Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 12 de 12
Filtrar
1.
Luminescence ; 39(4): e4719, 2024 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-38637113

RESUMO

The luminescence properties of erbium and yttrium co-doped cadmium difluoride with three different concentrations of yttrium were investigated. First, we synthesized single crystal samples with good optical quality using the Bridgman technique. From the optical absorption spectra, recorded at room temperature, both in the ultraviolet-visible and infrared spectral ranges, Judd-Ofelt analysis was performed based on yttrium concentrations to predict the radiative properties of Er3+ luminescent ions. For the 10% optimum concentration of yttrium, a detailed photoluminescence investigation was carried out. We mainly explored green, red, and near-infrared fluorescence under different excitation wavelengths and presented their highlight spectroscopic characteristics. The desired transitions had relatively high emission cross-sections both under visible and near-infrared excitation. Optical gain followed a similar trend. Furthermore, the dynamic fluorescence study showed a significant increase in the measured lifetime under an 800 nm infrared excitation. The upconversion process under an 800 nm excitation produced quantum efficiency greater than 100% due to the contribution of more than one energy transfer mechanism.


Assuntos
Érbio , Luminescência , Íons , Fluorescência , Érbio/química , Ítrio/química
2.
Cell Physiol Biochem ; 33(4): 967-81, 2014.
Artigo em Inglês | MEDLINE | ID: mdl-24714055

RESUMO

BACKGROUND: The cardioprotective effect of anaesthetic preconditioning as measured by reduction of ischaemia-reperfusion (I/R) injury is a well described phenomenon. However little is known about the impact on the myocardial proteome. We therefore investigated proteome dynamics at different experimental time points of a preconditioning protocol. METHODS: Using an in vivo rat model of desflurane-induced preconditioning (DES-PC) cardiac tissue proteomes were analysed by a gel-based comparative approach. Treatment-dependent protein alterations were assessed by intra-group comparisons. Proteins were identified by mass-spectrometry. RESULTS: A total of 40 protein spots were altered during the 30-minutes lasting preconditioning protocol. None of the proteins was differentially regulated consistently at all experimental time points. Interestingly, 1) the repeated administration of desflurane mostly accounted for proteome alterations during DES-PC, 2) the majority of altered protein species showed a decrease in abundance, 3) these changes primarily affected metabolic proteins involved in NADH/NAD(+) redox balance, calcium homeostasis and acidosis and 4) protein alterations were not exclusively due to expression changes but also represented modifications of specific protein isoforms. CONCLUSION: DES-PC evokes dynamic alterations in the cardiac proteome which substantiate a tight regulation of bioenergetic proteins. Unique protein modifications may play a more important role in the preconditioning response.


Assuntos
Anestésicos Inalatórios/farmacologia , Coração/efeitos dos fármacos , Precondicionamento Isquêmico Miocárdico , Isoflurano/análogos & derivados , Miocárdio/metabolismo , Proteoma/metabolismo , Animais , Cromatografia Líquida de Alta Pressão , Desflurano , Eletroforese em Gel Bidimensional , Isoflurano/farmacologia , Isoformas de Proteínas/análise , Isoformas de Proteínas/metabolismo , Ratos , Espectrometria de Massas por Ionização por Electrospray , Remodelação Ventricular/efeitos dos fármacos
3.
Anesth Analg ; 113(4): 730-7, 2011 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-21642605

RESUMO

BACKGROUND: Obesity is a significant risk factor for surgical site infections (SSIs), for poorly understood reasons. SSIs are a major cause of morbidity, prolonged hospitalization, and increased health care cost. Drug disposition in general is frequently altered in the obese. Preoperative antibiotic administration, achieving adequate tissue concentrations at the time of incision, is an essential strategy to prevent SSIs. Nonetheless, there is little information regarding antibiotic concentrations in obese surgical patients. This investigation tested the hypothesis that the prophylactic antibiotic cefoxitin may have delayed and/or diminished tissue penetration in the obese. METHODS: Plasma and tissue concentrations of cefoxitin were determined in obese patients undergoing abdominal and pelvic surgery (body mass index 43 ± 10 kg/m(2), n = 14, 2 g cefoxitin) and in normal-weight patients and healthy volunteers (body mass index 20 ± 2 kg/m(2), n = 13, 1 g cefoxitin). Tissue concentrations were measured using a microdialysis probe in the subcutaneous layer of the abdomen, and in adipose tissue excised at the time of incision and wound closure. RESULTS: Plasma concentrations and area under the concentration-time curve (AUC) were approximately 2-fold higher in the obese patients because of the 2-fold-higher dose. Dose-normalized concentrations were higher, although AUCs were not significantly different. Measured and dose-normalized subcutaneous cefoxitin concentrations and AUCs in the obese patients were significantly lower than in the normal-weight subjects. There was an inverse relationship between cefoxitin tissue penetration (AUC(tissue)/AUC(plasma) ratio) and body mass index. Tissue penetration was substantially lower in the obese patients (0.08 ± 0.07 vs 0.37 ± 0.26, P < 0.05). Adipose tissue cefoxitin concentrations in obese patients were only 7.8 ± 7.3 and 2.7 ± 1.4 µg/g, respectively, at incision and closure, below the minimum inhibitory concentration of 8 and 16 µg/mL, respectively, for aerobic and anaerobic microorganisms. CONCLUSION: Obese surgical patients have impaired tissue penetration of the prophylactic antibiotic cefoxitin, and inadequate tissue concentrations despite increased clinical dose (2 g). Inadequate tissue antibiotic concentrations may be a factor in the increased risk of SSIs in obese surgical patients. Additional studies are needed to define doses achieving adequate tissue concentrations.


Assuntos
Antibacterianos/farmacocinética , Antibioticoprofilaxia , Cefoxitina/farmacocinética , Obesidade/complicações , Procedimentos Cirúrgicos Operatórios/efeitos adversos , Infecção da Ferida Cirúrgica/prevenção & controle , Abdome/cirurgia , Tecido Adiposo/metabolismo , Tecido Adiposo/cirurgia , Adolescente , Adulto , Antibacterianos/administração & dosagem , Antibacterianos/sangue , Área Sob a Curva , Índice de Massa Corporal , Estudos de Casos e Controles , Cefoxitina/administração & dosagem , Cefoxitina/sangue , Feminino , Humanos , Masculino , Testes de Sensibilidade Microbiana , Microdiálise , Pessoa de Meia-Idade , Missouri , Obesidade/diagnóstico , Obesidade/metabolismo , Pelve/cirurgia , Medição de Risco , Fatores de Risco , Infecção da Ferida Cirúrgica/etiologia , Infecção da Ferida Cirúrgica/metabolismo , Distribuição Tecidual , Resultado do Tratamento , Adulto Jovem
4.
Anesthesiology ; 113(6): 1289-98, 2010 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-21068666

RESUMO

BACKGROUND: Sevoflurane induces preconditioning (SevoPC). The effect of aprotinin and the involvement of endothelial nitric-oxide synthase (NOS) on SevoPC are unknown. We investigated (1) whether SevoPC is strengthened by multiple preconditioning cycles, (2) whether SevoPC is blocked by aprotinin, and (3) whether endothelial NOS plays a crucial role in SevoPC. METHODS: Anesthetized male Wistar rats were randomized to 15 groups (each n = 6) and underwent 25-min regional myocardial ischemia and 2-h reperfusion. Controls were not treated further. Preconditioning groups inhaled 1 minimum alveolar concentration of sevoflurane for 5 min (SEVO-I), twice for 5 min each (SEVO-II), three times for 5 min each (SEVO-III), or six times for 5 min each (SEVO-VI). Aprotinin was administered with and without sevoflurane. Involvement of endothelial NOS was determined with the nonspecific NOS blocker N-nitro-l-arginine-methyl-ester, the specific neuronal NOS blocker 7-nitroindazole, and the specific inducible NOS blocker aminoguanidine. RESULTS: SevoPC reduced infarct size in all protocols (SEVO-I, 42 ± 6%; SEVO-II, 33 ± 4%; SEVO-III, 11 ± 5%; SEVO-VI, 16 ± 4%; all P < 0.001 vs. control, 67 ± 3%) and was least after three and six cycles of sevoflurane (P < 0.001 vs. SEVO-II and -I, respectively). Aprotinin alone had no effect on infarct size but blocked SevoPC. N-nitro-l-arginine-methyl-ester abolished SevoPC (67 ± 4%; P < 0.05 vs. SEVO-III). Aminoguanidine and 7-nitroindazole blocked SevoPC only partially (25 ± 6 and 31 ± 6%, respectively; P < 0.05 vs. SEVO-III and control). SevoPC induced endothelial NOS phosphorylation, which was abrogated by aprotinin. CONCLUSION: SevoPC is strengthened by multiple preconditioning cycles, and phosphorylation of endothelial NOS is a crucial step in mediating SevoPC. These effects are abolished by aprotinin.


Assuntos
Anestésicos Inalatórios/farmacologia , Aprotinina/farmacologia , Cardiotônicos , Hemostáticos/farmacologia , Precondicionamento Isquêmico Miocárdico , Éteres Metílicos/farmacologia , Infarto do Miocárdio/patologia , Miocárdio/enzimologia , Óxido Nítrico Sintase Tipo III/metabolismo , Anestésicos Inalatórios/administração & dosagem , Animais , Western Blotting , Relação Dose-Resposta a Droga , Inibidores Enzimáticos/farmacologia , Guanidinas/farmacologia , Hemodinâmica/efeitos dos fármacos , Masculino , Éteres Metílicos/administração & dosagem , Traumatismo por Reperfusão Miocárdica/patologia , Traumatismo por Reperfusão Miocárdica/prevenção & controle , NG-Nitroarginina Metil Éster/farmacologia , Óxido Nítrico Sintase Tipo III/antagonistas & inibidores , Fosforilação , Alvéolos Pulmonares/efeitos dos fármacos , Ratos , Ratos Wistar , Sevoflurano
5.
Artif Organs ; 30(12): 965-8, 2006 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-17181839

RESUMO

Major experimental surgery on laboratory animals requires adequate anesthesia and ventilation to keep the animal alive throughout the procedure. A ventilator is a machine that helps the anesthesized animal breathe through an endotracheal tube by pumping a volume of gas (oxygen, air, or other gaseous mixtures), comparable with the normal tidal volume, into the animal's lungs. There are two main categories of ventilators for small laboratory rodents: volume-controlled and pressure-controlled ones. The volume-controlled ventilator injects a preset volume into the animal's lungs, no matter the airways' resistance (with the peak inspiratory pressure allowed to vary), while the pressure ventilator controls the inspiratory pressure and allows the inspiratory volume to vary. Here we show a rat pressure ventilator with a simple expiratory valve that allows gas delivery through electronic expiration control and offers easy pressure monitoring and frequency change during ventilation.


Assuntos
Monitorização Fisiológica/instrumentação , Ventiladores Mecânicos , Animais , Desenho de Equipamento , Pressão , Troca Gasosa Pulmonar , Ventilação Pulmonar , Ratos
6.
J Am Assoc Lab Anim Sci ; 45(6): 54-7, 2006 Nov.
Artigo em Inglês | MEDLINE | ID: mdl-17089993

RESUMO

Experimental induction of ventricular fibrillation in animals yields valuable information about this deadly arrhythmia. Human adult or pediatric defibrillators and their paddles can be used easily in larger animals such as dogs and pigs, but these animals are more difficult to house and handle, and available biochemical assays may be limited. In contrast, rats are easy and relatively inexpensive to house and handle, and numerous biochemical tests are available. However, in most cases, even pediatric electrodes are impractical for use in rats. Proper placement of defibrillation electrodes on the thorax requires that the electrical axis of the heart be situated between the defibrillator paddles. The most common approach to defibrillation in rats uses 2 electrodes: one is built into a board that underlies and touches the rat's back, and another is positioned manually on the anterior thorax. The aim of this study was to produce electrodes that are 1) easy to handle, 2) specifically designed for rats, 3) efficiently deliver defibrillation shocks along the electric axis of the heart, and 4) can be used for both in vivo defibrillation and on isolated heart preparations.


Assuntos
Desfibriladores , Modelos Animais de Doenças , Cardioversão Elétrica/instrumentação , Ratos , Fibrilação Ventricular/terapia , Animais , Desenho de Equipamento , Masculino , Ratos Sprague-Dawley
7.
Eur J Pharmacol ; 539(1-2): 1-9, 2006 Jun 06.
Artigo em Inglês | MEDLINE | ID: mdl-16716295

RESUMO

Xenon elicits preconditioning of the myocardium via protein kinase C-epsilon. We determined the implication of (1) the mitochondrial adenosinetriphosphate dependent potassium (K(ATP)) channels and (2) the 3'phosphatidylinositol-dependent kinase-1 (PDK-1) in activating protein kinase C-epsilon. For infarct size measurements, anaesthetized rats were subjected to 25 min of coronary artery occlusion followed by 120 min of reperfusion. Rats received xenon 70% during three 5-min periods before ischaemia with or without the K(ATP) channel blocker 5-hydroxydecanoate or Wortmannin as PI3K/PDK-1 inhibitor. For Western blot, hearts were excised at five time points after xenon preconditioning (Control, 15, 25, 35, 45 min). Infarct size was reduced from 42+/-6% (mean+/-S.D.) to 27+/-8% after xenon preconditioning (P<0.05). Western blot revealed an increased activation of PKC-epsilon after 45 min and of PDK-1 after 25 min during xenon preconditioning. 5-hydroxydecanoate and Wortmannin blocked both effects. PKC-epsilon is activated downstream of mitochondrial K(ATP) channels and PDK-1. Both pathways are functionally involved in xenon preconditioning.


Assuntos
Trifosfato de Adenosina/fisiologia , Anestésicos Inalatórios/farmacologia , Precondicionamento Isquêmico Miocárdico , Miocárdio/enzimologia , Canais de Potássio/fisiologia , Proteína Quinase C-épsilon/fisiologia , Proteínas Serina-Treonina Quinases/fisiologia , Xenônio/farmacologia , Proteínas Quinases Dependentes de 3-Fosfoinositídeo , Animais , Ativação Enzimática , Masculino , Mitocôndrias Cardíacas/metabolismo , Infarto do Miocárdio/metabolismo , Infarto do Miocárdio/prevenção & controle , Miocárdio/patologia , Ratos , Ratos Wistar , Transdução de Sinais
8.
Anesthesiology ; 103(6): 1174-82, 2005 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-16306729

RESUMO

BACKGROUND: For nitrous oxide, a preconditioning effect on the heart has yet not been investigated. This is important because nitrous oxide is commonly used in combination with volatile anesthetics, which are known to precondition the heart. The authors aimed to clarify (1) whether nitrous oxide preconditions the heart, (2) how it affects protein kinase C (PKC) and tyrosine kinases (such as Src) as central mediators of preconditioning, and (3) whether isoflurane-induced preconditioning is influenced by nitrous oxide. METHODS: For infarct size measurements, anesthetized rats were subjected to 25 min of coronary artery occlusion followed by 120 min of reperfusion. Rats received nitrous oxide (60%), isoflurane (1.4%) or isoflurane-nitrous oxide (1.4%/60%) during three 5-min periods before index ischemia (each group, n = 7). Control animals remained untreated for 45 min. Additional hearts (control, 60% nitrous oxide alone%, and isoflurane-nitrous oxide [0.6%/60%, in equianesthetic doses]) were excised for Western blot of PKC-epsilon and Src kinase (each group, n = 4). RESULTS: Nitrous oxide had no effect on infarct size (59.1 +/- 15.2% of the area at risk vs. 51.1 +/- 10.9% in controls). Isoflurane (1.4%) and isoflurane-nitrous oxide (1.4%/60%) reduced infarct size to 30.9 +/- 10.6 and 28.7 +/- 11.8% (both P < 0.01). Nitrous oxide (60%) had no effect on phosphorylation (2.3 +/- 1.8 vs. 2.5 +/- 1.7 in controls, average light intensity, arbitrary units) and translocation (7.0 +/- 4.3 vs. 7.4 +/- 5.2 in controls) of PKC-epsilon. Src kinase phosphorylation was not influenced by nitrous oxide (4.6 +/- 3.9 vs. 5.0 +/- 3.8; 3.2 +/- 2.2 vs. 3.5 +/- 3.0). Isoflurane-nitrous oxide (0.6%/60%, in equianesthetic doses) induced PKC-epsilon phosphorylation (5.4 +/- 1.9 vs. 2.8 +/- 1.5; P < 0.001) and translocation to membrane regions (13.8 +/- 13.0 vs. 6.7 +/- 2.0 in controls; P < 0.05). CONCLUSIONS: Nitrous oxide is the first inhalational anesthetic without preconditioning effect on the heart. However, isoflurane-induced preconditioning and PKC-epsilon activation are not influenced by nitrous oxide.


Assuntos
Tonsila do Cerebelo/fisiologia , Anestésicos Inalatórios/farmacologia , Memória/efeitos dos fármacos , Éteres Metílicos/farmacologia , Animais , Aprendizagem da Esquiva/efeitos dos fármacos , Desflurano , Agonistas de Aminoácidos Excitatórios/toxicidade , Halotano/farmacologia , Hiperalgesia/induzido quimicamente , Hiperalgesia/psicologia , Isoflurano/análogos & derivados , Isoflurano/farmacologia , Masculino , N-Metilaspartato/toxicidade , Ratos , Ratos Sprague-Dawley , Sevoflurano
9.
Anesth Analg ; 101(4): 934-941, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16192499

RESUMO

UNLABELLED: Delta1-opioid receptor agonists can induce cardioprotection by early and late preconditioning (LPC). Morphine (MO) is commonly used for pain treatment during acute coronary syndromes. We investigated whether MO can induce myocardial protection by LPC and whether a nuclear transcription factor kappaB (NF-kappaB)-dependent intracellular signaling pathway is involved. Rats were subjected to 25 min of regional ischemia and 2 h of reperfusion 24 h after treatment with saline (NaCl; 0.9% 5 mL), lipopolysaccharide of Escherichia coli (LPS; 1 mg/kg), or MO (3 mg/kg). LPS is a trigger of LPC and served as positive control. Naloxone (NAL) was used to investigate the role of opioid receptors in LPC and was given before NaCl, LPS, or MO application (trigger phase) or before ischemia-reperfusion (mediator phase). Infarct size (percentage area at risk) was 59% +/- 9%, 51% +/- 6%, or 53% +/- 10% in the NaCl, NAL-NaCl, and NaCl-NAL groups, respectively. Pretreatment with MO reduced infarct size to 20% +/- 6% after 24 h (MO-24h), and this effect was abolished by NAL in the trigger (NAL-MO, 53% +/- 14%) and in the mediator (MO-NAL, 60% +/- 8%) phases. Pretreatment with LPS reduced infarct size to 23% +/- 8%. NAL administration in the trigger phase had no effect on infarct size (NAL-LPS 30% +/- 16%), whereas NAL during the mediator phase of LPC abolished the LPS-induced cardioprotection (LPS-NAL 54% +/- 8%). The role of NF-kappaB in morphine-induced LPC was investigated by Western blot and electrophoretic mobility shift assay. Morphine and LPS treatment increased phosphorylation of the inhibitory protein kappaB, leading to an increased activity of NF-kappaB. Thus, MO induces LPC similarly to LPS and it is likely that this cardioprotection is mediated at least in part by activation of NF-kappaB. Opioid receptors are involved as mediators in both MO- and LPS-induced LPC but as triggers only in MO-induced LPC. IMPLICATIONS: Like lipopolysaccharide, morphine induces late preconditioning and activation of nuclear transcription factor-kappaB. Opioid receptors are involved as mediators in both morphine- and lipopolysaccharide-induced late preconditioning but as triggers only in morphine-induced late preconditioning.


Assuntos
Coração/efeitos dos fármacos , Precondicionamento Isquêmico Miocárdico , Morfina/farmacologia , NF-kappa B/fisiologia , Receptores Opioides/fisiologia , Animais , Pressão Sanguínea/efeitos dos fármacos , Ensaio de Desvio de Mobilidade Eletroforética , Frequência Cardíaca/efeitos dos fármacos , Lipopolissacarídeos/farmacologia , Masculino , Infarto do Miocárdio/prevenção & controle , Fosforilação , Substâncias Protetoras/farmacologia , Ratos , Ratos Wistar
10.
Br J Pharmacol ; 146(3): 445-55, 2005 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-16086037

RESUMO

We previously demonstrated that the anesthetic gas xenon exerts cardioprotection by preconditioning in vivo via activation of protein kinase C (PKC)-epsilon and p38 mitogen-activated protein kinase (MAPK). P38 MAPK interacts with the actin cytoskeleton via the MAPK-activated protein kinase-2 (MAPKAPK-2) and heat-shock protein 27 (HSP27). The present study further elucidated the underlying molecular mechanism of xenon-induced preconditioning (Xe-PC) by focusing on a potential link of xenon to the cytoskeleton. Anesthetized rats received either xenon (Xe-PC, n = 6) or the volatile anesthetic isoflurane (Iso-PC, n = 6) during three 5-min periods interspersed with two 5-min and one final 10-min washout period. Control rats (n = 6) remained untreated for 45 min. Additional rats were either pretreated with the PKC inhibitor Calphostin C (0.1 mg kg(-1)) or with the p38 MAPK inhibitor SB203580 (1 mg kg(-1)) with and without anesthetic preconditioning (each, n = 6). Hearts were excised for immunohistochemistry of F-actin fibers and phosphorylated HSP27. Phosphorylation of MAPKAPK-2 and HSP27 were assessed by Western blot. HSP27 and actin colocalization were investigated by co-immunoprecipitation. Xe-PC induced phosphorylation of MAPKAPK-2 (control 1.0 +/- 0.2 vs Xe-PC 1.6 +/- 0.1, P < 0.05) and HSP27 (control 5.0 +/- 0.5 vs Xe-PC 9.8 +/- 1.0, P < 0.001). Both effects were blocked by Calphostin C and SB203580. Xe-PC enhanced translocation of HSP27 to the particulate fraction and increased F-actin polymerization. F-actin and pHSP27 were colocalized after Xe-PC. Xe-PC activates MAPKAPK-2 and HSP27 downstream of PKC and p38 MAPK. These data link Xe-PC to the cytoskeleton, revealing new insights into the mechanisms of Xe-PC in vivo.


Assuntos
Anestésicos Inalatórios/farmacologia , Proteínas de Choque Térmico/metabolismo , Precondicionamento Isquêmico Miocárdico , Proteínas de Neoplasias/metabolismo , Proteínas Serina-Treonina Quinases/metabolismo , Xenônio/farmacologia , Animais , Citoesqueleto/efeitos dos fármacos , Citoesqueleto/metabolismo , Proteínas de Choque Térmico HSP27 , Peptídeos e Proteínas de Sinalização Intracelular , Isoflurano/farmacologia , Masculino , Miocárdio/metabolismo , Fosforilação/efeitos dos fármacos , Ratos , Ratos Wistar , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
11.
Br J Pharmacol ; 144(1): 123-32, 2005 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-15644876

RESUMO

Xenon is an anesthetic with minimal hemodynamic side effects, making it an ideal agent for cardiocompromised patients. We investigated if xenon induces pharmacological preconditioning (PC) of the rat heart and elucidated the underlying molecular mechanisms. For infarct size measurements, anesthetized rats were subjected to 25 min of coronary artery occlusion followed by 120 min of reperfusion. Rats received either the anesthetic gas xenon, the volatile anesthetic isoflurane or as positive control ischemic preconditioning (IPC) during three 5-min periods before 25-min ischemia. Control animals remained untreated for 45 min. To investigate the involvement of protein kinase C (PKC) and p38 mitogen-activated protein kinase (MAPK), rats were pretreated with the PKC inhibitor calphostin C (0.1 mg kg(-1)) or the p38 MAPK inhibitor SB203580 (1 mg kg(-1)). Additional hearts were excised for Western blot and immunohistochemistry. Infarct size was reduced from 50.9+/-16.7% in controls to 28.1+/-10.3% in xenon, 28.6+/-9.9% in isoflurane and to 28.5+/-5.4% in IPC hearts. Both, calphostin C and SB203580, abolished the observed cardioprotection after xenon and isoflurane administration but not after IPC. Immunofluorescence staining and Western blot assay revealed an increased phosphorylation and translocation of PKC-epsilon in xenon treated hearts. This effect could be blocked by calphostin C but not by SB203580. Moreover, the phosphorylation of p38 MAPK was induced by xenon and this effect was blocked by calphostin C. In summary, we demonstrate that xenon induces cardioprotection by PC and that activation of PKC-epsilon and its downstream target p38 MAPK are central molecular mechanisms involved. Thus, the results of the present study may contribute to elucidate the beneficial cardioprotective effects of this anesthetic gas.


Assuntos
Anestésicos Inalatórios/farmacologia , Coração/fisiologia , Precondicionamento Isquêmico Miocárdico , Proteína Quinase C/metabolismo , Xenônio/farmacologia , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo , Animais , Western Blotting , Ativação Enzimática , Inibidores Enzimáticos/farmacologia , Técnica Direta de Fluorescência para Anticorpo , Coração/efeitos dos fármacos , Imidazóis/farmacologia , Imuno-Histoquímica , Isoflurano/farmacologia , Masculino , Microscopia Confocal , Reperfusão Miocárdica , Naftalenos/farmacologia , Proteína Quinase C/antagonistas & inibidores , Proteína Quinase C-épsilon , Piridinas/farmacologia , Ratos , Ratos Wistar
12.
Anesthesiology ; 101(6): 1372-80, 2004 Dec.
Artigo em Inglês | MEDLINE | ID: mdl-15564945

RESUMO

BACKGROUND: Activation of protein kinase C epsilon (PKC-epsilon) and extracellular signal-regulated kinase 1 and 2 (ERK1/2) are important for cardioprotection by preconditioning. The present study investigated the time dependency of PKC-epsilon and ERK1/2 activation during desflurane-induced preconditioning in the rat heart. METHODS: Anesthetized rats were subjected to regional myocardial ischemia and reperfusion, and infarct size was measured by triphenyltetrazoliumchloride staining (percentage of area at risk). In three groups, desflurane-induced preconditioning was induced by two 5-min periods of desflurane inhalation (1 minimal alveolar concentration), interspersed with two 10-min periods of washout. Three groups did not undergo desflurane-induced preconditioning. The rats received 0.9% saline, the PKC blocker calphostin C, or the ERK1/2 inhibitor PD98059 with or without desflurane preconditioning (each group, n = 7). Additional hearts were excised at four different time points with or without PKC or ERK1/2 blockade: without further treatment, after the first or the second period of desflurane-induced preconditioning, or at the end of the last washout phase (each time point, n = 4). Phosphorylated cytosolic PKC-epsilon and ERK1/2, and membrane translocation of PKC-epsilon were determined by Western blot analysis (average light intensity). RESULTS: Desflurane significantly reduced infarct size from 57.2 +/- 4.7% in controls to 35.2 +/- 16.7% (desflurane-induced preconditioning, mean +/- SD, P < 0.05). Both calphostin C and PD98059 abolished this effect (58.8 +/- 13.2% and 64.2 +/- 15.4% respectively, both P < 0.05 versus desflurane-induced preconditioning). Cytosolic phosphorylated PKC-epsilon reached its maximum after the second desflurane-induced preconditioning and returned to baseline after the last washout period. Both calphostin C and PD98059 inhibited PKC-epsilon activation. ERK1/2 phosphorylation reached its maximum after the first desflurane-induced preconditioning and returned to baseline after the last washout period. Calphostin C had no effect on ERK1/2 phosphorylation. CONCLUSIONS: Both, PKC and ERK1/2 mediate desflurane-induced preconditioning. PKC-epsilon and ERK1/2 are both activated in a time dependent manner during desflurane-induced preconditioning, but ERK1/2 activation during desflurane-induced preconditioning is not PKC dependent. Moreover, ERK1/2 blockade abolished PKC-epsilon activation, suggesting ERK-dependent activation of PKC-epsilon during desflurane-induced preconditioning.


Assuntos
Anestésicos Inalatórios/farmacologia , Coração/efeitos dos fármacos , Isoflurano/análogos & derivados , Isoflurano/farmacologia , Proteínas Quinases Ativadas por Mitógeno/metabolismo , Infarto do Miocárdio/induzido quimicamente , Miocárdio/enzimologia , Proteína Quinase C/metabolismo , Animais , Western Blotting , Membrana Celular/efeitos dos fármacos , Membrana Celular/enzimologia , Citosol/efeitos dos fármacos , Citosol/enzimologia , Desflurano , Ativação Enzimática/efeitos dos fármacos , Hemodinâmica/efeitos dos fármacos , Cinética , Masculino , Infarto do Miocárdio/patologia , Miocárdio/patologia , Fosforilação , Proteína Quinase C-épsilon , Ratos , Ratos Wistar
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA