Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 23
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
J Clin Med ; 11(13)2022 Jul 01.
Artigo em Inglês | MEDLINE | ID: mdl-35807112

RESUMO

BACKGROUND: Bimaxillary orthognathic surgery bears the risk of severe postoperative airway complications. There are no clear recommendations for immediate postoperative follow-up and monitoring. OBJECTIVE: to identify potential risk factors for prolonged mechanical ventilation and delayed extubation in patients undergoing bimaxillary orthognathic surgery. METHODS: The data of all consecutive patients undergoing bimaxillary surgery between May 2012 and October 2019 were analyzed in a single-center retrospective cohort study. The clinical data were evaluated regarding baseline characteristics and potential factors linked with delayed extubation. RESULTS: A total of 195 patients were included; 54.9% were female, and the median age was 23 years (IQR 5). The median body mass index was 23.1 (IQR 8). Nine patients (4.6%) were of American Society of Anesthesiologists Physical Status Classification System III or higher. The median duration of mechanical ventilation in the intensive care unit was 280 min (IQR, 526 min). Multivariable analysis revealed that premedication with benzodiazepines (odds ratio (OR) 2.60, 95% confidence interval (0.99; 6.81)), the male sex (OR 2.43, 95% confidence interval (1.10; 5.36)), and the duration of surgery (OR 1.54, 95% confidence interval (1.07; 2.23)) were associated with prolonged mechanical ventilation. By contrast, total intravenous anesthesia was associated with shorter ventilation time (OR 0.19, 95% confidence interval (0.09; 0.43)). CONCLUSION: premedication with benzodiazepines, the male sex, and the duration of surgery might be considered to be independent risk factors for delayed extubation in patients undergoing bimaxillary surgery.

2.
J Stomatol Oral Maxillofac Surg ; 123(5): e556-e562, 2022 10.
Artigo em Inglês | MEDLINE | ID: mdl-35272089

RESUMO

BACKGROUND: Microvascular surgery has become a standardized technique for reconstruction of large tissue defects in Head and Neck Reconstructive Surgery. However, the main dreaded complications are thrombosis of blood vessels or major bleeding after surgery. Several different anticoagulation protocols have been established in the last decades to overcome these problems with varying degrees of success. METHODS: Over a period of six years, a standardized anticoagulation protocol including acetylsalicylic acid (ASA) and unfractionated heparin (UFH) for direct intraoperative and postoperative administration was established, optimized and compared to a previously used non-standardized protocol. A total of 178 flap surgeries were included in the development and optimization process of the protocol. RESULTS: ASA significantly increased the risk of complications when used for longer than 72 h (OR = 2.52; p = 0.002; 95% CI 1.39-4.59). Administration of UFH reduced flap loss (bolus: OR 0.68; p = 0.47; 95% CI 0.24-1.93; continuous UFH administration: OR = 0.61; p = 0.33; 95% CI 0.22-1.66), however doses greater than 500 IU/ h of UFH as continuous infusion increased the risk of complications. Reduction in ischemia time had no effect on the occurrence of complications. CONCLUSION: Anticoagulation regimes in microvascular surgery can influence the postoperative complication rate. The optimal protocol should consist of a combination of ASA and UFH for the intraoperative and direct postoperative phase. Prolonged administration of ASA as well as doses >500 IU/ h of UFH are to be avoided due to the increased complication rate.


Assuntos
Retalhos de Tecido Biológico , Heparina , Anticoagulantes/efeitos adversos , Aspirina/efeitos adversos , Heparina/efeitos adversos , Humanos , Isquemia/epidemiologia , Isquemia/cirurgia , Complicações Pós-Operatórias/epidemiologia , Complicações Pós-Operatórias/etiologia , Complicações Pós-Operatórias/prevenção & controle
3.
JAMA ; 313(21): 2133-41, 2015 Jun 02.
Artigo em Inglês | MEDLINE | ID: mdl-26024502

RESUMO

IMPORTANCE: No interventions have yet been identified to reduce the risk of acute kidney injury in the setting of cardiac surgery. OBJECTIVE: To determine whether remote ischemic preconditioning reduces the rate and severity of acute kidney injury in patients undergoing cardiac surgery. DESIGN, SETTING, AND PARTICIPANTS: In this multicenter trial, we enrolled 240 patients at high risk for acute kidney injury, as identified by a Cleveland Clinic Foundation score of 6 or higher, between August 2013 and June 2014 at 4 hospitals in Germany. We randomized them to receive remote ischemic preconditioning or sham remote ischemic preconditioning (control). All patients completed follow-up 30 days after surgery and were analyzed according to the intention-to-treat principle. INTERVENTIONS: Patients received either remote ischemic preconditioning (3 cycles of 5-minute ischemia and 5-minute reperfusion in one upper arm after induction of anesthesia) or sham remote ischemic preconditioning (control), both via blood pressure cuff inflation. MAIN OUTCOMES AND MEASURES: The primary end point was the rate of acute kidney injury defined by Kidney Disease: Improving Global Outcomes criteria within the first 72 hours after cardiac surgery. Secondary end points included use of renal replacement therapy, duration of intensive care unit stay, occurrence of myocardial infarction and stroke, in-hospital and 30-day mortality, and change in acute kidney injury biomarkers. RESULTS: Acute kidney injury was significantly reduced with remote ischemic preconditioning (45 of 120 patients [37.5%]) compared with control (63 of 120 patients [52.5%]; absolute risk reduction, 15%; 95% CI, 2.56%-27.44%; P = .02). Fewer patients receiving remote ischemic preconditioning received renal replacement therapy (7 [5.8%] vs 19 [15.8%]; absolute risk reduction, 10%; 95% CI, 2.25%-17.75%; P = .01), and remote ischemic preconditioning reduced intensive care unit stay (3 days [interquartile range, 2-5]) vs 4 days (interquartile range, 2-7) (P = .04). There was no significant effect of remote ischemic preconditioning on myocardial infarction, stroke, or mortality. Remote ischemic preconditioning significantly attenuated the release of urinary insulinlike growth factor-binding protein 7 and tissue inhibitor of metalloproteinases 2 after surgery (remote ischemic preconditioning, 0.36 vs control, 0.97 ng/mL2/1000; difference, 0.61; 95% CI, 0.27-0.86; P < .001). No adverse events were reported with remote ischemic preconditioning. CONCLUSIONS AND RELEVANCE: Among high-risk patients undergoing cardiac surgery, remote ischemic preconditioning compared with no ischemic preconditioning significantly reduced the rate of acute kidney injury and use of renal replacement therapy. The observed reduction in the rate of acute kidney injury and the need for renal replacement warrants further investigation. TRIAL REGISTRATION: German Clinical Trials Register Identifier: DRKS00005333.


Assuntos
Injúria Renal Aguda/prevenção & controle , Procedimentos Cirúrgicos Cardíacos , Precondicionamento Isquêmico , Complicações Pós-Operatórias/prevenção & controle , Proteínas de Fase Aguda/urina , Idoso , Biomarcadores/sangue , Biomarcadores/urina , Creatinina/sangue , Feminino , Humanos , Unidades de Terapia Intensiva , Tempo de Internação , Lipocalina-2 , Lipocalinas/urina , Masculino , Pessoa de Meia-Idade , Proteínas Proto-Oncogênicas/urina , Terapia de Substituição Renal/estatística & dados numéricos
4.
PLoS One ; 8(10): e77258, 2013.
Artigo em Inglês | MEDLINE | ID: mdl-24167567

RESUMO

Ischemic and traumatic brain injury is associated with increased risk for death and disability. The inhibition of penumbral tissue damage has been recognized as a target for therapeutic intervention, because cellular injury evolves progressively upon ATP-depletion and loss of ion homeostasis. In patients, thiopental is used to treat refractory intracranial hypertension by reducing intracranial pressure and cerebral metabolic demands; however, therapeutic benefits of thiopental-treatment are controversially discussed. In the present study we identified fundamental neuroprotective molecular mechanisms mediated by thiopental. Here we show that thiopental inhibits global protein synthesis, which preserves the intracellular energy metabolite content in oxygen-deprived human neuronal SK-N-SH cells or primary mouse cortical neurons and thus ameliorates hypoxic cell damage. Sensitivity to hypoxic damage was restored by pharmacologic repression of eukaryotic elongation factor 2 kinase. Translational inhibition was mediated by calcium influx, activation of the AMP-activated protein kinase, and inhibitory phosphorylation of eukaryotic elongation factor 2. Our results explain the reduction of cerebral metabolic demands during thiopental treatment. Cycloheximide also protected neurons from hypoxic cell death, indicating that translational inhibitors may generally reduce secondary brain injury. In conclusion our study demonstrates that therapeutic inhibition of global protein synthesis protects neurons from hypoxic damage by preserving energy balance in oxygen-deprived cells. Molecular evidence for thiopental-mediated neuroprotection favours a positive clinical evaluation of barbiturate treatment. The chemical structure of thiopental could represent a pharmacologically relevant scaffold for the development of new organ-protective compounds to ameliorate tissue damage when oxygen availability is limited.


Assuntos
Hipnóticos e Sedativos/farmacologia , Neurônios/metabolismo , Fator 2 de Elongação de Peptídeos/metabolismo , Biossíntese de Proteínas/efeitos dos fármacos , Tiopental/farmacologia , Animais , Lesões Encefálicas/tratamento farmacológico , Lesões Encefálicas/metabolismo , Lesões Encefálicas/patologia , Isquemia Encefálica/tratamento farmacológico , Isquemia Encefálica/metabolismo , Isquemia Encefálica/patologia , Morte Celular/efeitos dos fármacos , Hipóxia Celular/efeitos dos fármacos , Linhagem Celular , Quinase do Fator 2 de Elongação/metabolismo , Humanos , Camundongos , Neurônios/patologia , Oxigênio/metabolismo
5.
J Pharmacol Exp Ther ; 347(3): 781-93, 2013 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-24049063

RESUMO

Oxygen deprivation during ischemic or hemorrhagic stroke results in ATP depletion, loss of ion homeostasis, membrane depolarization, and excitotoxicity. Pharmacologic restoration of cellular energy supply may offer a promising concept to reduce hypoxic cell injury. In this study, we investigated whether carbimazole, a thionamide used to treat hyperthyroidism, reduces neuronal cell damage in oxygen-deprived human SK-N-SH cells or primary cortical neurons. Our results revealed that carbimazole induces an inhibitory phosphorylation of eukaryotic elongation factor 2 (eEF2) that was associated with a marked inhibition of global protein synthesis. Translational inhibition resulted in significant bioenergetic savings, preserving intracellular ATP content in oxygen-deprived neuronal cells and diminishing hypoxic cellular damage. Phosphorylation of eEF2 was mediated by AMP-activated protein kinase and eEF2 kinase. Carbimazole also induced a moderate calcium influx and a transient cAMP increase. To test whether translational inhibition generally diminishes hypoxic cell damage when ATP availability is limiting, the translational repressors cycloheximide and anisomycin were used. Cycloheximide and anisomycin also preserved ATP content in hypoxic SK-N-SH cells and significantly reduced hypoxic neuronal cell damage. Taken together, these data support a causal relation between the pharmacologic inhibition of global protein synthesis and efficient protection of neurons from ischemic damage by preservation of high-energy metabolites in oxygen-deprived cells. Furthermore, our results indicate that carbimazole or other translational inhibitors may be interesting candidates for the development of new organ-protective compounds. Their chemical structure may be used for computer-assisted drug design or screening of compounds to find new agents with the potential to diminish neuronal damage under ATP-limited conditions.


Assuntos
Antitireóideos/farmacologia , Carbimazol/farmacologia , Hipóxia Celular/efeitos dos fármacos , Neurônios/efeitos dos fármacos , Inibidores da Síntese de Proteínas , Proteínas Quinases Ativadas por AMP/metabolismo , Trifosfato de Adenosina/metabolismo , Autorradiografia , Western Blotting , Cálcio/metabolismo , Caspases/metabolismo , Linhagem Celular , Sobrevivência Celular/efeitos dos fármacos , AMP Cíclico/metabolismo , Quinase do Fator 2 de Elongação/metabolismo , Metabolismo Energético/efeitos dos fármacos , Humanos , Fosforilação
6.
Int J Biochem Cell Biol ; 45(2): 201-12, 2013 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-23041477

RESUMO

Carbon monoxide (CO) is an endogenous gaseous transmitter that exerts antiproliferative effects in many cell types, but effects of CO on the translational machinery are not described. We examined the effects of the carbon monoxide releasing molecule-2 (CORM-2) on critical steps in translational signaling and global protein synthesis in pancreatic stellate cells (PSCs), the most prominent collagen-producing cells in the pancreas, whose activation is associated with pancreatic fibrosis. PSCs were isolated from rat pancreatic tissue and incubated with CORM-2. CORM-2 prevented the decrease in the phosphorylation of eukaryotic elongation factor 2 (eEF2) caused by serum. By contrast, the activation dependent phosphorylation of initiation factor 4E-binding protein 1 (4E-BP1) was inhibited by CORM-2 treatment. The phosphorylation of eukaryotic initiation factor 2α (eIF2α) and eukaryotic initiation factor 4E (eIF4E) were not affected by CORM-2 treatment. In consequence, CORM-2 mediated eEF2 phosphorylation and inactivation of 4E-BP1 suppressed global protein synthesis. These observations were associated with inhibition of phosphatidylinositol 3-kinase-Akt-mammalian target of rapamycin (PI3K-Akt-mTOR) signaling and increased intracellular calcium and cAMP levels. The CORM-2 mediated inhibition of protein synthesis resulted in downregulation of cyclin D1 and cyclin E expression, a subsequent decline in the phosphorylation of the retinoblastoma tumor suppressor protein (Rb) and cell growth arrest at the G(0)/G(1) phase checkpoint of the cell cycle. Our results suggest the therapeutic application of CO releasing molecules such as CORM-2 for the treatment of fibrosis, inflammation, cancer, or other pathologic states associated with excessive protein synthesis or hyperproliferation. However, prolonged exogenous application of CO might also have negative effects on cellular protein homeostasis.


Assuntos
Compostos Organometálicos/farmacologia , Elongação Traducional da Cadeia Peptídica/efeitos dos fármacos , Fator 2 de Elongação de Peptídeos/metabolismo , Inibidores da Síntese de Proteínas/farmacologia , Animais , Sinalização do Cálcio , Monóxido de Carbono/metabolismo , Monóxido de Carbono/farmacologia , Células Cultivadas , AMP Cíclico/metabolismo , Ciclina D1/metabolismo , Ciclina E/metabolismo , Pontos de Checagem da Fase G1 do Ciclo Celular/efeitos dos fármacos , Heme Oxigenase-1/metabolismo , Sistema de Sinalização das MAP Quinases , Masculino , Compostos Organometálicos/metabolismo , Células Estreladas do Pâncreas/efeitos dos fármacos , Células Estreladas do Pâncreas/metabolismo , Fosfatidilinositol 3-Quinases/metabolismo , Fosforilação , Cultura Primária de Células , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Inibidores da Síntese de Proteínas/metabolismo , Proteínas Proto-Oncogênicas c-akt/metabolismo , Ratos , Ratos Wistar , Proteína do Retinoblastoma/metabolismo , Serina-Treonina Quinases TOR/metabolismo
7.
PLoS One ; 7(9): e46479, 2012.
Artigo em Inglês | MEDLINE | ID: mdl-23029526

RESUMO

PURPOSE: Ischemia and reperfusion injury (I/R) of neuronal structures and organs is associated with increased morbidity and mortality due to neuronal cell death. We hypothesized that inhalation of carbon monoxide (CO) after I/R injury ('postconditioning') would protect retinal ganglion cells (RGC). METHODS: Retinal I/R injury was performed in Sprague-Dawley rats (n = 8) by increasing ocular pressure (120 mmHg, 1 h). Rats inhaled room air or CO (250 ppm) for 1 h immediately following ischemia or with 1.5 and 3 h latency. Retinal tissue was harvested to analyze Bcl-2, Bax, Caspase-3, HO-1 expression and phosphorylation of the nuclear transcription factor (NF)-κB, p38 and ERK-1/2 MAPK. NF-κB activation was determined and inhibition of ERK-1/2 was performed using PD98059 (2 mg/kg). Densities of fluorogold prelabeled RGC were analyzed 7 days after injury. Microglia, macrophage and Müller cell activation and proliferation were evaluated by Iba-1, GFAP and Ki-67 staining. RESULTS: Inhalation of CO after I/R inhibited Bax and Caspase-3 expression (Bax: 1.9 ± 0.3 vs. 1.4 ± 0.2, p = 0.028; caspase-3: 2.0 ± 0.2 vs. 1.5 ± 0.1, p = 0.007; mean ± S.D., fold induction at 12 h), while expression of Bcl-2 was induced (1.2 ± 0.2 vs. 1.6 ± 0.2, p = 0.001; mean ± S.D., fold induction at 12 h). CO postconditioning suppressed retinal p38 phosphorylation (p = 0.023 at 24 h) and induced the phosphorylation of ERK-1/2 (p<0.001 at 24 h). CO postconditioning inhibited the expression of HO-1. The activation of NF-κB, microglia and Müller cells was potently inhibited by CO as well as immigration of proliferative microglia and macrophages into the retina. CO protected I/R-injured RGC with a therapeutic window at least up to 3 h (n = 8; RGC/mm(2); mean ± S.D.: 1255 ± 327 I/R only vs. 1956 ± 157 immediate CO treatment, vs. 1830 ± 109 1.5 h time lag and vs. 1626 ± 122 3 h time lag; p<0.001). Inhibition of ERK-1/2 did not counteract the CO effects (RGC/mm(2): 1956 ± 157 vs. 1931 ± 124, mean ± S.D., p = 0.799). CONCLUSION: Inhaled CO, administered after retinal ischemic injury, protects RGC through its strong anti-apoptotic and anti-inflammatory effects.


Assuntos
Anti-Inflamatórios/administração & dosagem , Apoptose/efeitos dos fármacos , Monóxido de Carbono/administração & dosagem , Fármacos Neuroprotetores/administração & dosagem , Traumatismo por Reperfusão/tratamento farmacológico , Retinite/prevenção & controle , Administração por Inalação , Animais , Caspase 3/genética , Caspase 3/metabolismo , Movimento Celular/efeitos dos fármacos , Ativação Enzimática , Feminino , Expressão Gênica/efeitos dos fármacos , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Isquemia/tratamento farmacológico , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Neuroglia/efeitos dos fármacos , Neuroglia/fisiologia , Fosforilação , Processamento de Proteína Pós-Traducional/efeitos dos fármacos , Proteínas Proto-Oncogênicas c-bcl-2/genética , Proteínas Proto-Oncogênicas c-bcl-2/metabolismo , Ratos , Ratos Sprague-Dawley , Retina/efeitos dos fármacos , Retina/patologia , Células Ganglionares da Retina/efeitos dos fármacos , Células Ganglionares da Retina/patologia , Vasos Retinianos/patologia , Proteína X Associada a bcl-2/genética , Proteína X Associada a bcl-2/metabolismo
8.
Pancreas ; 41(1): 74-83, 2012 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-21934550

RESUMO

OBJECTIVES: Pancreatic stellate cells (PSCs) play a crucial role during pancreatic fibrosis development. Hydrogen sulfide (H2S) is a recently discovered gaseous transmitter, whose role in PSCs has not been explored yet. In the present study, we examined the effects of sodium hydrosulfide (NaHS), an H2S donor, on rat PSCs and elucidated the mechanisms involved. METHODS: Primary PSCs were isolated from rat pancreatic tissue. Lactate dehydrogenase and caspase assays were performed to detect cell death. Pancreatic stellate cell proliferation was determined by cell count analyses, bromodeoxyuridine incorporation, and flow cytometry. The role of heme oxygenase-1 (HO-1) was assessed by pharmacological HO inhibition and transfection of HO-1 small interfering RNA. Pancreatic stellate cell migration was determined by a wound healing assay, and PSC contraction was assessed by a gel contraction assay. α-Smooth muscle actin, collagen type I, and fibronectin messenger RNAs were analyzed by real-time polymerase chain reaction. RESULTS: NaHS inhibited PSC proliferation at nontoxic concentrations. This was associated with HO-1-mediated repression of extracellular signal-regulated kinase 1/2 signaling. NaHS suppressed PSC migration and activation as well as extracellular matrix synthesis. CONCLUSIONS: The results of the present study indicate that NaHS inhibits key cell functions of PSCs. Administration of H(2)S-releasing compounds might represent a novel strategy in the treatment of pancreatic fibrosis.


Assuntos
Movimento Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Sulfeto de Hidrogênio/farmacologia , Células Estreladas do Pâncreas/efeitos dos fármacos , Actinas/genética , Animais , Western Blotting , Células Cultivadas , Colágeno Tipo I/genética , Matriz Extracelular/metabolismo , Fibronectinas/genética , Expressão Gênica/efeitos dos fármacos , Heme Oxigenase-1/genética , Heme Oxigenase-1/metabolismo , Sulfeto de Hidrogênio/metabolismo , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Células Estreladas do Pâncreas/metabolismo , Interferência de RNA , Ratos , Ratos Wistar , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Sulfetos/metabolismo , Sulfetos/farmacologia
9.
Curr Pharm Biotechnol ; 13(6): 813-8, 2012 May.
Artigo em Inglês | MEDLINE | ID: mdl-22201611

RESUMO

Carbon monoxide (CO), often referred to as the silent killer, is a colorless, odorless and tasteless gas. It combines with hemoglobin to produce carboxyhemoglobin, which is ineffective for delivering oxygen to animal and human tissues. On the other hand, CO is endogenously produced in the body as a byproduct of heme degradation catalyzed by the heme oxygenase (HO) enzymes. In the past decade, evidence has accumulated to suggest important physiological roles for CO in mammalian tissues. In the pancreas, modulation of endogenous CO production or administration of exogenous CO may represent a therapeutic option for the treatment of endocrine and exocrine pancreatic disorders. In cell culture, CO exerts anti-diabetic effects and brief exposure of purified mouse islets to CO ameliorates functional performance after transplantation. Recent advances include the observation that CO carriers possess potent anti-proliferative effects in an in vitro model of pancreatic fibrosis. In vivo, CO confers tissue protection in animal models of pancreatic disease, including those with hyperglycemia and inflammatory injury of the gland. However, there are still a number of unanswered questions surrounding its physiological and pathophysiological relevance and the preferred route of CO administration in the pancreas still remains to be settled. This brief review focuses on the roles, effects and mechanisms of action of CO in the pancreas.


Assuntos
Monóxido de Carbono/metabolismo , Pâncreas/metabolismo , Animais , Monóxido de Carbono/farmacologia , Heme Oxigenase (Desciclizante)/metabolismo , Humanos , Hiperglicemia/metabolismo , Pancreatite/metabolismo , Traumatismo por Reperfusão/metabolismo
10.
World J Gastroenterol ; 17(37): 4184-90, 2011 Oct 07.
Artigo em Inglês | MEDLINE | ID: mdl-22072849

RESUMO

AIM: To characterize the inductive effects of isoflurane (ISO) on hepatic heme oxygenase-1 (HO-1) in an animal model of hepatic steatosis. METHODS: Lean (LEAN) and obese (FAT) Zucker rats were randomized into 4 groups: 1: LEAN + pentobarbital sodium (PEN); 2: LEAN + ISO; 3: FAT + PEN; 4: FAT + ISO. The animals were mechanically ventilated for 6 h. In vitro analyses of liver tissue included determination of HO-1 mRNA and protein expression as well as measurement of HO enzyme activity and immunohistochemical analyses. RESULTS: Compared to PEN treatment, ISO administration profoundly induced hepatic HO-1 mRNA and protein expression and significantly increased HO enzyme activity in lean Zucker rats. In contrast, no difference in HO-1 gene expression was observed after ISO or PEN anesthesia in obese Zucker rats. CONCLUSION: The present study demonstrates that ISO is an inducer of hepatic HO-1 gene expression in non-steatotic organs but failed to upregulate HO-1 in steatotic livers.


Assuntos
Anestésicos Inalatórios/farmacologia , Indução Enzimática , Fígado Gorduroso/enzimologia , Heme Oxigenase-1/metabolismo , Isoflurano/farmacologia , Fígado/efeitos dos fármacos , Fígado/enzimologia , Animais , Fígado Gorduroso/genética , Expressão Gênica , Glutationa Transferase/sangue , Heme Oxigenase-1/genética , Masculino , RNA Mensageiro/genética , RNA Mensageiro/metabolismo , Distribuição Aleatória , Ratos , Ratos Zucker
11.
Eur J Pharmacol ; 670(1): 58-66, 2011 Nov 16.
Artigo em Inglês | MEDLINE | ID: mdl-21925493

RESUMO

The carbon monoxide releasing molecule tricarbonyldichlororuthenium (CORM-2) displays protective actions like carbon monoxide. The molecular mechanism underlying this effect remains controversial. We hypothesized that CORM-2 mediates cytoprotection via induction of heat shock proteins through activation of p38 mitogen-activated kinase. Embryonic bovine lung cells were incubated with CORM-2. Apoptosis was induced by staurosporine and analyzed by flow cytometry following annexin-V staining, caspase-3 activity assay, and by Western Blot for caspase-3 cleavage. Heat shock response was assessed by DNA-binding activity of heat shock factor 1 and by reporter gene activity. Cells were transfected with siRNA targeting p38 isoforms. Data were analyzed with ANOVA and post-hoc Holm-Sidak test. CORM-2 inhibited staurosporine-induced apoptosis (% annexin-V positive cells: staurosporine = 60 ± 4% vs. CORM-2 10 µM = 48 ± 4%, CORM-2 25 µM=42 ± 5%, CORM-2 50 µM = 40 ± 4% and CORM-2 100 µM = 38 ± 2%, mean ± S.D., P<0.001; caspase-3 activity: staurosporine=92 ± 15 RFUs vs. CORM-2 50 µM=60 ± 14 RFUs, mean ± S.D. P<0.001). CORM-2 induced phosphorylation of p38 MAPK, but not of JNK and ERK1/2. CORM-2 induced DNA-binding of heat shock factor 1 and elicited a 4-fold induction of gene activity (P<0.05). Incubation with the Hsp inhibitors KNK437 attenuated and 17-AAG abolished the anti-apoptotic effect of CORM-2 (P<0.001). p38 inhibition and silencing of p38ß attenuated the anti-apoptotic effect of CORM-2 (P<0.05), most likely by abolishing CORM-2-induced HSF-1 binding activity. These findings suggest that CORM-2-mediated cytoprotection is caused by induction of the heat shock response and by p38 activation. Furthermore, the p38ß isoform activation may represent an upstream mechanism of heat shock response induction.


Assuntos
Dióxido de Carbono/química , Dióxido de Carbono/farmacologia , Citoproteção/efeitos dos fármacos , Resposta ao Choque Térmico/efeitos dos fármacos , Pulmão/citologia , Proteína Quinase 11 Ativada por Mitógeno/metabolismo , Compostos Organometálicos/química , Animais , Apoptose/efeitos dos fármacos , Bovinos , Ativação Enzimática/efeitos dos fármacos , Inativação Gênica , Pulmão/efeitos dos fármacos , Pulmão/enzimologia , Proteína Quinase 11 Ativada por Mitógeno/deficiência , Proteína Quinase 11 Ativada por Mitógeno/genética , Estaurosporina/farmacologia
12.
Mol Vis ; 17: 1275-86, 2011.
Artigo em Inglês | MEDLINE | ID: mdl-21633713

RESUMO

PURPOSE: Retinal ischemia/reperfusion (I/R) injury plays an important role in the pathophysiology of various ocular diseases. Retinal ganglion cells (RGCs) are particularly vulnerable to ischemia. Hydrogen sulfide (H(2)S) was recently shown to be neuroprotective in the brain and retina due to its antiapoptotic effects. Rapid preconditioning of retinal neurons by inhaled H(2)S before I/R injury may reduce apoptosis in the rat retina. METHODS: I/R injury was created on the left eye of rats (n=8) with or without inhaled H(2)S preconditioning (80 ppm) for one hour before ischemia. Densities of fluorogold prelabeled RGCs were analyzed 7 days after injury in retinal whole mounts. Retinal tissue was harvested to analyze protein expression of heat shock protein (HSP)-90 and the mitogen-activated protein kinases (MAPKs) c-jun N-terminal kinase (JNK), extracellular signal-regulated kinase (ERK)1/2 and p38 to elucidate a possible pathway of neuroprotection. DNA binding activity of the transcription factors nuclear factor-kappa-light-chain-enhancer of activated B-cells (NF-κB), cyclic adenosine monophosphate response element binding protein (CREB), and heat shock element (HSE), as well as caspase-3 cleavage and activity, were determined. Retinal sections were further assessed using anti-glial fibrillary acidic protein staining. RESULTS: RGC death after I/R injury decreased by 41.5% after H(2)S preconditioning compared to room air (p<0.001). H(2)S inhalation before ischemia reduced caspase-3 cleavage (p<0.001) and attenuated caspase-3 activity (p<0.001). Furthermore, HSP-90 expression was significantly elevated in the retina after H(2)S preconditioning. NF-κB but not CREB or HSE showed specific, H2S-dependent regulation, as well as the MAPKs ERK1/2 and JNK but not p38. CONCLUSIONS: H(2)S preconditioning mediates antiapoptotic effects in retinal I/R injury, thus exhibiting neuroprotection. Based on these observations, H(2)S could represent a novel and promising therapeutic agent to counteract neuronal injuries in the eye. Further studies are needed to prove H(2)S's neuroprotective propensity using a postconditioning approach.


Assuntos
Apoptose/efeitos dos fármacos , Sulfeto de Hidrogênio/administração & dosagem , Precondicionamento Isquêmico/métodos , Fármacos Neuroprotetores/administração & dosagem , Traumatismo por Reperfusão/fisiopatologia , Vasos Retinianos/fisiopatologia , Administração por Inalação , Animais , Inibidores de Caspase , Morte Celular/efeitos dos fármacos , DNA/metabolismo , Feminino , Proteína Glial Fibrilar Ácida/metabolismo , Proteínas de Choque Térmico HSP90/metabolismo , Masculino , Proteínas Quinases Ativadas por Mitógeno/metabolismo , NF-kappa B/metabolismo , Ratos , Ratos Sprague-Dawley , Traumatismo por Reperfusão/patologia , Retina/metabolismo , Células Ganglionares da Retina/efeitos dos fármacos , Vasos Retinianos/efeitos dos fármacos , Fatores de Tempo , Distribuição Tecidual
13.
Anesth Analg ; 112(2): 282-91, 2011 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-21156982

RESUMO

BACKGROUND: Administration of inhaled carbon monoxide before organ ischemic injury exerts protective effects in animal models. Because there are no data showing that this also works after an ischemic insult, our objective in this study was to investigate whether inhaled carbon monoxide attenuates cardiopulmonary bypass (CPB)-induced lung injury in a pig model. METHODS: Animals were randomized to a SHAM group (n = 5), a SHAM group plus inhaled carbon monoxide (n = 5), standard CPB (n = 10), and to CPB plus inhaled carbon monoxide (n = 10). Carbon monoxide (250 ppm) was given for 1 hour after termination of CPB. Lung biopsies were obtained before CPB, immediately after separation from CPB, and for 5 hours after termination of CPB to determine expression of pulmonary heat shock proteins 70 and 90, cytokines, alveolar macrophage infiltration, and fluorogenic caspase-3 activity. RESULTS: At 5 hours after CPB, administration of inhaled carbon monoxide was associated with reduced pulmonary expression of the inflammatory cytokines tumor necrosis factor (CPB + CO 521 ± 77 vs CPB 821 ± 97 pg · mL(-1), P < 0.001) and interleukin-6 (304 ± 81 vs 860 ± 153 pg · mL(-1), P < 0.001), increased pulmonary expression of the cytoprotective heat shock protein 70 (CPB + CO 79 ± 14 vs CPB 36 ± 9 ng · mL(-1), P < 0.001) and the antiinflammatory cytokine interleukin-10 (CPB + CO 278 ± 40 vs CPB 63 ± 20 pg · mL(-1), P < 0.001), and with reduced pulmonary apoptotic protein caspase-3 activity (CPB + CO 0.73 ± 0.11 vs CPB 0.99 ± 0.1 RFU, P < 0.05). Carbon monoxide administration was associated with reduced histological evidence of lung injury and alveolar macrophage infiltration (78 ± 39 vs 145 ± 34 counts per field of vision, P < 0.001). CONCLUSIONS: These results suggest that administration of low concentrations of carbon monoxide after CPB ("postconditioning") protects the lung from CPB-related lung injury.


Assuntos
Monóxido de Carbono/administração & dosagem , Ponte Cardiopulmonar/efeitos adversos , Isquemia/prevenção & controle , Lesão Pulmonar/prevenção & controle , Pulmão/efeitos dos fármacos , Administração por Inalação , Animais , Apoptose/efeitos dos fármacos , Biópsia , Caspase 3/metabolismo , Modelos Animais de Doenças , Proteínas de Choque Térmico HSP70/metabolismo , Hemodinâmica/efeitos dos fármacos , Mediadores da Inflamação/metabolismo , Interleucina-10/metabolismo , Interleucina-6/metabolismo , Isquemia/etiologia , Isquemia/imunologia , Isquemia/patologia , Isquemia/fisiopatologia , Pulmão/irrigação sanguínea , Pulmão/imunologia , Pulmão/patologia , Pulmão/fisiopatologia , Lesão Pulmonar/etiologia , Lesão Pulmonar/imunologia , Lesão Pulmonar/patologia , Lesão Pulmonar/fisiopatologia , Macrófagos/efeitos dos fármacos , Macrófagos/imunologia , Suínos , Fatores de Tempo , Fator de Necrose Tumoral alfa/metabolismo
14.
Int J Biochem Cell Biol ; 42(11): 1876-83, 2010 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-20727416

RESUMO

Sevoflurane is a potent non-toxic inducer of the hepatoprotective enzyme heme oxygenase-1 (HO-1). So far, little is known about the underlying molecular mechanism. Therefore the aim of this study was to characterize the respective signal transduction pathway and in particular to elucidate the role of Kupffer cells in this context. Rats were treated with or without sevoflurane. The effects on hepatic HO-1 gene expression, mitogen-activated protein kinases and transcription factors were studied by Northern and Western blot analyses, immunostaining, electrophoretic mobility shift assays, and enzymatic activity assays. Kupffer cells were depleted by administration of clodronate liposomes in vivo to characterize their role in HO-1 signal transduction. In additional in vitro experiments, HO-1 mRNA expression in primary rat hepatocytes and HepG2 cells was assessed. Sevoflurane up-regulated HO-1 gene expression in pericentral hepatocytes and increased HO enzyme activity in vivo. This was associated with activation of ERK1/2 and activator protein-1. We identified c-jun/AP-1, JunD, c-fos, and Fra-1 as active subunits of the activator protein-1 complex. Administration of clodronate liposomes to rats led to depletion of Kupffer cells without affecting sevoflurane induced HO-1 expression. Moreover, sevoflurane up-regulated HO-1 mRNA in primary rat hepatocytes but not in HepG2 cells. Our results suggest that sevoflurane induced HO-1 gene expression in pericentral hepatocytes does not depend on Kupffer cells and is associated with activation of ERK1/2 and activator protein-1. Since we could recently demonstrate significant hepatoprotective effects of HO-1 induced by isoflurane, the present results may help to establish new concepts in hepatic organ protection.


Assuntos
Heme Oxigenase-1/metabolismo , Células de Kupffer/metabolismo , Fígado/metabolismo , Éteres Metílicos/farmacologia , Proteína Quinase 1 Ativada por Mitógeno/metabolismo , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Fator de Transcrição AP-1/metabolismo , Animais , Northern Blotting , Western Blotting , Células Cultivadas , Ensaio de Desvio de Mobilidade Eletroforética , Heme Oxigenase-1/genética , Células Hep G2 , Hepatócitos/efeitos dos fármacos , Hepatócitos/metabolismo , Humanos , Imuno-Histoquímica , Fígado/efeitos dos fármacos , Fígado/enzimologia , Masculino , Proteína Quinase 1 Ativada por Mitógeno/genética , Proteína Quinase 3 Ativada por Mitógeno/genética , Reação em Cadeia da Polimerase , Ratos , Ratos Sprague-Dawley , Sevoflurano , Fator de Transcrição AP-1/genética
15.
Anesth Analg ; 111(1): 29-37, 2010 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-20519418

RESUMO

BACKGROUND: Cardiopulmonary bypass (CPB) may be associated with acute kidney injury (AKI). Inhaled carbon monoxide (CO) is cyto- and organ-protective. We hypothesized that pretreatment with inhaled CO prevents CPB-associated AKI. METHODS: Pigs (n = 38) were nonrandomly assigned to SHAM, standard CPB, pretreatment with inhaled CO (250 ppm, 1 hour) before SHAM or CPB, to pretreatment with quercetin (an inhibitor of the heat shock response), and to pretreatment with SnPPIX (an inhibitor of endogenously derived CO), before CO inhalation and CPB. The primary outcome variables were markers of AKI (urea, uric acid, creatinine, cystatin C, neutrophil gelatinase-associated lipocalin, interleukin-6, tumor necrosis factor-alpha), which were determined 120 minutes after CPB. Secondary outcome variables were heat shock protein (HSP)-70 and heme oxygenase-1 protein expressions as indicators of CO-mediated heat shock response. RESULTS: Pretreatment with inhaled CO attenuated (all P < 0.001) CPB-associated, (1) increases in serum concentrations of cystatin C (64 +/- 14 vs 28 +/- 9 ng/mL), neutrophil gelatinase-associated lipocalin (391 +/- 65 vs 183 +/- 56 ng/mL), renal tumor necrosis factor-alpha (450 +/- 73 vs 179 +/- 110 pg/mL), and interleukin-6 (483 +/- 102 vs 125 +/- 67 pg/mL); (2) increase in renal caspase-3 activity (550 +/- 66 vs 259 +/- 52 relative fluorescent units); and (3) histological evidence of AKI. These effects were accompanied by activation of HSP-70 (196 +/- 64 vs 554 +/- 149 ng/mL, P < 0.001). Pretreatment with the heat shock response inhibitor quercetin counteracted the CO-associated biochemical and histological renoprotective effects (all P < 0.001), whereas the heme oxygenase inhibitor SnPPIX only partially counteracted the CO-associated renoprotection and the activation of the heat shock response. CONCLUSIONS: CO treatment before CPB was associated with evidence of renoprotection, demonstrated by fewer histological injuries and decreased cystatin C concentrations. The findings that the antiinflammatory and antiapoptotic effects of CO were accompanied by activation of HSP-70, which in turn were reversed by quercetin, suggest that renoprotection by pretreatment with inhaled CO before CPB is mediated by activation of the renal heat shock response.


Assuntos
Monóxido de Carbono/farmacologia , Ponte Cardiopulmonar/efeitos adversos , Proteínas de Choque Térmico/biossíntese , Resposta ao Choque Térmico/efeitos dos fármacos , Resposta ao Choque Térmico/fisiologia , Nefropatias/etiologia , Nefropatias/prevenção & controle , Doença Aguda , Administração por Inalação , Animais , Monóxido de Carbono/administração & dosagem , Monóxido de Carbono/sangue , Caspase 3/biossíntese , Heme Oxigenase (Desciclizante)/biossíntese , Hemodinâmica/efeitos dos fármacos , Interleucina-6/biossíntese , Rim/patologia , Nefropatias/patologia , Testes de Função Renal , RNA/biossíntese , RNA/genética , Suínos , Fator de Necrose Tumoral alfa/biossíntese
16.
Mol Pharmacol ; 77(4): 660-9, 2010 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-20053955

RESUMO

Proliferation of pancreatic stellate cells (PSCs) plays a cardinal role during fibrosis development. Therefore, the suppression of PSC growth represents a therapeutic option for the treatment of pancreatic fibrosis. It has been shown that up-regulation of the enzyme heme oxygenase-1 (HO-1) could exert antiproliferative effects on PSCs, but no information is available on the possible role of carbon monoxide (CO), a catalytic byproduct of the HO metabolism, in this process. In the present study, we have examined the effect of CO releasing molecule-2 (CORM-2) liberated CO on PSC proliferation and have elucidated the mechanisms involved. Using primary rat PSCs, we found that CORM-2 inhibited PSC proliferation at nontoxic concentrations by arresting cells at the G(0)/G(1) phase of the cell cycle. This effect was associated with activation of p38 mitogen-activated protein kinase (MAPK) signaling, induction of HO-1 protein, and up-regulation of the cell cycle inhibitor p21(Waf1/Cip1). The p38 MAPK inhibitor 4-(4-flurophenyl)-2-(4-methylsulfinylphenyl)-5-(4-pyridyl)imidazole (SB203580) abolished the inhibitory effect of CORM-2 on PSC proliferation and prevented both CORM-2-induced HO-1 and p21(Waf1/Cip1) up-regulation. Treatment with tin protoporphyrin IX, an HO inhibitor, or transfection of HO-1 small interfering RNA abolished the inductive effect of CORM-2 on p21(Waf1/Cip1) and reversed the suppressive effect of CORM-2 on PSC growth. The ability of CORM-2 to induce cell cycle arrest was abrogated in p21(Waf1/Cip1)-silenced cells. Taken together, our results suggest that CORM-2 inhibits PSC proliferation by activation of the p38/HO-1 pathway. These findings may indicate a therapeutic potential of CO carriers in the treatment of pancreatic fibrosis.


Assuntos
Heme Oxigenase-1/fisiologia , Compostos Organometálicos/farmacologia , Pâncreas/efeitos dos fármacos , Transdução de Sinais/fisiologia , Proteínas Quinases p38 Ativadas por Mitógeno/fisiologia , Animais , Monóxido de Carbono/farmacologia , Ciclo Celular/efeitos dos fármacos , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Inibidor de Quinase Dependente de Ciclina p21/análise , Sistema de Sinalização das MAP Quinases , Masculino , Compostos Organometálicos/uso terapêutico , Pâncreas/citologia , Ratos , Ratos Wistar
17.
J Pharmacol Exp Ther ; 327(3): 863-71, 2008 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-18784349

RESUMO

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.


Assuntos
Proliferação de Células/efeitos dos fármacos , Proteína Quinase 3 Ativada por Mitógeno/metabolismo , Pâncreas/citologia , Transdução de Sinais/efeitos dos fármacos , Animais , Células Cultivadas , Curcumina/farmacologia , Heme Oxigenase-1/genética , Heme Oxigenase-1/fisiologia , Fator de Crescimento Derivado de Plaquetas/farmacologia , Ratos , Ativação Transcricional/efeitos dos fármacos
18.
Anesthesiology ; 108(6): 1025-36, 2008 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-18497603

RESUMO

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.


Assuntos
Antimetabólitos/farmacologia , Monóxido de Carbono/farmacologia , Ponte Cardiopulmonar , Citocinas/metabolismo , Inflamação/prevenção & controle , Pulmão/efeitos dos fármacos , Animais , Antimetabólitos/administração & dosagem , Apoptose/efeitos dos fármacos , Gasometria , Northern Blotting , Monóxido de Carbono/administração & dosagem , Carboxihemoglobina/análise , Carboxihemoglobina/metabolismo , Ponte Cardiopulmonar/efeitos adversos , Caspase 3/metabolismo , Ensaio de Imunoadsorção Enzimática , Inflamação/etiologia , Interleucina-10/metabolismo , Interleucina-1beta/metabolismo , Pulmão/citologia , Pulmão/metabolismo , Modelos Animais , Oxigênio/sangue , Suínos , Fator de Necrose Tumoral alfa/metabolismo
19.
Pancreas ; 36(4): 377-84, 2008 May.
Artigo em Inglês | MEDLINE | ID: mdl-18437084

RESUMO

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.


Assuntos
Heme Oxigenase-1/metabolismo , Microcirculação/fisiologia , Pâncreas/irrigação sanguínea , Pancreatite/enzimologia , Traumatismo por Reperfusão/enzimologia , Animais , Capilares/enzimologia , Endotélio Vascular/fisiologia , Endotélio Vascular/fisiopatologia , Leucócitos/fisiologia , Masculino , Microcirculação/efeitos dos fármacos , Pancreatite/induzido quimicamente , Protoporfirinas/toxicidade , RNA/genética , RNA/isolamento & purificação , Ratos , Ratos Sprague-Dawley
20.
Anesth Analg ; 106(4): 1150-60, table of contents, 2008 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-18349187

RESUMO

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.


Assuntos
Anestésicos Inalatórios/farmacologia , Ativação Enzimática/efeitos dos fármacos , Éteres Metílicos/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Apoptose/efeitos dos fármacos , Caspases/efeitos dos fármacos , Caspases/metabolismo , Desflurano , Humanos , Inflamação/fisiopatologia , Isoflurano/análogos & derivados , Isoflurano/farmacologia , Células Jurkat , Proteínas de Neoplasias/metabolismo , Sevoflurano
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...