peribronchiolar lymphoid infiltration at 14 days. B cells and T cells were present in approximately equal numbers, with CD4+ T cells predominating over CD8+ T cells by 3- to 28-fold. Treatment with H57 resulted in near-complete prevention of the lymphocytic inflammatory infiltrate and increased luciferase activity throughout the 56-day study period, in association with TCR modulation and T cell depletion. Thus, anti-TCR mAb decreases inflammation and prolongs gene expression following adenovirus-mediated gene transfer.


Asunto(s)
Adenoviridae/genética , Técnicas de Transferencia de Gen , Vectores Genéticos , Neumonía/prevención & control , Receptores de Antígenos de Linfocitos T/inmunología , Animales , Anticuerpos Monoclonales , Formación de Anticuerpos , Epitelio/inmunología , Femenino , Intubación Intratraqueal , Luciferasas/genética , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Factores de Tiempo
10.
Cardiovasc Res ; 36(2): 205-15, 1997 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9463632

RESUMEN

OBJECTIVE: Peroxynitrite and hydroxyl radical, reactive oxidants produced during reperfusion, are potent triggers of DNA single strand breakage. DNA injury triggers the activation of the nuclear enzyme poly (ADP-ribose) synthetase (PARS), which contributes to cellular energetic depletion. Using 3-aminobenzamide, an inhibitor of PARS, we investigated the role of PARS in the pathogenesis of myocardial reperfusion injury in a rat model. METHODS AND RESULTS: Occlusion of the left main coronary artery (one hour) followed by reperfusion (one hour) in the anesthetized rat caused severe cardiac necrosis, neutrophil infiltration, and increased plasma creatine phosphokinase activity. There was significant peroxynitrite production during reperfusion, as indicated by a massive increase in nitrotyrosine in the necrotic myocardium. Reperfusion was also associated with a significant loss of myocardial ATP. In vivo administration of the PARS inhibitor 3-aminobenzamide (10 mg/kg i.v.) to rats subjected to myocardial ischemia and reperfusion, reduced myocardial infarct size and blunted the increase in plasma creatine phosphokinase activity and myeloperoxidase activity in infarcted hearts. In addition, 3-aminobenzamide partially preserved the myocardial ATP levels. In vitro, pharmacological inhibition of PARS also ameliorated peroxynitrite-induced cytotoxicity in rat cardiac myocytes and human endothelial cells. CONCLUSION: 3-aminobenzamide has significant protective effects in myocardial reperfusion injury. We hypothesize that activation of PARS activation plays a role in the pathophysiology of acute myocardial infarction.


Asunto(s)
Benzamidas/uso terapéutico , Inhibidores Enzimáticos/uso terapéutico , Daño por Reperfusión Miocárdica/prevención & control , Inhibidores de Poli(ADP-Ribosa) Polimerasas , Adenosina Trifosfato/metabolismo , Animales , Células Cultivadas , Creatina Quinasa/sangre , Endotelio Vascular/efectos de los fármacos , Endotelio Vascular/patología , Técnicas In Vitro , Masculino , Daño por Reperfusión Miocárdica/metabolismo , Daño por Reperfusión Miocárdica/patología , Miocardio/enzimología , Miocardio/metabolismo , Miocardio/patología , Peroxidasa/metabolismo , Ratas , Ratas Wistar , Tirosina/análogos & derivados , Tirosina/metabolismo
11.
Hum Gene Ther ; 6(4): 457-67, 1995 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-7612702

RESUMEN

To evaluate the role of cell-mediated immunity during gene transfer to the respiratory epithelium, the time course of luciferase activity was assessed after intratracheal administration of Av1Luc1, an E1a-E3-deleted adenoviral (Ad5) vector expressing firefly luciferase, to FVB/N, BALB/c and BALB/c-nu/nu adult mice. Adenovirus-mediated luciferase activity was rapidly lost from the respiratory tract between 2 and 14 days after treatment of both FVB/N and BALB/c wild-type mice. In the wild-type mice, loss of luciferase activity was associated with an early inflammatory response consisting of infiltration with macrophages and polymorphonuclear leukocytes and a more prolonged response characterized by lymphocytic infiltration. In the immune-deficient nu/nu mice, luciferase activity was maintained at higher levels than in immune-competent mice after exposure to virus and was associated with a distinct pattern of inflammation, consisting primarily of macrophages and polymorphonuclear cells but lacking the lymphocytic infiltrates typical of the inflammation in wild-type mice. Adenoviral DNA was rapidly cleared from the lungs of both nu/nu and wild-type mice. Markedly increased expression of proliferating cell nuclear antigen (PCNA) was observed in bronchiolar and alveolar epithelial cells and in inflammatory cells after exposure to Av1LUc1. The proliferative response of the respiratory epithelium was more extensive and persistent in wild-type than in nu/nu mice. To assess further the impact of the immune system on adenovirus-mediated gene expression, cotton rats treated with cyclosporin A or dexamethasone were exposed to Av1Luc1. Both agents decreased lung inflammation and significantly increased lung luciferase activity. The loss of lung luciferase activity is dependent, in part, on the immune-mediated clearance of respiratory epithelial cells, which may limit the extent and duration of gene expression with recombinant adenoviral vectors.


Asunto(s)
Infecciones por Adenoviridae/inmunología , Adenovirus Humanos/fisiología , Virus Defectuosos/fisiología , Técnicas de Transferencia de Gen , Vectores Genéticos/fisiología , Pulmón/virología , Neumonía Viral/inmunología , Replicación Viral , Infecciones por Adenoviridae/patología , Infecciones por Adenoviridae/virología , Adenovirus Humanos/genética , Adenovirus Humanos/inmunología , Adenovirus Humanos/aislamiento & purificación , Animales , Secuencia de Bases , Ciclosporina/farmacología , Virus Defectuosos/genética , Virus Defectuosos/inmunología , Virus Defectuosos/aislamiento & purificación , Virus Defectuosos/patogenicidad , Dexametasona/farmacología , Femenino , Regulación Viral de la Expresión Génica , Genes Reporteros , Vectores Genéticos/efectos adversos , Vectores Genéticos/genética , Vectores Genéticos/inmunología , Vectores Genéticos/aislamiento & purificación , Inmunidad Celular , Inmunocompetencia , Luciferasas/análisis , Luciferasas/genética , Pulmón/inmunología , Pulmón/patología , Macrófagos/inmunología , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos , Ratones Desnudos , Datos de Secuencia Molecular , Neutrófilos/inmunología , Neumonía Viral/patología , Neumonía Viral/virología , Antígeno Nuclear de Célula en Proliferación/análisis , Proteínas Recombinantes de Fusión/análisis , Sigmodontinae
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA