Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
J Clin Med ; 11(13)2022 Jul 01.
Article in English | MEDLINE | ID: mdl-35807112

ABSTRACT

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.
JAMA ; 313(21): 2133-41, 2015 Jun 02.
Article in English | MEDLINE | ID: mdl-26024502

ABSTRACT

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.


Subject(s)
Acute Kidney Injury/prevention & control , Cardiac Surgical Procedures , Ischemic Preconditioning , Postoperative Complications/prevention & control , Acute-Phase Proteins/urine , Aged , Biomarkers/blood , Biomarkers/urine , Creatinine/blood , Female , Humans , Intensive Care Units , Length of Stay , Lipocalin-2 , Lipocalins/urine , Male , Middle Aged , Proto-Oncogene Proteins/urine , Renal Replacement Therapy/statistics & numerical data
3.
PLoS One ; 8(10): e77258, 2013.
Article in English | MEDLINE | ID: mdl-24167567

ABSTRACT

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.


Subject(s)
Hypnotics and Sedatives/pharmacology , Neurons/metabolism , Peptide Elongation Factor 2/metabolism , Protein Biosynthesis/drug effects , Thiopental/pharmacology , Animals , Brain Injuries/drug therapy , Brain Injuries/metabolism , Brain Injuries/pathology , Brain Ischemia/drug therapy , Brain Ischemia/metabolism , Brain Ischemia/pathology , Cell Death/drug effects , Cell Hypoxia/drug effects , Cell Line , Elongation Factor 2 Kinase/metabolism , Humans , Mice , Neurons/pathology , Oxygen/metabolism
4.
J Pharmacol Exp Ther ; 347(3): 781-93, 2013 Dec.
Article in English | MEDLINE | ID: mdl-24049063

ABSTRACT

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.


Subject(s)
Antithyroid Agents/pharmacology , Carbimazole/pharmacology , Cell Hypoxia/drug effects , Neurons/drug effects , Protein Synthesis Inhibitors , AMP-Activated Protein Kinases/metabolism , Adenosine Triphosphate/metabolism , Autoradiography , Blotting, Western , Calcium/metabolism , Caspases/metabolism , Cell Line , Cell Survival/drug effects , Cyclic AMP/metabolism , Elongation Factor 2 Kinase/metabolism , Energy Metabolism/drug effects , Humans , Phosphorylation
5.
PLoS One ; 7(9): e46479, 2012.
Article in English | MEDLINE | ID: mdl-23029526

ABSTRACT

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.


Subject(s)
Anti-Inflammatory Agents/administration & dosage , Apoptosis/drug effects , Carbon Monoxide/administration & dosage , Neuroprotective Agents/administration & dosage , Reperfusion Injury/drug therapy , Retinitis/prevention & control , Administration, Inhalation , Animals , Caspase 3/genetics , Caspase 3/metabolism , Cell Movement/drug effects , Enzyme Activation , Female , Gene Expression/drug effects , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Ischemia/drug therapy , Male , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Neuroglia/drug effects , Neuroglia/physiology , Phosphorylation , Protein Processing, Post-Translational/drug effects , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Rats , Rats, Sprague-Dawley , Retina/drug effects , Retina/pathology , Retinal Ganglion Cells/drug effects , Retinal Ganglion Cells/pathology , Retinal Vessels/pathology , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
6.
Pancreas ; 41(1): 74-83, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21934550

ABSTRACT

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.


Subject(s)
Cell Movement/drug effects , Cell Proliferation/drug effects , Hydrogen Sulfide/pharmacology , Pancreatic Stellate Cells/drug effects , Actins/genetics , Animals , Blotting, Western , Cells, Cultured , Collagen Type I/genetics , Extracellular Matrix/metabolism , Fibronectins/genetics , Gene Expression/drug effects , Heme Oxygenase-1/genetics , Heme Oxygenase-1/metabolism , Hydrogen Sulfide/metabolism , MAP Kinase Signaling System/drug effects , Male , Mitogen-Activated Protein Kinases/metabolism , Pancreatic Stellate Cells/metabolism , RNA Interference , Rats , Rats, Wistar , Reverse Transcriptase Polymerase Chain Reaction , Sulfides/metabolism , Sulfides/pharmacology
7.
Curr Pharm Biotechnol ; 13(6): 813-8, 2012 May.
Article in English | MEDLINE | ID: mdl-22201611

ABSTRACT

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.


Subject(s)
Carbon Monoxide/metabolism , Pancreas/metabolism , Animals , Carbon Monoxide/pharmacology , Heme Oxygenase (Decyclizing)/metabolism , Humans , Hyperglycemia/metabolism , Pancreatitis/metabolism , Reperfusion Injury/metabolism
8.
World J Gastroenterol ; 17(37): 4184-90, 2011 Oct 07.
Article in English | MEDLINE | ID: mdl-22072849

ABSTRACT

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.


Subject(s)
Anesthetics, Inhalation/pharmacology , Enzyme Induction , Fatty Liver/enzymology , Heme Oxygenase-1/metabolism , Isoflurane/pharmacology , Liver/drug effects , Liver/enzymology , Animals , Fatty Liver/genetics , Gene Expression , Glutathione Transferase/blood , Heme Oxygenase-1/genetics , Male , RNA, Messenger/genetics , RNA, Messenger/metabolism , Random Allocation , Rats , Rats, Zucker
9.
Eur J Pharmacol ; 670(1): 58-66, 2011 Nov 16.
Article in English | MEDLINE | ID: mdl-21925493

ABSTRACT

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.


Subject(s)
Carbon Dioxide/chemistry , Carbon Dioxide/pharmacology , Cytoprotection/drug effects , Heat-Shock Response/drug effects , Lung/cytology , Mitogen-Activated Protein Kinase 11/metabolism , Organometallic Compounds/chemistry , Animals , Apoptosis/drug effects , Cattle , Enzyme Activation/drug effects , Gene Silencing , Lung/drug effects , Lung/enzymology , Mitogen-Activated Protein Kinase 11/deficiency , Mitogen-Activated Protein Kinase 11/genetics , Staurosporine/pharmacology
10.
Mol Vis ; 17: 1275-86, 2011.
Article in English | MEDLINE | ID: mdl-21633713

ABSTRACT

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.


Subject(s)
Apoptosis/drug effects , Hydrogen Sulfide/administration & dosage , Ischemic Preconditioning/methods , Neuroprotective Agents/administration & dosage , Reperfusion Injury/physiopathology , Retinal Vessels/physiopathology , Administration, Inhalation , Animals , Caspase Inhibitors , Cell Death/drug effects , DNA/metabolism , Female , Glial Fibrillary Acidic Protein/metabolism , HSP90 Heat-Shock Proteins/metabolism , Male , Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Rats , Rats, Sprague-Dawley , Reperfusion Injury/pathology , Retina/metabolism , Retinal Ganglion Cells/drug effects , Retinal Vessels/drug effects , Time Factors , Tissue Distribution
11.
Anesth Analg ; 112(2): 282-91, 2011 Feb.
Article in English | MEDLINE | ID: mdl-21156982

ABSTRACT

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.


Subject(s)
Carbon Monoxide/administration & dosage , Cardiopulmonary Bypass/adverse effects , Ischemia/prevention & control , Lung Injury/prevention & control , Lung/drug effects , Administration, Inhalation , Animals , Apoptosis/drug effects , Biopsy , Caspase 3/metabolism , Disease Models, Animal , HSP70 Heat-Shock Proteins/metabolism , Hemodynamics/drug effects , Inflammation Mediators/metabolism , Interleukin-10/metabolism , Interleukin-6/metabolism , Ischemia/etiology , Ischemia/immunology , Ischemia/pathology , Ischemia/physiopathology , Lung/blood supply , Lung/immunology , Lung/pathology , Lung/physiopathology , Lung Injury/etiology , Lung Injury/immunology , Lung Injury/pathology , Lung Injury/physiopathology , Macrophages/drug effects , Macrophages/immunology , Swine , Time Factors , Tumor Necrosis Factor-alpha/metabolism
12.
Anesth Analg ; 111(1): 29-37, 2010 Jul.
Article in English | MEDLINE | ID: mdl-20519418

ABSTRACT

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.


Subject(s)
Carbon Monoxide/pharmacology , Cardiopulmonary Bypass/adverse effects , Heat-Shock Proteins/biosynthesis , Heat-Shock Response/drug effects , Heat-Shock Response/physiology , Kidney Diseases/etiology , Kidney Diseases/prevention & control , Acute Disease , Administration, Inhalation , Animals , Carbon Monoxide/administration & dosage , Carbon Monoxide/blood , Caspase 3/biosynthesis , Heme Oxygenase (Decyclizing)/biosynthesis , Hemodynamics/drug effects , Interleukin-6/biosynthesis , Kidney/pathology , Kidney Diseases/pathology , Kidney Function Tests , RNA/biosynthesis , RNA/genetics , Swine , Tumor Necrosis Factor-alpha/biosynthesis
13.
Mol Pharmacol ; 77(4): 660-9, 2010 Apr.
Article in English | MEDLINE | ID: mdl-20053955

ABSTRACT

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.


Subject(s)
Heme Oxygenase-1/physiology , Organometallic Compounds/pharmacology , Pancreas/drug effects , Signal Transduction/physiology , p38 Mitogen-Activated Protein Kinases/physiology , Animals , Carbon Monoxide/pharmacology , Cell Cycle/drug effects , Cell Proliferation/drug effects , Cells, Cultured , Cyclin-Dependent Kinase Inhibitor p21/analysis , MAP Kinase Signaling System , Male , Organometallic Compounds/therapeutic use , Pancreas/cytology , Rats , Rats, Wistar
14.
J Pharmacol Exp Ther ; 327(3): 863-71, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18784349

ABSTRACT

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.


Subject(s)
Cell Proliferation/drug effects , Mitogen-Activated Protein Kinase 3/metabolism , Pancreas/cytology , Signal Transduction/drug effects , Animals , Cells, Cultured , Curcumin/pharmacology , Heme Oxygenase-1/genetics , Heme Oxygenase-1/physiology , Platelet-Derived Growth Factor/pharmacology , Rats , Transcriptional Activation/drug effects
15.
Anesthesiology ; 108(6): 1025-36, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18497603

ABSTRACT

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.


Subject(s)
Antimetabolites/pharmacology , Carbon Monoxide/pharmacology , Cardiopulmonary Bypass , Cytokines/metabolism , Inflammation/prevention & control , Lung/drug effects , Animals , Antimetabolites/administration & dosage , Apoptosis/drug effects , Blood Gas Analysis , Blotting, Northern , Carbon Monoxide/administration & dosage , Carboxyhemoglobin/analysis , Carboxyhemoglobin/metabolism , Cardiopulmonary Bypass/adverse effects , Caspase 3/metabolism , Enzyme-Linked Immunosorbent Assay , Inflammation/etiology , Interleukin-10/metabolism , Interleukin-1beta/metabolism , Lung/cytology , Lung/metabolism , Models, Animal , Oxygen/blood , Swine , Tumor Necrosis Factor-alpha/metabolism
16.
Pancreas ; 36(4): 377-84, 2008 May.
Article in English | MEDLINE | ID: mdl-18437084

ABSTRACT

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.


Subject(s)
Heme Oxygenase-1/metabolism , Microcirculation/physiology , Pancreas/blood supply , Pancreatitis/enzymology , Reperfusion Injury/enzymology , Animals , Capillaries/enzymology , Endothelium, Vascular/physiology , Endothelium, Vascular/physiopathology , Leukocytes/physiology , Male , Microcirculation/drug effects , Pancreatitis/chemically induced , Protoporphyrins/toxicity , RNA/genetics , RNA/isolation & purification , Rats , Rats, Sprague-Dawley
17.
J Pharmacol Exp Ther ; 324(3): 1037-44, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18055877

ABSTRACT

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.


Subject(s)
Antithyroid Agents/pharmacology , I-kappa B Kinase/antagonists & inhibitors , NF-kappa B/antagonists & inhibitors , rac1 GTP-Binding Protein/antagonists & inhibitors , Antithyroid Agents/chemistry , Humans , I-kappa B Kinase/metabolism , Jurkat Cells , Leukocytes, Mononuclear/drug effects , Leukocytes, Mononuclear/metabolism , NF-kappa B/metabolism , Sulfhydryl Compounds/chemistry , Sulfhydryl Compounds/pharmacology , rac1 GTP-Binding Protein/metabolism
18.
Mol Pharmacol ; 72(6): 1647-56, 2007 Dec.
Article in English | MEDLINE | ID: mdl-17878268

ABSTRACT

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.


Subject(s)
Immunosuppressive Agents/pharmacology , NFATC Transcription Factors/antagonists & inhibitors , NFATC Transcription Factors/physiology , Signal Transduction/physiology , T-Lymphocytes/physiology , Thiobarbiturates/pharmacology , Transcription Factor AP-1/antagonists & inhibitors , Transcription Factor AP-1/physiology , Cell Proliferation/drug effects , Humans , Immunosuppressive Agents/chemistry , Jurkat Cells , Signal Transduction/drug effects , Structure-Activity Relationship , T-Lymphocytes/drug effects , Thiobarbiturates/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...