Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 28
Filtrar
Más filtros












Base de datos
Intervalo de año de publicación
1.
ANZ J Surg ; 91(7-8): 1509-1514, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-33576122

RESUMEN

BACKGROUND: Over the last 2 decades, outcomes for oesophageal cancer have improved due to advances in surgical and oncological practice. Optimizing outcomes by centralization of oesophagectomy to high-volume centres has been observed. The aim of this study was to establish if technical and oncological outcomes after oesophagectomy in southern New Zealand are comparable to recent benchmarks. METHODS: Consecutive patients undergoing oesophagectomy for cancer and benign pathology at Dunedin Hospital from 1995 to 2019 were prospectively audited. For malignant cases, histology was obtained retrospectively along with details of neo-adjuvant and adjuvant therapy. The primary outcome was disease-specific survival, stratified by time, resection margin, and TNM staging. Secondary outcomes included mortality and morbidity of oesophagectomy. Complications were graded using the Clavien-Dindo classification. RESULTS: Oesophagectomy was performed in 108 patients, and 99 patients had surgery for oesophageal malignancy. The median survival was 35.3 (95% confidence interval (CI) 30.0-93.4) months and the 5-year survival overall was 41.7%. Comparing survival in patients undergoing oesophagectomy up to 2006 and afterwards showed an improvement in 5-year survival (30.3%, 95% CI (14.2-60.0) versus 47.8%, 95% CI (32.5, not reached), respectively, P = 0.041). There were two perioperative deaths (1.8%), six clinical anastomotic leaks (5.5%), four anastomotic strictures (3.7%) and five chylothoraces (4.6%). CONCLUSION: This 25-year survey of oesophagectomy in southern New Zealand audits the results of a low volume centre, where a variety of neo-adjuvant treatments have been used. Despite this, perioperative morbidity, mortality and survival are comparable to those achieved by international high-volume centres.


Asunto(s)
Neoplasias Esofágicas , Esofagectomía , Fuga Anastomótica , Neoplasias Esofágicas/cirugía , Esofagectomía/efectos adversos , Humanos , Nueva Zelanda/epidemiología , Estudios Retrospectivos
2.
PLoS One ; 12(9): e0184393, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28934227

RESUMEN

Hemorrhage following whole-body γ-irradiation in a combined injury (CI) model increases mortality compared to whole-body γ-irradiation alone (RI). The decreased survival in CI is accompanied by increased bone marrow injury, decreased hematocrit, and alterations of miRNA in the kidney. In this study, our aim was to examine cytokine homeostasis, susceptibility to systemic bacterial infection, and intestinal injury. More specifically, we evaluated the interleukin-6 (IL-6)-induced stress proteins including C-reactive protein (CRP), complement 3 (C3), Flt-3 ligand, and corticosterone. CD2F1 male mice received 8.75 Gy 60Co gamma photons (0.6 Gy/min, bilateral) which was followed by a hemorrhage of 20% of the blood volume. In serum, RI caused an increase of IL-1, IL-2, IL-3, IL-5, IL-6, IL-12, IL-13, IL-15, IL-17A, IL-18, G-CSF, CM-CSF, eotaxin, IFN-γ, MCP-1, MIP, RANTES, and TNF-α, which were all increased by hemorrhage alone, except IL-9, IL-17A, and MCP-1. Nevertheless, CI further elevated RI-induced increases of these cytokines except for G-CSF, IFN- γ and RANTES in serum. In the ileum, hemorrhage in the CI model significantly enhanced RI-induced IL-1ß, IL-3, IL-6, IL-10, IL-12p70, IL-13, IL-18, and TNF-α concentrations. In addition, Proteus mirabilis Gram(-) was found in only 1 of 6 surviving RI mice on Day 15, whereas Streptococcus sanguinis Gram(+) and Sphingomonas paucimobilis Gram(-) were detected in 2 of 3 surviving CI mice (with 3 CI mice diseased due to inflammation and infection before day 15) at the same time point. Hemorrhage in the CI model enhanced the RI-induced increases in C3 and decreases in CRP concentrations. However, hemorrhage alone did not alter the basal levels, but hemorrhage in the CI model displayed similar increases in Flt-3 ligand levels as RI did. Hemorrhage alone altered the basal levels of corticosterone early after injury, which then returned to the baseline, but in RI mice and CI mice the increased corticosterone concentration remained elevated throughout the 15 day study. CI increased 8 miRNAs and decreased 10 miRNAs in serum, and increased 16 miRNA and decreased 6 miRNAs in ileum tissue. Among the altered miRNAs, CI increased miR-34 in the serum and ileum which targeted an increased phosphorylation of ERK, p38, and increased NF-κB, thereby leading to increased iNOS expression and activation of caspase-3 in the ileum. Further, let-7g/miR-98 targeted the increased phosphorylation of STAT3 in the ileum, which is known to bind to the iNOS gene. These changes may correlate with cell death in the ileum of CI mice. The histopathology displayed blunted villi and villus edema in RI and CI mice. Based on the in silico analysis, miR-15, miR-99, and miR-100 were predicted to regulate IL-6 and TNF. These results suggest that CI-induced alterations of cytokines/chemokines, CRP, and C3 cause a homeostatic imbalance and may contribute to the pathophysiology of the gastrointestinal injury. Inhibitory intervention in these responses may prove therapeutic for CI and improve recovery of the ileal morphologic damage.


Asunto(s)
Caspasa 3/metabolismo , Complemento C3/metabolismo , Citocinas/metabolismo , Hemorragia/metabolismo , MicroARNs/metabolismo , Irradiación Corporal Total/efectos adversos , Animales , Apoptosis/fisiología , Apoptosis/efectos de la radiación , Infecciones Bacterianas/etiología , Infecciones Bacterianas/metabolismo , Infecciones Bacterianas/mortalidad , Infecciones Bacterianas/patología , Proteína C-Reactiva/metabolismo , Radioisótopos de Cobalto/efectos adversos , Corticosterona/metabolismo , Hemorragia/complicaciones , Hemorragia/mortalidad , Hemorragia/patología , Íleon/metabolismo , Íleon/microbiología , Íleon/patología , Íleon/efectos de la radiación , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Mucosa Intestinal/efectos de la radiación , Masculino , Ratones , Traumatismos Experimentales por Radiación/complicaciones , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/mortalidad , Traumatismos Experimentales por Radiación/patología , Distribución Aleatoria , Tirosina Quinasa 3 Similar a fms/metabolismo
3.
Mediators Inflamm ; 2017: 7582079, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28408792

RESUMEN

Ionizing radiation combined with trauma tissue injury (combined injury, CI) results in greater mortality and H-ARS than radiation alone (radiation injury, RI), which includes thrombocytopenia. The aim of this study was to determine whether increases in numbers of thrombocytes would improve survival and mitigate H-ARS after CI. We observed in mice that WBC and platelets remained very low in surviving RI animals that were given 9.5 Gy 60Co-γ-photon radiation, whereas only lymphocytes and basophils remained low in surviving CI mice that were irradiated and then given skin wounds. Numbers of RBC and platelets, hemoglobin concentrations, and hematocrit values remained low in surviving RI and CI mice. CI induced 30-day mortality higher than RI. Radiation delayed wound healing by approximately 14 days. Treatment with a thrombopoietin receptor agonist, Alxn4100TPO, after CI improved survival, mitigated body-weight loss, and reduced water consumption. Though this therapy delayed wound-healing rate more than in vehicle groups, it greatly increased numbers of platelets in sham, wounded, RI, and CI mice; it significantly mitigated decreases in WBC, spleen weights, and splenocytes in CI mice and decreases in RBC, hemoglobin, hematocrit values, and splenocytes and splenomegaly in RI mice. The results suggest that Alxn4100TPO is effective in mitigating CI.


Asunto(s)
Anticuerpos Monoclonales/uso terapéutico , Traumatismos por Radiación/tratamiento farmacológico , Traumatismos por Radiación/terapia , Receptores de Trombopoyetina/antagonistas & inhibidores , Animales , Anticuerpos Monoclonales Humanizados , Peso Corporal/efectos de los fármacos , Peso Corporal/efectos de la radiación , Femenino , Rayos gamma , Ratones , Receptores de Trombopoyetina/metabolismo , Irradiación Corporal Total/métodos , Cicatrización de Heridas/efectos de los fármacos , Cicatrización de Heridas/efectos de la radiación , Heridas y Lesiones/tratamiento farmacológico
4.
Int J Radiat Biol ; 91(9): 690-702, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25994812

RESUMEN

PURPOSE: A combination therapy for combined injury (CI) using a non-specific immunomodulator, synthetic trehalose dicorynomycolate and monophosphoryl lipid A (STDCM-MPL), was evaluated to augment oral antimicrobial agents, levofloxacin (LVX) and amoxicillin (AMX), to eliminate endogenous sepsis and modulate cytokine production. MATERIALS AND METHODS: Female B6D2F(1)/J mice received 9.75 Gy cobalt-60 gamma-radiation and wound. Bacteria were isolated and identified in three tissues. Incidence of bacteria and cytokines were compared between treatment groups. RESULTS: Results demonstrated that the lethal dose for 50% at 30 days (LD(50/30)) of B6D2F(1)/J mice was 9.42 Gy. Antimicrobial therapy increased survival in radiation-injured (RI) mice. Combination therapy increased survival after RI and extended survival time but did not increase survival after CI. Sepsis began five days earlier in CI mice than RI mice with Gram-negative species predominating early and Gram-positive species increasing later. LVX plus AMX eliminated sepsis in CI and RI mice. STDCM-MPL eliminated Gram-positive bacteria in CI and most RI mice but not Gram-negative. Treatments significantly modulated 12 cytokines tested, which pertain to wound healing or elimination of infection. CONCLUSIONS: Combination therapy eliminates infection and prolongs survival time but does not assure CI mouse survival, suggesting that additional treatment for proliferative-cell recovery is required.


Asunto(s)
Antiinfecciosos/administración & dosificación , Citocinas/biosíntesis , Factores Inmunológicos/administración & dosificación , Sepsis/tratamiento farmacológico , Sepsis/inmunología , Amoxicilina/administración & dosificación , Animales , Infecciones Bacterianas/tratamiento farmacológico , Infecciones Bacterianas/inmunología , Infecciones Bacterianas/microbiología , Quimiocinas/biosíntesis , Factores Cordón/administración & dosificación , Modelos Animales de Enfermedad , Quimioterapia Combinada , Femenino , Rayos gamma/efectos adversos , Sustancias de Crecimiento/biosíntesis , Levofloxacino/administración & dosificación , Lípido A/administración & dosificación , Lípido A/análogos & derivados , Ratones , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Traumatismos Experimentales por Radiación/inmunología , Traumatismos Experimentales por Radiación/microbiología , Sepsis/microbiología , Piel/lesiones , Infección de Heridas/tratamiento farmacológico , Infección de Heridas/inmunología , Infección de Heridas/microbiología
5.
Comp Med ; 64(6): 456-63, 2014 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-25527026

RESUMEN

In the absence of supportive care, exposing Göttingen minipigs to γ-radiation doses of less than 2 Gy achieves lethality due to hematopoietic acute radiation syndrome. Doses of 2 to 5 Gy are associated with an accelerated hematopoietic syndrome, characterized by villus blunting and fusion, the beginning of sepsis, and a mild transient reduction in plasma citrulline concentration. We exposed male Göttingen minipigs (age, 5 mo; weight, 9 to 11 kg) to γ-radiation doses of 5 to 12 Gy (total body; (60)Co, 0.6 Gy/min) to test whether these animals exhibit classic gastrointestinal acute radiation syndrome (GI-ARS). After exposure, the minipigs were monitored for 10 d by using clinical signs, CBC counts, and parameters associated with the development of the gastrointestinal syndrome. Göttingen minipigs exposed to γ radiation of 5 to 12 Gy demonstrate a dose-dependent occurrence of all parameters classically associated with acute GI-ARS. These results suggest that Göttingen minipigs may be a suitable model for studying GI-ARS after total body irradiation, but the use of supportive care to extend survival beyond 10 d is recommended. This study is the first step toward determining the feasibility of using Göttingen minipigs in testing the efficacy of candidate drugs for the treatment of GI-ARS after total body irradiation.


Asunto(s)
Síndrome de Radiación Aguda/patología , Modelos Animales de Enfermedad , Rayos gamma/efectos adversos , Enfermedades Gastrointestinales/patología , Porcinos Enanos , Animales , Citrulina/sangre , Relación Dosis-Respuesta en la Radiación , Determinación de Punto Final , Técnicas Histológicas , Modelos Lineales , Masculino , Porcinos
6.
Oxid Med Cell Longev ; 2014: 215858, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25374650

RESUMEN

Exposure to ionizing radiation alone (RI) or combined with traumatic tissue injury (CI) is a crucial life-threatening factor in nuclear and radiological events. In our laboratory, mice exposed to (60)Co-γ-photon radiation (9.5 Gy, 0.4 Gy/min, bilateral) followed by 15% total-body-surface-area skin wounds (R-W CI) or burns (R-B CI) experienced an increment of ≥18% higher mortality over a 30-day observation period compared to RI alone. CI was accompanied by severe leukocytopenia, thrombocytopenia, erythropenia, and anemia. At the 30th day after injury, numbers of WBC and platelets still remained very low in surviving RI and CI mice. In contrast, their RBC, hemoglobin, and hematocrit were recovered towards preirradiation levels. Only RI induced splenomegaly. RI and CI resulted in bone-marrow cell depletion. In R-W CI mice, ghrelin (a hunger-stimulating peptide) therapy increased survival, mitigated body-weight loss, accelerated wound healing, and increased hematocrit. In R-B CI mice, ghrelin therapy increased survival and numbers of neutrophils, lymphocytes, and platelets and ameliorated bone-marrow cell depletion. In RI mice, this treatment increased survival, hemoglobin, and hematocrit and inhibited splenomegaly. Our novel results are the first to suggest that ghrelin therapy effectively improved survival by mitigating CI-induced leukocytopenia, thrombocytopenia, and bone-marrow injury or the RI-induced decreased hemoglobin and hematocrit.


Asunto(s)
Quemaduras/tratamiento farmacológico , Ghrelina/farmacología , Traumatismos Experimentales por Radiación/sangre , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Irradiación Corporal Total/efectos adversos , Heridas y Lesiones/sangre , Heridas y Lesiones/tratamiento farmacológico , Animales , Quemaduras/sangre , Radioisótopos de Cobalto , Femenino , Rayos gamma/efectos adversos , Leucopenia/tratamiento farmacológico , Leucopenia/etiología , Ratones , Distribución Aleatoria , Esplenomegalia/tratamiento farmacológico , Esplenomegalia/etiología , Trombocitopenia/tratamiento farmacológico , Trombocitopenia/etiología , Cicatrización de Heridas/efectos de los fármacos
7.
PLoS One ; 9(11): e114078, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25423021

RESUMEN

The purpose of this study was to elucidate the role of gamma-tocotrienol (GT3)-mobilized progenitors in mitigating damage to mice exposed to a supralethal dose of cobalt-60 gamma-radiation. CD2F1 mice were transfused 24 h post-irradiation with whole blood or isolated peripheral blood mononuclear cells (PBMC) from donors that had received GT3 72 h prior to blood collection and recipient mice were monitored for 30 days. To understand the role of GT3-induced granulocyte colony-stimulating factor (G-CSF) in mobilizing progenitors, donor mice were administered a neutralizing antibody specific to G-CSF or its isotype before blood collection. Bacterial translocation from gut to heart, spleen and liver of irradiated recipient mice was evaluated by bacterial culture on enriched and selective agar media. Endotoxin in serum samples also was measured. We also analyzed the colony-forming units in the spleens of irradiated mice. Our results demonstrate that whole blood or PBMC from GT3-administered mice mitigated radiation injury when administered 24 h post-irradiation. Furthermore, administration of a G-CSF antibody to GT3-injected mice abrogated the efficacy of blood or PBMC obtained from such donors. Additionally, GT3-mobilized PBMC inhibited the translocation of intestinal bacteria to the heart, spleen, and liver, and increased colony forming unit-spleen (CFU-S) numbers in irradiated mice. Our data suggests that GT3 induces G-CSF, which mobilizes progenitors and these progenitors mitigate radiation injury in recipient mice. This approach using mobilized progenitor cells from GT3-injected donors could be a potential treatment for humans exposed to high doses of radiation.


Asunto(s)
Cromanos/farmacología , Protectores contra Radiación/farmacología , Células Madre/efectos de la radiación , Vitamina E/análogos & derivados , Animales , Radioisótopos de Cobalto/toxicidad , Masculino , Ratones , Células Madre/efectos de los fármacos , Vitamina E/farmacología
8.
Oxid Med Cell Longev ; 2014: 481392, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-24738019

RESUMEN

Exposure to ionizing radiation alone (radiation injury, RI) or combined with traumatic tissue injury (radiation combined injury, CI) is a crucial life-threatening factor in nuclear and radiological accidents. As demonstrated in animal models, CI results in greater mortality than RI. In our laboratory, we found that B6D2F1/J female mice exposed to (60)Co-γ-photon radiation followed by 15% total-body-surface-area skin burns experienced an increment of 18% higher mortality over a 30-day observation period compared to irradiation alone; that was accompanied by severe cytopenia, thrombopenia, erythropenia, and anemia. At the 30th day after injury, neutrophils, lymphocytes, and platelets still remained very low in surviving RI and CI mice. In contrast, their RBC, hemoglobin, and hematocrit were similar to basal levels. Comparing CI and RI mice, only RI induced splenomegaly. Both RI and CI resulted in bone marrow cell depletion. It was observed that only the RI mice treated with pegylated G-CSF after RI resulted in 100% survival over the 30-day period, and pegylated G-CSF mitigated RI-induced body-weight loss and depletion of WBC and platelets. Peg-G-CSF treatment sustained RBC balance, hemoglobin levels, and hematocrits and inhibited splenomegaly after RI. The results suggest that pegylated G-CSF effectively sustained animal survival by mitigating radiation-induced cytopenia, thrombopenia, erythropenia, and anemia.


Asunto(s)
Plaquetas/patología , Quemaduras/complicaciones , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Polietilenglicoles/química , Radiación Ionizante , Esplenomegalia/tratamiento farmacológico , Irradiación Corporal Total , Animales , Plaquetas/efectos de los fármacos , Plaquetas/efectos de la radiación , Peso Corporal/efectos de los fármacos , Peso Corporal/efectos de la radiación , Células de la Médula Ósea/efectos de los fármacos , Células de la Médula Ósea/metabolismo , Células de la Médula Ósea/patología , Quemaduras/sangre , Quemaduras/tratamiento farmacológico , Recuento de Células , Femenino , Factor Estimulante de Colonias de Granulocitos/farmacología , Ratones , Tamaño de los Órganos/efectos de los fármacos , Bazo/efectos de los fármacos , Bazo/patología , Esplenomegalia/sangre , Esplenomegalia/complicaciones , Análisis de Supervivencia
9.
Int J Radiat Biol ; 90(5): 363-72, 2014 May.
Artículo en Inglés | MEDLINE | ID: mdl-24524283

RESUMEN

PURPOSE: To characterize acute radiation syndrome (ARS) sequelae at doses intermediate between the bone marrow (H-ARS) and full gastrointestinal (GI-ARS) syndrome. METHODS: Male minipigs, approximately 5 months old, 9-12 kg in weight, were irradiated with Cobalt-60 (total body, bilateral gamma irradiation, 0.6 Gy/min). Endpoints were 10-day survival, gastrointestinal histology, plasma citrulline, bacterial translocation, vomiting, diarrhea, vital signs, systemic inflammatory response syndrome (SIRS), febrile neutropenia (FN). RESULTS: We exposed animals to doses (2.2-5.0 Gy) above those causing H-ARS (1.6-2.0 Gy), and evaluated development of ARS. Compared to what was observed during H-ARS (historical data: Moroni et al. 2011a , 2011c ), doses above 2 Gy produced signs of increasingly severe pulmonary damage, faster deterioration of clinical conditions, and faster increases in levels of C-reactive protein (CRP). In the range of 4.6-5.0 Gy, animals died by day 9-10; signs of the classic GI syndrome, as measured by diarrhea, vomiting and bacterial translocation, did not occur. At doses above 2 Gy we observed transient reduction in circulating citrulline levels, and animals exhibited earlier depletion of blood elements and faster onset of SIRS and FN. CONCLUSIONS: An accelerated hematopoietic subsyndrome (AH-ARS) is observed at radiation doses between those producing H-ARS and GI-ARS. It is characterized by early onset of SIRS and FN, and greater lung damage, compared to H-ARS.


Asunto(s)
Tracto Gastrointestinal/efectos de la radiación , Hematopoyesis/efectos de la radiación , Dosis de Radiación , Traumatismos Experimentales por Radiación/patología , Porcinos Enanos , Animales , Células de la Médula Ósea/efectos de la radiación , Inflamación/etiología , Masculino , Traumatismos Experimentales por Radiación/complicaciones , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/fisiopatología , Análisis de Supervivencia , Porcinos , Síndrome , Factores de Tiempo
10.
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
11.
PLoS One ; 8(3): e58389, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23520506

RESUMEN

Radiation combined injury (CI) is a radiation injury (RI) combined with other types of injury, which generally leads to greater mortality than RI alone. A spectrum of specific, time-dependent pathophysiological changes is associated with CI. Of these changes, the massive release of pro-inflammatory cytokines, severe hematopoietic and gastrointestinal losses and bacterial sepsis are important treatment targets to improve survival. Ciprofloxacin (CIP) is known to have immunomodulatory effect besides the antimicrobial activity. The present study reports that CIP ameliorated pathophysiological changes unique to CI that later led to major mortality. B6D2F1/J mice received CI on day 0, by RI followed by wound trauma, and were treated with CIP (90 mg/kg p.o., q.d. within 2 h after CI through day 10). At day 10, CIP treatment not only significantly reduced pro-inflammatory cytokine and chemokine concentrations, including interleukin-6 (IL-6) and KC (i.e., IL-8 in human), but it also enhanced IL-3 production compared to vehicle-treated controls. Mice treated with CIP displayed a greater repopulation of bone marrow cells. CIP also limited CI-induced apoptosis and autophagy in ileal villi, systemic bacterial infection, and IgA production. CIP treatment led to LD(0/10) compared to LD(20/10) for vehicle-treated group after CI. Given the multiple beneficial activities of CIP shown in our experiments, CIP may prove to be a useful therapeutic drug for CI.


Asunto(s)
Antiinfecciosos/farmacología , Apoptosis , Autofagia , Ciprofloxacina/farmacología , Citocinas/sangre , Íleon , Traumatismos Experimentales por Radiación , Piel/lesiones , Heridas y Lesiones , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Autofagia/efectos de los fármacos , Autofagia/efectos de la radiación , Infecciones Bacterianas/sangre , Infecciones Bacterianas/tratamiento farmacológico , Infecciones Bacterianas/etiología , Infecciones Bacterianas/patología , Femenino , Íleon/metabolismo , Íleon/patología , Ratones , Traumatismos Experimentales por Radiación/sangre , Traumatismos Experimentales por Radiación/complicaciones , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Traumatismos Experimentales por Radiación/patología , Irradiación Corporal Total , Heridas y Lesiones/sangre , Heridas y Lesiones/complicaciones , Heridas y Lesiones/tratamiento farmacológico , Heridas y Lesiones/patología
12.
Cell Biosci ; 3(1): 36, 2013 Sep 16.
Artículo en Inglés | MEDLINE | ID: mdl-24499553

RESUMEN

BACKGROUND: Our previous research demonstrated that one subcutaneous injection of 17-Dimethylaminoethylamino-17-demethoxygeldanamycin (17-DMAG) 24 hours (h) before irradiation (8.75 Gy) increased mouse survival by 75%. However, the protective mechanism of 17-DMAG is currently unknown. The present study aimed to investigate whether oral administration of 17-DMAG was also radioprotective and the potential role it may play in radioprotection. RESULTS: A single dose of orally pre-administered (24, 48, or 72 h) 17-DMAG (10 mg/kg) increased irradiated mouse survival, reduced body weight loss, improved water consumption, and decreased facial dropsy, whereas orally post-administered 17-DMAG failed. Additional oral doses of pre-treatment did not improve 30-day survival. The protective effect of multiple pre-administrations (2-3 times) of 17-DMAG at 10 mg/kg was equal to the outcome of a single pre-treatment. In 17-DMAG-pretreated mice, attenuation of bone marrow aplasia in femurs 30 days after irradiation with recovered expressions of cluster of differentiation 34, 44 (CD34, CD44), and survivin in bone marrow cells were observed. 17-DMAG also elevated serum granulocyte-colony stimulating factor (G-CSF), decreased serum fms-related tyrosine kinase 3 ligand, and reduced white blood cell depletion. 17-DMAG ameliorated small intestinal histological damage, promoted recovery of villus heights and intestinal crypts including stem cells, where increased leucine-rich repeat-containing G-protein coupled receptor 5 (Lgr5) was found 30 days after irradiation. CONCLUSIONS: 17-DMAG is a potential radioprotectant for bone marrow and small intestine that results in survival improvement.

13.
Int J Radiat Biol ; 89(5): 334-45, 2013 May.
Artículo en Inglés | MEDLINE | ID: mdl-23270472

RESUMEN

PURPOSE: The objective of this study was to elucidate the action of α-tocopherol succinate (TS)- and AMD3100-mobilized progenitors in mitigating radiation-induced injuries. MATERIAL AND METHODS: CD2F1 mice were exposed to a high dose of radiation and then transfused intravenously with 5 million peripheral blood mononuclear cells (PBMC) from TS- and AMD3100-injected mice after irradiation. Intestinal and splenic tissues were harvested after irradiation and cells of those tissues were analyzed for markers of apoptosis and mitosis. Bacterial translocation from gut to heart, spleen, and liver in TS-treated and irradiated mice was evaluated by bacterial culture. RESULTS: We observed that the infusion of PBMC from TS- and AMD3100-injected mice significantly inhibited apoptosis, increased cell proliferation in the analyzed tissues of recipient mice, and inhibited bacterial translocation to various organs compared to mice receiving cells from vehicle-mobilized cells. This study further supports our contention that the infusion of TS-mobilized progenitor-containing PBMC acts as a bridging therapy by inhibiting radiation-induced apoptosis, enhancing cell proliferation, and inhibiting bacterial translocation in irradiated mice. CONCLUSIONS: We suggest that this novel bridging therapeutic approach that involves the infusion of TS-mobilized hematopoietic progenitors following acute radiation injury might be applicable to humans as well.


Asunto(s)
Movimiento Celular/efectos de los fármacos , Intestinos/efectos de los fármacos , Intestinos/efectos de la radiación , Protectores contra Radiación/farmacología , Células Madre/citología , Irradiación Corporal Total/efectos adversos , alfa-Tocoferol/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/efectos de la radiación , Bacterias/efectos de los fármacos , Bacterias/efectos de la radiación , Movimiento Celular/efectos de la radiación , Proliferación Celular/efectos de los fármacos , Proliferación Celular/efectos de la radiación , Roturas del ADN/efectos de los fármacos , Roturas del ADN/efectos de la radiación , Endotoxinas/sangre , Rayos gamma/efectos adversos , Intestinos/citología , Intestinos/microbiología , Yeyuno/citología , Yeyuno/efectos de los fármacos , Yeyuno/efectos de la radiación , Masculino , Ratones , Traumatismos por Radiación/prevención & control , Bazo/citología , Bazo/efectos de los fármacos , Bazo/efectos de la radiación , Células Madre/efectos de los fármacos , Células Madre/efectos de la radiación
14.
Cell Biosci ; 2(1): 20, 2012 Jun 11.
Artículo en Inglés | MEDLINE | ID: mdl-22686656

RESUMEN

BACKGROUND: Wounding following whole-body γ-irradiation (radiation combined injury, RCI) increases mortality. Wounding-induced increases in radiation mortality are triggered by sustained activation of inducible nitric oxide synthase pathways, persistent alteration of cytokine homeostasis, and increased susceptibility to bacterial infection. Among these factors, cytokines along with other biomarkers have been adopted for biodosimetric evaluation and assessment of radiation dose and injury. Therefore, wounding could complicate biodosimetric assessments. RESULTS: In this report, such confounding effects were addressed. Mice were given 60Co γ-photon radiation followed by skin wounding. Wound trauma exacerbated radiation-induced mortality, body-weight loss, and wound healing. Analyses of DNA damage in bone-marrow cells and peripheral blood mononuclear cells (PBMCs), changes in hematology and cytokine profiles, and fundamental clinical signs were evaluated. Early biomarkers (1 d after RCI) vs. irradiation alone included significant decreases in survivin expression in bone marrow cells, enhanced increases in γ-H2AX formation in Lin+ bone marrow cells, enhanced increases in IL-1ß, IL-6, IL-8, and G-CSF concentrations in blood, and concomitant decreases in γ-H2AX formation in PBMCs and decreases in numbers of splenocytes, lymphocytes, and neutrophils. Intermediate biomarkers (7 - 10 d after RCI) included continuously decreased γ-H2AX formation in PBMC and enhanced increases in IL-1ß, IL-6, IL-8, and G-CSF concentrations in blood. The clinical signs evaluated after RCI were increased water consumption, decreased body weight, and decreased wound healing rate and survival rate. Late clinical signs (30 d after RCI) included poor survival and wound healing. CONCLUSION: Results suggest that confounding factors such as wounding alters ionizing radiation dose assessment and agents inhibiting these responses may prove therapeutic for radiation combined injury and reduce related mortality.

15.
Radiat Res ; 177(2): 133-45, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22013885

RESUMEN

The purpose of this study was to elucidate the role of α-tocopherol succinate (α-TS) in protecting mice from gastrointestinal syndrome induced by total-body irradiation. CD2F1 mice were injected subcutaneously with 400 mg/kg of α-TS and exposed to different doses of (60)Co γ radiation, and 30-day survival was monitored. Jejunum sections were analyzed for crypts and villi, PUMA (p53 upregulated modulator of apoptosis), and apoptosis (terminal deoxynucleotidyl transferase dUTP nick end labeling - TUNEL). The crypt regeneration in irradiated mice was evaluated by 5-bromo-2-deoxyuridine (BrdU). Bacterial translocation from gut to heart, spleen and liver in α-TS-treated and irradiated mice was evaluated by bacterial culture on sheep blood agar, colistin-nalidixic acid, and xylose-lysine-desoxycholate medium. Our results demonstrate that α-TS enhanced survival in a significant number of mice irradiated with 9.5, 10, 11 and 11.5 Gy (60)Co γ radiation when administered 24 h before radiation exposure. α-TS also protected the intestinal tissue of irradiated mice in terms of crypt and villus number, villus length and mitotic figures. TS treatment decreased the number of TUNEL- and PUMA-positive cells and increased the number of BrdU-positive cells in jejunum compared to vehicle-treated mice. Further, α-TS inhibited gut bacterial translocation to the heart, spleen and liver in irradiated mice. Our data suggest that α-TS protects mice from radiation-induced gastrointestinal damage by inhibiting apoptosis, promoting regeneration of crypt cells, and inhibiting translocation of gut bacteria.


Asunto(s)
Enfermedades Gastrointestinales/prevención & control , Tracto Gastrointestinal/efectos de los fármacos , Traumatismos por Radiación/etiología , Traumatismos por Radiación/prevención & control , Irradiación Corporal Total/efectos adversos , alfa-Tocoferol/administración & dosificación , Animales , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Enfermedades Gastrointestinales/etiología , Tracto Gastrointestinal/diagnóstico por imagen , Tracto Gastrointestinal/patología , Masculino , Ratones , Dosis de Radiación , Protección Radiológica , Protectores contra Radiación/administración & dosificación , Radiografía , Tasa de Supervivencia , Resultado del Tratamiento
16.
Radiat Res ; 173(3): 319-32, 2010 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-20199217

RESUMEN

Abstract Although it is documented that concurrent wounding increases mortality from radiation injury, the molecular mechanism of combined injury is unknown. In this study, mice were exposed to gamma radiation followed by skin wounding. Wound trauma exacerbated radiation-induced mortality, reducing the LD(50/30) from 9.65 Gy to 8.95 Gy. Analyses of histopathology, inducible nitric oxide synthase (iNOS), and serum cytokines were performed on mouse ileum and skin at various times after 9.75 Gy and/or wounding. In the ileum, the villi were significantly shortened 3 days postirradiation but not after wounding; combined injury resulted in decreased villus width and tunica muscularis thickness. The skin of mice subjected to combined injury was less cellular and had a smaller healing bud than the skin of mice subjected to wounding alone. Combined injury significantly delayed wound closure times; it also prolonged the increased levels of iNOS protein in the skin and ileum. iNOS up-regulation was correlated with increases in transcription factors, including NF-kappaB and NF-IL6. The increase in NF-IL6 may be due to increases in cytokines, including IL-1beta, -6, -8, -9, -10 and -13, G-CSF, eotaxin, INF-gamma, MCP-1, MIP-1alpha and MIP-1beta. Combined injury resulted in early detection of bacteria in the blood of the heart and liver, whereas radiation alone resulted in later detection of bacteria; only a transient bacteremia occurred after wounding alone. Results suggest that enhancement of iNOS, cytokines and bacterial infection triggered by combined injury may contribute to mortality. Agents that inhibit these responses may prove to be therapeutic for combined injury and may reduce related mortality.


Asunto(s)
Infecciones Bacterianas , Citocinas/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/microbiología , Transducción de Señal/efectos de la radiación , Animales , Peso Corporal/efectos de la radiación , Citocinas/sangre , Ingestión de Líquidos/efectos de la radiación , Inducción Enzimática/efectos de la radiación , Femenino , Regulación Enzimológica de la Expresión Génica/efectos de la radiación , Corazón/microbiología , Corazón/efectos de la radiación , Íleon/metabolismo , Íleon/efectos de la radiación , Interleucina-6/metabolismo , Hígado/microbiología , Hígado/efectos de la radiación , Ratones , Mortalidad , FN-kappa B/metabolismo , Óxido Nítrico Sintasa de Tipo II/biosíntesis , ARN Mensajero/genética , ARN Mensajero/metabolismo , Traumatismos Experimentales por Radiación/patología , Traumatismos Experimentales por Radiación/fisiopatología , Piel/lesiones , Piel/metabolismo , Piel/fisiopatología , Piel/efectos de la radiación , Factores de Tiempo , Cicatrización de Heridas/efectos de la radiación
17.
Health Phys ; 98(2): 145-52, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20065676

RESUMEN

Combined injuries, which are expected after a radiation dispersal device release or nuclear weapon detonation, are the combination of radiation exposure and tissue injuries from blast and thermal energy. To determine the impact of such trauma, mice were used to (1) evaluate the consequences of skin tissue injuries after various qualities and doses of radiation and (2) document substances that increase survival from radiation injury. Female 12- to 20-wk-old mice weighing 23 +/- 3 g received dorsal skin burns or wounds (15% total body skin surface) under methoxyflurane anesthesia before or after irradiation in this study approved by the Armed Forces Radiobiology Research Institute (AFRRI) Institutional Animal Care and Use Committee. Methoxyflurane is analgesic up to 48 h after injury. The radiations used in these studies included Co gamma photons (1.25 MeV) and research-reactor-produced neutrons with an average energy of 0.96 MeV in either an enriched-field [n/(n + gamma) = 0.95] configuration at 4.2 kW or a mixed-field [n/(n + gamma) = 0.67] configuration operated at 45 kW. Dose rates averaged 0.4 Gy/min. Endpoints included survival, LD50/30s (lethal dose to produce 50% mortality in 30 d), dose modifying factors, relative biological effectiveness values, tissue alterations, susceptibility to bacterial challenge, and countermeasure efficacies. Countermeasures evaluated included S-3-(3-methylaminopropylamino) propylthiophosphorothioic acid (WR-151327), antibiotics, immune modulators, and bone marrow transplantation. Of these treatments, survival was improved by WR-151327, antibiotics, synthetic trehalose discorynomycolate, and bone marrow transplantation. Because trauma to irradiated personnel and medical countermeasures may affect biodosimetric measurements, it will be necessary to quickly determine radiation dose in order to implement appropriate therapy.


Asunto(s)
Quemaduras/fisiopatología , Traumatismo Múltiple/fisiopatología , Traumatismos por Radiación/fisiopatología , Tasa de Supervivencia , Irradiación Corporal Total , Animales , Relación Dosis-Respuesta en la Radiación , Femenino , Ratones , Dosis de Radiación , Análisis de Supervivencia
18.
J Med Microbiol ; 54(Pt 12): 1157-1162, 2005 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-16278429

RESUMEN

Biological agents and ionizing radiation lead to more severe clinical outcomes than either insult alone. This study investigated the survival of non-irradiated and (60)Co-gamma-irradiated mice given therapy for inhalation anthrax with ciprofloxacin (CIP) or a clinically relevant mixture of clarithromycin (CLR) and its major human microbiologically important metabolite 14-hydroxy clarithromycin (14-OH CLR). All B6D2F1/J 10-week-old female mice were inoculated intratracheally with 3 x 10(8) c.f.u. of Bacillus anthracis Sterne spores 4 days after the non-lethal 7 Gy dose of (60)Co gamma radiation. Twenty-one days of treatment with CLR/14-OH CLR, 150 mg kg(-1) twice daily, or CIP, 16.5 mg kg(-1) twice daily, began 24 h after inoculation. Pharmacokinetics indicate that the area under the curve (AUC) for 14-OH CLR on the concentration-versus-time graph was slightly higher in gamma-irradiated than non-irradiated animals. Neither drug was able to increase survival in gamma-irradiated animals. CIP and CLR/14-OH CLR therapies in non-irradiated animals increased survival from 49 % (17/35 mice) in buffer-treated animals to 94 % (33/35) and 100 %, respectively (P < 0.001). B. anthracis Sterne only was isolated from 25-50 % of treated mice with or without irradiation. Mixed infections with B. anthracis Sterne were present in 50-71 % of gamma-irradiated mice but only in 5-10 % of mice without irradiation.


Asunto(s)
Carbunco/tratamiento farmacológico , Carbunco/radioterapia , Ciprofloxacina/uso terapéutico , Claritromicina/uso terapéutico , Animales , Antibacterianos/uso terapéutico , Ciprofloxacina/sangre , Ciprofloxacina/farmacocinética , Claritromicina/sangre , Claritromicina/farmacocinética , Cobalto , Quimioterapia Combinada , Femenino , Rayos gamma , Ratones
19.
Mol Cell Biochem ; 272(1-2): 29-34, 2005 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16010969

RESUMEN

New, safe antimicrobial agents are needed to prevent and overcome severe bacterial, viral, and fungal infections. Based on our previous experience and that of others, we postulated that herbal essential oils, such as those of origanum, and monolaurin offer such possibilities. We examined in vitro the cidal and/or static effects of oil of origanum, several other essential oils, and monolaurin on Staphylococcus aureus, Bacillus anthracis Sterne, Escherichia coli, Klebsiella pneumoniae, Helicobacter pylori, and Mycobacterium terrae. Origanum proved cidal to all tested organisms with the exception of B. anthracis Sterne in which it was static. Monolaurin was cidal to S. aureus and M. terrae but not to E. coli and K. pneumoniae. Unlike the other two gram-negative organisms, H. pylori were extremely sensitive to monolaurin. Similar to origanum, monolaurin was static to B. anthracis Sterne. Because of their longstanding safety record, origanum and/or monolaurin, alone or combined with antibiotics, might prove useful in the prevention and treatment of severe bacterial infections, especially those that are difficult to treat and/or are antibiotic resistant.


Asunto(s)
Antibacterianos/farmacología , Glicéridos/farmacología , Bacterias Gramnegativas/efectos de los fármacos , Bacterias Grampositivas/efectos de los fármacos , Lauratos/farmacología , Aceites Volátiles/farmacología , Origanum/química , Aceites de Plantas/farmacología , Glicéridos/análisis , Lauratos/análisis , Pruebas de Sensibilidad Microbiana , Monoglicéridos
20.
Comp Med ; 54(2): 185-92, 2004 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-15134365

RESUMEN

Klebsiella pneumoniae is a common cause of nosocomially acquired pneumonia in immunocompromised patients. Previously, we established a pneumonia model using Klebsiella pneumoniae in B6D2F1/J mice sublethally irradiated with 7-Gy 60Co gamma-radiation and inoculated intratracheally. In the study reported here, we investigated survival of mice following 10 days of antimicrobial therapy with ceftriaxone, gentamicin, gatifloxacin, and a ceftriaxone-gentamicin combination given once daily. Survival was significantly prolonged in response to all therapies. However, survival of mice was 95% when treated with the ceftriaxone-gentamicin combination followed by ceftriaxone alone (75%), and gatifloxacin (80%), whereas survival for controls was 0%. In addition, resistance to any of the treatments did not develop during the study. We conclude that an immunocompromised status does not alter the Infectious Disease Society of America's primary recommendation for treating community-acquired K. pneumoniae pneumonia using a third-generation cephalosporin, with or without an aminoglycoside.


Asunto(s)
Antibacterianos/uso terapéutico , Huésped Inmunocomprometido , Infecciones por Klebsiella/tratamiento farmacológico , Neumonía Bacteriana/tratamiento farmacológico , Animales , Ceftriaxona/uso terapéutico , Combinación de Medicamentos , Femenino , Fluoroquinolonas/uso terapéutico , Gatifloxacina , Gentamicinas/uso terapéutico , Humanos , Klebsiella pneumoniae/metabolismo , Ratones , Ratones Endogámicos , Tasa de Supervivencia , Irradiación Corporal Total
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...