Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
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
2.
Int J Mol Sci ; 18(8)2017 Aug 03.
Artículo en Inglés | MEDLINE | ID: mdl-28771181

RESUMEN

Nuclear industrial accidents and the detonation of nuclear devices cause a variety of damaging factors which, when their impacts are combined, produce complicated injuries challenging for medical treatment. Thus, trauma following acute ionizing irradiation (IR) can deteriorate the IR-induced secondary reactive metabolic and inflammatory impacts to dose-limiting tissues, such as bone marrow/lymphatic, gastrointestinal tissues, and vascular endothelial tissues, exacerbating the severity of the primary injury and decreasing survival from the exposure. Previously we first reported that ghrelin therapy effectively improved survival by mitigating leukocytopenia, thrombocytopenia, and bone-marrow injury resulting from radiation combined with burn trauma. This study was aimed at investigating whether radiation combined with burn trauma induced the cerebro-vascular impairment and intracranial hemorrhage that could be reversed by ghrelin therapy. When B6D2F1 female mice were exposed to 9.5 Gy Cobalt-60 γ-radiation followed by 15% total skin surface burn, cerebro-vascular impairment and intracranial hemorrhage as well as platelet depletion were observed. Ghrelin treatment after irradiation combined with burn trauma significantly decreased platelet depletion and brain hemorrhage. The results suggest that ghrelin treatment is an effective therapy for ionizing radiation combined with burn trauma.


Asunto(s)
Quemaduras/tratamiento farmacológico , Rayos gamma/efectos adversos , Ghrelina/farmacología , Hemorragias Intracraneales/tratamiento farmacológico , Traumatismos Experimentales por Radiación/tratamiento farmacológico , Animales , Quemaduras/metabolismo , Quemaduras/patología , Femenino , Hemorragias Intracraneales/metabolismo , Hemorragias Intracraneales/patología , Ratones , Traumatismos Experimentales por Radiación/metabolismo , Traumatismos Experimentales por Radiación/patología
3.
J Cell Mol Med ; 19(5): 1133-50, 2015 May.
Artículo en Inglés | MEDLINE | ID: mdl-25721260

RESUMEN

The bone marrow stroma constitutes the marrow-blood barrier, which sustains immunochemical homoeostasis and protection of the haematopoietic tissue in sequelae of systemic bacterial infections. Under these conditions, the bone marrow stromal cells affected by circulating bacterial pathogens shall elicit the adaptive stress-response mechanisms to maintain integrity of the barrier. The objective of this communication was to demonstrate (i) that in vitro challenge of mesenchymal stromal cells, i.e. colony-forming unit fibroblasts (CFU-F), with Staphylococcus epidermidis can activate the autophagy pathway to execute antibacterial defence response, and (ii) that homoeostatic shift because of the bacteria-induced stress includes the mitochondrial remodelling and sequestration of compromised organelles via mitophagy. Implication of Drp1 and PINK1-PARK2-dependent mechanisms in the mitophagy turnover of the aberrant mitochondria in mesenchymal stromal cells is investigated and discussed.


Asunto(s)
Autofagia/fisiología , Células Madre Mesenquimatosas/metabolismo , Mitocondrias/metabolismo , Staphylococcus epidermidis/crecimiento & desarrollo , Animales , Autofagia/genética , Células Cultivadas , Dinaminas/genética , Dinaminas/metabolismo , Expresión Génica , Interacciones Huésped-Patógeno , Immunoblotting , Lisosomas/metabolismo , Lisosomas/ultraestructura , Células Madre Mesenquimatosas/microbiología , Células Madre Mesenquimatosas/ultraestructura , Microscopía Confocal , Mitocondrias/ultraestructura , Mitofagia/genética , Mitofagia/fisiología , Fagosomas/metabolismo , Fagosomas/ultraestructura , Proteínas Quinasas/genética , Proteínas Quinasas/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/genética , Staphylococcus epidermidis/fisiología , Ubiquitina-Proteína Ligasas/genética , Ubiquitina-Proteína Ligasas/metabolismo
4.
Biometals ; 25(1): 203-17, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22089858

RESUMEN

Although iron is known to be a component of the pathogenesis and/or maintenance of acute lung injury (ALI) in experimental animals and human subjects, the majority of these studies have focused on disturbances in iron homeostasis in the airways resulting from exposure to noxious gases and particles. Considerably less is known about the effect of increased plasma levels of redox-reactive non-transferrin bound iron (NTBI) and its impact on pulmonary endothelium. Plasma levels of NTBI can increase under various pathophysiological conditions, including those associated with ALI, and multiple mechanisms are in place to affect the [Fe(2+)]/[Fe(3+)] redox steady state. It is well accepted, however, that intracellular transport of NTBI occurs after reduction of [Fe(3+)] to [Fe(2+)] (and is mediated by divalent metal transporters). Accordingly, as an experimental model to investigate mechanisms mediating vascular effects of redox reactive iron, rat pulmonary artery endothelial cells (RPAECs) were subjected to pulse treatment (10 min) with [Fe(2+)] nitriloacetate (30 µM) in the presence of pyrithione, an iron ionophore, to acutely increase intracellular labile pool of iron. Cellular iron influx and cell shape profile were monitored with time-lapse imaging techniques. Exposure of RPAECs to [Fe(2+)] resulted in: (i) an increase in intracellular iron as detected by the iron sensitive fluorophore, PhenGreen; (ii) depletion of cell glutathione; and (iii) nuclear translocation of stress-response transcriptional factors Nrf2 and NFkB (p65). The resulting iron-induced cell alterations were characterized by cell polarization and formation of membrane cuplike and microvilli-like projections abundant with ICAM-1, caveolin-1, and F-actin. The iron-induced re-arrangements in cytoskeleton, alterations in focal cell-cell interactions, and cell buckling were accompanied by decrease in electrical resistance of RPAEC monolayer. These effects were partially eliminated in the presence of N,N'-bis (2-hydroxybenzyl) ethylenediamine-N,N'-diacetic acid, an iron chelator, and Y27632, a Rho-kinase inhibitor. Thus acute increases in labile iron in cultured pulmonary endothelium result in structural remodeling (and a proinflammatory phenotype) that occurs via post-transcriptional mechanisms regulated in a redox sensitive fashion.


Asunto(s)
Células Endoteliales/efectos de los fármacos , Células Endoteliales/fisiología , Hierro/farmacología , Arteria Pulmonar/citología , Animales , Caveolina 1/metabolismo , Células Cultivadas , Impedancia Eléctrica , Células Endoteliales/citología , Glutatión/metabolismo , Humanos , Molécula 1 de Adhesión Intercelular/metabolismo , Hierro/metabolismo , Ratas , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo
5.
Biometals ; 24(2): 279-89, 2011 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-21229381

RESUMEN

Extracellular iron has been implicated in the pathogenesis of post-injury organ failure. However, the source(s) and biochemical species of this iron have not been identified. Based upon evidence that distant organ injury results from an increase in intestinal permeability, we looked for ferrous iron in mesenteric lymph in anesthetized rats undergoing hemorrhage and fluid resuscitation (H/R). Ferrous iron increased in lymph from 4.7 nmol/mg of protein prior to hemorrhage to 86.6 nmol/mg during resuscitation. Utilizing immuno-spin trapping in protein fractions that were rich in iron, we tentatively indentified protein carrier(s) of ferrous iron by MALDI-TOF MS. One of the identified proteins was the metalloproteinase (MMP) inhibitor, TIMP-2. Antibody to TIMP-2 immunoprecipitated 74% of the ferrozine detectable iron in its protein fraction. TIMP-2 binds iron in vitro at pH 6.3, which is typical of conditions in the mesentery during hemorrhage, but it retains the ability to inhibit the metalloproteases MMP-2 and MMP-9. In summary, there is a large increase in extracellular ferrous iron in the gut in H/R demonstrating dysregulation of iron homeostasis. We have identified, for the first time, the binding of extracellular iron to TIMP-2.


Asunto(s)
Hemorragia/metabolismo , Hierro/metabolismo , Inhibidor Tisular de Metaloproteinasa-2/metabolismo , Animales , Cromatografía en Gel , Espectroscopía de Resonancia por Spin del Electrón , Concentración de Iones de Hidrógeno , Inmunoprecipitación , Masculino , Inhibidores de la Metaloproteinasa de la Matriz , Ratas , Ratas Sprague-Dawley , Resucitación , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Inhibidor Tisular de Metaloproteinasa-2/farmacología
6.
Radiat Res ; 196(1): 1-16, 2021 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-33979447

RESUMEN

Exposure to ionizing radiation, mechanical trauma, toxic chemicals or infections, or combinations thereof (i.e., combined injury) can induce organic injury to brain tissues, the structural disarrangement of interactive networks of neurovascular and glial cells, as well as on arrays of the paracrine and systemic destruction. This leads to subsequent decline in cognitive capacity and decompensation of mental health. There is an ongoing need for improvement in mitigating and treating radiation- or combined injury-induced brain injury. Cranial irradiation per se can cause a multifactorial encephalopathy that occurs in a radiation dose- and time-dependent manner due to differences in radiosensitivity among the various constituents of brain parenchyma and vasculature. Of particular concern are the radiosensitivity and inflammation susceptibility of: 1. the neurogenic and oligodendrogenic niches in the subependymal and hippocampal domains; and 2. the microvascular endothelium. Thus, cranial or total-body irradiation can cause a plethora of biochemical and cellular disorders in brain tissues, including: 1. decline in neurogenesis and oligodendrogenesis; 2. impairment of the blood-brain barrier; and 3. ablation of vascular capillary. These changes, along with cerebrovascular inflammation, underlie different stages of encephalopathy, from the early protracted stage to the late delayed stage. It is evident that ionizing radiation combined with other traumatic insults such as penetrating wound, burn, blast, systemic infection and chemotherapy, among others, can exacerbate the radiation sequelae (and vice versa) with increasing severity of neurogenic and microvascular patterns of radiation brain damage.


Asunto(s)
Lesiones Encefálicas/etiología , Encéfalo/efectos de la radiación , Circulación Cerebrovascular/efectos de la radiación , Traumatismos por Radiación/etiología , Radiación Ionizante , Radioterapia/efectos adversos , Animales , Encéfalo/irrigación sanguínea , Encéfalo/patología , Relación Dosis-Respuesta en la Radiación , Humanos
7.
J Pathol ; 219(2): 242-52, 2009 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-19681094

RESUMEN

Macroautophagy (mAG) is a lysosomal mechanism of degradation of cell self-constituents damaged due to variety of stress factors, including ionizing irradiation. Activation of mAG requires expression of mAG protein Atg8 (LC3) and conversion of its form I (LC3-I) to form II (LC3-II), mediated by redox-sensitive Atg4 protease. We have demonstrated upregulation of this pathway in the innate host defense Paneth cells of the small intestine (SI) due to ionizing irradiation and correlation of this effect with induction of pro-oxidant inducible nitric oxide synthase (iNOS). CD2F1 mice were exposed to 9.25 Gy gamma-ionizing irradiation. Small intestinal specimens were collected during 7 days after ionizing irradiation. Assessment of ionizing irradiation-associated alterations in small intestinal crypt and villus cells and activation of the mAG pathway was conducted using microscopical and biochemical techniques. Analysis of iNOS protein and the associated formation of nitrites and lipid peroxidation products was performed using immunoblotting and biochemical analysis, and revealed increases in iNOS protein, nitrate levels and oxidative stress at day 1 following ionizing irradiation. Increase in immunoreactivity of LC3 protein in the crypt cells was observed at day 7 following ionizing irradiation. This effect predominantly occurred in the CD15-positive Paneth cells and was associated with accumulation of LC3-II isoform. The formation of autophagosomes in Paneth cells was confirmed by transmission electron microscopy (TEM). Up-regulation of LC3 pathway in the irradiated SI was accompanied by a decreased protein-protein interaction between LC3 and chaperone heat shock protein 70. A high-level of LC3-immunoreactivity in vacuole-shaped structures was spatially co-localized with immunoreactivity of 3-nitro-tyrosine. The observed effects were diminished in iNOS knockout B6.129P2-NOS2(tm1Lau)/J mice subjected to the same treatments. We postulate that the observed up-regulation of mAG in the irradiated small intestine is at least in part mediated by the iNOS signalling mechanism.


Asunto(s)
Autofagia/efectos de la radiación , Rayos gamma , Intestino Delgado/efectos de la radiación , Células de Paneth/efectos de la radiación , Regulación hacia Arriba/efectos de la radiación , Animales , Etiquetado Corte-Fin in Situ , Intestino Delgado/ultraestructura , Masculino , Ratones , Ratones Endogámicos , Ratones Noqueados , Microscopía Confocal , Microscopía Electrónica de Rastreo , Óxido Nítrico Sintasa de Tipo II/fisiología , Estrés Oxidativo/efectos de la radiación , Células de Paneth/ultraestructura , Irradiación Corporal Total
8.
J Cell Mol Med ; 13(2): 373-87, 2009 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-18793351

RESUMEN

Autophagy is an intracellular process in which a cell digests its own constituents via lysosomal degradative pathway. Though autophagy has been shown in several cardiac diseases like heart failure, hypertrophy and ischaemic cardiomyopathy, the role and the regulation of autophagy is still largely unknown. Bcl-2-associated athanogene (BAG-1) is a multifunctional pro-survival molecule that binds with Hsp70/Hsc70. In this study, myocardial adaptation to ischaemia by repeated brief episodes of ischaemia and reperfusion (I/R) prior to lethal I/R enhanced the expression of autophagosomal membrane specific protein light chain 3 (LC3)-II, and Beclin-1, a molecule involved in autophagy and BAG-1. Autophagosomes structures were found in the adapted myocardium through electron microscopy. Co-immunoprecipitation and co-immunofluorescence analyses revealed that LC3-II was bound with BAG-1. Inhibition of autophagy by treating rats with Wortmannin (15 microg/kg; intraperitoneally) abolished the ischaemic adaptation-induced induction of LC3-II, Beclin-1, BAG-1 and cardioprotection. Intramyocardial injection of BAG-1 siRNA attenuated the induction of LC3-II, and abolished the cardioprotection achieved by adaptation. Furthermore, hypoxic adaptation in cardiac myoblast cells induced LC3-II and BAG-1. BAG-1 siRNA treatment attenuated hypoxic adaptation-induced LC3-II and BAG-1, and abolished improvement in cardiac cell survival and reduction of cell death. These results clearly indicate that myocardial protection elicited by adaptation is mediated at least in part via up-regulation of autophagy in association with BAG-1 protein.


Asunto(s)
Autofagia/fisiología , Cardiotónicos/metabolismo , Proteínas de Unión al ADN/metabolismo , Isquemia/metabolismo , Miocardio/metabolismo , Factores de Transcripción/metabolismo , Androstadienos/metabolismo , Animales , Proteínas Reguladoras de la Apoptosis/metabolismo , Beclina-1 , Células Cultivadas , Proteínas de Unión al ADN/genética , Precondicionamiento Isquémico Miocárdico , Masculino , Proteínas Asociadas a Microtúbulos/genética , Proteínas Asociadas a Microtúbulos/metabolismo , Miocardio/ultraestructura , ARN Interferente Pequeño/metabolismo , Ratas , Ratas Sprague-Dawley , Factores de Transcripción/genética , Wortmanina
9.
Free Radic Biol Med ; 43(3): 397-407, 2007 Aug 01.
Artículo en Inglés | MEDLINE | ID: mdl-17602955

RESUMEN

A recent study showed that cardiac adaptation could potentiate translocation of thioredoxin-1 (Trx-1) into the nucleus, which then interacted with Ref-1, resulting in a survival signal. Here, we present evidence that such adaptation also causes nuclear translocation of Ref-1, which is almost completely inhibited when the hearts were pretreated with antisense Ref-1 that also abolished the cardioprotective adaptive response. Significant amounts of NFkappaB and Nrf2 were found to be associated with Ref-1 when the nuclear extract obtained from the left ventricle was immunoprecipitated with Ref-1. Such Ref-1-NFkappaB and Ref-1-Nrf2 interactions were significantly inhibited with antisense Ref-1. However, immunoprecipitation of nuclear extract with NFkappaB showed that the association of Trx-1 with NFkappaB is increased in the adapted heart, which was again significantly blocked by antisense Ref-1. Nrf2 was also associated with NFkappaB; however, such association appeared to be independent of Ref-1. In contrast, myocardial adaptation to ischemia inhibited the ischemia reperfusion-induced loss of Nrf2 from the nucleus, which was inhibited by antisense Ref-1. The nuclear translocation and activation of Ref-1 appeared to generate a survival signal as evidenced by the increased phosphorylation of Akt that was inhibited with antisense Ref-1. Finally, confocal microscopy confirmed the results of immunoblotting, clearly showing the nuclear translocation of Ref-1 and nuclear 3D colocalization of Ref-1 with NFkappaB in the adapted heart and its inhibition with antisense Ref-1. Our results show that PC potentiates a survival signal through the phosphorylation of Akt by causing nuclear translocation and activation of Ref-1, where significant interaction among NFkappaB and Ref-1, Trx-1, and Nrf2 appears to regulate Ref-1-induced survival signal.


Asunto(s)
ADN-(Sitio Apurínico o Apirimidínico) Liasa/fisiología , Daño por Reperfusión Miocárdica/fisiopatología , Animales , Núcleo Celular/metabolismo , Supervivencia Celular/fisiología , Activación Enzimática , Masculino , Malondialdehído/metabolismo , Miocardio/metabolismo , Factor 2 Relacionado con NF-E2/metabolismo , FN-kappa B/metabolismo , Oligodesoxirribonucleótidos Antisentido/farmacología , Fosforilación , Transporte de Proteínas/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/metabolismo , Ratas , Ratas Sprague-Dawley , Tiorredoxinas/metabolismo
10.
Antioxid Redox Signal ; 9(4): 483-95, 2007 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-17280489

RESUMEN

Transmigration of phagocytic leukocytes (PLCs) from the peripheral blood into injured lung requires a conversion of the microvascular endothelial cells (ECs) to the proinflammatory phenotypes and spatiotemporal interplay of different types of cell adhesion molecules (CAMs) on PLC and endothelium. The present report is focused on involvement of iron-dependent redox signaling in spatial coordination of lung CAM due to either a pulmonary trauma or endotracheal iron administration in rats. Redox alterations, deposition of 3-nitrotyrosine, expression of VE-cadherin, ICAM-1, and the PLC integrins, and the status of thioredoxin, Ref-1, NF-kappaB and Nrf2 redox-sensitive elements in the alveolar microvasculature were assessed with EPR spectroscopy, immunobloting, and confocal microscopy. We demonstrated for the first time in vivo that the presence of catalytically active iron, deposition of myeloperoxidase, and induction of the oxidative stress in the lung-injury models were accompanied by (a) downregulation of VE-cadherin, (b) upregulation and polarization of ICAM-1 and the PLC integrins, and (c) nuclear translocation and interaction of thioredoxin, Ref-1, and NF-kappaB and complex structural changes in EC and PLC at the sites of their contacts. The studies suggested that a part of the proinflammatory action of iron in the lung resulted from its stimulation of the redox-sensitive factors.


Asunto(s)
Moléculas de Adhesión Celular/metabolismo , Hierro/metabolismo , Enfermedades Pulmonares Intersticiales/metabolismo , Pulmón/metabolismo , Animales , Antígenos CD/metabolismo , Cadherinas/metabolismo , ADN-(Sitio Apurínico o Apirimidínico) Liasa/metabolismo , Espectroscopía de Resonancia por Spin del Electrón , Células Endoteliales/efectos de los fármacos , Células Endoteliales/metabolismo , Immunoblotting , Integrinas/metabolismo , Molécula 1 de Adhesión Intercelular/metabolismo , Hierro/farmacología , Leucocitos/efectos de los fármacos , Leucocitos/metabolismo , Pulmón/efectos de los fármacos , Pulmón/patología , Enfermedades Pulmonares Intersticiales/inducido químicamente , Masculino , Microcirculación/efectos de los fármacos , Microscopía Confocal , FN-kappa B/metabolismo , Oxidación-Reducción , Peroxidasa/metabolismo , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/metabolismo , Alveolos Pulmonares/patología , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos , Tiorredoxinas/metabolismo
11.
Toxicol Sci ; 95(1): 289-96, 2007 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-17060374

RESUMEN

Blast overpressure (BOP), also known as high energy impulse noise, is a damaging outcome of explosive detonations and firing of weapons. Exposure to BOP shock waves alone results in injury predominantly to the hollow organ systems such as auditory, respiratory, and gastrointestinal systems. In recent years, the hazards of BOP that once were confined to military and professional settings have become a global societal problem as terrorist bombings and armed conflicts involving both military and civilian populations increased significantly. We have previously investigated the effects of single BOP exposures at different peak pressures. In this study, we examined the effects of repeated exposure to a low-level BOP and whether the number of exposures or time after exposure would alter the injury outcome. We exposed deeply anesthetized rats to simulated BOP at 62 +/- 2 kPa peak pressure. The lungs were examined immediately after one exposure (1 + 0), or 1 h after one (1 + 1), two (2 + 1), or three (3 + 1) consecutive exposures at 3-min interval. In one group of animals, we examined the effects of repeated exposure on lung weight, methemoglobin, transferrin, antioxidants, and lipid peroxidation. In a second group, the lungs were fixed inflated at 25 cm water, sectioned, and examined histologically after one to three repeated exposures, or after one exposure at 1, 6, and 24 h. We found that single BOP exposure causes notable changes after 1 h, and that repeating BOP exposure did not add markedly to the effect of the first one. However, the effects increased significantly with time from 1 to 24 h. These observations have biological and occupational implications, and emphasize the need for protection from low-level BOP, and for prompt treatment within the first hour following BOP exposure.


Asunto(s)
Traumatismos por Explosión/metabolismo , Traumatismos por Explosión/patología , Explosiones , Pulmón/metabolismo , Pulmón/patología , Estrés Oxidativo , Presión del Aire , Animales , Antioxidantes/metabolismo , Ácido Ascórbico/metabolismo , Peroxidación de Lípido , Lesión Pulmonar , Masculino , Metahemoglobina/metabolismo , Personal Militar , Enfermedades Profesionales , Tamaño de los Órganos , Ratas , Ratas Sprague-Dawley , Organismos Libres de Patógenos Específicos , Factores de Tiempo , Transferrina/metabolismo , Vitamina E/metabolismo
12.
J Appl Physiol (1985) ; 100(4): 1267-77, 2006 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-16339342

RESUMEN

The results of previous inhibitor studies suggest that there is some increase in nitric oxide (NO) production from constitutive NO synthase in early hemorrhage (H), but the magnitude of NO production early after H has not been previously assessed. It is generally believed that only modest production rates are possible from the constitutively expressed NO synthases. To study this, anesthetized male Sprague-Dawley rats were subjected to 90 min of isobaric (40 mmHg) H. During this period of time, the dynamics of accumulation of NO intermediates in the arterial blood was assessed using electron paramagnetic resonance spectroscopy, chemiluminescence, fluorescence imaging, and mass spectrometry. Electron paramagnetic resonance-detectable NO adducts were also measured with spin traps in blood plasma and red blood cells. H led to an increase in the concentration of hemoglobin-NO from 0.9 +/- 0.2 to 4.8 +/- 0.7 microM. This accumulation was attenuated by a nonselective inhibitor of NO synthase, NG-nitro-L-argininemethyl ester (L-NAME), but not by NG-nitro-D-argininemethyl ester (D-NAME) or 1400W. Administration of L-NAME (but not 1400W or D-NAME) during H produced a short-term increase in mean arterial pressure ( approximately 90%). In H, the level of N oxides in red blood cells increased sevenfold. S-nitrosylation of plasma proteins was revealed with "biotin switch" techniques. The results provide compelling evidence that there is brisk production of NO in early H. The results indicate that the initial compensatory response to H is more complicated than previously realized, and it involves an orchestrated balance between intense vasoconstrictor and vasodilatory components.


Asunto(s)
Hemorragia/sangre , Óxido Nítrico/sangre , S-Nitrosotioles/sangre , Animales , Presión Sanguínea/efectos de los fármacos , Ceruloplasmina/metabolismo , Inhibidores Enzimáticos/farmacología , Eritrocitos/metabolismo , Hemoglobina Glucada , Hemorragia/enzimología , Hemorragia/fisiopatología , Masculino , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , Óxido Nítrico Sintasa/metabolismo , Ratas , Ratas Sprague-Dawley , Flujo Sanguíneo Regional , Factores de Tiempo
13.
Antioxidants (Basel) ; 4(1): 134-52, 2015 Feb 27.
Artículo en Inglés | MEDLINE | ID: mdl-26785342

RESUMEN

The biological effects of high-dose total body ionizing irradiation [(thereafter, irradiation (IR)] are attributed to primary oxidative breakage of biomolecule targets, mitotic, apoptotic and necrotic cell death in the dose-limiting tissues, clastogenic and epigenetic effects, and cascades of functional and reactive responses leading to radiation sickness defined as the acute radiation syndrome (ARS). The range of remaining and protracted injuries at any given radiation dose as well as the dynamics of post-IR alterations is tissue-specific. Therefore, functional integrity of the homeostatic tissue barriers may decline gradually within weeks in the post-IR period culminating with sepsis and failure of organs and systems. Multiple organ failure (MOF) leading to moribundity is a common sequela of the hemotapoietic form of ARS (hARS). Onset of MOF in hARS can be presented as "two-hit phenomenon" where the "first hit" is the underlying consequences of the IR-induced radiolysis in cells and biofluids, non-septic inflammation, metabolic up-regulation of pro-oxidative metabolic reactions, suppression of the radiosensitive hematopoietic and lymphoid tissues and the damage to gut mucosa and vascular endothelium. While the "second hit" derives from bacterial translocation and spread of the bacterial pathogens and inflammagens through the vascular system leading to septic inflammatory, metabolic responses and a cascade of redox pro-oxidative and adaptive reactions. This sequence of events can create a ground for development of prolonged metabolic, inflammatory, oxidative, nitrative, and carbonyl, electrophilic stress in crucial tissues and thus exacerbate the hARS outcomes. With this perspective, the redox mechanisms, which can mediate the IR-induced protracted oxidative post-translational modification of proteins, oxidation of lipids and carbohydrates and their countermeasures in hARS are subjects of the current review. Potential role of ubiquitous, radioresistant mesenchymal stromal cells in the protracted responses to IR and IR-related septicemia is also discussed.

14.
Toxicology ; 189(1-2): 63-74, 2003 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-12821283

RESUMEN

High-energy impulse noise (BLAST) is a physical event characterized by an abrupt rise in atmospheric pressure above ambient lasting for a very short period, but potentially causing significant material and biological damage. Exposure to high-level BLAST can be destructive and lethal. Low-level BLAST similar to what is encountered repeatedly by military personnel during training and combat from detonation of munitions and firing of large caliber weapons, and during occupational use of explosives and some heavy machinery, can also cause significant injury. Globally, civilians are increasingly exposed to BLAST resulting from terrorist bombings or abandoned unmarked mines following numerous wars and conflicts. We have shown previously in several animal models that exposure to non-lethal BLAST results in pathological changes, mostly to the hollow organs characterized in the lungs, the most sensitive organ, by rupture of alveolar septa, and pulmonary hemorrhage and edema. These events potentially can cause alveolar flooding, respiratory insufficiency and adult respiratory distress syndrome (ARDS), leading to varying degrees of hypoxia, antioxidant depletion and oxidative damage. We have also observed progressive formation of nitric oxide in blood and other tissues. The totality of these observations supports our general hypothesis that exposure to BLAST can lead to antioxidant depletion and oxidative damage. Understanding the mechanism(s) of BLAST-induced oxidative stress may have important implications that include a potential beneficial role for antioxidants as a prophylaxis or as secondary treatment of injury after exposure alongside other protective and therapeutic modalities. In addition, it suggests a role for endogenous nitric oxide in the injury. This report reviews experimental evidence of BLAST-induced antioxidant depletion, and the potential benefit from antioxidant supplementation before exposure.


Asunto(s)
Antioxidantes/metabolismo , Traumatismos por Explosión/patología , Explosiones , Lesión Pulmonar , Animales , Antioxidantes/uso terapéutico , Ácido Ascórbico/metabolismo , Ácido Ascórbico/uso terapéutico , Traumatismos por Explosión/tratamiento farmacológico , Modelos Animales de Enfermedad , Glutatión/metabolismo , Humanos , Pulmón/metabolismo , Pulmón/patología , Estrés Oxidativo/efectos de los fármacos , Estrés Oxidativo/fisiología , Vitamina E/metabolismo , Vitamina E/uso terapéutico
15.
Toxicology ; 179(3): 257-66, 2002 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-12270597

RESUMEN

The p53 protein is widely regarded as an important sensor of genotoxic damage in cells, and mutations in p53 are the most frequent observed in human cancers. Rapid assays for evaluating the potential of a chemical or physical agent to alter the transcriptional regulatory role of p53 may therefore serve as useful tools in toxicological research. In this study, the use of enhanced green fluorescent protein (EGFP) as a live cell reporter to assess the transactivation response of p53 to chemical and physical agents was evaluated. A stable murine bone marrow stromal cell line (D2XRIIGFP24) expressing EGFP under control of p53 response elements was established. D2XRIIGFP24 cells displayed low constitutive background fluorescence which was significantly enhanced in response to exposure to agents that induced p53 protein levels. Increases in EGFP fluorescence in response to oxidative and nitrosative stress as well as UVC irradiation were dose-dependent, detectable within 3 h of exposure and correlated closely with the amount of p53 protein accumulated within the cell. The results demonstrate the potential for rapid and early detection of p53 transactivation using the EGFP reporter approach and indicate this approach is adaptable to a variety of fluorescent assay techniques and a useful cell model for molecular toxicology research.


Asunto(s)
Células de la Médula Ósea/citología , Línea Celular , Células Clonales , Pruebas de Mutagenicidad/métodos , Estrés Fisiológico/genética , Proteína p53 Supresora de Tumor/metabolismo , Animales , Ensayo Cometa , Relación Dosis-Respuesta a Droga , Relación Dosis-Respuesta en la Radiación , Genes Reporteros , Proteínas Fluorescentes Verdes , Peróxido de Hidrógeno/toxicidad , Cinética , Proteínas Luminiscentes/metabolismo , Ratones , Óxido Nítrico/toxicidad , Estrés Oxidativo , Células del Estroma , Activación Transcripcional , Proteína p53 Supresora de Tumor/análisis , Proteína p53 Supresora de Tumor/genética , Rayos Ultravioleta/efectos adversos
16.
J Cell Sci Ther ; 5(6)2014.
Artículo en Inglés | MEDLINE | ID: mdl-34457993

RESUMEN

The aim of this study was to investigate whether treatment with mesenchymal stem cells (MSCs) could improve survival after radiation combined injury. Bone marrow MSCs (BMSCs) were isolated from femurs of B6D2F1/J female mice and were expanded and cultivated in hypoxic conditions (5% O2, 10% CO2, 85% N2) over 30 days. BMSCs were transfused to mice 24 hr after combined injury due to 60Co-γ-photon irradiation (9.25 and 9.75 Gy, 0.4 Gy/min, bilateral) followed by skin wounding (CI). Water consumption, body weight, wound healing, and survival tallies were monitored during observation period. Mice subjected to CI experienced a dramatic moribundity over a 30-day observation period. Thus, CI (9.25 Gy)-animal group was characterized by 40% mortality rate while CI (9.75 Gy)-animal group had 100% mortality rate. CI-induced sickness was accompanied by body weight loss, increased water intake, and delayed wound healing. At the 30th day post-injury, bone marrow cell depletion still remained in surviving CI mice. Treatment of CI (9.25 Gy)-animal group with BMSCs led to an increase in 30-day survival rate by 30%, attenuated body weight loss, accelerated wound healing rate, and ameliorated bone-marrow cell depletion. Our novel results are the first to suggest that BMSC therapy is efficacious to sustain animal survival after CI.

17.
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
18.
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
19.
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
20.
Oxid Med Cell Longev ; 2013: 186795, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23710283

RESUMEN

Acute bacterial inflammation is accompanied by excessive release of bacterial toxins and production of reactive oxygen and nitrogen species (ROS and RNS), which ultimately results in redox stress. These factors can induce damage to components of tissue barriers, including damage to ubiquitous mesenchymal stromal cells (MSCs), and thus can exacerbate the septic multiple organ dysfunctions. The mechanisms employed by MSCs in order to survive these stress conditions are still poorly understood and require clarification. In this report, we demonstrated that in vitro treatment of MSCs with lipopolysaccharide (LPS) induced inflammatory responses, which included, but not limited to, upregulation of iNOS and release of RNS and ROS. These events triggered in MSCs a cascade of responses driving adaptive remodeling and resistance to a "self-inflicted" oxidative stress. Thus, while MSCs displayed high levels of constitutively present adaptogens, for example, HSP70 and mitochondrial Sirt3, treatment with LPS induced a number of adaptive responses that included induction and nuclear translocation of redox response elements such as NFkB, TRX1, Ref1, Nrf2, FoxO3a, HO1, and activation of autophagy and mitochondrial remodeling. We propose that the above prosurvival pathways activated in MSCs in vitro could be a part of adaptive responses employed by stromal cells under septic conditions.


Asunto(s)
Inflamación/patología , Lipopolisacáridos/farmacología , Células Madre Mesenquimatosas/metabolismo , Sepsis/patología , Animales , Núcleo Celular/efectos de los fármacos , Núcleo Celular/metabolismo , Técnica del Anticuerpo Fluorescente , Inflamación/metabolismo , Células Madre Mesenquimatosas/efectos de los fármacos , Células Madre Mesenquimatosas/ultraestructura , Ratones , Mitocondrias/efectos de los fármacos , Mitocondrias/metabolismo , Mitocondrias/ultraestructura , FN-kappa B/efectos de los fármacos , FN-kappa B/metabolismo , FN-kappa B/ultraestructura , Óxido Nítrico/metabolismo , Óxido Nítrico Sintasa de Tipo II/metabolismo , Oxidación-Reducción/efectos de los fármacos , Fagosomas/efectos de los fármacos , Fagosomas/metabolismo , Fagosomas/ultraestructura , Especies de Nitrógeno Reactivo/metabolismo , Especies Reactivas de Oxígeno/metabolismo , Elementos de Respuesta/genética , Sepsis/metabolismo , Tiorredoxinas/metabolismo , Factor de Transcripción ReIA/metabolismo , Transcripción Genética/efectos de los fármacos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA