Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 17 de 17
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
Am J Physiol Gastrointest Liver Physiol ; 306(5): G445-53, 2014 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-24407593

RESUMEN

Intestinal intussusception (ISS) commonly causes intestinal obstruction in children. One mechanism that has been proposed to cause ISS is inflammation-induced alteration of intestinal motility. We investigated whether innate inflammatory factors or altered motility is required for induction of ISS by LPS. We compared rates of ISS among BALB/c and C57BL/6 mice, mice lacking lymphocytes or depleted of phagocytes, or mice with defects in the Toll-like receptor 4 (TLR4) signaling pathway following administration of LPS or the Ca(2+) analog MnCl2. At 6 or 2 h after administration of LPS or MnCl2, respectively, mice underwent image analysis to assess intestinal contraction rate or laparotomy to identify ISS. LPS-induced ISS (LPS-ISS) was observed in BALB/c mice, but not in C57BL/6 mice or any BALB/c mice with disruptions of TLR4 signaling. LPS-induced serum TNF-α, IL-6, and nitric oxide (NO) and intestinal NO levels were similar in BALB/c and C57BL/6 mice. The rate of LPS-ISS was significantly reduced in phagocyte-depleted, but not lymphocyte-deficient, mice. Intestinal contraction rates were reduced in LPS-ISS-susceptible BALB/c mice, but not in LPS-ISS-resistant C57BL/6 or TLR4 mutant mice, suggesting a role for reduced intestinal contraction rate in LPS-ISS susceptibility. This was tested with MnCl2, a Ca(2+) antagonist that reduced intestinal contraction rates and induced ISS, irrespective of mouse strain. Therefore, LPS-ISS is initiated by innate immune signaling that requires TLR4 and phagocytes but may be independent of TNF-α, IL-6, and NO levels. Furthermore, alteration of intestinal motility, specifically, reduced intestinal contraction rate, is a key factor in the development of ISS.


Asunto(s)
Cloruros/toxicidad , Motilidad Gastrointestinal/efectos de los fármacos , Inmunidad Innata/fisiología , Intususcepción/inducido químicamente , Lipopolisacáridos/toxicidad , Proteínas de Fase Aguda/genética , Proteínas de Fase Aguda/metabolismo , Animales , Proteínas Portadoras/genética , Proteínas Portadoras/metabolismo , Femenino , Regulación de la Expresión Génica/inmunología , Inmunidad Innata/efectos de los fármacos , Intususcepción/inmunología , Intususcepción/metabolismo , Receptores de Lipopolisacáridos/genética , Receptores de Lipopolisacáridos/metabolismo , Masculino , Compuestos de Manganeso , Glicoproteínas de Membrana/genética , Glicoproteínas de Membrana/metabolismo , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Ratones SCID , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo
2.
J Trauma ; 68(1): 69-75, 2010 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-20065760

RESUMEN

BACKGROUND: Although ketamine has many beneficial effects in a rat model of noninfectious inflammation with lipopolysaccharide (LPS), its effects on gut ileus are unknown. We hypothesized that ketamine would improve LPS-induced ileus and therefore examined its effects on gastric emptying and intestinal transit as well as duodenogastric bile reflux and associated gastric bleeding. METHODS: Male rats received saline or ketamine (7 mg/kg ip) 1 hour before saline or LPS (20 mg/kg ip) for 5 hours. Thirty minutes before killing, rats received orogastric rhodamine B isothiocyanate-labeled dextran and 5 minutes later fluorescein isothiocyanate-labeled dextran via a duodenal catheter. GI contents were collected for dye, bile acid, and hemoglobin (index of bleeding) determinations. RESULTS: LPS significantly impaired intestinal transit and increased duodenogastric bile reflux and gastric luminal hemoglobin content. Ketamine improved intestinal transit, prevented LPS-induced bile reflux, and diminished gastric bleeding. In mechanistic studies, ketamine also attenuated LPS-induced upregulation of the proinflammatory genes inducible nitric oxide synthase and cyclo-oxygenase-2 in the stomach but preserved expression of the anti-inflammatory gene heme-oxygenase-1 (Western blot). CONCLUSIONS: These data suggest that ketamine may prevent LPS-induced gastric bleeding by decreasing bile reflux through improved intestinal transit or by local changes in nitric oxide, prostaglandin, and carbon monoxide metabolism.


Asunto(s)
Reflujo Duodenogástrico/fisiopatología , Hemorragia Gastrointestinal/fisiopatología , Ketamina/farmacología , Animales , Antiinflamatorios/farmacología , Ciclooxigenasa 2/metabolismo , Reflujo Duodenogástrico/inducido químicamente , Escherichia coli , Vaciamiento Gástrico/efectos de los fármacos , Mucosa Gástrica/metabolismo , Contenido Digestivo/química , Hemorragia Gastrointestinal/inducido químicamente , Hemorragia Gastrointestinal/diagnóstico , Hemorragia Gastrointestinal/tratamiento farmacológico , Tránsito Gastrointestinal/efectos de los fármacos , Hemo-Oxigenasa 1/metabolismo , Hemoglobinas/análisis , Hipnóticos y Sedantes/farmacología , Lipopolisacáridos , Masculino , Óxido Nítrico Sintasa de Tipo II/metabolismo , Ratas , Ratas Sprague-Dawley
3.
J Clin Invest ; 118(4): 1491-501, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18340377

RESUMEN

Priapism, abnormally prolonged penile erection in the absence of sexual excitation, is associated with ischemia-mediated erectile tissue damage and subsequent erectile dysfunction. It is common among males with sickle cell disease (SCD), and SCD transgenic mice are an accepted model of the disorder. Current strategies to manage priapism suffer from a poor fundamental understanding of the molecular mechanisms underlying the disorder. Here we report that mice lacking adenosine deaminase (ADA), an enzyme necessary for the breakdown of adenosine, displayed unexpected priapic activity. ADA enzyme therapy successfully corrected the priapic activity both in vivo and in vitro, suggesting that it was dependent on elevated adenosine levels. Further genetic and pharmacologic evidence demonstrated that A2B adenosine receptor-mediated (A2BR-mediated) cAMP and cGMP induction was required for elevated adenosine-induced prolonged penile erection. Finally, priapic activity in SCD transgenic mice was also caused by elevated adenosine levels and A2BR activation. Thus, we have shown that excessive adenosine accumulation in the penis contributes to priapism through increased A2BR signaling in both Ada -/- and SCD transgenic mice. These findings provide insight regarding the molecular basis of priapism and suggest that strategies to either reduce adenosine or block A2BR activation may prove beneficial in the treatment of this disorder.


Asunto(s)
Adenosina/metabolismo , Erección Peniana , Pene/metabolismo , Receptor de Adenosina A2B/metabolismo , Transducción de Señal , Adenosina/farmacología , Adenosina Desaminasa/deficiencia , Adenosina Desaminasa/genética , Adenosina Desaminasa/metabolismo , Animales , AMP Cíclico/biosíntesis , GMP Cíclico/biosíntesis , Fibrosis/genética , Fibrosis/metabolismo , Fibrosis/patología , Masculino , Ratones , Ratones Noqueados , Miocitos del Músculo Liso/efectos de los fármacos , Erección Peniana/efectos de los fármacos , Pene/anatomía & histología , Pene/irrigación sanguínea , Transducción de Señal/efectos de los fármacos , Factores de Tiempo
4.
Crit Care Med ; 35(2): 538-43, 2007 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17205008

RESUMEN

INTRODUCTION: Acute edema induced by resuscitation and mesenteric venous hypertension impairs intestinal transit and contractility and reduces intestinal stiffness. Pretreatment with hypertonic saline (HS) can prevent these changes. Changes in tissue stiffness have been shown to trigger signaling cascades via stress fiber formation. We proposed that acute intestinal edema leads to a decrease in intestinal transit that may be mediated by changes in stiffness, leading to stress fiber formation and decreased intestinal transit. Furthermore, HS administration will abolish these detrimental effects of edema. RESULTS: Intestinal edema causes a significant increase in tissue water and a significant decrease in intestinal transit and stiffness compared with sham. HS reversed these changes to sham levels. In addition, tissue edema led to significant stress fiber formation and decreased numbers of focal contacts. HS preserved tissue stiffness, prevented stress fiber formation, and was associated with improved intestinal function. CONCLUSION: HS eliminates intestinal tissue edema formation and improves intestinal transit. In addition, the action of HS may be mediated through its preservation of tissue stiffness, which leads to prevention of signaling via stress fiber formation, leading to preserved intestinal function. Finally, intestinal edema may provide a novel physiologic model for examining stiffness and stress fiber signaling.


Asunto(s)
Modelos Animales de Enfermedad , Edema/tratamiento farmacológico , Tránsito Gastrointestinal/efectos de los fármacos , Enfermedades Intestinales/tratamiento farmacológico , Solución Salina Hipertónica/uso terapéutico , Enfermedad Aguda , Animales , Enfermedades Intestinales/fisiopatología , Masculino , Ratas , Ratas Sprague-Dawley , Recuperación de la Función
5.
J Surg Res ; 138(1): 106-10, 2007 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-17161426

RESUMEN

BACKGROUND: Acute intestinal edema adversely affects intestinal transit, permeability, and contractility. Current resuscitation modalities, while effective, are associated with development of acute intestinal edema. Knowledge of levels of tissue edema would allow clinicians to monitor intestinal tissue water and may help prevent the detrimental effects of edema. However, there is no simple method to measure intestinal tissue water without biopsy. We sought to develop a tissue impedance analyzer to measure tissue edema, without the need for invasive biopsy. METHODS: Oscillating voltage input was applied to the analyzer circuit and an oscilloscope measured the voltage output across any load. Rats were randomized to three groups: sham, mild edema (80 mL/kg of NS resuscitation), and severe edema (80 mL/kg of NS resuscitation with intestinal venous hypertension). Intestinal edema was measured by wet-to-dry tissue weight ratio. Bowel impedance was measured and converted to capacitance using a standard curve. RESULTS: Acute intestinal edema causes a significant increase in bowel capacitance. This capacitance can be used to predict tissue water concentration. CONCLUSION: Using an impedance analyzer circuit, it is possible to measure intestinal edema reliably and quickly. This may prove to be a useful tool in the resuscitation of critically ill patients.


Asunto(s)
Edema/diagnóstico , Impedancia Eléctrica , Electrónica Médica/instrumentación , Electrónica Médica/métodos , Intestinos/fisiopatología , Enfermedad Aguda , Animales , Edema/fisiopatología , Capacidad Eléctrica , Electrónica Médica/normas , Diseño de Equipo , Intestinos/patología , Masculino , Tamaño de los Órganos , Ratas , Ratas Sprague-Dawley , Reproducibilidad de los Resultados , Índice de Severidad de la Enfermedad
6.
J Trauma ; 61(1): 66-73; discussion 73-4, 2006 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-16832251

RESUMEN

BACKGROUND: Hypertonic saline (HS) resuscitation prevents neutrophil mediated injury after shock. The optimal dose is not known, but appears as a result of osmotic stress. We hypothesized that a dose dependent effect exists related to increasing tonicity and that the optimal gut protective dose would provide better protection against remote organ injury than large volume isotonic crystalloids. METHODS: In experiment 1, rats were assigned to controls (sham/no resuscitation, sham/4 mL/kg 7.5% HS, superior mesenteric artery occlusion [SMAO]/no resuscitation), SMAO/equal volume (4 mL/kg 0.9% NS, 4 mL/kg 2.5% HS, 4 mL/kg 5% HS, 4 mL/kg 7.5% HS and 4 mL/kg 10% HS) or SMAO/equal sodium (33 mL/kg 0.9% NS, 12 mL/kg 2.5% HS, 6 mL/kg 5% HS, 4 mL/kg 7.5% HS, and 3 mL/kg 10% HS). In experiment 2, rats were assigned to the same control groups, and to either SMAO/NS (33 mL/kg 0.9% NS, equal salt load) or SMAO/HS (4 mL/kg 7.5% HS). The SMAO was clamped for 60 minutes and boluses given 5 minutes before clamp removal. After 6 hours of reperfusion, ileum and lungs were harvested for analysis of histologic injury, myeloperoxidase (MPO) as an index of neutrophil mediated injury, and serum ALT and AST drawn as markers of liver injury. RESULTS: In experiment 1, equal volume and equal sodium decreased injury and inflammation with increasing tonicity in a dose dependent fashion, with the optimal effect seen at 7.5%. In experiment 2, NS resuscitation resulted in minimal improvement of SMAO-induced lung injury and inflammation or increases in serum ALT and AST whereas HS resuscitation significantly decreased these parameters. CONCLUSION: The protective effect of HS is related to increased tonicity. While NS had little effect on SMAO-induced remote organ injury, optimal dose HS resuscitation was quite protective. This supports the growing evidence that HS protection may be because of its gut protective effects.


Asunto(s)
Fluidoterapia/métodos , Daño por Reperfusión/prevención & control , Solución Salina Hipertónica/farmacología , Choque Hemorrágico/terapia , Circulación Esplácnica/efectos de los fármacos , Análisis de Varianza , Animales , Soluciones Cristaloides , Relación Dosis-Respuesta a Droga , Íleon/irrigación sanguínea , Íleon/efectos de los fármacos , Íleon/patología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Soluciones Isotónicas/farmacología , Soluciones Isotónicas/uso terapéutico , Pulmón/irrigación sanguínea , Pulmón/efectos de los fármacos , Pulmón/patología , Masculino , Ratas , Ratas Sprague-Dawley , Solución Salina Hipertónica/administración & dosificación
7.
Crit Care Med ; 34(6): 1713-8, 2006 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-16625118

RESUMEN

OBJECTIVE: We have shown that acute edema induced by mesenteric venous hypertension (MV-HTN) impairs intestinal transit and reduces the standardized engineering measures of intestinal stiffness (elastic modulus) and residual stress (opening angle). We hypothesized that hypertonic saline (7.5%) would reverse these detrimental effects of acute edema. DESIGN: Laboratory study. SETTING: University laboratory. SUBJECTS: Male Sprague Dawley rats (270-330 g). INTERVENTIONS: Rats were randomized to five groups: sham, MV-HTN alone, MV-HTN with 4 mL/kg normal saline resuscitation (equal volume), MV-HTN with 33 mL/kg normal saline resuscitation (equal salt), and MV-HTN with 4 mL/kg hypertonic saline. Intestinal edema was measured by wet to dry tissue weight ratio. A duodenal catheter was placed and, 30 mins before death, fluorescein isothiocyanate Dextran was injected. At death, dye concentrations were measured to determine intestinal transit. Segments of distal ileum were hung to a fixed point in a tissue bath and to a force displacement transducer and stretched in increments of 1 mm; we recorded the new length and the corresponding force from the force displacement transducer to determine elastic modulus. Next, two transverse cuts were made yielding a 1- to 2-mm thick ring-shaped segment of tissue which was then cut radially to open the ring. Then the opening angle was measured. MEASUREMENTS AND MAIN RESULTS: MV-HTN, MV-HTN with 4 mL/kg normal saline, and MV-HTN with 33 mL/kg normal saline caused a significant increase in tissue edema and a significant decrease in intestinal transit, stiffness, and residual stress compared with sham. Hypertonic saline significantly lessened the effect of edema on intestinal transit and prevented the changes in stiffness and residual stress. CONCLUSIONS: Hypertonic saline prevented intestinal tissue edema. In addition, hypertonic saline improved intestinal transit, possibly through more efficient transmission of muscle force through stiffer intestinal tissue.


Asunto(s)
Edema/prevención & control , Ileus/prevención & control , Resucitación/métodos , Solución Salina Hipertónica/uso terapéutico , Animales , Modelos Animales de Enfermedad , Edema/etiología , Edema/metabolismo , Ileus/etiología , Ileus/metabolismo , Masculino , Concentración Osmolar , Ratas , Ratas Sprague-Dawley , Solución Salina Hipertónica/administración & dosificación , Sodio/sangre , Presión Venosa
8.
Surgery ; 139(1): 54-60, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16364718

RESUMEN

BACKGROUND: The late phase of post-traumatic multisystem organ failure is associated with sepsis from organisms that normally reside within the gut's lumen. Morphine, a commonly employed analgesic in injured patients, is associated with intestinal stasis, bacterial overgrowth, and translocation when administered to rats. N-formyl-methionyl-leucyl-phenylalanine (FMLP), a toxic product of gram-negative organisms, provokes an increase in mucosal permeability when infused into the ileal lumen of this species. The current study was designed to examine the effects of morphine on FMLP perturbation of the mucosal barrier of the ileum of rats and mice to an impermeant macromolecule, dextran 4400. The potential role of mucosal mast cells in the response to either agent alone or in combination was examined. METHODS: Intact and isolated segments of distal ileum of naïve and sensitized (Trichinella spiralis and egg albumin) Sprague-Dawley rats were exposed to FMLP with or without morphine or doxantrazole, a mast cell-stabilizing agent. Isolated segments of distal ileum of mast cell-deficient mice also were studied. RESULTS: Mucosal exposure of distal ileal mucosa (intact and isolated, and naive and sensitized) to FMLP was associated with an increase in permeability to dextran 4400, which was completely ablated by morphine and doxantrazole. Sensitization was associated with a prolongation of the FMLP response. Ilea of mast cell-deficient mice (but not their wild type litter mates) were unresponsive to FMLP. CONCLUSIONS: Morphine antagonizes the provocative effect of FMLP on the mucosal barrier to dextran 4400 of the ilea of rats and mice. Intestinal mucosal mast cells play a central role in the process.


Asunto(s)
Analgésicos Opioides/farmacología , Íleon/metabolismo , Mucosa Intestinal/metabolismo , Mastocitos/fisiología , Morfina/farmacología , Aminas/metabolismo , Animales , Citocinas/metabolismo , Dextranos/farmacocinética , Inmunización , Técnicas In Vitro , Masculino , Mastocitos/efectos de los fármacos , Ratones , Ratones Mutantes , N-Formilmetionina Leucil-Fenilalanina/antagonistas & inhibidores , N-Formilmetionina Leucil-Fenilalanina/farmacología , Permeabilidad/efectos de los fármacos , Ratas/inmunología , Ratas/parasitología , Ratas Sprague-Dawley , Tioxantenos/farmacología , Trichinella spiralis/inmunología , Xantonas/farmacología
9.
J Trauma ; 59(5): 1092-8, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16385285

RESUMEN

BACKGROUND: We have previously demonstrated that hypertonic saline (HS) resuscitation decreased inflammation and mucosal injury after mesenteric ischemia/reperfusion (I/R). In contrast to I/R cell necrosis, apoptosis provides controlled cell death that minimizes inflammation. We therefore hypothesized that HS resuscitation after mesenteric I/R would induce apoptosis and decrease mucosal injury. METHODS: Rats underwent 60 minutes of superior mesenteric artery occlusion (SMAO) and then received no resuscitation or resuscitation with 4 mL/kg of HS, 4 mL/kg of lactated Ringer's (LR) solution (equal volume), or 32 mL/kg of LR solution (equal salt load). Rats were killed at 6 hours of reperfusion, and ileum was harvested for analysis. DNA fragmentation (apoptosis) was assessed by terminal deoxynucleotidyl transferase-mediated dUTP-biotin nick end-labeling (TUNEL) and mucosal injury by histology (Chiu score 0-5). Caspase-3 (proapoptotic mediator) and Bcl-xL (antiapoptotic mediator) protein expression were analyzed by Western immunoblot. RESULTS: SMAO with no resuscitation, SMAO with 4 mL/kg of LR, and SMAO with 32 mL/kg of LR increased apoptosis (quantitated by TUNEL) and I/R-induced mucosal injury (quantitated by Chiu score). This was associated with an increase to similar levels in both proapoptotic caspase-3 and antiapoptotic Bcl-xL protein expression. Moreover, SMAO with 4 mL/kg of HS further increased apoptosis but decreased mucosal injury. This was associated with a differential expression of proapoptotic caspase-3 over antiapoptotic Bcl-xL. CONCLUSION: HS resuscitation after mesenteric I/R significantly increased ileal mucosal apoptosis while decreasing mucosal injury and may represent a novel mechanism by which HS resuscitation after mesenteric I/R reduces inflammation and imparts protection to the gut.


Asunto(s)
Mucosa Intestinal/fisiopatología , Daño por Reperfusión/terapia , Animales , Apoptosis , Caspasa 3 , Caspasas/metabolismo , Íleon/fisiopatología , Etiquetado Corte-Fin in Situ , Masculino , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/fisiopatología , Solución Salina Hipertónica/uso terapéutico
10.
Shock ; 24(5): 462-9, 2005 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-16247333

RESUMEN

Cyclooxygenase (COX)-2 has been identified as an important mediator elaborated during ischemia/reperfusion, with pro- and anti-inflammatory properties having been reported. As the role of COX-2 in the small intestine remains unclear, we hypothesized that COX-2 expression would mediate mesenteric ischemia/reperfusion-induced gut injury, inflammation, and impaired transit and that these deleterious effects could be reversed by the selective COX-2 inhibitor, N-[2-(cyclohexyloxy)-4-nitrophenyl] methanesulphanamide (NS-398). Additionally, we sought to determine the role of peroxisome proliferator-activated receptor gamma (PPARgamma) in mediating protection by NS-398 in this model. Rats underwent sham surgery or were pretreated with NS-398 (3, 10, or 30 mg/kg) intraperitoneally 1 h before 60 min of superior mesenteric artery occlusion and 30 min to 6 h of reperfusion. In some experiments, NS-398 (30 mg/kg) was administered postischemia. Ileum was harvested for COX-2 mRNA and protein, PGE2, myeloperoxidase (inflammation), histology (injury), intestinal transit and PPARgamma protein expression, and DNA-binding activity. COX-2 expression and PGE2 production increased after mesenteric ischemia/reperfusion and were associated with gut inflammation, injury, and impaired transit. Inhibition of COX-2 by NS-398 (30 mg/kg, but not 3 or 10 mg/kg) not only reversed the deleterious effects of COX-2, but additionally induced expression and nuclear translocation of PPARgamma. NS-398 given postischemia was equally protective. In conclusion, COX-2 may function as a proinflammatory mediator in a rodent model of mesenteric ischemia/reperfusion. Reversal of gut inflammation, injury, and impaired transit by high-dose NS-398 is associated with PPAR activation, suggesting a potential role for PPAR-gamma in shock-induced gut protection.


Asunto(s)
Ciclooxigenasa 2/metabolismo , Mesenterio/patología , PPAR gamma/metabolismo , Daño por Reperfusión/metabolismo , Transporte Activo de Núcleo Celular , Animales , Núcleo Celular/metabolismo , Ciclooxigenasa 1/metabolismo , Inhibidores de la Ciclooxigenasa/farmacología , Citoplasma/metabolismo , Citosol/metabolismo , ADN/química , Cartilla de ADN/química , Dinoprostona/metabolismo , Relación Dosis-Respuesta a Droga , Tracto Gastrointestinal/patología , Inflamación , Intestino Delgado/patología , Isquemia/patología , Masculino , Nitrobencenos/farmacología , Peroxidasa/metabolismo , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Sulfonamidas/farmacología , Factores de Tiempo
11.
Folia Med Cracov ; 46(1-2): 75-82, 2005.
Artículo en Inglés | MEDLINE | ID: mdl-17037289

RESUMEN

Changes of intestinal motility and transit due to morphine administration are attributed to its central and peripheral action, however coexistence of both central and peripheral mu receptors in morphine dependence hasn't been clearly demonstrated so far. Our purpose was the evaluation of the effect of either CNS or ENS mu receptor blockade on intestinal motility in morphine dependent rats. Twenty male rats were subjected to chronic subcutaneous morphine infusions for 72 hrs. On each day motility indices (MI) were analysed in the animals' duodenum and ascending colon before and after either intraperitoneal (IP) or intracerebroventricular (ICV) mu antagonist (CTOP) administration. Tolerance of the intestinal motility to morphine developed in both the duodenum and the ascending colon after 72 hrs of the infusion. Dependence was observed at 24 hrs and maintained at 48 and 72 hrs of morphine administration. On each day no differences of MI reaction to mu blockade were visible regarding the IP or ICV CTOP administration. Our results suggest the involvement of both the CNS and the ENS mu opioid receptors in mechanisms of the intestinal tolerance to and dependence upon morphine.


Asunto(s)
Sistema Nervioso Central/fisiopatología , Motilidad Gastrointestinal/efectos de los fármacos , Dependencia de Morfina/fisiopatología , Morfina/farmacología , Sistema Nervioso Periférico/fisiopatología , Receptores Opioides mu/metabolismo , Animales , Sistema Nervioso Central/efectos de los fármacos , Modelos Animales de Enfermedad , Masculino , Sistema Nervioso Periférico/efectos de los fármacos , Ratas , Ratas Sprague-Dawley , Receptores Opioides mu/efectos de los fármacos
12.
J Trauma ; 56(4): 749-58; discussion 758-9, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15187737

RESUMEN

BACKGROUND: Hypertonic saline (HTS) has been shown to modulate the inflammatory response after shock. We have previously demonstrated that heme oygenase-1 (HO-1) induction is protective against gut dysfunction in models of shock-induced gut ischemia/reperfusion (I/R). We therefore hypothesized that HTS prevents gut inflammation, injury, and impaired transit by inducing HO-1 in a model of gut I/R. METHODS: Rats underwent 60 minutes of superior mesenteric artery occlusion (SMAO) and then were resuscitated with 4 mL/kg of HTS, an equal volume of lactated Ringer's (LR) solution (4 mL/kg, low volume), or equal salt LR solution (32 mL/kg, high volume) and compared with SMAO alone or shams. A separate group was pretreated with the HO-1 blocker Sn protoporphyrin IX (SNP IX) before SMAO plus HTS. At 6 hours of reperfusion, transit was determined and ileum harvested for HO-1 (anti-inflammatory) and inducible nitric oxide synthase (proinflammatory) immunoreactivity, myeloperoxidase (MPO) activity, and histologic injury. Data are expressed as mean +/- SEM (analysis of variance). RESULTS: Intestinal transit was severely impaired after SMAO (2.5 +/- 0.1), improved with low- and high-volume LR solution (3.2 +/- 0.2 and 3.1 +/- 0.1, not significant), but returned to sham (4.6 +/- 0.2) with HTS (4.8 +/- 0.2). Pretreatment with SNP abrogated this protective effect. Myeloperoxidase activity was significantly increased by SMAO (SMAO, 2.3 +/- 0.3; sham, 0.4 +/- 0.05), lessened by low- and high-volume LR solution (1.5 +/- 0.3 and 1.7 +/- 0.4), but returned to sham levels with HTS (1.0 +/- 0.01). Activity with SNP IX pretreatment was significantly increased (4.04 +/- 0.8). Mucosal injury followed a similar pattern. Inducible nitric oxide synthase was increased by SMAO and low- and high-volume LR solution (0.8 +/- 0.2, 0.8 +/- 0.03, and 0.8 +/- 0.02, respectively; sham, 0.5 +/- 0.02), but significantly reduced by HTS (0.7 +/- 0.02). HO-1 was induced by SMAO and low- and high-volume LR solution (0.33 +/- 0.02, 0.32 +/- 0.03, and 0.37 +/- 0.4, respectively; sham, 0.0 +/- 0.0), but was further increased with HTS (0.49 +/- 0.04). CONCLUSION: HTS resuscitation protects against inflammation, injury, and impaired intestinal transit after gut I/R in part by inducing HO-1. This is a novel mechanism of HO-1 protection.


Asunto(s)
Inducción Enzimática/efectos de los fármacos , Hemo Oxigenasa (Desciclizante)/biosíntesis , Inflamación/prevención & control , Daño por Reperfusión/prevención & control , Solución Salina Hipertónica/uso terapéutico , Animales , Hemo Oxigenasa (Desciclizante)/metabolismo , Hemo-Oxigenasa 1 , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/lesiones , Masculino , Óxido Nítrico Sintasa/biosíntesis , Óxido Nítrico Sintasa/metabolismo , Óxido Nítrico Sintasa de Tipo II , Ratas , Ratas Sprague-Dawley , Resucitación/métodos , Choque/enzimología , Choque/metabolismo , Choque/terapia
13.
Shock ; 20(6): 483-92, 2003 12.
Artículo en Inglés | MEDLINE | ID: mdl-14625470

RESUMEN

Abdominal compartment syndrome (ACS) has emerged to be a significant problem in patients who develop postinjury multiple organ failure (MOF). Current laboratory research suggests that ACS could be a second hit for the development of MOF. Recent studies demonstrate that ACS is an independent predictor of MOF and that the prevention of ACS decreases the incidence of MOF. The Trauma Research Centers at the University of Colorado and the University of Texas-Houston Medical School are focused on defining the role of the gut in postinjury MOF. Because ACS is a plausible modifiable risk factor, our interest has been to 1) describe the epidemiology of ACS, 2) build prediction models, 3) provide strategies for prevention and treatment of ACS, and 4) develop relevant laboratory models. This review summarizes our findings.


Asunto(s)
Abdomen , Síndromes Compartimentales/etiología , Síndromes Compartimentales/prevención & control , Insuficiencia Multiorgánica/diagnóstico , Traumatismo Múltiple , Adulto , Síndromes Compartimentales/diagnóstico , Sistema Digestivo/patología , Edema , Femenino , Humanos , Modelos Logísticos , Masculino , Modelos Teóricos , Perfusión , Presión , Factores de Riesgo , Vejiga Urinaria/patología
14.
J Surg Res ; 115(1): 48-55, 2003 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-14572772

RESUMEN

PURPOSE: Gut ischemia/reperfusion (I/R) elicits an inflammatory response that impairs intestinal transit. We have previously shown that regional intraischemic hypothermia (IH) protects against moderate gut I/R-induced mucosal injury, is associated with decreased NF-kappaB activity and inducible nitric oxide synthase induction and preserves heme oxygenase-1 (HO-1) expression. HO-1 provides cytoprotection in various models of oxidant stress. We, therefore, tested the hypothesis that IH protects against gut I/R-induced impaired intestinal transit via HO-1 induction. MATERIALS AND METHODS: At laparotomy (lap), Sprague-Dawley rats had duodenal catheters placed followed by sham or gut I/R (superior mesenteric artery occlusion for 75 min) with or without regional IH (15 degrees C). Each animal was placed on a heating blanket maintaining systemic normothermia (37 degrees C). At 12 or 24 h of reperfusion, small intestinal transit was determined by quantitating the distribution of a tracer (FITC dextran) in the intestine 30 min after instillation (expressed as geometric center of distribution). Ileal samples were obtained for histology and HO-1 expression, assessed by Western immunoblot at 12 and 24 h of reperfusion. In separate experiments, rats were pretreated with an HO-1 inhibitor Sn protoporphyrin IX (25 mumol/kg, ip), 1 h before superior mesenteric artery occlusion and transit measured as above. RESULTS: Rats treated with I/R had increased histological injury and impaired intestinal transit at both 12 and 24 h compared with sham. Rats treated with I/R+IH exhibited histological injury and transit comparable with sham controls. I/R induced HO-1 expression at 12 and 24 h of reperfusion and IH augmented this I/R-induced HO-1 expression. Sn protoporphyrin IX abrogated IH protection against histological injury and impaired transit. CONCLUSION: We conclude that intraischemic regional hypothermia protects against histological injury and impaired intestinal transit caused by severe gut I/R injury. Hypothermic protection under these conditions is in part due to HO-1 expression.


Asunto(s)
Tránsito Gastrointestinal , Hemo Oxigenasa (Desciclizante)/análisis , Hipotermia Inducida , Intestinos/irrigación sanguínea , Daño por Reperfusión/prevención & control , Animales , Western Blotting , Constricción , Inhibidores Enzimáticos/farmacología , Hemo Oxigenasa (Desciclizante)/antagonistas & inhibidores , Hemo Oxigenasa (Desciclizante)/biosíntesis , Hemo-Oxigenasa 1 , Íleon/enzimología , Íleon/patología , Intestino Delgado/fisiopatología , Masculino , Arteria Mesentérica Superior , Metaloporfirinas/farmacología , Protoporfirinas/farmacología , Ratas , Ratas Sprague-Dawley , Daño por Reperfusión/patología , Daño por Reperfusión/fisiopatología
16.
Gastroenterology ; 123(3): 810-6, 2002 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-12198707

RESUMEN

BACKGROUND & AIMS: Gastrointestinal function may be impaired after severe injury, hampering tolerance to enteral nutrition. The purpose of this study was to determine how different sodium-coupled solutes modulate gut function after ischemia/reperfusion (I/R) in a rodent model. METHODS: At laparotomy, rats had jejunal sacs filled with (glucose + alanine), glucose, glutamine, alanine, or mannitol (osmotic control), followed by superior mesenteric artery clamping for 60 minutes and 30 minutes of reperfusion. After reperfusion, sacs were harvested for morphologic examination, adenosine triphosphate (ATP) assay, or mounted in an Ussing chamber. RESULTS: Small intestinal epithelial absorptive capacity, as assessed by changes in short-circuit current in response to glucose, after gut I/R, was decreased by alanine or (glucose + alanine) but not glucose or glutamine alone and correlated with changes in tissue ATP. Gut I/R caused a significant morphologic injury that was worsened by alanine or (glucose + alanine) but lessened by glucose or glutamine alone. CONCLUSIONS: During gut I/R, alanine can enhance gut injury, deplete energy (ATP), and impair gut absorption, whereas glucose or glutamine is protective against injury and can maintain absorptive capacity and ATP. These results suggest that solutes (such as alanine), which further stress an already metabolically stressed bowel, should be cautiously administered to critically ill patients whereas those solutes that contribute to energy production (such as glucose and glutamine) may be safely continued.


Asunto(s)
Aminoácidos/farmacología , Glucosa/farmacología , Mucosa Intestinal/patología , Isquemia/metabolismo , Yeyuno/irrigación sanguínea , Yeyuno/patología , Daño por Reperfusión/metabolismo , Adenosina Trifosfato/metabolismo , Alanina/metabolismo , Alanina/farmacología , Aminoácidos/metabolismo , Animales , Transporte Biológico/efectos de los fármacos , Combinación de Medicamentos , Glucosa/metabolismo , Glutamina/metabolismo , Glutamina/farmacología , Mucosa Intestinal/irrigación sanguínea , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Yeyuno/efectos de los fármacos , Yeyuno/metabolismo , Ratas , Ratas Sprague-Dawley , Sodio/metabolismo
17.
Am J Physiol Gastrointest Liver Physiol ; 282(6): G1059-68, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12016132

RESUMEN

Mesenteric ischemia-reperfusion (I/R) injury to the intestine is a common and often devastating clinical occurrence for which there are few therapeutic options. alpha-Melanocyte-stimulating hormone (alpha-MSH) is a tridecapeptide released by the pituitary gland and immunocompetent cells that exerts anti-inflammatory actions and abrogates postischemic injury to the kidneys and brainstem of rodents. To test the hypothesis that alpha-MSH would afford similar protection in the postischemic small intestine, we analyzed the effects of this peptide on intestinal transit, histology, myeloperoxidase activity, and nuclear factor-kappaB (NF-kappaB) activation after 45 min of superior mesenteric artery occlusion and

Asunto(s)
Intestino Delgado/fisiología , Daño por Reperfusión/tratamiento farmacológico , alfa-MSH/farmacología , Animales , Motilidad Gastrointestinal , Mucosa Intestinal/metabolismo , Obstrucción Intestinal/tratamiento farmacológico , Obstrucción Intestinal/prevención & control , Intestino Delgado/irrigación sanguínea , Masculino , FN-kappa B/metabolismo , Péptidos/metabolismo , Ratas , Ratas Sprague-Dawley , Circulación Esplácnica
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA