Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 33
Filtrar
Más filtros

Tipo del documento
País de afiliación
Intervalo de año de publicación
1.
Mol Pharmacol ; 75(3): 667-76, 2009 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-19103761

RESUMEN

Calcium-dependent cell death occurs in neurodegenerative diseases and ischemic or traumatic brain injury. We analyzed whether thioureylenes can act in a neuroprotective manner by pharmacological suppression of calcium-dependent pathological pathways. In human neuroblastoma (SK-N-SH) cells, thioureylenes (thiopental, carbimazole) inhibited the calcium-dependent neuronal protein phosphatase (PP)-2B, the activation of the proapoptotic transcription factor nuclear factor of activated T-cells, BAD-induced initiation of caspase-3, and poly-(ADP-ribose)-polymerase cleavage. Caspase-3-independent cell death was attenuated by carbimazole and the protein kinase C (PKC) delta inhibitor rottlerin by a PP-2B-independent mechanism. Neuroprotective effects were mediated by the redox-active sulfur of thioureylenes. Furthermore, we observed that the route of calcium mobilization was differentially linked to caspase-dependent or independent cell death and that BAD dephosphorylation did not necessarily induce intrinsic caspase activation. In addition, a new 30- to 35-kDa caspase-3 fragment with an unknown function was identified. In organotypic hippocampal slice cultures, thioureylenes inhibited caspase-3 activation or reduced N-methyl-d-aspartate and kainic acid receptor-mediated cell death that was independent of caspase-3. Because prolonged inhibition of caspase-3 resulted in caspase-independent cellular damage, different types of cell death must be taken under therapeutic consideration. Here we show that thioureylenes in combination with PKCdelta inhibitors might represent a promising therapeutic approach to attenuate neuronal damage.


Asunto(s)
Calcio/metabolismo , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Tiouracilo/farmacología , Animales , Apoptosis/efectos de los fármacos , Apoptosis/fisiología , Calcio/antagonistas & inhibidores , Muerte Celular/efectos de los fármacos , Muerte Celular/fisiología , Línea Celular Tumoral , Células Cultivadas , Compuestos Heterocíclicos/química , Compuestos Heterocíclicos/farmacología , Humanos , Ratones , Neuronas/patología , Tiouracilo/análogos & derivados
2.
Endocrinology ; 149(3): 1314-22, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18063677

RESUMEN

Recent evidence suggests the existence of a stem cell-like subpopulation of cells in hematological and solid tumor entities, which determine the malignant phenotype of a given tumor through their proliferative potential and chemotherapy resistance. A recently used technique for the isolation of this cell population is through exclusion of the vital dye Hoechst 33342, which defines the so-called side population (SP). Herein we demonstrate the presence of SP cells in a variety of adrenal specimens, including primary cultures of human adrenocortical tumors and normal adrenal glands as well as established human and murine adrenocortical cancer cell lines by fluorescence-activated cell sorter analysis and confocal microscopy. On a functional level, SP cells from the human adrenocortical tumor cell line NCI h295R revealed an expression pattern consistent with a less differentiated phenotype, including lower expression of steroidogenic enzymes such as steroid acute regulatory protein (StAR) and side-chain cleavage enzyme (P450scc) in comparison with non-SP cells. However, proliferation between SP and non-SP cells did not differ (105.6 +/- 18.1 vs. 100.0 +/- 3.5%). Furthermore, re-sorting and tracing experiments revealed the capacity for both cell types to give rise to the original SP- and non-SP-containing cell population. Similarly to the baseline growth kinetics, no survival benefit was evident in SP cells after treatment with cytotoxic agents commonly used in adrenocortical carcinomas. Taken together, these findings provide evidence that Hoechst dye exclusion, in contrast to what has been reported for other tumor entities, is not a major tumor stem cell defining marker in adrenocortical NCI h295R tumor cells.


Asunto(s)
Neoplasias de la Corteza Suprarrenal/patología , Carcinoma Corticosuprarrenal/patología , Células Madre Neoplásicas/citología , Neoplasias de la Corteza Suprarrenal/enzimología , Glándulas Suprarrenales/citología , Carcinoma Corticosuprarrenal/enzimología , Antineoplásicos/farmacología , Antineoplásicos/uso terapéutico , Ciclo Celular/fisiología , Diferenciación Celular/fisiología , Línea Celular Tumoral , Proliferación Celular , Enzima de Desdoblamiento de la Cadena Lateral del Colesterol/metabolismo , Colorantes , Resistencia a Antineoplásicos/fisiología , Humanos , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/enzimología , Fenotipo , Fosfoproteínas/metabolismo , Células Tumorales Cultivadas
3.
J Pharmacol Exp Ther ; 327(3): 863-71, 2008 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-18784349

RESUMEN

Activation of pancreatic stellate cells (PSCs) is the key process in the development of pancreatic fibrosis, a common feature of chronic pancreatitis and pancreatic cancer. In recent studies, curcumin has been shown to inhibit PSC proliferation via an extracellular signal-regulated kinase (ERK)1/2-dependent mechanism. In addition, curcumin is a potent inducer of the cytoprotective enzyme heme oxygenase-1 (HO-1) in other cell types. Therefore, the aims of this study were to 1) characterize the effect of curcumin on HO-1 gene expression in PSCs, 2) explore whether HO-1 induction contributes to the inhibitory effect of curcumin on PSC proliferation, and 3) clarify the involvement of the mitogen-activated protein kinase (MAPK) family in this context. Cultured rat PSCs were incubated with curcumin and assessed for HO-1 up-regulation by Northern blot analysis, immunoblotting, and activity assays. The effect of HO-1 on platelet-derived growth factor (PDGF)-induced PSC proliferation and MAPK activation was determined by immunoblotting, cell proliferation assays, and cell count analyses. Curcumin induced HO-1 gene expression in PSCs in a time- and dose-dependent manner and inhibited PDGF-mediated ERK1/2 phosphorylation and PSC proliferation. These effects were blocked by treatment of PSCs with tin protoporphyrin IX, an HO inhibitor, or transfection of HO-1 small interfering RNA. Our data provide evidence that HO-1 induction contributes to the inhibitory effect of curcumin on PSC proliferation. Therefore, therapeutic up-regulation of HO-1 could represent a mode for inhibition of PSC proliferation and thus may provide a novel strategy in the prevention of pancreatic fibrosis.


Asunto(s)
Proliferación Celular/efectos de los fármacos , Proteína Quinasa 3 Activada por Mitógenos/metabolismo , Páncreas/citología , Transducción de Señal/efectos de los fármacos , Animales , Células Cultivadas , Curcumina/farmacología , Hemo-Oxigenasa 1/genética , Hemo-Oxigenasa 1/fisiología , Factor de Crecimiento Derivado de Plaquetas/farmacología , Ratas , Activación Transcripcional/efectos de los fármacos
4.
J Pharmacol Exp Ther ; 324(3): 1037-44, 2008 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-18055877

RESUMEN

Thionamides, inhibitors of the thyroid peroxidase-mediated iodination, are clinically used in the treatment of hyperthyroidism. However, the use of antithyroid drugs is associated with immunomodulatory effects, and recent studies with thionamide-related heterocyclic thioderivates demonstrated direct anti-inflammatory and immunosuppressive properties. Using primary human T-lymphocytes, we show that the heterocyclic thionamides carbimazole and propylthiouracil inhibit synthesis of the proinflammatory cytokines tumor necrosis factor (TNF)alpha and interferon (IFN)gamma. In addition, DNA binding of nuclear factor (NF)-kappaB, a proinflammatory transcription factor that regulates both TNFalpha and IFNgamma synthesis, and NF-kappaB-dependent reporter gene expression were reduced. Abrogation of NF-kappaB activity was accompanied by reduced phosphorylation and proteolytic degradation of inhibitor of kappaB (IkappaB)alpha, the inhibitory subunit of the NF-kappaB complex. Carbimazole inhibited NF-kappaB via the small GTPase Rac-1, whereas propylthiouracil inhibited the phosphorylation of IkappaBalpha by its kinase inhibitor of kappaB kinase alpha. Methimazole had no effect on NF-kappaB induction, demonstrating that drug potency correlated with the chemical reactivity of the thionamide-associated sulfur group. Taken together, our data demonstrate that thioureylenes with a common, heterocyclic structure inhibit inflammation and immune function via the NF-kappaB pathway. Our results may explain the observed remission of proinflammatory diseases upon antithyroid therapy in hyperthyroid patients. The use of related thioureylenes may provide a new therapeutic basis for the development and application of anti-inflammatory compounds.


Asunto(s)
Antitiroideos/farmacología , Quinasa I-kappa B/antagonistas & inhibidores , FN-kappa B/antagonistas & inhibidores , Proteína de Unión al GTP rac1/antagonistas & inhibidores , Antitiroideos/química , Humanos , Quinasa I-kappa B/metabolismo , Células Jurkat , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/metabolismo , FN-kappa B/metabolismo , Compuestos de Sulfhidrilo/química , Compuestos de Sulfhidrilo/farmacología , Proteína de Unión al GTP rac1/metabolismo
5.
J Pharmacol Exp Ther ; 325(1): 217-25, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18218830

RESUMEN

Barbiturates, which are used for the treatment of intracranial hypertension after severe head injury, have been associated with anti-inflammatory side effects. Although all barbiturates inhibit T-cell function, only thiobarbiturates markedly reduce the activation of the transcription factor nuclear factor-kappaB (NF-kappaB). Various pharmacologic inhibitors of the NF-kappaB pathway are concomitant nonthermal inducers of the heat shock response (HSR), a cellular defense system that is associated with protection of cells and organs. We hypothesize that thiopental mediates cytoprotection by inducing the HSR. Human CD3(+) T lymphocytes were incubated with thiopental, pentobarbital, etomidate, ketamine, midazolam, or propofol. Human Jurkat T cells were transfected with small interfering RNA (siRNA) targeting heat 70-kDa shock protein (hsp 70) before thiopental incubation. Apoptosis was induced by staurosporine. DNA binding activity of HSF-1 was analyzed by electrophoretic mobility shift assay; mRNA expression of hsp27, -32, -70, and -90 was analyzed by Northern blot, and protein expression of hsp70 was analyzed by Western blot and flow cytometry after fluorescein isothiocyanate (FITC)-hsp70-antibody staining. Apoptosis was assessed by flow cytometry after annexin V-FITC or annexin V-phycoerythrin staining. Activity of caspase-3 was measured by fluorogenic caspase activity assay. Thiopental induced hsp27, -70, and -90 but not hsp32 mRNA expression as well as hsp70 protein expression. Thiopental dose-dependently activated the DNA binding activity of HSF-1, whereas other substances investigated had no effect. In addition, pretreatment with thiopental significantly attenuated staurosporine-induced apoptosis and caspase-like activity. Transfection with hsp70-siRNA before thiopental treatment reduced this attenuation. Thiopental specifically and differentially induces a heat shock response, and it mediates cytoprotection via the expression of hsp70 in human T lymphocytes.


Asunto(s)
Apoptosis/efectos de los fármacos , Proteínas HSP70 de Choque Térmico/genética , Linfocitos T/efectos de los fármacos , Tiopental/farmacología , Complejo CD3 , Regulación de la Expresión Génica/efectos de los fármacos , Proteínas HSP70 de Choque Térmico/análisis , Respuesta al Choque Térmico/efectos de los fármacos , Humanos , Sustancias Protectoras/farmacología , ARN Mensajero/análisis , Linfocitos T/citología
6.
Anesthesiology ; 108(6): 1025-36, 2008 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-18497603

RESUMEN

BACKGROUND: Cardiopulmonary bypass (CPB) is associated with pulmonary inflammation and dysfunction. This may lead to acute lung injury and acute respiratory distress syndrome with increased morbidity and mortality. The authors hypothesized that inhaled carbon monoxide before initiation of CPB would reduce inflammatory response in the lungs. METHODS: In a porcine model, a beating-heart CPB was used. The animals were either randomized to a control group, to standard CPB, or to CPB plus carbon monoxide. In the latter group, lungs were ventilated with 250 ppm inhaled carbon monoxide in addition to standard ventilation before CPB. Lung tissue samples were obtained at various time points, and pulmonary cytokine levels were determined. RESULTS: Hemodynamic parameters were largely unaffected by CPB or carbon monoxide inhalation. There were no significant differences in cytokine expression in mononuclear cells between the groups throughout the experimental time course. Compared with standard CPB animals, carbon monoxide significantly suppresses tumor necrosis factor-alpha and interleukin-1beta levels (P < 0.05) and induced the antiinflammatory cytokine interleukin 10 (P < 0.001). Carbon monoxide inhalation modulates effector caspase activity in lung tissue during CPB. CONCLUSIONS: The results demonstrate that inhaled carbon monoxide significantly reduces CPB-induced inflammation via suppression of tumor necrosis factor alpha, and interleukin-1beta expression and elevation of interleukin 10. Apoptosis induced by CPB was associated with caspase-3 activation and was significantly attenuated by carbon monoxide treatment. Based on the observations of this study, inhaled carbon monoxide could represent a potential new therapeutic modality for counteracting CPB-induced lung injury.


Asunto(s)
Antimetabolitos/farmacología , Monóxido de Carbono/farmacología , Puente Cardiopulmonar , Citocinas/metabolismo , Inflamación/prevención & control , Pulmón/efectos de los fármacos , Animales , Antimetabolitos/administración & dosificación , Apoptosis/efectos de los fármacos , Análisis de los Gases de la Sangre , Northern Blotting , Monóxido de Carbono/administración & dosificación , Carboxihemoglobina/análisis , Carboxihemoglobina/metabolismo , Puente Cardiopulmonar/efectos adversos , Caspasa 3/metabolismo , Ensayo de Inmunoadsorción Enzimática , Inflamación/etiología , Interleucina-10/metabolismo , Interleucina-1beta/metabolismo , Pulmón/citología , Pulmón/metabolismo , Modelos Animales , Oxígeno/sangre , Porcinos , Factor de Necrosis Tumoral alfa/metabolismo
7.
Anesth Analg ; 106(4): 1150-60, table of contents, 2008 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-18349187

RESUMEN

BACKGROUND: Modulation of the inflammatory stress response by anesthesia may be responsible for an increased susceptibility to infectious complications, such as wound infection or pneumonia. Sevoflurane, a specific inhibitor of activator protein-1, an immediate early transcription factor, induces apoptosis in T-cells. Because p38 can be involved either in pro- or antiapoptotic processes, we examined whether the sevoflurane-induced apoptosis is mediated by p38 activation in Jurkat T-cells. METHODS: Jurkat T-cells were exposed to different concentrations of sevoflurane, isoflurane, or desflurane in vitro. Phosphorylation of mitogen-activated protein (MAP) kinases, upstream kinases, downstream activating transcription factor 2 ATF-2, and caspase-3 processing were evaluated by Western blot. p38 kinase activity was evaluated after immunoprecipitation and phosphorylation of the substrate ATF-2 using Western blot. Apoptosis was assessed using flow cytometry after staining with green fluorescent protein-annexin V. RESULTS: While desflurane had no effect, sevoflurane and isoflurane induced p38 phosphorylation with sevoflurane inducing p38 kinase activity. Sevoflurane did not affect the MAP kinases ERK and JNK. Sevoflurane exposure also induced phosphorylation of apoptosis signal-regulating kinase-1 (ASK1), MAP kinase kinases 3 and 6 (MKK3/MKK6), and ATF-2. Pretreatment of cells with the general caspase inhibitor Z-VAD.fmk did not prevent the sevoflurane-induced phosphorylation of p38. Isoflurane- and sevoflurane-mediated caspase-3 processing and apoptosis could not be abolished by pretreatment with the specific p38 inhibitors SB202190 and SB203580. CONCLUSIONS: Sevoflurane is a specific activator of the apoptosis signal-regulating kinase-1-, MKK3/MKK6-p38 MAP kinase cascade in Jurkat T-cells. Our data suggest that sevoflurane-induced p38 activation is not affected by caspase activation. Furthermore, sevoflurane-induced apoptosis is not dependent on p38 MAP kinase activation.


Asunto(s)
Anestésicos por Inhalación/farmacología , Activación Enzimática/efectos de los fármacos , Éteres Metílicos/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Apoptosis/efectos de los fármacos , Caspasas/efectos de los fármacos , Caspasas/metabolismo , Desflurano , Humanos , Inflamación/fisiopatología , Isoflurano/análogos & derivados , Isoflurano/farmacología , Células Jurkat , Proteínas de Neoplasias/metabolismo , Sevoflurano
8.
Mol Pharmacol ; 72(6): 1647-56, 2007 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-17878268

RESUMEN

Treatment of hyperthyroidism by thionamides is associated with immunomodulatory effects, but the mechanism of thionamide-induced immunosuppression is unclear. Here we show that thionamides directly inhibit interleukin-2 cytokine expression, proliferation, and the activation (CD69 expression) of primary human T lymphocytes. Inhibition of immune function was associated with a repression of DNA binding of the cooperatively acting immunoregulatory transcription factors activator protein 1 (AP-1) and nuclear factor of activated T-cells (NFAT). Likewise, thionamides block the GTPase p21Ras, the mitogen-activated protein kinases, and impair the calcineurin/calmodulin-dependent NFAT dephosphorylation and nuclear translocation. The potency of inhibition correlated with the chemical reactivity of the thionamide-associated sulfur group. Taken together, our data demonstrate that thio-derivates with a common heterocyclic thioureylene-structure mediate a direct suppression of immune functions in T-cells via inhibition of the AP-1/NFAT pathway. Our observations may also explain the clinical and pathological resolution of some secondary, calcineurin, and mitogen-activated protein kinase-associated diseases upon thionamide treatment in hyperthyroid patients. This offers a new therapeutic basis for the development and application of heterocyclic thio-derivates.


Asunto(s)
Inmunosupresores/farmacología , Factores de Transcripción NFATC/antagonistas & inhibidores , Factores de Transcripción NFATC/fisiología , Transducción de Señal/fisiología , Linfocitos T/fisiología , Tiobarbitúricos/farmacología , Factor de Transcripción AP-1/antagonistas & inhibidores , Factor de Transcripción AP-1/fisiología , Proliferación Celular/efectos de los fármacos , Humanos , Inmunosupresores/química , Células Jurkat , Transducción de Señal/efectos de los fármacos , Relación Estructura-Actividad , Linfocitos T/efectos de los fármacos , Tiobarbitúricos/química
9.
Int J Biochem Cell Biol ; 39(12): 2278-88, 2007.
Artículo en Inglés | MEDLINE | ID: mdl-17689131

RESUMEN

The involvement of p38 in fundamental physiological processes and the fact that deregulation often leads to disease indicates the potential impact of p38 dependent mechanisms. Here we demonstrate a new pathway that includes the induction of the mitogen activated protein kinase p38 by protein kinase C and results in a specific phosphorylation of c-Jun in T-lymphocytes. P38 directly phosphorylates c-Jun within its transactivation domain at serine 63 and serine 73 and thus posttranscriptionally affects the presence of DNA-bound phosphorylated c-Jun, a prerequisite for activator protein 1 dependent gene transcription. Moreover, DNA-binding activity of c-Fos, FosB, and JunB were also dependent on the p38 protein kinase activity, whereas JunD, Fra-1 and Fra-2 were not affected. Although we show that stress induced mitogen activated protein kinases share c-Jun as a substrate for phosphorylation, p38 mediated effects could not be rescued by the c-Jun N-terminal kinases. This demonstrates that the protein kinase p38 plays a unique and non-redundant role in posttranslational c-Jun regulation. The induction of a p38 dependent c-Jun phosphorylation was comparable in both CD4(+) and CD8(+) T-cells, proposing a ubiquitous pathway that is not linked to T-cell subtype and effector function. In contrast, ATF-2 was predominantly phosphorylated in CD8(+) T-cells. Different cell lines show p38-dependent c-Jun phosphorylation upon phorbol ester induction but there is evidence that the simian virus 40 large T-antigen may interfere with this pathway.


Asunto(s)
Proteínas Proto-Oncogénicas c-jun/metabolismo , Factor de Transcripción AP-1/metabolismo , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Animales , Anticuerpos/farmacología , Antígenos CD28/inmunología , Complejo CD3/inmunología , Línea Celular , Línea Celular Tumoral , ADN/metabolismo , Dactinomicina/farmacología , Inhibidores Enzimáticos/farmacología , Humanos , Isoenzimas/antagonistas & inhibidores , Isoenzimas/metabolismo , Proteínas Quinasas JNK Activadas por Mitógenos/antagonistas & inhibidores , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Ratones , Células 3T3 NIH , Fosforilación/efectos de los fármacos , Unión Proteica/efectos de los fármacos , Proteína Quinasa C/metabolismo , Proteínas Proto-Oncogénicas c-fos/metabolismo , Serina/metabolismo , Subgrupos de Linfocitos T/metabolismo , Linfocitos T/efectos de los fármacos , Linfocitos T/metabolismo , Acetato de Tetradecanoilforbol/análogos & derivados , Acetato de Tetradecanoilforbol/farmacología , Proteínas Quinasas p38 Activadas por Mitógenos/antagonistas & inhibidores
10.
Cell Oncol (Dordr) ; 40(6): 639-650, 2017 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-28808957

RESUMEN

BACKGROUND: Solid tumors contain various components that together form the tumor microenvironment. Cancer associated fibroblasts (CAFs) are capable of secreting and responding to signaling molecules and growth factors. Due to their role in tumor development, CAFs are considered as potential therapeutic targets. A prominent tumor-associated signaling molecule is transforming growth factor ß (TGFß), an inducer of epithelial-to-mesenchymal transition (EMT). The differential action of TGFß on CAFs and ETCs (epithelial tumor cells) has recently gained interest. Here, we aimed to investigate the effects of TGFß on CAFs and ETCs at the proteomic level. METHODS: We established a 2D co-culture system of differentially fluorescently labeled CAFs and ETCs and stimulated this co-culture system with TGFß. The respective cell types were separated using FACS and subjected to quantitative analyses of individual proteomes using mass spectrometry. RESULTS: We found that TGFß treatment had a strong impact on the proteome composition of CAFs, whereas ETCs responded only marginally to TGFß. Quantitative proteomic analyses of the different cell types revealed up-regulation of extracellular matrix (ECM) proteins in TGFß treated CAFs. In addition, we found that the TGFß treated CAFs exhibited increased N-cadherin levels. CONCLUSIONS: From our data we conclude that CAFs respond to TGFß treatment by changing their proteome composition, while ETCs appear to be rather resilient.


Asunto(s)
Fibroblastos Asociados al Cáncer/citología , Fibroblastos Asociados al Cáncer/efectos de los fármacos , Técnicas de Cocultivo/métodos , Factor de Crecimiento Transformador beta/farmacología , Línea Celular Tumoral , Proliferación Celular/efectos de los fármacos , Transición Epitelial-Mesenquimal/efectos de los fármacos , Células HCT116 , Humanos , Proteómica , Microambiente Tumoral
11.
Expert Rev Mol Diagn ; 4(5): 619-29, 2004 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-15347256

RESUMEN

Breath tests are attractive since they are noninvasive and can be repeated frequently in the dynamically changing state of critically ill patients. Volatile organic compounds can be produced anywhere in the body and are transported via the bloodstream and exhaled through the lung. They can reflect physiologic or pathologic biochemical processes such as lipid peroxidation, liver disease, renal failure, allograft rejection, and dextrose or cholesterol metabolism. This review describes the diagnostic potential of endogenous organic volatile substances in the breath of critically ill patients. Since many of these patients require ventilatory support, aspects of breath analysis under mechanical ventilation will be addressed. Analytical procedures, problems concerning the physiologic meaning of breath markers and future developments will be discussed.


Asunto(s)
Biomarcadores/análisis , Pruebas Respiratorias , Enfermedad Crítica , Colesterol/metabolismo , Diseño de Equipo , Rechazo de Injerto , Humanos , Hidrocarburos/análisis , Hidrocarburos/metabolismo , Peroxidación de Lípido , Hepatopatías/metabolismo , Modelos Teóricos , Insuficiencia Renal/metabolismo , Respiración Artificial/instrumentación , Volatilización
12.
Pancreas ; 36(4): 377-84, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18437084

RESUMEN

OBJECTIVES: Microcirculatory derangements caused by ischemia and reperfusion (I/R) play a pivotal role in acute and graft pancreatitis. The inducible enzyme heme oxygenase 1 (HO-1) has been shown to decrease I/R injury by modulation of capillary perfusion in other organs. It was the aim of this study to evaluate the effect of HO-1 induction on pancreatic microcirculation after I/R. METHODS: Rats were randomized into 4 groups: (1) sham controls; (2) 1-hour ischemia and 2-hour reperfusion (I/R); (3) I/R + cobalt protoporphyrin (CoPP), an HO-1 inducer; and (4) I/R + CoPP + tin protoporphyrin, an HO inhibitor. Functional capillary density (FCD) and leukocyte endothelium interaction were analyzed using intravital microscopy during reperfusion. Expression of HO-1 mRNA, HO-1 protein, and HO activity were assessed by Northern blot, Western blot, and an HO activity assay. RESULTS: Functional capillary density decreased significantly in the I/R group as compared with sham controls. Cobalt protoporphyrin treatment increased FCD to control values. In contrast, HO inhibition in CoPP-pretreated animals lowered FCD and increased leukocyte endothelium interaction significantly. Cobalt protoporphyrin administration increased HO-1 mRNA, protein, and HO activity, whereas activity of the enzyme was reduced after injection of tin protoporphyrin. CONCLUSIONS: Heme oxygenase 1 plays a beneficial role in pancreatic microcirculatory derangements after I/R. This could be of therapeutic relevance after pancreas transplantation and other forms of postischemic pancreatitis.


Asunto(s)
Hemo-Oxigenasa 1/metabolismo , Microcirculación/fisiología , Páncreas/irrigación sanguínea , Pancreatitis/enzimología , Daño por Reperfusión/enzimología , Animales , Capilares/enzimología , Endotelio Vascular/fisiología , Endotelio Vascular/fisiopatología , Leucocitos/fisiología , Masculino , Microcirculación/efectos de los fármacos , Pancreatitis/inducido químicamente , Protoporfirinas/toxicidad , ARN/genética , ARN/aislamiento & purificación , Ratas , Ratas Sprague-Dawley
13.
Nitric Oxide ; 18(1): 61-9, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17999922

RESUMEN

Nitric oxide plays a crucial role in the maintenance of liver function and integrity. During stress, the inducible heme oxygenase-1 protein and its reaction products, including carbon monoxide, also exert potent hepatoprotective effects. We investigated a potential relationship between endogenous nitric oxide synthesis and the hepatic regulation of heme oxygenase-1. Inhibition of nitric oxide synthesis in vivo by injection of l-NAME led to a dose-dependent induction of heme oxygenase-1 mRNA, protein and activity in the rat liver, whereas did not affect the expression of other heat shock proteins. The effect of l-NAME was demonstrated by hemodynamic changes within the liver circulation as measured by ultrasonic flow probes. Inhibition of nitric oxide synthase led to a decline in hepatic arterial and portal venous blood flow, and subsequently caused liver cell damage. In contrast, the combined administration of l-NAME and the nitric oxide-independent intestinal vasodilator dihydralazine completely restored portal venous flow, abolished the liver cell damage, and prevented the upregulation of heme oxygenase-1, despite inhibition of nitric oxide production. In conclusion, nitric oxide deficiency upregulates hepatic heme oxygenase-1, which is reversible by maintaining hepatic blood flow. This interdependence has important implications for the development of therapeutic strategies aimed at modulating the activity of these hepatoprotective mediator systems.


Asunto(s)
Eritrocitos/metabolismo , Hemo-Oxigenasa 1/metabolismo , Hígado/enzimología , Óxido Nítrico/metabolismo , Nitritos/metabolismo , Animales , Células Cultivadas , Relación Dosis-Respuesta a Droga , Activación Enzimática/efectos de los fármacos , Proteínas de Choque Térmico/efectos de los fármacos , Proteínas de Choque Térmico/metabolismo , Hemo-Oxigenasa 1/biosíntesis , Hemo-Oxigenasa 1/efectos de los fármacos , Hígado/metabolismo , Masculino , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico/sangre , Óxido Nítrico/deficiencia , Nitritos/sangre , Nitroarginina/farmacología , Oxidación-Reducción , ARN Mensajero/efectos de los fármacos , ARN Mensajero/metabolismo , Ratas , Ratas Sprague-Dawley , Regulación hacia Arriba/efectos de los fármacos
14.
Ann Surg ; 245(6): 931-42, 2007 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-17522519

RESUMEN

OBJECTIVE: It was the aim of this study to characterize the influence of isoflurane-induced heme oxygenase-1 (HO-1) expression on hepatocellular integrity after ischemia and reperfusion. SUMMARY BACKGROUND DATA: Abundant experimental data characterize HO-1 as one of the most powerful inducible enzymes that contribute to the protection of the liver and other organs after harmful stimuli. Therapeutic strategies aimed at utilizing the protective effects of HO-1 are hampered by the fact that most pharmacological inducers of this enzyme perturb organ function by themselves and are not available for use in patients because of their toxicity and undesirable or unknown side effects. METHODS: Rats were pretreated with isoflurane before induction of partial hepatic ischemia (1 hour) and reperfusion (1 hour). At the end of each experiment, blood and liver tissue were obtained for molecular biologic, histologic, and immunohistochemical analyses. RESULTS: Isoflurane pretreatment increased hepatic HO-1 mRNA, HO-1 protein, HO enzyme activity, and decreased plasma levels of AST, ALT, and alpha-GST. Histologic analysis of livers obtained from isoflurane-pretreated rats showed a reduction of necrotic areas, particularly in the perivenular region, the predominant site of isoflurane-induced HO-1 expression. In addition, sinusoidal congestion that could otherwise be observed after ischemia/reperfusion was inhibited by the anesthetic. Furthermore, isoflurane augmented hepatic microvascular blood flow and lowered the malondialdehyde content within the liver compared with control animals. Administration of tin protoporphyrin IX inhibited HO activity and abolished the isoflurane-induced protective effects. CONCLUSIONS: This study provides first evidence that pretreatment with the nontoxic and clinically approved anesthetic isoflurane induces hepatic HO-1 expression, and thereby protects rat livers from ischemia/reperfusion injury.


Asunto(s)
Anestésicos/farmacología , Hemo-Oxigenasa 1/metabolismo , Isoflurano/farmacología , Hígado/irrigación sanguínea , Hígado/enzimología , Daño por Reperfusión/enzimología , Daño por Reperfusión/prevención & control , Análisis de Varianza , Animales , Velocidad del Flujo Sanguíneo , Northern Blotting , Western Blotting , Técnicas para Inmunoenzimas , Masculino , Malondialdehído/metabolismo , Microcirculación , Ratas , Ratas Sprague-Dawley
15.
Crit Care Med ; 34(3): 815-22, 2006 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-16521277

RESUMEN

OBJECTIVE: Impaired hepatic perfusion after hemorrhagic shock frequently results in hepatocellular dysfunction associated with increased mortality. This study characterizes the effect of the vasodilators dihydralazine and urapidil on hepatocellular perfusion and integrity after hemorrhagic shock and resuscitation. DESIGN: Prospective, randomized, controlled experimental study. SETTING: University experimental laboratory. SUBJECTS: Male Sprague-Dawley rats. INTERVENTIONS: To register systemic and regional hepatic hemodynamics, rats (n=6 per group) were instrumented and randomly assigned to the following groups: shock+vehicle; shock+dihydralazine (1.5 mg/kg); or shock+urapidil (3 mg/kg). After 1 hr of hemorrhagic shock, animals were resuscitated for 5 hrs and mean arterial pressure was maintained at 70+/-5 mm Hg by administration of dihydralazine or urapidil. To evaluate hepatic heme oxygenase-1 expression and liver injury (determination of levels of alanine and aspartate aminotransferase [ALT, AST] and histology), an additional series of experiments with six animals per group was performed. At the end of each experiment, animals were killed and blood and liver tissue was obtained for subsequent analyses. MEASUREMENTS AND MAIN RESULTS: Dihydralazine increased cardiac output and portal and hepatic microvascular flow (p<.05) and reduced liver injury after shock (lower ALT and AST levels [p<.05]; improvement of histopathological changes). In contrast, urapidil had no effect on portal flow or liver injury. Hepatic heme oxygenase-1 mRNA expression was upregulated in animals subjected to hemorrhagic shock but did not differ among experimental groups. CONCLUSIONS: Dihydralazine increases nutritive portal and hepatic microvascular flow and limits liver injury after hemorrhagic shock. This protective effect appears to be the result of increased cardiac output and increased portal flow. These findings may offer a new strategy for hepatic protection after hemorrhagic shock.


Asunto(s)
Antihipertensivos/uso terapéutico , Dihidralazina/uso terapéutico , Hemo-Oxigenasa 1/efectos de los fármacos , Fallo Hepático Agudo/prevención & control , Choque Hemorrágico/tratamiento farmacológico , Análisis de Varianza , Animales , Antihipertensivos/farmacología , Dihidralazina/farmacología , Hemo-Oxigenasa 1/metabolismo , Hemodinámica/efectos de los fármacos , Circulación Hepática/efectos de los fármacos , Masculino , Microcirculación , Piperazinas/farmacología , Distribución Aleatoria , Ratas , Ratas Sprague-Dawley
16.
Anesthesiology ; 104(1): 101-9, 2006 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-16394696

RESUMEN

BACKGROUND: The heme oxygenase pathway represents a major cell and organ protective system in the liver. The authors recently showed that isoflurane and sevoflurane up-regulate the inducible isoform heme oxygenase 1 (HO-1). Because the activating cascade remained unclear, it was the aim of this study to identify the underlying mechanism of this effect. METHODS: Rats were anesthetized with pentobarbital intravenously or with isoflurane per inhalation (2.3 vol%). Kupffer cell function was inhibited by dexamethasone or gadolinium chloride. Nitric oxide synthases were inhibited by either N(omega)-nitro-L-arginine methyl ester or S-methyl thiourea. N-acetyl-cysteine served as an antioxidant, and diethyldithiocarbamate served as an inhibitor of cytochrome P450 2E1. Protein kinase C and phospholipase A2 were inhibited by chelerythrine or quinacrine, respectively. HO-1 was analyzed in liver tissue by Northern blot, Western blot, immunostaining, and enzymatic activity assay. RESULTS: In contrast to pentobarbital, isoflurane induced HO-1 after 4-6 h in hepatocytes in the pericentral region of the liver. The induction was prevented in the presence of dexamethasone (P < 0.05) and gadolinium chloride (P < 0.05). Inhibition of nitric oxide synthases or reactive oxygen intermediates did not affect isoflurane-mediated HO-1 up-regulation. In contrast, chelerythrine (P < 0.05) and quinacrine (P < 0.05) resulted in a blockade of HO-1 induction. CONCLUSION: The up-regulation of HO-1 by isoflurane in the liver is restricted to parenchymal cells and depends on Kupffer cell function. The induction is independent of nitric oxide or reactive oxygen species but does involve protein kinase C and phospholipase A2.


Asunto(s)
Anestésicos por Inhalación/farmacología , Hemo-Oxigenasa 1/biosíntesis , Isoflurano/farmacología , Hígado/enzimología , Acetilcisteína/farmacología , Animales , Antiinflamatorios/farmacología , Northern Blotting , Western Blotting , Dexametasona/farmacología , Ditiocarba/farmacología , Inducción Enzimática/efectos de los fármacos , Inhibidores Enzimáticos/farmacología , Gadolinio/farmacología , Inmunohistoquímica , Isotiuronio/análogos & derivados , Isotiuronio/farmacología , Hígado/efectos de los fármacos , Masculino , NG-Nitroarginina Metil Éster/farmacología , Óxido Nítrico Sintasa/antagonistas & inhibidores , ARN/análisis , ARN/biosíntesis , Ratas , Ratas Sprague-Dawley , Transducción de Señal/efectos de los fármacos
17.
Anesthesiology ; 102(6): 1147-57, 2005 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-15915027

RESUMEN

BACKGROUND: Volatile anesthetics modulate lymphocyte function during surgery, and this compromises postoperative immune competence. The current work was undertaken to examine whether volatile anesthetics induce apoptosis in human T lymphocytes and what apoptotic signaling pathway might be used. METHODS: Effects of sevoflurane, isoflurane, and desflurane were studied in primary human CD3 T lymphocytes and Jurkat T cells in vitro. Apoptosis and mitochondrial membrane potential were assessed using flow cytometry after green fluorescent protein-annexin V and DiOC6-fluorochrome staining. Activity and proteolytic processing of caspase 3 was measured by cleaving of the fluorogenic effector caspase substrate Ac-DEVD-AMC and by anti-caspase-3 Western blotting. Release of mitochondrial cytochrome c was studied after cell fractionation using anti-cytochrome c Western blotting and enzyme-linked immunosorbent assays. RESULTS: Sevoflurane and isoflurane induced apoptosis in human T lymphocytes in a dose-dependent manner. By contrast, desflurane did not exert any proapoptotic effects. The apoptotic signaling pathway used by sevoflurane involved disruption of the mitochondrial membrane potential and release of cytochrome c from mitochondria to the cytosol. In addition, the authors observed a proteolytic cleavage of the inactive p32 procaspase 3 to the active p17 fragment, increased caspase-3-like activity, and cleavage of the caspase-3 substrate poly-ADP-ribose-polymerase. Sevoflurane-induced apoptosis was blocked by the general caspase inhibitor Z-VAD.fmk. Death signaling was not mediated via the Fas/CD95 receptor pathway because neither anti-Fas/CD95 receptor antagonism nor FADD deficiency or caspase-8 deficiency were able to attenuate sevoflurane-mediated apoptosis. CONCLUSION: Sevoflurane and isoflurane induce apoptosis in T lymphocytes via increased mitochondrial membrane permeability and caspase-3 activation, but independently of death receptor signaling.


Asunto(s)
Anestésicos por Inhalación/farmacología , Apoptosis/efectos de los fármacos , Caspasas/metabolismo , Mitocondrias/efectos de los fármacos , Linfocitos T/efectos de los fármacos , Apoptosis/fisiología , Caspasa 3 , Células Cultivadas , Humanos , Células Jurkat , Mitocondrias/enzimología , Linfocitos T/enzimología , Volatilización
18.
Clin Chem Lab Med ; 40(6): 587-94, 2002 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-12211653

RESUMEN

Free radical-mediated inflammatory processes account for a great portion of morbidity and mortality in critically ill patients. The purpose of this study was to determine two plasma peroxidation markers and three volatile markers related to lipid peroxidation, metabolic activity and cholesterol metabolism, and to explore relationships between the different markers and patients' clinical status. Substances were analyzed in whole blood and in exhaled air in patients with head injury, acute respiratory distress syndrome (ARDS) and in those being at risk of developing ARDS. These results were compared with the baseline measurements in healthy individuals. Additionally, patients were assessed according to their inflammatory status. Concentrations of malondialdehyde and thiobarbituric acid-reactive substances in plasma as well as pentane concentrations in breath increased with increasing inflammatory status. Although these compounds are generated through peroxidation of fatty acids, concentrations of these markers were significantly different in patient groups. Isoprene concentrations were lowest in the ARDS group. Acetone concentrations were not different between patient groups. We conclude that for the assessment of lipid peroxidation and other inflammatory reactions a set of parameters has to be defined. More detailed insights into inflammatory processes can be obtained when the volatile markers and the serum markers are considered together.


Asunto(s)
Enfermedad Crítica/clasificación , Hemiterpenos , Peroxidación de Lípido , Peróxidos Lipídicos/análisis , Sustancias Reactivas al Ácido Tiobarbitúrico/análisis , Acetona/análisis , Adulto , Biomarcadores/análisis , Pruebas Respiratorias , Butadienos/análisis , Traumatismos Craneocerebrales/sangre , Traumatismos Craneocerebrales/fisiopatología , Femenino , Humanos , Peróxidos Lipídicos/sangre , Masculino , Malondialdehído/análisis , Pentanos/análisis , Valores de Referencia , Respiración Artificial , Síndrome de Dificultad Respiratoria/sangre , Síndrome de Dificultad Respiratoria/fisiopatología
19.
Clin Microbiol Infect ; 5(6): 355-363, 1999 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-11856280

RESUMEN

OBJECTIVE: To determine the potential of laboratory services in identifying cross-transmission of multiresistant Gram-negative bacilli (MR GNB) and Staphylococcus aureus in adult intensive care units by routine typing of clinical isolates. METHODS: Over a 12-month period, isolates with indistinguishable PCR fingerprints were traced back to the source patients and their epidemiologic relationships were investigated. Possible episodes of cross-transmission were ascertained, and the validity of antibiograms in identifying the same cluster assessed. RESULTS: Of 3503 specimens received by the microbiological laboratory during 5372 patient days, 1295 cultures showed bacterial growth. Of these, 132 were primary isolates of MR GNB and 92 were primary isolates of S. aureus. Thirty-two MR GNB isolates (24%) shared fingerprints with one or more other isolates. Indistinguishable isolates from epidemiologically related patients suggested 17 episodes of cross-transmission. The positive and negative predictive values of antibiogram-based identification of these episodes were 19% and 72% respectively. S aureus displayed limited genetic diversity. The two most frequent genotypes contained 19 and 16 isolates, of which the majority appeared to be epidemiologically unrelated. CONCLUSIONS: Endemic transmission of MR GNB occurs mainly between two patients and remains unrecognized by conventional laboratory investigation. Rapid genetic typing methods identify patients involved in cross-transmission and give an insight into the population dynamics of MR GNB on adult intensive care units.

20.
J Hematother Stem Cell Res ; 12(1): 71-82, 2003 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-12662438

RESUMEN

The rapid efflux of the fluorescent DNA-binding dye Hoechst 33342 identifies a rare, so-called side population (SP), which rapidly expels the dye, can reconstitute the bone marrow (BM) of lethally irradiated mice, and has proven negative for most lineage markers including CD34. Because SP cells from human cell sources, such as mobilized peripheral blood [apheresis products (AP)], cord blood (CB), or BM have not been extensively characterized to date, we sought to analyze SP cells from various cell sources. We detected murine SP cells with a median frequency of 0.04% (n = 23) and a 52-fold colony-forming units (CFU) increase compared to unsorted cells (p = 0.028). The median frequency of human SP cells was 0.02% (n = 90), with highest numbers in donor AP, and lower in CB and BM. Human SP cells were mostly CD34(-) and lineage marker-negative. These showed no enrichment in CFU before expansion; however, they displayed a CFU increase after 5-7 days of cytokine-supported suspension culture (10.7-fold at day 5, 7.2-fold at day 7; n = 17) that was significant compared to both input (day 0) SP and to non-SP cells before and after expansion (p < 0.05). SP cells demonstrated a significant long-term culture-initiating cell (LTC-IC) increase of 167-fold (n = 17) as compared to non-SP cells (p = 0.002), with the highest numbers from AP specimens. We conclude that human primitive hematopoietic cells can be isolated via Hoechst staining and that SP cells of various human sources show substantial differences and represent a rare CD34(-) population with stem cell potential.


Asunto(s)
Antígenos CD34/metabolismo , Bencimidazoles/farmacología , Colorantes Fluorescentes/farmacología , Animales , Anticuerpos Monoclonales/metabolismo , Antígenos CD34/biosíntesis , Eliminación de Componentes Sanguíneos , Células de la Médula Ósea/metabolismo , División Celular , Linaje de la Célula , Separación Celular , Células Cultivadas , Citocinas/metabolismo , Sangre Fetal/metabolismo , Citometría de Flujo , Células Madre Hematopoyéticas/metabolismo , Humanos , Masculino , Ratones , Ratones Endogámicos C57BL , Microscopía Fluorescente , Células Madre/citología , Células Madre/metabolismo , Factores de Tiempo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA