Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
Más filtros











Base de datos
Intervalo de año de publicación
1.
J Leukoc Biol ; 107(3): 445-453, 2020 03.
Artículo en Inglés | MEDLINE | ID: mdl-32017192

RESUMEN

Macrophages can change their physiology in response to microenvironmental signals. This differentiation into classically activated M1 or alternatively activated M2 macrophages is known as polarization. In this study, we isolated bone marrow-derived macrophages from ß2m-deficient (deficient in both MHC class Ia and Ib) and Kb Db -deficient (deficient only in MHC class Ia) mice and found that ß2m-deficient macrophages showed a significantly lower M2b polarization efficiency. In addition, the absence of constitutive MHC class Ib expression decreased the stability of the Notch-1 intracellular domain. Finally, we found that ß2m-deficient mice exposed to irradiation showed reduced bacterial translocation and sepsis severity. Overall, our study demonstrates that MHC class Ib molecules are essential for M2b macrophage polarization and suggests that MHC class Ib molecules play an important role during infection-induced innate immunity.


Asunto(s)
Polaridad Celular , Rayos gamma , Antígenos de Histocompatibilidad Clase I/metabolismo , Macrófagos/patología , Macrófagos/efectos de la radiación , Sepsis/inmunología , Animales , Traslocación Bacteriana/efectos de la radiación , Polaridad Celular/efectos de la radiación , Enterococcus faecalis/fisiología , Femenino , Ratones Endogámicos C57BL , Sepsis/microbiología , Transducción de Señal/efectos de la radiación , Microglobulina beta-2/deficiencia , Microglobulina beta-2/metabolismo
2.
J Immunol ; 204(5): 1255-1262, 2020 03 01.
Artículo en Inglés | MEDLINE | ID: mdl-31941655

RESUMEN

Gut bacteria-associated sepsis is a serious concern in patients with gastrointestinal acute radiation syndrome (GIARS). In our previous studies, gut bacteria-associated sepsis caused high mortality rates in mice exposed to 6-9 Gy of γ-rays. IL-12+CD38+ iNOS+ Mϕ (M1Mϕ) located in the bacterial translocation site (mesenteric lymph nodes [MLNs]) of unirradiated mice were characterized as host defense antibacterial effector cells. However, cells isolated from the MLNs of GIARS mice were mostly CCL1+IL-10+LIGHT+miR-27a+ Mϕ (M2bMϕ, inhibitor cells for the M1Mϕ polarization). Reduced long noncoding RNA Gas5 and increased miR-222 expression in MLN-Mϕ influenced by the irradiation were shown to be associated with M2bMϕ polarization. In this study, the mortality of mice exposed to 7 Gy of γ-rays (7 Gy GIARS mice) was completely controlled after the administration of glycyrrhizin (GL), a major active ingredient in licorice root (Glycyrrhiza glabra). Bacterial translocation and subsequent sepsis were minimal in 7 Gy GIARS mice treated with GL. Increased Gas5 RNA level and decreased miR-222 expression were shown in MLN-Mϕ isolated from 7 Gy GIARS mice treated with GL, and these macrophages did not display any properties of M2bMϕ. These results indicate that gut bacteria-associated sepsis in 7 Gy GIARS mice was controlled by the GL through the inhibition of M2bMϕ polarization at the bacteria translocation site. Expression of Ccl1, a gene required for M2bMϕ survival, is silenced in the MLNs of 7 Gy GIARS mice because of Gas5 RNA, which is increased in these cells after the suppression of miR-222 (a Gas5 RNA expression inhibitor) by the GL.


Asunto(s)
Bacterias/inmunología , Infecciones Bacterianas , Fenómenos Fisiológicos Bacterianos , Traslocación Bacteriana , Rayos gamma/efectos adversos , Ácido Glicirrínico/farmacología , Intestinos , Macrófagos , MicroARNs/inmunología , ARN Largo no Codificante/inmunología , Traumatismos Experimentales por Radiación , Animales , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/patología , Infecciones Bacterianas/prevención & control , Fenómenos Fisiológicos Bacterianos/efectos de los fármacos , Fenómenos Fisiológicos Bacterianos/inmunología , Fenómenos Fisiológicos Bacterianos/efectos de la radiación , Traslocación Bacteriana/efectos de los fármacos , Traslocación Bacteriana/inmunología , Traslocación Bacteriana/efectos de la radiación , Intestinos/inmunología , Intestinos/microbiología , Macrófagos/inmunología , Macrófagos/microbiología , Macrófagos/patología , Masculino , Ratones , Ratones Endogámicos BALB C , Traumatismos Experimentales por Radiación/inmunología , Traumatismos Experimentales por Radiación/microbiología , Traumatismos Experimentales por Radiación/patología , Traumatismos Experimentales por Radiación/prevención & control , Sepsis/inmunología , Sepsis/microbiología , Sepsis/patología , Sepsis/prevención & control
3.
Toxicol Appl Pharmacol ; 329: 40-47, 2017 08 15.
Artículo en Inglés | MEDLINE | ID: mdl-28526636

RESUMEN

Radiation-induced enteritis is a major side effect in cancer patients undergoing abdominopelvic radiotherapy. Radiation exposure produces an uncontrolled inflammatory cascade and epithelial cell loss leading to impaired epithelial barrier function. The goal of this study was to determine the effect of rebamipide on regeneration of the intestinal epithelia after radiation injury. The abdomens of C57BL/6 mice were exposed to 13Gy of irradiation (IR) and then the mice were treated with rebamipide. Upon IR, intestinal epithelia were destroyed structurally at the microscopic level and bacterial translocation was increased. The intestinal damage reached a maximum level on day 6 post-IR and intestinal regeneration occurred thereafter. We found that rebamipide significantly ameliorated radiation-induced intestinal injury. In mice treated with rebamipide after IR, intestinal barrier function recovered and expression of the tight junction components of the intestinal barrier were upregulated. Rebamipide administration reduced radiation-induced intestinal mucosal injury. The levels of proinflammatory cytokines and matrix metallopeptidase 9 (MMP9) were significantly reduced upon rebamipide administration. Intestinal cell proliferation and ß-catenin expression also increased upon rebamipide administration. These data demonstrate that rebamipide reverses impairment of the intestinal barrier by increasing intestinal cell proliferation and attenuating the inflammatory response by inhibiting MMP9 and proinflammatory cytokine expression in a murine model of radiation-induced enteritis.


Asunto(s)
Alanina/análogos & derivados , Enteritis/prevención & control , Células Epiteliales/efectos de los fármacos , Mucosa Intestinal/efectos de los fármacos , Quinolonas/farmacología , Traumatismos Experimentales por Radiación/prevención & control , Protectores contra Radiación/farmacología , Alanina/farmacología , Animales , Traslocación Bacteriana/efectos de los fármacos , Traslocación Bacteriana/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Citocinas/metabolismo , Citoprotección , Relación Dosis-Respuesta a Droga , Enteritis/metabolismo , Enteritis/patología , Células Epiteliales/metabolismo , Células Epiteliales/patología , Células Epiteliales/efectos de la radiación , Mediadores de Inflamación/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Mucosa Intestinal/efectos de la radiación , Masculino , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones Endogámicos C57BL , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/patología , Uniones Estrechas/efectos de los fármacos , Uniones Estrechas/metabolismo , Uniones Estrechas/patología , Uniones Estrechas/efectos de la radiación , Factores de Tiempo , beta Catenina/metabolismo
4.
Radiat Res ; 180(6): 649-57, 2013 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-24294967

RESUMEN

We recently demonstrated that natural delta-tocotrienol (DT3) significantly enhanced survival in total-body irradiated (TBI) mice, and protected mouse bone marrow cells from radiation-induced damage through Erk activation-associated mTOR survival pathways. Here, we further evaluated the effects and mechanisms of DT3 on survival of radiation-induced mouse acute gastrointestinal syndrome. DT3 (75-100 mg/kg) or vehicle was administered as a single subcutaneous injection to CD2F1 mice 24 h before 10-12 Gy (60)Co total-body irradiation at a dose rate of 0.6 Gy/min and survival was monitored. In a separate group of mice, jejunum sections were stained with hematoxylin and eosin and the surviving crypts in irradiated mice were counted. Apoptosis in intestinal epithelial cells was measured by terminal deoxynucleotidyl transferase-mediated deoxyuridine triphosphate nick-end labeling (TUNEL) staining and bacterial translocation from gut to heart, spleen and liver in irradiated mice were evaluated. DT3 (75 mg/kg) significantly enhanced survival in mice that received 10, 10.5, 11 or 12 Gy TBI. Administration of DT3 protected intestinal tissue, decreased apoptotic cells in jejunum and inhibited gut bacterial translocation in irradiated mice. Furthermore, DT3 significantly inhibited radiation-induced production of pro-inflammatory factors interleukin-1ß and -6 and suppressed expression of protein tyrosine kinase 6 (PTK6), a stress-induced kinase that promotes apoptosis in mouse intestinal cells. Our data demonstrate that administration of DT3 protected mice from radiation-induced gastrointestinal system damage.


Asunto(s)
Tracto Gastrointestinal/lesiones , Traumatismos Experimentales por Radiación/prevención & control , Protectores contra Radiación/farmacología , Vitamina E/análogos & derivados , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Traslocación Bacteriana/efectos de los fármacos , Traslocación Bacteriana/efectos de la radiación , Proteínas Portadoras/metabolismo , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/efectos de la radiación , Radioisótopos de Cobalto/efectos adversos , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Activación Enzimática/efectos de la radiación , Células Epiteliales/efectos de los fármacos , Células Epiteliales/efectos de la radiación , Rayos gamma/efectos adversos , Tracto Gastrointestinal/citología , Tracto Gastrointestinal/efectos de los fármacos , Tracto Gastrointestinal/efectos de la radiación , Yeyuno/citología , Yeyuno/efectos de los fármacos , Yeyuno/efectos de la radiación , Masculino , Ratones , Proteínas de Microfilamentos , Fotones/efectos adversos , Proteínas Tirosina Quinasas/metabolismo , Análisis de Supervivencia , Vitamina E/farmacología
5.
J Immunol ; 189(1): 296-303, 2012 Jul 01.
Artículo en Inglés | MEDLINE | ID: mdl-22664870

RESUMEN

The influence of whole-body gamma-irradiation on the antibacterial host defense against Enterococcus faecalis translocation was investigated. Mice irradiated with or without 5 Gy [(137)Cs] gamma-rays were orally infected with 10(6) CFU/mouse E. faecalis. The pathogen was detected in the mesenteric lymph nodes (MLNs) of irradiated mice 1-4 d postinfection, whereas E. faecalis was not isolated from MLNs of normal mice. All irradiated mice died within 5 d of infection, whereas no mortality was shown in normal mice infected with the pathogen. Irradiated mice inoculated with normal mouse MLN macrophages (M) were shown to be resistant against the infection, although the same mice inoculated with irradiated mouse MLNM (I-MLNM) died postinfection. I-MLNM were identified as IL-10(+)IL-12(-)CCL1(+)LIGHT(+) M (M2bM) and were shown to be inhibitory on M conversion from resident M to IL-10(-)IL-12(+)M (M1M). M2bM were demonstrated in MLNs of mice 10-35 d after gamma-irradiation. M1M were not induced by E. faecalis Ag in cultures of I-MLNM, whereas normal mouse MLNM were converted to M1M in response to the Ag stimulation. After treatment with CCL1 antisense oligodeoxynucleotides, M2bM disappeared in MLNs of irradiated mice, and M1M were generated in MLNs of these mice following E. faecalis stimulation. These results indicate that M2bM presented in the I-MLNM populations were responsible for the impaired resistance of mice irradiated with gamma-rays to bacterial translocation and subsequent sepsis. E. faecalis translocation and subsequent sepsis may be controlled immunologically by the intervention of M2bM present in MLNs.


Asunto(s)
Bacteriemia/inmunología , Traslocación Bacteriana/inmunología , Enterococcus faecalis/inmunología , Infecciones por Bacterias Grampositivas/inmunología , Macrófagos/inmunología , Macrófagos/microbiología , Animales , Bacteriemia/microbiología , Bacteriemia/patología , Traslocación Bacteriana/efectos de la radiación , Enterococcus faecalis/efectos de la radiación , Rayos gamma , Infecciones por Bacterias Grampositivas/patología , Macrófagos/efectos de la radiación , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos NOD , Ratones Noqueados , Ratones SCID , Factores de Tiempo
6.
Shock ; 38(2): 170-6, 2012 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-22576002

RESUMEN

Sepsis is the leading cause of mortality in intensive care units. Early detection and intervention are critical to prevent death. The acute radiation syndrome is characterized by damage of the gastrointestinal and hematopoietic systems. Translocation of intestinal microflora combined with immune system compromise may lead to septicemia and death. This work examined the utility of procalcitonin, a clinical sepsis biomarker, in a mouse model of radiation toxicity. C57/BL6 mice were exposed to total body irradiation (TBI). Intestinal mucosal permeability was measured in vivo, and liver bacterial load and plasma levels of procalcitonin (PCT), lipopolysaccharide (LPS), and LPS-binding protein were measured at baseline and at 3.5, 7, and 10 days after TBI. The value of early PCT in predicting subsequent lethality was determined by receiver operating characteristic analysis. Four days after TBI, a dose-dependent increase in permeability of the intestinal mucosa was observed, whereas bacterial translocation was present from day 7 onward. There was a high positive correlation between bacterial translocation and all sepsis biomarkers, with PCT exhibiting the strongest correlation. Moreover, plasma PCT levels were elevated already from day 3.5 onward, whereas LPS was elevated from day 7 and LPS-binding protein only 10 days after TBI. Receiver operating characteristic analysis revealed that PCT levels measured 3.5 days after TBI predicted lethality at 10 days. These data demonstrate the value of PCT as an early biomarker in radiation-induced bacteremia for mouse studies and suggest that clinical results from other septic conditions may apply to postradiation septicemia in humans.


Asunto(s)
Bacteriemia/diagnóstico , Carga Bacteriana , Calcitonina/sangre , Precursores de Proteínas/sangre , Traumatismos Experimentales por Radiación/diagnóstico , Irradiación Corporal Total/efectos adversos , Proteínas de Fase Aguda , Animales , Carga Bacteriana/efectos de la radiación , Traslocación Bacteriana/efectos de la radiación , Biomarcadores/sangre , Péptido Relacionado con Gen de Calcitonina , Proteínas Portadoras/sangre , Ensayo de Inmunoadsorción Enzimática , Fluorescencia , Mucosa Intestinal/efectos de la radiación , Lipopolisacáridos/sangre , Hígado/microbiología , Hepatopatías/microbiología , Glicoproteínas de Membrana/sangre , Ratones , Ratones Endogámicos C57BL , Permeabilidad/efectos de la radiación , Curva ROC
7.
Radiat Res ; 171(6): 698-707, 2009 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-19580476

RESUMEN

Somatostatin analogs ameliorate intestinal injury after localized irradiation. This study investigated whether SOM230, a novel, metabolically stable analog with broad receptor affinity, reduces intestinal injury and lethality in mice exposed to total-body irradiation (TBI). Male CD2F1 mice were exposed to 7-15 Gy TBI. Twice-daily administration of SOM230 (1, 4 or 10 mg/kg per day) or vehicle was started either 2 days before or 4 h after TBI and continued for either 14 or 21 days. Parameters of intestinal and hematopoietic radiation injury, bacterial translocation, and circulating cytokine levels were assessed. Animal survival was monitored for up to 30 days. SOM230 increased survival (P < 0.001) and prolonged survival time (P < 0.001) whether administration was initiated before or after TBI. There was no benefit from administration for 21 compared to 14 days. The survival benefit of SOM230 was completely reversed by co-administration of pancreatic enzymes (P = 0.009). Consistent with the presumed non-cytoprotective mechanism of action, SOM230 did not influence hematopoietic injury or intestinal crypt lethality. However, SOM230 preserved mucosal surface area (P < 0.001) and reduced bacterial translocation in a dose-dependent manner (P < 0.001). Circulating IL-12 levels were reduced in SOM230-treated mice (P = 0.007). No toxicity from SOM230 was observed. SOM230 enhances animal survival whether administration begins before or after TBI; i.e., it is effective both as a protector and as a mitigator. The mechanism likely involves reduction of intraluminal pancreatic enzymes. Because of its efficacy and favorable safety profile, SOM230 is a promising countermeasure against radiation and should undergo further development.


Asunto(s)
Mucosa Intestinal/efectos de la radiación , Páncreas/enzimología , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Protectores contra Radiación/administración & dosificación , Somatostatina/análogos & derivados , Irradiación Corporal Total , Animales , Traslocación Bacteriana/efectos de los fármacos , Traslocación Bacteriana/efectos de la radiación , Células Sanguíneas/efectos de los fármacos , Células Sanguíneas/efectos de la radiación , Radioisótopos de Cesio/toxicidad , Quimiocina CXCL9/sangre , Citrulina/sangre , Relación Dosis-Respuesta en la Radiación , Interleucina-12/sangre , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Yeyuno/efectos de los fármacos , Yeyuno/patología , Yeyuno/efectos de la radiación , Estimación de Kaplan-Meier , Hígado/efectos de los fármacos , Hígado/microbiología , Hígado/efectos de la radiación , Masculino , Ratones , Páncreas/efectos de los fármacos , Páncreas/efectos de la radiación , Traumatismos Experimentales por Radiación/mortalidad , Distribución Aleatoria , Somatostatina/administración & dosificación , Dosimetría Termoluminiscente , Resultado del Tratamiento
8.
J Clin Invest ; 117(8): 2197-204, 2007 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-17657310

RESUMEN

Lymphodepletion with total body irradiation (TBI) increases the efficacy of adoptively transferred tumor-specific CD8(+) T cells by depleting inhibitory lymphocytes and increasing homeostatic cytokine levels. We found that TBI augmented the function of adoptively transferred CD8(+) T cells in mice genetically deficient in all lymphocytes, indicating the existence of another TBI mechanism of action. Additional investigation revealed commensal gut microflora in the mesenteric lymph nodes and elevated LPS levels in the sera of irradiated mice. These findings correlated with increased dendritic cell activation and heightened levels of systemic inflammatory cytokines. Reduction of host microflora using antibiotics, neutralization of serum LPS using polymyxin B, or removal of LPS signaling components using mice genetically deficient in CD14 and TLR4 reduced the beneficial effects of TBI on tumor regression. Conversely, administration of microbial ligand-containing serum or ultrapure LPS from irradiated animals to nonirradiated antibody-lymphodepleted mice enhanced CD8(+) T cell activation and improved tumor regression. Administration of ultrapure LPS to irradiated animals further enhanced the number and function of the adoptively transferred cells, leading to long-term cure of mice with large B16F10 tumors and enhanced autoimmune vitiligo. Thus, disruption of the homeostatic balance between the host and microbes can enhance cell-based tumor immunotherapy.


Asunto(s)
Traslado Adoptivo , Traslocación Bacteriana/inmunología , Linfocitos T CD8-positivos/inmunología , Neoplasias Experimentales/inmunología , Transducción de Señal/inmunología , Receptor Toll-Like 4/inmunología , Irradiación Corporal Total , Animales , Antibacterianos/farmacología , Enfermedades Autoinmunes/inmunología , Enfermedades Autoinmunes/microbiología , Enfermedades Autoinmunes/patología , Autoinmunidad/efectos de los fármacos , Autoinmunidad/inmunología , Traslocación Bacteriana/efectos de la radiación , Linfocitos T CD8-positivos/patología , Linfocitos T CD8-positivos/trasplante , Línea Celular Tumoral , Citocinas/inmunología , Intestinos/inmunología , Intestinos/microbiología , Intestinos/patología , Receptores de Lipopolisacáridos/inmunología , Lipopolisacáridos/inmunología , Lipopolisacáridos/farmacología , Activación de Linfocitos/efectos de los fármacos , Activación de Linfocitos/inmunología , Depleción Linfocítica , Ratones , Trasplante de Neoplasias , Neoplasias Experimentales/microbiología , Neoplasias Experimentales/patología , Neoplasias Experimentales/terapia , Polimixina B/farmacología , Transducción de Señal/efectos de los fármacos , Vitíligo/inmunología , Vitíligo/microbiología , Vitíligo/patología
9.
Int J Radiat Biol ; 83(6): 375-82, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17487677

RESUMEN

PURPOSE: Both enteric infection and exposure to ionizing radiation are associated with increased intestinal permeability. However, the combined effect of irradiation and enteric infection has not been described. We combined infection of mice with the enteric pathogen, Citrobacter rodentium, with exposure to ionizing radiation and assessed the impact on colonic epithelial ion transport, permeability and bacterial translocation. MATERIALS AND METHODS: Mice were infected with C. rodentium and then received whole-body exposure to 5 Gray gamma-radiation 7 days later. Three days post-irradiation, mice were euthanized and colons removed. Control groups included sham-infected mice that were irradiated and mice that were infected, but not irradiated. RESULTS: Macroscopic damage score and colonic wall thickness were increased by C. rodentium infection, but these parameters were not exacerbated by irradiation. Infection caused an increase in myeloperoxidase activity that was reduced by irradiation. Irradiation reduced the secretory response to electrical field stimulation, forskolin and carbachol; these changes were not altered by infection with C. rodentium. None of the treatments caused an increase in permeability to 51Cr-ethylenediaminetetraacetic acid (EDTA). However, combined infection and irradiation synergistically increased bacterial translocation to mesenteric lymph nodes, liver, spleen and blood. CONCLUSIONS: Although the combination of irradiation and infection did not exacerbate the individual effects of these challenges on ion secretion and mucosal permeability to 51Cr-EDTA, it dramatically increased susceptibility to bacterial translocation and bacteremia. These results have important implications for patients who develop an enteric infection during the course of abdominopelvic radiotherapy.


Asunto(s)
Traslocación Bacteriana/efectos de la radiación , Citrobacter rodentium/fisiología , Citrobacter rodentium/efectos de la radiación , Colitis/microbiología , Colon/microbiología , Colon/efectos de la radiación , Infecciones por Enterobacteriaceae/microbiología , Animales , Susceptibilidad a Enfermedades/microbiología , Relación Dosis-Respuesta en la Radiación , Masculino , Ratones , Ratones Endogámicos C57BL , Dosis de Radiación , Radiación Ionizante
10.
J Surg Res ; 89(2): 121-5, 2000 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-10729239

RESUMEN

BACKGROUND: Recent studies indicated that glutamine and arginine support the mucosal barrier in several ways. This experimental study hypothesized that administration of glutamine- and arginine-enriched diets before abdominal radiation therapy would provide a radioprotective effect on intestinal mucosa, and this would augment the therapeutic effectiveness provided by postirradiation administration. MATERIALS AND METHODS: A rat model of radiation enteritis was designed with a single dose of 1100 cGy to the abdomen. Thirty-five rats were randomized into five groups of seven. A 7-day glutamine-enriched diet for Group I and a 7-day arginine-enriched diet for Group II were administered both pre- and postradiation. For Groups III and IV, the same glutamine and arginine diets were given, respectively, postradiation only. Group V was fed a glutamine- and arginine-free diet and was the control group. The rats underwent laparotomy for culture of mesenteric lymph nodes and removal of segments of ileum, jejenum, and colon for microscopic examination. RESULTS: Bacterial translocation was significantly higher in Group V (P < 0.05), while intestinal villus count and villus height were significantly higher in all of the groups fed glutamine and arginine when compared with the control group (P < 0.0001 and P < 0.05, respectively). CONCLUSION: Both arginine- and glutamine-enriched diets have protective effects on gut mucosa in the postirradiation state; however, pre- and postirradiation administration together does not provide superior protection versus postradiation administration alone.


Asunto(s)
Arginina/administración & dosificación , Enteritis/dietoterapia , Enteritis/prevención & control , Glutamina/administración & dosificación , Traumatismos Experimentales por Radiación/dietoterapia , Traumatismos Experimentales por Radiación/prevención & control , Animales , Arginina/uso terapéutico , Traslocación Bacteriana/efectos de la radiación , Dieta , Glutamina/uso terapéutico , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/microbiología , Intestinos/efectos de los fármacos , Intestinos/patología , Intestinos/efectos de la radiación , Microvellosidades/efectos de los fármacos , Microvellosidades/patología , Microvellosidades/efectos de la radiación , Ratas , Ratas Sprague-Dawley
11.
Acta Oncol ; 37(2): 179-85, 1998.
Artículo en Inglés | MEDLINE | ID: mdl-9636013

RESUMEN

Growth hormone stimulates the growth of intestinal mucosa and may reduce the severity of injury caused by radiation. Male Wistar rats underwent abdominal irradiation (12 Gy) and were treated with either human growth hormone (hGH) or saline, and sacrificed at day 4 or 7 post-irradiation. Bacterial translocation, and the ileal mucosal thickness, proliferation, and disaccharidase activity were assessed. Mortality was 65% in irradiated animals, whereas hGH caused a decrement (29%, p < 0.05). Bacterial translocation was also reduced by hGH (p < 0.05). Treating irradiated rats with hGH prevented body weight loss (p < 0.05). Mucosal thickness increased faster in irradiated hGH-treated animals. The proliferative index showed an increment in hGH-treated animals (p < 0.05). Giving hGH to irradiated rats prevented decrease in sucrose activity, and increment in lactase activity. In conclusion, giving hGH to irradiated rats promotes the adaptative process of the intestine and acute radiation-related negative effects, including mortality, bacterial translocation, and weight loss.


Asunto(s)
Abdomen/efectos de la radiación , Traslocación Bacteriana/efectos de los fármacos , Hormona de Crecimiento Humana/farmacología , Mucosa Intestinal/efectos de la radiación , Animales , Traslocación Bacteriana/efectos de la radiación , División Celular , Mucosa Intestinal/microbiología , Mucosa Intestinal/patología , Masculino , Antígeno Nuclear de Célula en Proliferación/análisis , Traumatismos Experimentales por Radiación/mortalidad , Traumatismos Experimentales por Radiación/patología , Ratas , Ratas Wistar
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA