Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 19 de 19
Filtrar
1.
Anesth Analg ; 124(1): 52-60, 2017 01.
Artículo en Inglés | MEDLINE | ID: mdl-27861438

RESUMEN

BACKGROUND: Metoclopramide and domperidone are prokinetic and antiemetic substances often used in clinical practice. Although domperidone has a more favorable side effect profile and is considered the first-line agent, severe cardiac side effects were reported during the administration of both substances. Cardiac Na channels are common targets of therapeutics inducing cardiotoxicity. Therefore, the aim of this study was to investigate whether the differential cardiotoxicities of metoclopramide and domperidone correlate with the block of Na channels. METHODS: Effects of metoclopramide and domperidone on the human α-subunit Nav1.5 expressed in human embryonic kidney 293 cells and on Na currents in neonatal rat cardiomyocytes were investigated by means of whole-cell patch clamp recordings. RESULTS: Tonic block of resting Nav1.5 channels was more potent for domperidone (IC50 85 ± 25 µM; 95% confidence interval [CI], 36-134) compared with metoclopramide (IC50 458 ± 28 µM; 95% CI, 403-513). Both agents induced use-dependent block at 10 and 1 Hz, stabilized fast and slow inactivation, and delayed recovery from inactivation. However, metoclopramide induced considerably smaller effects compared with domperidone. Na currents in rat cardiomyocytes displayed tonic and use-dependent block by both substances, and in this system, domperidone (IC50 312 ± 15 µM; 95% CI, 22-602) and metoclopramide (IC50 250 ± 30 µM; 95% CI, 191-309) induced a similar degree of tonic block. CONCLUSIONS: Our data demonstrate that the clinically relevant cardiotoxicity of domperidone and metoclopramide corresponds to a rather potent and local anesthetic-like inhibition of cardiac Na channels including Nav1.5. These data suggest that Nav1.5 might be a hitherto unrecognized molecular mechanism of some cardiovascular side effects, for example, malignant arrhythmias of prokinetic and antiemetic agents.


Asunto(s)
Antieméticos/toxicidad , Domperidona/toxicidad , Metoclopramida/toxicidad , Miocitos Cardíacos/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.5/efectos de los fármacos , Sodio/metabolismo , Bloqueadores del Canal de Sodio Activado por Voltaje/toxicidad , Animales , Animales Recién Nacidos , Sitios de Unión , Cardiotoxicidad , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Potenciales de la Membrana , Miocitos Cardíacos/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/genética , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Ratas Sprague-Dawley , Factores de Tiempo , Transfección
2.
J Neurosci ; 35(3): 1125-35, 2015 Jan 21.
Artículo en Inglés | MEDLINE | ID: mdl-25609627

RESUMEN

Slack (Slo2.2) is a sodium-activated potassium channel that regulates neuronal firing activities and patterns. Previous studies identified Slack in sensory neurons, but its contribution to acute and chronic pain in vivo remains elusive. Here we generated global and sensory neuron-specific Slack mutant mice and analyzed their behavior in various animal models of pain. Global ablation of Slack led to increased hypersensitivity in models of neuropathic pain, whereas the behavior in models of inflammatory and acute nociceptive pain was normal. Neuropathic pain behaviors were also exaggerated after ablation of Slack selectively in sensory neurons. Notably, the Slack opener loxapine ameliorated persisting neuropathic pain behaviors. In conclusion, Slack selectively controls the sensory input in neuropathic pain states, suggesting that modulating its activity might represent a novel strategy for management of neuropathic pain.


Asunto(s)
Hiperalgesia/metabolismo , Proteínas del Tejido Nervioso/metabolismo , Neuralgia/metabolismo , Canales de Potasio/metabolismo , Células Receptoras Sensoriales/metabolismo , Animales , Calor , Hiperalgesia/genética , Hiperalgesia/fisiopatología , Ratones , Ratones Noqueados , Proteínas del Tejido Nervioso/genética , Neuralgia/fisiopatología , Dimensión del Dolor , Umbral del Dolor/fisiología , Física , Canales de Potasio/genética , Canales de potasio activados por Sodio
3.
J Mol Cell Cardiol ; 97: 140-9, 2016 08.
Artículo en Inglés | MEDLINE | ID: mdl-27180262

RESUMEN

Initiation of cardiac excitation depends on a specialized group of cardiomyocytes at the venous pole of the heart, the sinoatrial node (SAN). The T-box transcription factor gene Tbx18 is expressed in the SAN myocardium and is required for formation of a large portion of the pacemaker. Previous studies suggested that Tbx18 is also sufficient to reprogram ventricular cardiomyocytes into SAN cells in rat, guinea-pig and pig hearts. To evaluate the consequences of misexpression of Tbx18 for imposing a nodal phenotype onto chamber myocardial cells in fetal mice, we used two independent conditional approaches with chamber-specific cre driver lines and an Hprt(Tbx18) misexpression allele. Myh6-Cre/+;Hprt(Tbx18/y) mice developed dilated atria with thickened walls, reduced right ventricles and septal defects that resulted in reduced embryonic and post-natal survival. Tagln-Cre/+;Hprt(Tbx18/y) mice exhibited slightly smaller hearts with rounded trabeculae that supported normal embryonic survival. Molecular analyses showed that the SAN gap junction and ion channel profile was not ectopically induced in chamber myocardium but the working myocardial gene program was partially inhibited in atria and ventricles of both misexpression models. Left atrial expression of Pitx2 was strongly repressed in Myh6-Cre/+;Hprt(Tbx18/y) embryos. We conclude that exclusion of Tbx18 expression from the developing atria and (right) ventricle is important to achieve normal cardiac left-right patterning and myocardial differentiation, and that Tbx18 is not sufficient to induce full SAN differentiation of chamber cardiomyocytes in fetal mice.


Asunto(s)
Regulación del Desarrollo de la Expresión Génica , Corazón/embriología , Miocardio/metabolismo , Nodo Sinoatrial/metabolismo , Proteínas de Dominio T Box/genética , Transcriptoma , Animales , Biomarcadores , Análisis por Conglomerados , Femenino , Feto , Perfilación de la Expresión Génica , Genes Letales , Insuficiencia Cardíaca/genética , Insuficiencia Cardíaca/metabolismo , Insuficiencia Cardíaca/patología , Masculino , Ratones , Ratones Transgénicos , Miocardio/patología
4.
Anesthesiology ; 124(5): 1153-65, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-26859646

RESUMEN

BACKGROUND: The relatively membrane-impermeable lidocaine derivative QX-314 has been reported to permeate the ion channels transient receptor potential vanilloid 1 (TRPV1) and transient receptor potential cation channel, subfamily A, member 1 (TRPA1) to induce a selective inhibition of sensory neurons. This approach is effective in rodents, but it also seems to be associated with neurotoxicity. The authors examined whether the human isoforms of TRPV1 and TRPA1 allow intracellular entry of QX-314 to mediate sodium channel inhibition and cytotoxicity. METHODS: Human embryonic kidney 293 (HEK-293) cells expressing wild-type or mutant human (h) TRPV1 or TRPA1 constructs as well as the sodium channel Nav1.7 were investigated by means of patch clamp and ratiometric calcium imaging. Cytotoxicity was examined by flow cytometry. RESULTS: Activation of hTRPA1 by carvacrol and hTRPV1 by capsaicin produced a QX-314-independent reduction of sodium current amplitudes. However, permeation of QX-314 through hTRPV1 or hTRPA1 was evident by a concentration-dependent, use-dependent inhibition of Nav1.7 activated at 10 Hz. Five and 30 mM QX-314 activated hTRPV1 via mechanisms involving the intracellular vanilloid-binding domain and hTRPA1 via unknown mechanisms independent of intracellular cysteins. Expression of hTRPV1, but not hTRPA1, was associated with a QX-314-induced cytotoxicity (viable cells 48 ± 5% after 30 mM QX-314) that was ameliorated by the TRPV1 antagonist 4-(3-chloro-2-pyridinyl)-N-[4-(1,1-dimethylethyl)phenyl]-1-piperazinecarboxamide (viable cells 81 ± 5%). CONCLUSIONS: The study data demonstrate that QX-314 directly activates and permeates the human isoforms of TRPV1 and TRPA1 to induce inhibition of sodium channels, but also a TRPV1-dependent cytotoxicity. These results warrant further validation of this approach in more intact preparations and may be valuable for the development of this concept into clinical practice.


Asunto(s)
Anestésicos Locales/farmacología , Canales de Calcio/efectos de los fármacos , Supervivencia Celular/efectos de los fármacos , Lidocaína/análogos & derivados , Proteínas del Tejido Nervioso/efectos de los fármacos , Bloqueadores de los Canales de Sodio/farmacología , Canales Catiónicos TRPV/efectos de los fármacos , Canales de Potencial de Receptor Transitorio/efectos de los fármacos , Calcio/metabolismo , Relación Dosis-Respuesta a Droga , Células HEK293 , Humanos , Lidocaína/farmacología , Canal de Sodio Activado por Voltaje NAV1.7/efectos de los fármacos , Proteínas del Tejido Nervioso/agonistas , Canal Catiónico TRPA1 , Canales Catiónicos TRPV/agonistas , Canales de Potencial de Receptor Transitorio/agonistas
5.
Artículo en Alemán | MEDLINE | ID: mdl-25723604

RESUMEN

Patients with opioid pretreatment can be divided into different groups.While patients after successful drug addiction treatment with or without drug replacement therapy usually not require an extensive perioperative pain therapy, patients with persistent chronic pain and patients with an existing opioid addiction regularly are challenging for the anesthetist. Important pathophysiological issues among the patients include opioid tolerance, opioid-induced hyperalgesia (OIH) as well as acute withdrawal symptomes. Pharmakokinetic properties of the opioid seems to be crucial the manifestation of an acute withdrawal syndrome following opioid administration, and thus the use of remifentanil has frequently been reported to induce withdrawal symptoms. While all established anesthetic procedures can be applied, regional anesthetic techniques should be included whenever possible. A common misstake when treating patients with a history of opioid abuse is an unwarranted restraint in using opioids. In patients with a ongoing opioid abuse, it may be efficient to apply methadone or buprenorphine even prior to surgery. While pregabalin and gabapentin are first line therapeutics for treatment of neuropathic pain, they also seem to be effective co-analgesics in patients suffering from chronic pain and undergo surgery. A similar statement applies to clonidine and dexmedetomidine, which probably induce analgesia by activation of the descending antinociceptive noradrenergic system. The intraoperative administration of S-ketamine is recommended for patients who either already have developed opioid tolerance or suffer from neuropathic pain, and by which postoperative pain is high and was already shown to be poorly adjusted. Other therapeutic options such as intraoperative administration of magnesium or lidocaine may be promising approaches.


Asunto(s)
Trastornos Relacionados con Opioides/complicaciones , Atención Perioperativa/métodos , Anestesia , Anestésicos , Humanos , Hiperalgesia/inducido químicamente , Manejo del Dolor
6.
Anesth Analg ; 118(6): 1238-45, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24755846

RESUMEN

BACKGROUND: The synthetic cannabinoid ajulemic acid has been demonstrated to alleviate pain in patients suffering from chronic neuropathic pain. Cannabinoids interact with several molecules within the pain circuit, including a potent inhibition of voltage-gated sodium channels. In this study, we closely characterized this property on neuronal and nonneuronal sodium channels. METHODS: The inhibition of sodium inward currents by ajulemic acid was studied in vitro. Human embryonic kidney 293t cells were used as the expression system for Nav1.2, 1.3, 1.4, 1.5, 1.5N406K, 1.5F1760A, and 1.7; Nav1.8 was transiently expressed in ND7/23 cells. Nav1.2, Nav1.3, and Nav 1.8 were from rats, and Nav1.4, Nav1.5, and Nav1.7 were of human origin. Sodium currents were analyzed by means of the whole cell patch-clamp technique. The investigated concentrations of ajulemic acid were 0.1, 0.3, 1, 3, 10, and 30 µmol/L. RESULTS: Ajulemic acid reversibly and concentration-dependently inhibited all voltage-gated sodium channel (Nav) isoforms investigated in this study, including Nav1.2, 1.3, 1.4, 1.5, 1.7, and 1.8. Tonic block of resting channels yielded half-maximal inhibitory concentration values between 2 and 9 µmol/L and was strongly enhanced on inactivated channels, suggesting state-dependent inhibition by ajulemic acid. Tonic block did not differ significantly when comparing Nav1.2 and Nav1.3, Nav1.4 and Nav1.5, and Nav1.7 and Nav1.8. Statistical analysis of other combinations of subunits (e.g., Nav1.2 and Nav1.4) by analysis of variance yielded a significant difference in block. Although we did not observe any relevant use-dependent block, ajulemic acid induced a strong hyperpolarizing shift of the voltage dependency of fast inactivation and modest shift of slow inactivation. The local anesthetic-insensitive Nav1.5 constructs N406K and F1760A displayed a preserved sensitivity to block by ajulemic acid. Finally, we found that low concentrations of ajulemic acid efficiently inhibited Navß4 peptide-mediated resurgent currents in Nav1.5. CONCLUSIONS: Our data suggest that block of sodium channels can be a relevant mechanism by which ajulemic acid alleviates neuropathic pain. The potent inhibition of resurgent currents and the preserved block on local anesthetic-insensitive channels indicates that ajulemic acid interacts with a conserved but yet unknown site of sodium channels.


Asunto(s)
Dronabinol/análogos & derivados , Bloqueadores de los Canales de Sodio , Canales de Sodio Activados por Voltaje/efectos de los fármacos , Algoritmos , Anestésicos Locales/metabolismo , Sitios de Unión , Relación Dosis-Respuesta a Droga , Dronabinol/farmacología , Células HEK293 , Humanos , Técnicas de Placa-Clamp , Canales de Sodio Activados por Voltaje/genética
7.
Eur J Anaesthesiol ; 31(11): 640-5, 2014 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-25275343

RESUMEN

BACKGROUND: Intra-articular injection of local anaesthetics, opioids and corticosteroids is frequently used to obtain perioperative analgesia following joint surgery. Although local anaesthetics were shown to induce chondrotoxicity, the safety profile regarding chondrotoxicity of other injected drugs is less clear. OBJECTIVE: Our objective was to investigate cytotoxicity of drugs used for intra-articular analgesia. DESIGN: An experimental in-vitro study. SETTING: Hannover Medical School, science laboratory, 2013. MATERIAL: Human cartilage cell line T/C 28-a2. INTERVENTION: Incubation of cells with different concentrations of bupivacaine, s-ketamine, morphine and dexamethasone for 1 h. MAIN OUTCOME MEASURES: Fraction of Annexin V positive and Annexin V and propidium iodide double positive cells after 1 h of incubation with tested drug measured by flow cytometry. RESULTS: Both morphine (0.1 to 10 µmol l) and dexamethasone (10 to 1000 µmol/l) failed to induce cytotoxicity after 1 h of exposure. The previously reported chondrotoxicity of bupivacaine (10 to 500 µmol l or 2.8 to 140 µg ml) was confirmed by a concentration-dependent increased staining with Annexin V and propidium iodide. Exposure to S-ketamine (10 to 500 µmol l) induced a significant late apoptotic and necrotic cell fraction at 10 µmol l or 2.4 µg ml. Concentrations of 100 and 500 µmol l induced a significant increase in early apoptotic cells. CONCLUSION: Morphine and dexamethasone showed no cytotoxic effects in our study and might thus be better alternatives to the clinically frequently applied bupivacaine. S-ketamine induced an intensive dose-dependent cytotoxic effect and should probably be avoided for intra-articular injection.


Asunto(s)
Bupivacaína/toxicidad , Condrocitos/efectos de los fármacos , Dexametasona/toxicidad , Ketamina/toxicidad , Morfina/toxicidad , Anestésicos Locales/administración & dosificación , Anestésicos Locales/toxicidad , Bupivacaína/administración & dosificación , Línea Celular , Supervivencia Celular/efectos de los fármacos , Supervivencia Celular/fisiología , Condrocitos/patología , Dexametasona/administración & dosificación , Relación Dosis-Respuesta a Droga , Humanos , Inyecciones Intraarticulares , Ketamina/administración & dosificación , Morfina/administración & dosificación
8.
Anesth Analg ; 117(5): 1101-8, 2013 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-24029851

RESUMEN

BACKGROUND: Systemic administration of lipid emulsions is an established treatment for local anesthetic intoxication. However, it is unclear by which mechanisms lipids achieve this function. The high cardiac toxicity of the lipophilic local anesthetic bupivacaine probably results from a long-lasting inhibition of the cardiac Na channel Nav1.5. In this study, we sought to determine whether lipid emulsions functionally interact with Nav1.5 or counteract inhibition by bupivacaine. METHODS: Human embryonic kidney cells expressing human Nav1.5 were investigated by whole-cell patch clamp. The effects of Intralipid® and Lipofundin® were explored on functional properties and on bupivacaine-induced inhibition. RESULTS: Intralipid and Lipofundin did not affect the voltage dependency of activation, but induced a small hyperpolarizing shift of the steady-state fast inactivation and impaired the recovery from fast inactivation. Lipofundin, but not Intralipid, induced a concentration-dependent but voltage-independent tonic block (42% ± 4% by 3% Lipofundin). The half-maximal inhibitory concentration (IC50) values for tonic block by bupivacaine (50 ± 4 µM) were significantly increased when lipids were coapplied (5% Intralipid: 196 ± 22 µM and 5% Lipofundin: 103 ± 8 µM). Use-dependent block by bupivacaine at 10 Hz was also reduced by both lipid emulsions. Moreover, the recovery of inactivated channels from bupivacaine-induced block was faster in the presence of lipids. CONCLUSIONS: Our data indicate that lipid emulsions reduce rather than increase availability of Nav1.5. However, both Intralipid and Lipofundin partly relieve Nav1.5 from block by bupivacaine. These effects are likely to involve not only a direct interaction of lipids with Nav1.5 but also the ability of lipid emulsions to absorb bupivacaine and thus reduce its effective concentration.


Asunto(s)
Anestésicos Locales/efectos adversos , Anestésicos Locales/farmacología , Bupivacaína/efectos adversos , Emulsiones Grasas Intravenosas/farmacología , Canal de Sodio Activado por Voltaje NAV1.5/efectos de los fármacos , Fosfolípidos/farmacología , Sorbitol/farmacología , Aceite de Soja/farmacología , Absorción , Relación Dosis-Respuesta a Droga , Combinación de Medicamentos , Electrofisiología , Emulsiones/farmacología , Células HEK293 , Humanos , Concentración 50 Inhibidora , Riñón/efectos de los fármacos , Lípidos/química , Técnicas de Placa-Clamp , Bloqueadores de los Canales de Sodio/farmacología
9.
Schweiz Monatsschr Zahnmed ; 123(10): 883-94, 2013.
Artículo en Francés, Alemán | MEDLINE | ID: mdl-24203487

RESUMEN

INTRODUCTION: Even if a mortal incident occurs rarely in a dentists daily routine there is a chance of it. The entire team has to face a great challenge then. Stress and incertainty are leading to mistakes and cost precious time which is not given for treating due to emergency. MATERIAL: Usual emergencies helped developing emergency kits which should make it easy for the practise team to act right and aim orientated in case of emergency. Therefore the combination for an as much efficient as possible state of those kits has priority. Through these the acting procedures turn out easier and more secure. RESULT: The management in case of emergency could be highly improved through the developing of emergency kits. The acting procedures turn out easier and more secure. The possibility for mistakes decreases.


Asunto(s)
Atención Odontológica , Tratamiento de Urgencia , Equipos y Suministros , Eficiencia , Humanos , Preparaciones Farmacéuticas
10.
J Exp Pharmacol ; 14: 353-365, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-36385942

RESUMEN

Introduction: Chloroquine (CQ) and its derivate hydroxychloroquine (HCQ) are successfully deployed for different diseases beyond the prophylaxis and treatment of malaria. Both substances exhibit antiviral properties and have been proposed for prophylaxis and treatment of COVID-19 caused by SARS-CoV-2. CQ and HCQ cause similar adverse events including life-threatening cardiac arrhythmia generally based on QT-prolongation, which is one of the most reported adverse events for both agents associated with the treatment of COVID-19. Various drugs known to induce QT-prolongation have been proven to exert local anesthetic (LA)-like properties regarding their impact on the cardiac Na+ channel Nav1.5. Inhibition of Nav1.5 is considered as the primary mechanism of cardiotoxicity caused by LAs. However, the mechanism of the arrhythmogenic effects of CQ and HCQ related to Nav1.5 has not yet been fully investigated. Therefore, the exact mechanism of how CQ and HCQ affect the sodium currents generated by Nav1.5 need to be further elucidated. Objective: This in vitro study aims to investigate the effects of CQ and HCQ on Nav1.5-generated sodium currents to identify possible LA-like mechanisms that might contribute to their arrhythmogenic properties. Methods: The effects of CQ and HCQ on Nav1.5-generated sodium currents by HEK-293 cells expressing either wild-type human Nav1.5 or mutant Nav1.5 F1760A are measured using the whole-cell patch-clamp technique. Results: Both agents induce a state-dependent inhibition of Nav1.5. Furthermore, CQ and HCQ produce a use-dependent block of Nav1.5 and a shift of fast and slow inactivation. Results of experiments investigating the effect on the LA-insensitive mutant Nav1.5-F1760A indicate that both agents at least in part employ the proposed LA-binding site of Nav1.5 to induce inhibition. Conclusion: This study demonstrated that CQ and HCQ exert LA-typical effects on Nav1.5 involving the proposed LA binding site, thus contributing to their arrhythmogenic properties.

11.
J Immunol ; 181(2): 1120-7, 2008 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-18606664

RESUMEN

Chemokines mediate the recruitment of leukocytes to the sites of inflammation. N-terminal truncation of chemokines by the protease dipeptidyl peptidase IV (DPPIV) potentially restricts their activity during inflammatory processes such as allergic reactions, but direct evidence in vivo is very rare. After demonstrating that N-terminal truncation of the chemokine CCL11/eotaxin by DPPIV results in a loss of CCR3-mediated intracellular calcium mobilization and CCR3 internalization in human eosinophils, we focused on the in vivo role of CCL11 and provide direct evidence for specific kinetic and rate-determining effects by DPPIV-like enzymatic activity on CCL11-mediated responses of eosinophils. Namely, it is demonstrated that i.v. administration of CCL11 in wild-type F344 rats leads to mobilization of eosinophils into the blood, peaking at 30 min. This mobilization is significantly increased in DPPIV-deficient F344 rats. Intradermal administration of CCL11 is followed by a dose-dependent recruitment of eosinophils into the skin and is significantly more effective in DPPIV-deficient F344 mutants as well as after pharmacological inhibition of DPPIV. Interestingly, CCL11 application leads to an up-regulation of DPPIV, which is not associated with negative feedback inhibition via DPPIV-cleaved CCL11((3-74)). These findings demonstrate regulatory effects of DPPIV for the recruitment of eosinophils. Furthermore, they illustrate that inhibitors of DPPIV have the potential to interfere with chemokine-mediated effects in vivo including but not limited to allergy.


Asunto(s)
Quimiocina CCL11/inmunología , Quimiotaxis de Leucocito , Dipeptidil Peptidasa 4/metabolismo , Eosinófilos/inmunología , Animales , Quimiocina CCL11/metabolismo , Dipeptidil Peptidasa 4/inmunología , Inhibidores de la Dipeptidil-Peptidasa IV , Regulación hacia Abajo , Inhibidores Enzimáticos/farmacología , Eosinófilos/metabolismo , Humanos , Isoleucina/análogos & derivados , Isoleucina/farmacología , Ratas , Ratas Endogámicas F344 , Ratas Mutantes , Receptores CCR3/metabolismo , Piel/inmunología , Linfocitos T/inmunología , Linfocitos T/metabolismo , Tiazoles/farmacología
12.
Neurosci Lett ; 661: 5-10, 2017 Nov 20.
Artículo en Inglés | MEDLINE | ID: mdl-28939386

RESUMEN

Proton-evoked activation of sensory neurons is counteracted by inhibition of voltage-gated Na+ channels, and the low acid-sensitivity of sensory neuron of the African naked mole-rat (ANMr) was reported to be due to a strong proton-evoked block of ANMrNav1.7. Here we aimed to reevaluate the role of the suggested negatively-charged motif in the ANMrNav1.7 domain IV P-loop for inhibition by protons. Patch clamp recordings were performed on the recombinant α-subunits Nav1.2-1.8. The insertion of the negatively charged motif (EKE) of ANMrNav1.7 into human Nav1.7 results in an increased proton-evoked tonic inhibition, but also in a reduced channel function. While the voltage-dependency of fast inactivation is changed in hNav1.7-EKE, pH 6.4 fails to induce a significant shift in both constructs. Proton-evoked inhibition of other channel α-subunits reveals a discrete differential inhibition among α-subunits with hNav1.7 displaying the lowest proton-sensitivity. The mutant hNav1.7-EKE displays a similar proton-sensitivity as Nav1.2, Nav1.3, Nav1.6 and Nav1.8. Overall, a correlation between proton-evoked inhibition and motif charge was not evident. Accordingly, a homology model of hNav1.7 shows that the EKE motif residues do not contribute to the pore lumen. Our data confirms that a negative charge of a postulated proton-motif encodes for a high proton-sensitivity when inserted into hNav1.7. However, a negatively charged motif is not a reliable predictor for a high proton-sensitivity in other α-subunits. Given the distance of the proton-motif from the pore mouth it seems unlikely that a blocking mechanism involving direct obstruction of the pore underlies the observed proton-evoked channel inhibition.


Asunto(s)
Canales de Sodio Activados por Voltaje/metabolismo , Células Cultivadas , Potenciales Evocados/efectos de los fármacos , Espacio Extracelular/efectos de los fármacos , Espacio Extracelular/metabolismo , Humanos , Potenciales de la Membrana/efectos de los fármacos , Técnicas de Placa-Clamp/métodos , Protones , Bloqueadores de los Canales de Sodio/farmacología , Canales de Sodio Activados por Voltaje/efectos de los fármacos
13.
Reg Anesth Pain Med ; 42(4): 462-468, 2017.
Artículo en Inglés | MEDLINE | ID: mdl-28394849

RESUMEN

BACKGROUND AND OBJECTIVES: Regional anesthesia includes application of local anesthetics (LAs) into the vicinity of peripheral nerves. Prolongation or improvement of nerve blocks with LAs can be accomplished by coapplication with adjuvants, including buprenorphine, ketamine, and clonidine. While the mechanisms mediating prolonged or improved LA-induced effects by adjuvants are poorly understood, we hypothesized that they are likely to increase LA-induced block of voltage-gated Na channels. In this study, we investigated the inhibitory effects of the LA bupivacaine alone and in combination with the adjuvants on neuronal Na channels. METHODS: Effects of bupivacaine, buprenorphine, ketamine, and clonidine on endogenous Na channels in ND7/23 neuroblastoma cells were investigated with whole-cell patch clamp. RESULTS: Bupivacaine, buprenorphine, ketamine, and clonidine are concentration- and state-dependent inhibitors of Na currents in ND7/23 cells. Tonic block of resting channels revealed an order of potency of bupivacaine (half-maximal inhibitory concentration [IC50] 178 ± 8 µM) > buprenorphine (IC50 172 ± 25) > clonidine (IC50 824 ± 55 µM) > ketamine (IC50 1377 ± 92 µM). Bupivacaine and buprenorphine, but not clonidine and ketamine, induced a strong use-dependent block at 10 Hz. Except for clonidine, all substances enhanced fast and slow inactivation. The combination of bupivacaine with one of the adjuvants resulted in a concentration-dependent potentiation bupivacaine-induced block. CONCLUSIONS: We demonstrate that buprenorphine, ketamine, and clonidine directly inhibit Na channels and that they potentiate the blocking efficacy of bupivacaine on Na channels. These data indicate that block of Na channels may account for the additive effects of adjuvants used for regional anesthesia.


Asunto(s)
Potenciales de Acción/efectos de los fármacos , Bupivacaína/administración & dosificación , Buprenorfina/administración & dosificación , Clonidina/administración & dosificación , Ketamina/administración & dosificación , Bloqueadores del Canal de Sodio Activado por Voltaje/administración & dosificación , Potenciales de Acción/fisiología , Anestésicos Locales/administración & dosificación , Línea Celular Tumoral , Relación Dosis-Respuesta a Droga , Sinergismo Farmacológico , Humanos
14.
Naunyn Schmiedebergs Arch Pharmacol ; 389(3): 315-25, 2016 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-26667357

RESUMEN

Propofol and dexmedetomidine are very commonly used sedative agents. However, several case reports demonstrated cardiovascular adverse effects of these two sedatives. Both substances were previously demonstrated to quite potently inhibit neuronal voltage-gated Na(+) channels. Thus, a possible molecular mechanism for some of their cardiac side effects is an inhibition of cardiac voltage gated Na(+) channels. In this study, we therefore explored the effects of propofol and dexmedetomidine on the cardiac predominant Na(+) channel α-subunit Nav1.5. Effects of propofol and dexmedetomidine were investigated on constructs of the human α-subunit Nav1.5 stably expressed in HEK-293 cells by means of whole-cell patch clamp recordings. Both agents induced a concentration-dependent tonic inhibition of Nav1.5. The calculated IC50 value for propofol was 228 ± 10 µM, and for dexmedetomidine 170 ± 20 µM. Tonic block only marginally increased on inactivated channels, and a weak use-dependent block at 10 Hz was observed for dexmedetomidine (16 ± 2 % by 100 µM). The voltage dependencies of fast and slow inactivation as well as the time course of recovery from inactivation were shifted by both propofol and dexmedetomidine. Propofol (IC50 126 ± 47 µM) and dexmedetomidine (IC50 182 ± 27 µM) blocked the persistent sodium current induced by veratradine. Finally, the local-anesthetic (LA)-insensitive mutant Nav1.5-F1760A exhibited reduced tonic and use-dependent block by both substances. Dexmedetomidine was generally more potent as compared to propofol. Propofol and dexmedetomidine seem to interact with the LA-binding site to inhibit the cardiac Na(+) channel Nav1.5 in a state-dependent manner. These data suggest that Nav1.5 is a hitherto unrecognized molecular component of some cardiovascular side effects of these sedative agents.


Asunto(s)
Dexmedetomidina/farmacología , Canal de Sodio Activado por Voltaje NAV1.5/fisiología , Propofol/farmacología , Bloqueadores de los Canales de Sodio/farmacología , Células HEK293 , Humanos , Hipnóticos y Sedantes/farmacología , Canal de Sodio Activado por Voltaje NAV1.5/genética , Técnicas de Placa-Clamp
15.
Eur J Pharmacol ; 789: 119-126, 2016 Oct 15.
Artículo en Inglés | MEDLINE | ID: mdl-27401036

RESUMEN

5-hydroxytryptamine 3 receptor (5-HT3 receptor) antagonists are administered for prevention and therapy of nausea and vomiting. Although regarded as safe therapeutics, they can also provoke arrhythmias by prolonging the QRS interval. However, the mechanisms mediating this cardiotoxicity are poorly understood. Here we investigated effects of 5-HT3 receptor antagonists on the cardiac Na(+) channel Nav1.5. We explored the interaction of dolasetron, tropisetron, granisetron and ondansetron on the human α-subunit Nav1.5 heterologously expressed in HEK293 cells. Sodium currents were explored by means of whole-cell patch clamp recordings. All four substances inhibited the Nav1.5 in a concentration and state-dependent manner. Dolasetron displayed the lowest blocking efficacy, and tropisetron was the most potent blocker with a half maximum blocking concentration of 18µM for tonic block of inactivated channels. Tropisetron was also the most potent use-dependent inhibitor, and it also induced a strong open -channel block. Both tonic and use-dependent block by tropisetron were abbreviated on the local-anesthetic insensitive mutant Nav1.5-F1760A. Co-administration of tropisetron and the local anesthetic bupivacaine or the hypnotic propofol augmented inhibition of Nav1.5. Our data demonstrate that 5-HT3 receptor antagonists induce a local-anesthetic like inhibition of Nav1.5, and that they display different blocking efficacies. Reports on a relevant cardiotoxicity of dolasetron as opposed to other 5-HT3 receptor antagonists do not seem to correlate with a block of Nav1.5. As inhibition of Nav1.5 was enhanced by propofol and bupivacaine however, it is possible that a combined administration of Na(+) channel blockers and 5-HT3 receptor antagonists can provoke arrhythmias.


Asunto(s)
Anestésicos Locales/farmacología , Miocardio/metabolismo , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Receptores de Serotonina 5-HT3/metabolismo , Antagonistas del Receptor de Serotonina 5-HT3/farmacología , Bloqueadores de los Canales de Sodio/farmacología , Sinergismo Farmacológico , Células HEK293 , Humanos , Mutación , Canal de Sodio Activado por Voltaje NAV1.5/genética , Neuronas/efectos de los fármacos , Neuronas/metabolismo , Sodio/metabolismo
16.
Eur J Pharmacol ; 783: 1-10, 2016 Jul 15.
Artículo en Inglés | MEDLINE | ID: mdl-27130441

RESUMEN

Duloxetine and venlafaxine are selective serotonin-norepinephrine-reuptake-inhibitors used as antidepressants and co-analgesics. While venlafaxine rather than duloxetine induce cardiovascular side-effects, neither of the substances are regarded cardiotoxic. Inhibition of cardiac Na(+)-channels can be associated with cardiotoxicity, and duloxetine was demonstrated to block neuronal Na(+)-channels. The aim of this study was to investigate if the non-life threatening cardiotoxicities of duloxetine and venlafaxine correlate with a weak inhibition of cardiac Na(+)-channels. Effects of duloxetine, venlafaxine and amitriptyline were examined on endogenous Na(+)-channels in neuroblastoma ND7/23 cells and on the α-subunits Nav1.5, Nav1.7 and Nav1.8 with whole-cell patch clamp recordings. Tonic block of the cardiac Na(+)-channel Nav1.5 and rat-cardiomyocytes (CM) revealed a higher potency for duloxetine (Nav 1.5 IC50 14±1µM, CM IC50 27±3µM) as compared to venlafaxine (Nav 1.5 IC50 671±26µM, CM IC50 452±34µM). Duloxetine was as potent as the cardiotoxic antidepressant amitriptyline (IC50 13±1µM). While venlafaxine almost failed to induce use-dependent block on Nav1.5 and cardiomyocytes, low concentrations of duloxetine (1, 10µM) induced prominent use-dependent block similar to amitriptyline. Duloxetine, but not venlafaxine stabilized fast and slow inactivation and delayed recovery from inactivation. Duloxetine induced an unselective inhibition of neuronal Na(+)-channels (IC50 ND7/23 23±1µM, Nav1.7 19±2µM, Nav1.8 29±2). Duloxetine, but not venlafaxine inhibits cardiac Na(+)-channels with a potency similar to amitriptyline. These data indicate that an inhibition of Na(+)-channels does not predict a clinically relevant cardiotoxicity.


Asunto(s)
Clorhidrato de Duloxetina/efectos adversos , Ventrículos Cardíacos/efectos de los fármacos , Canal de Sodio Activado por Voltaje NAV1.5/metabolismo , Neuronas/efectos de los fármacos , Inhibidores de Captación de Serotonina y Norepinefrina/efectos adversos , Bloqueadores de los Canales de Sodio/efectos adversos , Clorhidrato de Venlafaxina/efectos adversos , Animales , Células HEK293 , Ventrículos Cardíacos/citología , Humanos , Cinética , Miocitos Cardíacos/efectos de los fármacos , Miocitos Cardíacos/metabolismo , Neuronas/metabolismo , Ratas
17.
Lipids ; 51(11): 1241-1248, 2016 11.
Artículo en Inglés | MEDLINE | ID: mdl-27518386

RESUMEN

Lipid emulsions influence platelet aggregation and receptor expression. However, the effect on platelet function is not fully explained. Therefore, the aim of this study was to examine the influence of the lipids Lipofundin®, Lipidem® and ClinOleic® on surface expressions of P-selectin, GPIb and GPIIb/IIIa on platelets in vitro. Whole blood was incubated in two different concentrations (0.06 and 0.6 mg/ml) of LCT/MCT, n-3/LCT/MCT and LCT-MUFA for 30 min, followed by activation with TRAP-6 or ADP for flow-cytometric assay. Rates of P-selectin, GPIb and GPIIb/IIIa expression were analyzed. There was a significant increase in GPIIb/IIIa- and P-selectin-expression after incubation with LCT/MCT and n-3/LCT/MCT at the concentration of 0.6 mg/ml, without and after stimulation with TRAP-6 and ADP. GPIb was significantly decreased. Accordingly, LCT-MUFA had no effect on receptor expression of platelets in vitro. We demonstrated that LCT-MUFA did not activate receptor expression of platelets whereas LCT/MCT significantly increased platelet aggregation in vitro. This finding should be noted for parenteral nutrition of intensive care patients and, in the future, might provide further insight into the pathogenic pathways of acute thromboembolic events. However, prospectively designed clinical studies are needed to support our results.


Asunto(s)
Plaquetas/efectos de los fármacos , Emulsiones Grasas Intravenosas/farmacología , Selectina-P/metabolismo , Fosfolípidos/farmacología , Aceites de Plantas/farmacología , Complejo GPIIb-IIIa de Glicoproteína Plaquetaria/metabolismo , Complejo GPIb-IX de Glicoproteína Plaquetaria/metabolismo , Sorbitol/farmacología , Aceite de Soja/farmacología , Plaquetas/citología , Plaquetas/metabolismo , Combinación de Medicamentos , Emulsiones Grasas Intravenosas/química , Humanos , Aceite de Oliva/química , Aceite de Oliva/farmacología , Fosfolípidos/química , Aceites de Plantas/química , Agregación Plaquetaria/efectos de los fármacos , Sorbitol/química , Aceite de Soja/química , Triglicéridos/química , Triglicéridos/farmacología
18.
J Emerg Trauma Shock ; 7(2): 97-101, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24812454

RESUMEN

INTRODUCTION: The two-shot technique is an effective stopping power method. The precise mechanisms of action on the bone and soft-tissue structures of the skull; however, remain largely unclear. The aim of this study is to compare the terminal ballistics of the two-shot and single-shot techniques. MATERIALS AND METHODS: 40 fresh pigs' heads were randomly divided into 4 groups (n = 10). Either a single shot or two shots were fired at each head with a full metal jacket or a semi-jacketed bullet. Using thin-layer computed tomography and photography, the diameter of the destruction pattern and the fractures along the bullet path were then imaged and assessed. RESULTS: A single shot fired with a full metal jacket bullet causes minor lateral destruction along the bullet path. With two shots fired with a full metal jacket bullet, however, the maximum diameter of the bullet path is significantly greater (P < 0.05) than it is with a single shot fired with a full metal jacket bullet. In contrast, the maximum diameter with a semi-jacketed bullet is similar with the single-shot and two-shot techniques. CONCLUSION: With the two-shot technique, a full metal jacket bullet causes a destruction pattern that is comparable to that of a single shot fired with a semi-jacketed bullet.

19.
Naunyn Schmiedebergs Arch Pharmacol ; 387(10): 991-1000, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25012093

RESUMEN

Amylmetacresol and dichloro-benzylalcohol are ingredients of lozenges used for the treatment of sore throat. In a former in vitro study, a local anaesthetic-like effect of these substances has been described. Since amylmetacresol and dichloro-benzylalcohol are co-administered in over-the-counter lozenges, the intention of this study is to evaluate the in vitro effects of the combination of these compounds on the voltage-gated sodium channel. We analysed the block of inward sodium currents induced by the combination of amylmetacresol, dichloro-benzylalcohol and the local anaesthetic lidocaine. Tonic and use-dependent block and effects on the inactivated channel state of the neuronal sodium channel were examined. Therefore, the α-subunit of the voltage-gated NaV1.2 sodium channel was heterologously expressed in HEK 293 cells in vitro. Inward sodium currents were investigated in the whole-cell configuration of the patch-clamp technique. The combination of amylmetacresol and dichloro-benzylalcohol and the combination of amylmetacresol and lidocaine induced a block of resting and inactivated sodium channels both displaying a pronounced block at the inactivated channel state. In addition, the combination of all three compounds also resulted in a voltage-dependent block of inward sodium currents. While use-dependent block by co-application of amylmetacresol and dichloro-benzylalcohol was moderate (<20 %), lidocaine and amylmetacresol induced a robust use-dependent block (up to 50 %). This study demonstrates local anaesthetic-like effects of a combination of amylmetacresol and dichloro-benzylalcohol as established ingredients of lozenges. In the presence of amylmetacresol, dichloro-benzylalcohol and lidocaine, a prominent block of inward sodium currents is apparent.


Asunto(s)
Anestésicos Locales/administración & dosificación , Antiinfecciosos Locales/administración & dosificación , Lidocaína/administración & dosificación , Canal de Sodio Activado por Voltaje NAV1.2/fisiología , Faringitis , Bloqueadores de los Canales de Sodio/administración & dosificación , Administración Tópica , Alcoholes Bencílicos , Cresoles/administración & dosificación , Relación Dosis-Respuesta a Droga , Quimioterapia Combinada , Células HEK293 , Humanos , Neuronas/efectos de los fármacos , Neuronas/fisiología , Faringitis/tratamiento farmacológico
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA