Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 43
Filtrar
Más filtros

Banco de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Am J Transplant ; 16(5): 1503-15, 2016 05.
Artículo en Inglés | MEDLINE | ID: mdl-26602886

RESUMEN

Solid phase immunoassays (SPI) are now routinely used to detect HLA antibodies. However, the flow cytometric crossmatch (FCXM) remains the established method for assessing final donor-recipient compatibility. Since 2005 we have followed a protocol whereby the final allocation decision for renal transplantation is based on SPI (not the FCXM). Here we report long-term graft outcomes for 508 consecutive kidney transplants using this protocol. All recipients were negative for donor-specific antibody by SPI. Primary outcomes are graft survival and incidence of acute rejection within 1 year (AR <1 year) for FCXM+ (n = 54) and FCXM- (n = 454) recipients. Median follow-up is 7.1 years. FCXM+ recipients were significantly different from FCXM- recipients for the following risk factors: living donor (24% vs. 39%, p = 0.03), duration of dialysis (31.0 months vs. 13.5 months, p = 0.008), retransplants (17% vs. 7.3%, p = 0.04), % sensitized (63% vs. 19%, p = 0.001), and PRA >80% (20% vs. 4.8%, p = 0.001). Despite these differences, 5-year actual graft survival rates are 87% and 84%, respectively. AR <1 year occurred in 13% FCXM+ and 12% FCXM- recipients. Crossmatch status was not associated with graft outcomes in any univariate or multivariate model. Renal transplantation can be performed successfully, using SPI as the definitive test for donor-recipient compatibility.


Asunto(s)
Tipificación y Pruebas Cruzadas Sanguíneas , Rechazo de Injerto/diagnóstico , Asignación de Recursos para la Atención de Salud/métodos , Prueba de Histocompatibilidad/métodos , Isoanticuerpos/inmunología , Trasplante de Riñón , Obtención de Tejidos y Órganos , Linfocitos B/inmunología , Femenino , Citometría de Flujo/métodos , Estudios de Seguimiento , Rechazo de Injerto/prevención & control , Supervivencia de Injerto , Humanos , Masculino , Persona de Mediana Edad , Pronóstico , Estudios Retrospectivos , Factores de Riesgo , Donantes de Tejidos
2.
Transplant Proc ; 38(10): 3524-6, 2006 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17175321

RESUMEN

BACKGROUND: The occurrence of lymphocele formation following renal transplantation is variable, and the optimal approach to treatment remains undefined. Opening the peritoneum at the time of transplantation is one method of decreasing the incidence of lymphocele formation. The purpose of this study was to determine whether creating a peritoneal window at the time of transplantation decreases the incidence of lymphocele formation. METHODS: We performed a retrospective review of renal transplants conducted at our institution between 2002 and 2004. Records were reviewed to obtain details regarding opening of the peritoneum at the time of transplant and occurrence of lymphocele. Every patient underwent routine ultrasound imaging in the peri-operative period. Graft dysfunction secondary to the lymphocele was the primary indication for intervention. Data were analyzed by chi-square. RESULTS: During the initial transplant the peritoneum was opened in 35% of patients. The overall incidence of fluid collections, identified by ultrasound, was 24%. Opening the peritoneum did not decrease the incidence of lymphocele. However, more patients with a closed peritoneum required an intervention for a symptomatic lymphocele. In the 11 patients with an open peritoneum and a fluid collection, only one required an intervention. In patients whose peritoneum was left intact, 24% of fluid collections required intervention. Graft survival was equivalent. CONCLUSION: Creating a peritoneal window at the time of transplantation did not decrease the overall incidence of postoperative fluid collections. However, forming a peritoneal window at the time of transplantation did decrease the incidence of symptomatic lymphocele.


Asunto(s)
Trasplante de Riñón/métodos , Linfocele/prevención & control , Peritoneo/cirugía , Complicaciones Posoperatorias/prevención & control , Humanos , Incidencia , Linfocele/epidemiología , Complicaciones Posoperatorias/epidemiología , Estudios Retrospectivos
3.
Shock ; 12(1): 25-31, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10468048

RESUMEN

Inflammation induces the expression of angiogenic growth factors in tissues, which leads to microvascular growth. Bacterial lipopolysaccharide (LPS) provokes a transient inflammatory response in the heart and induces delayed cardiac resistance to post-ischemic contractile dysfunction. In this study, we examined: 1) the effects of LPS on myocardial expression of basic fibroblast growth factor (bFGF) and vascular endothelial growth factor (VEGF), 2) whether an increase in the density of myocardial microvessels follows the expression of angiogenic growth factors, and 3) the effect of LPS on myocardial resistance to infarction and its relationship with microvascular growth. Rats were treated with LPS (from Salmonella typhimurium, 0.5 mg/kg i.p.). The expression of bFGF and VEGF in the myocardium was examined at 6 and 12 h after LPS treatment by immunofluorescent staining. Myocardial capillary and arteriole densities were determined 3 days after LPS treatment by morphometry, using immunofluorescent staining of von Willebrand factor (a marker protein of endothelial cells) and alpha-smooth muscle actin (a marker protein of smooth muscle cells). To examine cardiac resistance to infarction, hearts were subjected to 40 min of regional ischemia and 2 h of reperfusion by reversible occlusion of left coronary artery at 3 days after LPS treatment. LPS induced cardiac bFGF and VEGF at 6 and 12 h after treatment. The expression of these growth factors was followed by an increase in myocardial capillary density (2032 +/- 78/mm2 vs. 1617 +/- 47/mm2 in saline control, P < 0.05), but not arteriole density, at 3 days. Meanwhile, infarct size was significantly reduced by LPS preconditioning (infarct/left ventricle 12.3 +/- 1.04% vs. 21.7 +/- 1.65% in saline control, 43% reduction, P < 0.05). These results suggest that LPS preconditioning induces cardiac bFGF and VEGF, and an increase in myocardial capillary density. This increased myocardial capillary density is associated with a reduced infarct size after in vivo regional ischemia-reperfusion.


Asunto(s)
Lipopolisacáridos/farmacología , Infarto del Miocardio/tratamiento farmacológico , Infarto del Miocardio/patología , Neovascularización Patológica/tratamiento farmacológico , Animales , Vasos Coronarios/efectos de los fármacos , Inflamación/tratamiento farmacológico , Inflamación/patología , Lipopolisacáridos/uso terapéutico , Masculino , Infarto del Miocardio/inmunología , Ratas , Ratas Sprague-Dawley
4.
Shock ; 10(6): 389-94, 1998 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-9872676

RESUMEN

Interleukin-10 (IL-10) protects animals from lethal endotoxemia. This beneficial effect is mediated, in part, by inhibition of inflammatory cytokine production, including tumor necrosis factor-alpha (TNF-alpha). Evidence suggests that IL-10 may inhibit activation of the transcription factor nuclear factor-kappaB (NF-kappaB) through an unknown mechanism. NF-kappaB activation in response to inflammatory signals is dependent upon degradation of its associated inhibitory peptide, inhibitory kappaB-alpha (IkappaB-alpha). We hypothesized that IL-10 prevents human monocyte NF-kappaB activation and resultant TNF-alpha production by stabilization of IkappaB-alpha. The purpose of this study was to determine the effect of IL-10 on lipopolysaccharide (LPS)-induced human monocyte TNF-alpha production, NF-kappaB activation, and IkappaB-alpha degradation. Monocytes were isolated from human donors. Cells were stimulated with endotoxin (LPS, 100 ng/mL) with and without human IL-10 (10 ng/mL). Following stimulation, TNF-alpha was measured in cell supernatants by ELISA, NF-kappaB activity by electrophoretic mobility shift assay, and IkappaB-alpha levels by Western blot. We observed that after LPS stimulation of human monocytes, TNF-alpha increased to 798+/-67 pg/mL (p < .001 versus control). IL-10 attenuated LPS-stimulated TNF-alpha production (297+/-54; p < .001 versus LPS alone). After LPS stimulation in human monocytes, IkappaB-alpha protein levels decreased, and NF-kappaB DNA binding increased. IL-10 pretreatment prevented LPS-induced decreases in IkappaB-alpha protein levels and attenuated NF-kappaB DNA binding. IL-10 appears to prevent activation of NF-kappaB by preserving IkappaB-alpha protein levels, leading to a reduction in TNF-alpha release.


Asunto(s)
Proteínas de Unión al ADN/metabolismo , Proteínas I-kappa B , Interleucina-10/farmacología , Monocitos/efectos de los fármacos , Monocitos/metabolismo , FN-kappa B/antagonistas & inhibidores , Animales , Secuencia de Bases , ADN/genética , ADN/metabolismo , Endotoxemia/prevención & control , Humanos , Técnicas In Vitro , Lipopolisacáridos/toxicidad , Inhibidor NF-kappaB alfa , FN-kappa B/genética , Factor de Necrosis Tumoral alfa/biosíntesis
5.
Shock ; 10(5): 309-18, 1998 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-9840644

RESUMEN

Pharmacological therapy of surgical disease often involves manipulating the physiologic balance between pro- and anti-inflammatory responses. Many agents target only one aspect of the inflammatory cascade. Originally identified as a protein elaborated by T-lymphocytes, IL-10 appears to globally inhibit cytokine production. The purpose of this manuscript is to examine the immunomodulatory and anti-inflammatory effects of interleukin-10 (IL-10) in an attempt to define the clinical utility of IL-10, both as a marker of and as a therapeutic strategy for intervention in inflammatory and immune-mediated diseases. IL-10 is elaborated from multiple sources and has diverse cellular effects to regulate immune and inflammatory responses. Accumulating evidence suggests that the anti-inflammatory influence of IL-10 observed at the cellular level may be manipulated to impact the immune and inflammatory-mediated responses associated with injury and sepsis, gastrointestinal and cardiovascular disease, and transplantation. In conclusion, IL-10 is an important mediator of immune and anti-inflammatory responses in surgical disease and, as such, has therapeutic promise as an immunomodulator and as an anti-inflammatory agent.


Asunto(s)
Interleucina-10/farmacología , Interleucina-10/fisiología , Sepsis/tratamiento farmacológico , Trasplante , Heridas y Lesiones/metabolismo , Animales , Enfermedades Cardiovasculares/tratamiento farmacológico , Enfermedades Cardiovasculares/metabolismo , Citocinas/metabolismo , Proteínas de Unión al ADN/metabolismo , Humanos , Inflamación/tratamiento farmacológico , Inflamación/metabolismo , Pancreatitis/tratamiento farmacológico , Pancreatitis/metabolismo , Proteínas Tirosina Quinasas/metabolismo , Factor de Transcripción STAT3 , Sepsis/inmunología , Sepsis/metabolismo , Transactivadores/metabolismo , Transcripción Genética , Heridas y Lesiones/tratamiento farmacológico , Heridas y Lesiones/inmunología
6.
Shock ; 14(1): 1-7, 2000 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10909885

RESUMEN

Chemokines are important mediators of inflammation. Animal studies suggest that inhibition of chemokine action results in a decrease in inflammation. Novel anti-inflammatory agents directed against chemokines are now available. Surgeons are uniquely positioned to treat multiple chemokine-mediated diseases. In this article, we review the biology and nomenclature of chemokines as well as their role in neutrophil migration. Further, the potential role of chemokines in various diseases related to surgical conditions, including adult respiratory distress syndrome, atherosclerosis, inflammatory bowel disease, and solid organ rejection, is reviewed. Finally, the idea that chemokines could be targets for novel therapeutic agents is discussed.


Asunto(s)
Quimiocinas/fisiología , Infiltración Neutrófila/fisiología , Complicaciones Posoperatorias/fisiopatología , Receptores de Quimiocina/fisiología , Animales , Arteriosclerosis/etiología , Arteriosclerosis/fisiopatología , Quimiocinas/biosíntesis , Quimiocinas/clasificación , Quimiocinas/genética , Regulación de la Expresión Génica/efectos de los fármacos , Rechazo de Injerto/etiología , Rechazo de Injerto/fisiopatología , Humanos , Quinasa I-kappa B , Enfermedades Inflamatorias del Intestino/etiología , Enfermedades Inflamatorias del Intestino/fisiopatología , Ratones , Ratones Noqueados , FN-kappa B/antagonistas & inhibidores , FN-kappa B/fisiología , Complicaciones Posoperatorias/etiología , Proteínas Serina-Treonina Quinasas/antagonistas & inhibidores , Receptores de Quimiocina/antagonistas & inhibidores , Síndrome de Dificultad Respiratoria/etiología , Síndrome de Dificultad Respiratoria/fisiopatología
7.
Ann N Y Acad Sci ; 874: 69-82, 1999 Jun 30.
Artículo en Inglés | MEDLINE | ID: mdl-10415522

RESUMEN

Overproduction of tumor necrosis factor-alpha (TNF-alpha) contributes to cardiac dysfunction associated with systemic or myocardial stress, such as endotoxemia and myocardial ischemia/reperfusion (I/R). Heat shock has been demonstrated to enhance cardiac functional resistance to I/R. However, the protective mechanisms remain unclear. The purpose of this study was to determine: (1) whether cardiac macrophages express heat shock protein 72 (HSP72) after heat shock, (2) whether induced cardiac HSP72 suppresses myocardial TNF-alpha production during I/R, and (3) whether preservation of postischemic myocardial function by heat shock is correlated with attenuated TNF-alpha production during I/R. Rats were subjected to heat shock (42 degrees C for 15 min) and 24 h recovery. Immunoblotting confirmed the expression of cardiac HSP72. Immunofluorescent staining detected HSP72 in cardiac interstitial cells including resident macrophages rather than myocytes. Global I/R caused a significant increase in myocardial TNF-alpha. The increase in myocardial TNF-alpha was blunted by prior heat shock and the reduced myocardial TNF-alpha level was correlated with improved cardiac functional recovery. This study demonstrates for the first time that heat shock induces HSP72 in cardiac resident macrophages and inhibits myocardial TNF-alpha production during I/R. These observations suggest that inhibition of myocardial TNF-alpha production may be a mechanism by which HSP72 protects the heart against postischemic dysfunction.


Asunto(s)
Calor , Isquemia Miocárdica/fisiopatología , Miocardio/metabolismo , Choque/metabolismo , Estrés Fisiológico/fisiopatología , Factor de Necrosis Tumoral alfa/antagonistas & inhibidores , Animales , Proteínas del Choque Térmico HSP72 , Corazón/fisiopatología , Proteínas de Choque Térmico/metabolismo , Proteínas de Choque Térmico/fisiología , Macrófagos/metabolismo , Masculino , Daño por Reperfusión Miocárdica/metabolismo , Miocardio/patología , Ratas , Ratas Sprague-Dawley
8.
Surgery ; 124(2): 318-26; discussion 326-7, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9706155

RESUMEN

BACKGROUND: Vessel injury provokes the release of proinflammatory cytokines and growth factors that influence vascular smooth muscle cell (VSMC) proliferation and migration. Produced by T lymphocytes, interleukin-10 (IL-10) and interferon-gamma (IFN-gamma) both have immunoregulatory functions and act on similar receptors, designated class II cytokine receptors. We hypothesized that the class II cytokine receptor participates in vascular remodeling by inhibiting VSMC proliferation. The purposes of this study were to determine the influence of class II cytokine receptor stimulation on (1) unstimulated, (2) cytokine-stimulated, and (3) growth factor-stimulated VSMC proliferation. METHODS: Human aortic VSMCs were isolated and cultured. VSMCs were treated with IL-10 or IFN with or without tumor necrosis factor-alpha (TNF-alpha) or basic fibroblast growth factor (FGF). Proliferation was quantified by colormetric assay. RESULTS: Compared to control, both TNF and FGF stimulated concentration-dependent VSMC proliferation (P < .005). IL-10 and IFN alone had no effect on unstimulated cell growth. With TNF or FGF stimulation, both IL-10, at a dose as low as 10 fg/ml, and IFN, at a dose as low as 1.0 U/ml, inhibited cell growth (P < .001). CONCLUSIONS: The class II cytokine receptor ligands, IL-10 and IFN, inhibit cytokine-(TNF) and growth factor-(FGF) induced VSMC proliferation. The class II cytokine receptor may provide a novel therapeutic target in regulating vessel wall remodeling after vascular injury.


Asunto(s)
Interferón gamma/inmunología , Interleucina-10/inmunología , Músculo Liso Vascular/química , Músculo Liso Vascular/citología , Receptores de Interferón/inmunología , Aorta Torácica/citología , División Celular/inmunología , Células Cultivadas , Factores de Crecimiento de Fibroblastos/farmacología , Humanos , Interferón gamma/farmacología , Interleucina-10/farmacología , Músculo Liso Vascular/efectos de los fármacos , Receptores de Interferón/agonistas , Receptores de Interferón/metabolismo , Linfocitos T/inmunología , Factor de Necrosis Tumoral alfa/farmacología , Receptor de Interferón gamma
9.
Surgery ; 124(2): 291-6; discussion 297, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9706151

RESUMEN

BACKGROUND: Oxidant stress caused by ischemia or endotoxemia induces myocardial dysfunction and cardiomyocyte death; however, mechanisms responsible remain unknown. We hypothesized that hydrogen peroxide (H2O2) induces myocardial dysfunction and cardiomyocyte death via P38 mitogen-activated protein kinase (MAPK)-mediated myocardial tumor necrosis factor (TNF) production. METHODS: Langendorff perfused rat hearts (6/group) were subjected to oxidant stress (H2O2 infusion; 300 mmol/L x 80 minutes), with and without prior infusion of a specific P38 kinase MAPK inhibitor (P38i = 1 mmol/L/min x 5 minutes) or TNF neutralization (20 mg TNF binding protein (BP)/min x 80 minutes). Developed pressure (DP), coronary flow, and end-diastolic pressure were continuously recorded. Myocardial creatine kinase (CK) loss was measured in the coronary effluent, and tissue TNF was measured in myocardial homogenates. RESULTS: Eighty minutes of H2O2 infusion induced a 6.5-fold increase in myocardial TNF production, which was associated with a 70% decrease in DP and increase in CK loss. P38 MAPK inhibition or TNF-BP decreased myocardial TNF production, cardiomyocyte death, and myocardial dysfunction. CONCLUSIONS: These results demonstrate that H2O2 alone induces myocardial TNF production. P38 MPAK is an oxidant-sensitive enzyme that mediates oxidant-induced myocardial TNF production, cardiac dysfunction, and cardiomyocyte death.


Asunto(s)
Proteínas Quinasas Dependientes de Calcio-Calmodulina/metabolismo , Cardiopatías/metabolismo , Peróxido de Hidrógeno/farmacología , Proteínas Quinasas Activadas por Mitógenos , Oxidantes/farmacología , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Creatina Quinasa/metabolismo , Activación Enzimática/efectos de los fármacos , Cardiopatías/inducido químicamente , Masculino , Miocardio/química , Miocardio/enzimología , Estrés Oxidativo/fisiología , Ratas , Ratas Sprague-Dawley , Función Ventricular Izquierda , Proteínas Quinasas p38 Activadas por Mitógenos
10.
Surgery ; 126(2): 135-41, 1999 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-10455875

RESUMEN

BACKGROUND: The purposes of this study were to (1) determine whether functional heat-shock protein 72 (HSP-72) may be delivered into the heart, (2) determine whether HSP-72 itself is protective against endotoxin (lipopolysaccharide [LPS]-induced cardiodepression, and (3) compare relative protection and time courses required for protection for thermally induced HSP-72 versus liposomally introduced HSP-72. METHODS: HSP-72 was introduced (liposomal HSP-72) or induced (heat shock, 42 degrees C x 15 minutes, 24 hours before) in rat heart before LPS administration (0.5 mg/kg intraperitoneal or ex vivo coronary infusion). Western blot analysis for HSP-72 was used to confirm its expression. Left ventricular developed pressure (Langendorff) was used as an index of cardiac function. RESULTS: Direct intracoronary perfusion of liposomal HSP-72 delivered functioning HSP-72 into the myocardium. LPS induced cardiodepression; however, heat shock pretreatment abolished LPS-induced contractile dysfunction. A direct connection was found between HSP-72 and protection derived from liposomal transfer experiments that similarly reduced LPS-induced cardiodepression. CONCLUSIONS: (1) HSP-72 prevents LPS-induced myocardial contractile dysfunction, (2) liposomal transfer of HSP-72 into the myocardium provides the first direct mechanistic connection between myocardial HSP-72 and protection against LPS, (3) HSP-72 induction requires 24 hours and liposomal transfer of HSP-72 requires 90 minutes, and (4) HSP-72 may offer a clinically acceptable means of protecting the heart.


Asunto(s)
Proteínas de Choque Térmico/administración & dosificación , Lipopolisacáridos/toxicidad , Contracción Miocárdica/efectos de los fármacos , Animales , Portadores de Fármacos , Proteínas HSP70 de Choque Térmico/biosíntesis , Proteínas HSP70 de Choque Térmico/genética , Proteínas del Choque Térmico HSP72 , Proteínas de Choque Térmico/biosíntesis , Calor , Liposomas , Masculino , Ratas , Ratas Sprague-Dawley
11.
Arch Surg ; 133(6): 667-9, 1998 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-9637469

RESUMEN

It is the regulation of gene expression that determines phenotype and cellular response. Several families of proteins control gene expression in cells and influence the pathogenesis of multiple organ failure, the acute phase response, atherosclerosis, and graft-vs-host disease. Understanding the basics of the regulation of gene transcription will allow the knowledgeable surgeon to target gene expression as a therapeutic modality in multiple diseases. We examine nuclear factor kappa B as an example of a transcription factor that is involved in multiple surgical diseases and has pharmacological inhibitors available to knowledgeable surgeons.


Asunto(s)
Regulación de la Expresión Génica/fisiología , Transcripción Genética/fisiología , Animales , Humanos , Inflamación/fisiopatología , FN-kappa B/genética , Heridas y Lesiones/fisiopatología
12.
Ann Thorac Surg ; 67(5): 1227-31; discussion 1231-2, 1999 May.
Artículo en Inglés | MEDLINE | ID: mdl-10355388

RESUMEN

BACKGROUND: Vessel injury results in an inflammatory response characterized by the elaboration of cytokines and growth factors, which ultimately influence vascular smooth muscle cell (VSMC) growth and contribute to atherogenesis. Nuclear factor-kappa B (NFkappaB) is a central transcription factor important in mediating stress and inflammatory-induced signals. We hypothesized that strategies aimed at inhibiting NFkappaB would abrogate mitogen-induced human VSMC proliferation. METHODS: Human aortic VSMC were stimulated with basic fibroblast growth factor (FGF) and tumor necrosis factor-alpha (TNF), and proliferation was quantified by a colormetric assay. The influence of NFkappaB on VSMC proliferation was examined by both nonspecific NFkappaB blockade with calpain inhibitor-1 (CI-1) and dexamethasone (Dex) and specific NFkappaB blockade with liposomal delivery of the NFkappaB inhibitory peptide, IkappaBalpha. RESULTS: FGF and TNF induced concentration-dependent VSMC proliferation (p < 0.002). Neither CI-1, Dex, nor liposomal IkappaBalpha influenced proliferation of unstimulated VSMC. However, both FGF- and TNF-stimulated VSMC proliferation was inhibited to the level of control with CI-1, Dex, and liposomal IkappaBalpha (p < 0.001). CONCLUSION: The mitogenic effect of FGF and TNF on human arterial VSMC may be prevented by inhibiting NFkappaB. Furthermore, liposomal delivery of endogenous inhibitory proteins such as IkappaBalpha may represent a novel, therapeutically accessible method for selective transcriptional suppression in the response to vascular injury.


Asunto(s)
Aorta/metabolismo , Proteínas de Unión al ADN/farmacología , Proteínas de Unión al ADN/fisiología , Proteínas I-kappa B , Músculo Liso Vascular/citología , FN-kappa B , Calpaína/antagonistas & inhibidores , División Celular/efectos de los fármacos , Células Cultivadas , Dexametasona/farmacología , Factores de Crecimiento de Fibroblastos/farmacología , Humanos , Liposomas , Mitógenos/farmacología , Músculo Liso Vascular/metabolismo , Inhibidor NF-kappaB alfa , FN-kappa B/antagonistas & inhibidores , FN-kappa B/farmacología , Transducción de Señal/fisiología , Factor de Necrosis Tumoral alfa/farmacología
13.
Ann Thorac Surg ; 65(4): 1065-70, 1998 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-9564929

RESUMEN

BACKGROUND: Ischemic stress and other protein kinase C (PKC)-linked receptor stimuli can induce rapid cardiac protection against ischemia-reperfusion injury. We and others have demonstrated that exogenous calcium (Ca2+) pretreatment confers PKC-mediated cardiac functional and infarct protection in animal models, but it remains unknown whether Ca2+ preconditioning confers similar postischemic functional protection in human myocardium, and, if so, whether the mechanism is mediated by PKC. We postulated that Ca2+ preconditioning confers ischemic tolerance to human myocardium by a PKC-dependent mechanism. METHODS: Human atrial trabeculae were suspended in organ baths and paced at 1 Hz, and force development was recorded. After 90 minutes of equilibration, all trabeculae were subjected to ischemia (45 minutes) and reperfusion (120 minutes). Exogenous CaCl2 (3.0 mmol/L for 5 minutes) or vehicle (saline solution) was administered before simulated ischemia, with or without concurrent PKC inhibition (bisindolylmaleimide I, 150 nmol/L). RESULTS: Ischemia-reperfusion resulted in decreased postischemic developed force, Ca2+ preconditioning protected human myocardium against ischemia-reperfusion injury (p < 0.05 versus control ischemia-reperfusion), and concurrent PKC inhibition abolished the salutary effect of Ca2+ preconditioning in human myocardium (p < 0.05 versus Ca2+ preconditioning). CONCLUSIONS: Preconditioning with Ca2+ represents a potent means of accessing PKC-mediated protection of the human myocardium against ischemia-reperfusion injury.


Asunto(s)
Calcio/uso terapéutico , Corazón/efectos de los fármacos , Animales , Calcio/administración & dosificación , Cloruro de Calcio/uso terapéutico , Estimulación Cardíaca Artificial , Modelos Animales de Enfermedad , Relación Dosis-Respuesta a Droga , Inhibidores Enzimáticos/farmacología , Atrios Cardíacos/efectos de los fármacos , Atrios Cardíacos/metabolismo , Humanos , Indoles/farmacología , Maleimidas/farmacología , Contracción Miocárdica/fisiología , Infarto del Miocardio/prevención & control , Isquemia Miocárdica/metabolismo , Isquemia Miocárdica/fisiopatología , Isquemia Miocárdica/prevención & control , Daño por Reperfusión Miocárdica/prevención & control , Miocardio/metabolismo , Proteína Quinasa C/antagonistas & inhibidores , Proteína Quinasa C/efectos de los fármacos , Proteína Quinasa C/metabolismo , Receptores de Cinasa C Activada , Receptores de Superficie Celular/antagonistas & inhibidores , Receptores de Superficie Celular/efectos de los fármacos , Receptores de Superficie Celular/metabolismo , Cloruro de Sodio , Factores de Tiempo
14.
Ann Thorac Surg ; 65(3): 868-74, 1998 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-9527242

RESUMEN

Women appear to be protected from cardiovascular disease until the onset of menopause. Considerable evidence supports the atheroprotective effects of endogenous and supplemental estrogens. The beneficial effects of estrogens on lipid metabolism cannot wholly explain this phenomenon. Accumulating data suggest that estrogen may act at the cellular and molecular level to influence atherogenesis. The purpose of this review is to examine lipid-independent mechanisms of estrogen-mediated atheroprotection after cardiovascular injury.


Asunto(s)
Enfermedad de la Arteria Coronaria/fisiopatología , Estrógenos/farmacología , Estrógenos/fisiología , Procedimientos Quirúrgicos Cardíacos , Adhesión Celular/fisiología , Enfermedad de la Arteria Coronaria/etiología , Citocinas/fisiología , Endotelio Vascular/fisiología , Matriz Extracelular/fisiología , Sustancias de Crecimiento/fisiología , Humanos , Leucocitos/fisiología , Músculo Liso Vascular/fisiología , Óxido Nítrico/fisiología , Prostaglandinas/fisiología
15.
Ann Thorac Surg ; 66(2): 313-7, 1998 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-9725362

RESUMEN

BACKGROUND: Inflammatory cytokine production contributes to lung injury after lung ischemia reperfusion and during lung transplant rejection. Although nitric oxide has been demonstrated to reduce lung injury associated with the adult respiratory distress syndrome, it remains unknown whether the mechanism of nitric oxide's beneficial effects involves reducing lung macrophage inflammatory cytokine production. The purpose of this study was to determine whether nitric oxide downregulates lung macrophage inflammatory cytokine production. METHODS: Lung macrophages were harvested by bronchoalveolar lavage (10(6) macrophage per milliliter from normal Sprague-Dawley rats, 6 animals per group) and treated under ex vivo tissue culture conditions with the nitric oxide releasing compound S-nitoso-N-acetyl-D, L-penicillamine (0, 10(-5) 10(-4), 10(-3), 10(-2) mol/L) before induction of inflammatory cytokines with endotoxin, (50 ng/mL for 24 hours). Supernatants were assayed for inflammatory cytokine production (tumor necrosis factor alpha, interleukin-1beta) by enzyme-linked immunosorbent assay. RESULTS: Continuous nitric oxide release by S-nitoso-N-acetyl-D, L-penicillamine decreased lung macrophage tumor necrosis factor-alpha and interleukin-1beta production in a dose-dependent fashion (6 rats per group; data were analyzed for significance [p < 0.05] using two-way analysis of variance with Tukey's post-hoc correction). CONCLUSIONS: Nitric oxide decreases inflammatory cytokine production by lung macrophage. The mechanism of nitric oxide's beneficial effects may be partially attributable to decreased production of inflammatory cytokines. Nitric oxide may serve an expanded role for reducing inflammatory cytokine production during acute lung injury, ischemia-reperfusion-induced inflammation, or lung transplant rejection.


Asunto(s)
Interleucina-1/biosíntesis , Macrófagos Alveolares/metabolismo , Óxido Nítrico/fisiología , Factor de Necrosis Tumoral alfa/biosíntesis , Animales , Células Cultivadas , Dimercaprol , Regulación hacia Abajo , Ensayo de Inmunoadsorción Enzimática , Masculino , Penicilamina/análogos & derivados , Penicilamina/farmacología , Ratas , Ratas Sprague-Dawley
17.
Arch Biochem Biophys ; 304(1): 163-9, 1993 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-8323281

RESUMEN

The protease encoded by the human immunodeficiency virus (HIV) is essential for the processing of viral polyproteins encoded by the gag and pol genes into mature viral proteins. The 99-residue protease from HIV-1 contains two cysteine residues (Cys-67 and Cys-95), both of which are usually conserved in viruses isolated from patients. Despite this conservation, neither residue is required for enzymatic activity. Certain site-specific cysteine mutants of HIV-1 protease are catalytically active, and the protease from HIV-2 lacks both cysteines. Copper is a potent inhibitor of HIV-1 protease, but not of mutants lacking cysteine (A. R. Karlström and R. L. Levine, 1991, Proc. Natl. Acad. Sci. USA 88, 5552-5556). The addition of copper to the protease at pH 5.5 induced aggregation of the protein, providing a possible basis for the inhibitory action of copper. However, addition of both copper and dithiothreitol still led to inhibition of activity but did not cause aggregation. These findings led to a study of the reactivity of the cysteine residues to 5,5'-dithiobis-(2-nitrobenzoic acid) (Ellman's reagent), a sulfhydryl compound which reacts with the ionized form of cysteine residues. At pH 6.2 in 6 M guanidine, no derivatization of cysteine residues occurred, consistent with the typical pKa of cysteine expected for the denatured protein. However, in the same buffer without guanidine, the native protease reacted rapidly with concomitant loss of proteolytic activity. Peptic mapping demonstrated that both Cys-67 and Cys-95 were derivatized. A catalytically active fusion protein of protease with protein A domains was then studied with the expectation that access to Cys-95 would be hindered. This was confirmed, with only Cys-67 reacting rapidly with Ellman's reagent. Enzymatic activity was again lost, indicating that derivatization of the surface-accessible Cys-67 was sufficient to inactivate the enzyme. The reactivity and accessibility of these residues suggest an interesting approach for the development of protease inhibitors which are not directed to the substrate-binding site.


Asunto(s)
Cisteína/química , Proteasa del VIH/química , Cobre/química , Ácido Ditionitrobenzoico/química , Proteasa del VIH/ultraestructura , Inhibidores de la Proteasa del VIH/química , Mapeo Peptídico , Desnaturalización Proteica , Estructura Terciaria de Proteína
18.
J Urol ; 162(1): 196-203, 1999 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-10379787

RESUMEN

Renal ischemia-reperfusion injury induces a cascade of events leading to cellular damage and organ dysfunction. Tumor necrosis factor-alpha (TNF), a potent proinflammatory cytokine, is released from the kidney in response to, and has been implicated in the pathogenesis of, renal ischemia-reperfusion injury. TNF induces glomerular fibrin deposition, cellular infiltration and vasoconstriction, leading to a reduction in glomerular filtration rate (GFR). The signaling cascade through which renal ischemia-reperfusion induces TNF production is beginning to be elucidated. Oxidants released following reperfusion activate p38 mitogen activated protein kinase (p38 MAP kinase) and the TNF transcription factor, NFkappaB, leading to subsequent TNF synthesis. In a positive feedback, proinflammatory fashion, binding of TNF to specific TNF membrane receptors can reactivate NFkappaB. This provides a mechanism by which TNF can upregulate its own expression as well as facilitate the expression of other genes pivotal to the inflammatory response. TNF receptor binding can also induce renal cell apoptosis, the major form of cell death associated with renal ischemia-reperfusion injury. Anti-TNF strategies targeting p38 MAP kinase, NFkappaB, and TNF itself are being investigated as methods of attenuating renal ischemic injury. The control of TNF production and activity represents a realistic goal for clinical medicine.


Asunto(s)
Riñón/irrigación sanguínea , Daño por Reperfusión/etiología , Factor de Necrosis Tumoral alfa/fisiología , Apoptosis , Humanos , Riñón/patología , Daño por Reperfusión/terapia
19.
Crit Care Med ; 28(9): 3314-31, 2000 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-11008997

RESUMEN

OBJECTIVE: To systematically review clinical trials in acute respiratory distress syndrome (ARDS). DATA SOURCES: Computerized bibliographic search of published research and citation review of relevant articles. STUDY SELECTION: All clinical trials of therapies for ARDS were reviewed. Therapies that have been compared in prospective, randomized trials were the focus of this analysis. DATA EXTRACTION: Data on population, interventions, and outcomes were obtained by review. Studies were graded for quality of scientific evidence. MAIN RESULTS: Lung protective ventilator strategy is supported by improved outcome in a single large, prospective trial and a second smaller trial. Other therapies for ARDS, including noninvasive positive pressure ventilation, inverse ratio ventilation, fluid restriction, inhaled nitric oxide, almitrine, prostacyclin, liquid ventilation, surfactant, and immune-modulating therapies, cannot be recommended at this time. Results of small trials using corticosteroids in late ARDS support the need for confirmatory large clinical trials. CONCLUSIONS: Lung protective ventilator strategy is the first therapy found to improve outcome in ARDS. Trials of prone ventilation and fluid restriction in ARDS and corticosteroids in late ARDS support the need for large, prospective, randomized trials.


Asunto(s)
Síndrome de Dificultad Respiratoria/terapia , Ensayos Clínicos como Asunto , Humanos , Estudios Prospectivos , Respiración Artificial , Resultado del Tratamiento
20.
J Surg Res ; 101(2): 210-5, 2001 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-11735278

RESUMEN

BACKGROUND: Local tumor necrosis factor (TNF)-alpha production by resident macrophages (M phi) contributes to posttraumatic tissue injury. Hypoxia decreases cellular cyclic adenosine monophosphate (cAMP) levels and enhances M phi secretion of TNF-alpha following lipopolysaccharide (LPS) stimulation. Thus, tissue hypoxia associated with trauma likely synergizes with proinflammatory mediators in the induction of M phi TNF-alpha production through an influence on cAMP generation or degradation. It is unclear whether elevation of cellular cAMP inhibits LPS-stimulated TNF-alpha production by hypoxic M phi. Moreover, it is unknown whether the synergism of hypoxia with LPS can be abrogated by promotion of cAMP generation or inhibition of cAMP degradation. METHODS: Rat peritoneal M phi were stimulated with Escherichia coli LPS (20 ng/ml) in a normoxic (room air with 5% CO(2)) or hypoxic (95% N(2) with 5% CO(2)) condition. TNF-alpha levels in cell-free supernatants were measured by enzyme-linked immunoassay. The beta-adrenoceptor agonist isoproterenol (ISP; 5.0 microM) and the adenylate cyclase activator forskolin (FSK; 50 microM) were applied to promote cAMP generation. The nonselective cyclic-3',5'-nucleotide phosphodiesterase (PDE) inhibitor 3-isobutyl-1-methylxanthine (IBMX; 1.0 mM) and the PDE III-specific inhibitor milrinone (200 microM) were used to inhibit cAMP degradation. The nondegradable cAMP analogue dibutyryl cAMP (dbcAMP; 100 microM) was applied to further determine the role of PDE. RESULTS. Although hypoxia alone had a minimal effect on TNF-alpha production, it dramatically enhanced LPS-stimulated TNF-alpha production (4.08 +/- 0.28 ng/10(6) cells in hypoxia plus LPS vs 1.63 +/- 0.26 ng/10(6) cells in LPS, 2.5-fold, P < 0.01). Promotion of cAMP generation by either ISP or FSK reduced TNF-alpha production by hypoxic cells. However, neither of these two agents abolished the synergism of hypoxia with LPS (1.68 +/- 0.13 ng/10(6) cells in ISP plus hypoxia plus LPS vs 0.55 +/- 0.04 ng/10(6) cells in ISP plus LPS, threefold; 1.17 +/- 0.03 ng/10(6) cells in FSK plus hypoxia plus LPS vs 0.33 +/- 0.02 ng/10(6) cells in FSK plus LPS, 3.5-fold; both P < 0.01). Inhibition of cAMP degradation with IBMX reduced TNF-alpha production in hypoxic cells and abrogated the synergism (0.31 +/- 0.11 ng/10(6) cells in IBMX plus hypoxia plus LPS vs 0.27 +/- 0.04 ng/10(6) cells in IBMX plus LPS, P > 0.05), and the PDE III inhibitor milrinone had a comparable effect. Moreover, dbcAMP also attenuated TNF-alpha production with abrogation of the synergistic effect of hypoxia (0.56 +/- 0.08 ng/10(6) cells in dbcAMP plus hypoxia plus LPS vs 0.46 +/- 0.04 ng/10(6) cells in dbcAMP plus LPS, P > 0.05). CONCLUSIONS: The results show that elevation of cellular cAMP, either by promotion of generation or by inhibition of degradation, suppresses LPS-stimulated TNF-alpha production in hypoxic M phi. It appears that hypoxia synergizes with LPS in the induction of M phi TNF-alpha production through PDE-mediated cAMP degradation. Inhibition of PDE may be a therapeutic approach for suppression of synergistic induction of M phi TNF-alpha production by hypoxia and LPS in posttraumatic tissue.


Asunto(s)
3',5'-AMP Cíclico Fosfodiesterasas/antagonistas & inhibidores , AMP Cíclico/fisiología , Inhibidores Enzimáticos/farmacología , Hipoxia/inmunología , Lipopolisacáridos/toxicidad , Macrófagos/metabolismo , Factor de Necrosis Tumoral alfa/biosíntesis , Animales , Masculino , Ratas , Ratas Sprague-Dawley
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA