Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 742
1.
Braz J Med Biol Res ; 56: e12073, 2023.
Article En | MEDLINE | ID: mdl-36722655

Procainamide (PA) and its in vivo metabolite, N-acetylprocainamide (NAPA), display some pharmacological differences. Although it is agreed that PA is a class IA antiarrhythmic, it has been reported that NAPA is a pure class III antiarrhythmic that affects only the repolarizing phase of the cardiac action potential. This last concept, observed exclusively in dogs, gained wide acceptance, appearing in classic pharmacology textbooks. However, evidence in species such as mice and rats indicates that NAPA can affect cardiac Na+ channels, which is unexpected for a pure class III antiarrhythmic drug. To further clarify this issue, the effects of PA (used as a reference drug) and NAPA on the maximum upstroke velocity (Vmax) and half-decay time (HDT) of the cardiac action potential were examined in the isolated right papillaris magnus of the guinea pig heart. Both PA and NAPA affected Vmax at lower concentrations than required to affect HDT, and NAPA had weaker effects on both variables. Thus, NAPA displayed typical class IA antiarrhythmic behavior. Therefore, the concept that NAPA is a pure class III antiarrhythmic drug is more species-dependent than previously envisioned. In addition, we demonstrated that the differential pharmacology of PA and NAPA is explainable, in molecular terms, by steric hindrance of the effects of NAPA and the greater number of potent aromatic-aromatic and cation π interactions with Na+ or K+ cardiac channels for PA.


Acecainide , Procainamide , Guinea Pigs , Animals , Dogs , Mice , Rats , Procainamide/pharmacology , Papillary Muscles , Anti-Arrhythmia Agents/pharmacology , Action Potentials
2.
J Med Chem ; 64(14): 10403-10417, 2021 07 22.
Article En | MEDLINE | ID: mdl-34185525

Epigenetic post-translational modifications are essential for human malaria parasite survival and progression through its life cycle. Here, we present new functionalized suberoylanilide hydroxamic acid (SAHA) derivatives that chemically combine the pan-histone deacetylase inhibitor SAHA with the DNA methyltransferase inhibitor procainamide. A three- or four-step chemical synthesis was designed starting from cheap raw materials. Compared to the single drugs, the combined molecules showed a superior activity in Plasmodium and a potent inhibition against human HDAC6, exerting no cytotoxicity in human cell lines. These new compounds are fully active in multidrug-resistant Plasmodium falciparum Cambodian isolates. They target transmission of the parasite by inducing irreversible morphological changes in gametocytes and inhibiting exflagellation. The compounds are slow-acting and have an additive antimalarial effect in combination with fast-acting epidrugs and dihydroartemisinin. The lead compound decreases parasitemia in mice in a severe malaria model. Taken together, this novel fused molecule offers an affordable alternative to current failing antimalarial therapy.


Antimalarials/pharmacology , Histone Deacetylase 6/antagonists & inhibitors , Histone Deacetylase Inhibitors/pharmacology , Hydroxamic Acids/pharmacology , Malaria, Falciparum/drug therapy , Plasmodium falciparum/drug effects , Procainamide/pharmacology , Antimalarials/chemical synthesis , Antimalarials/chemistry , Dose-Response Relationship, Drug , Drug Resistance, Multiple/drug effects , Histone Deacetylase 6/metabolism , Histone Deacetylase Inhibitors/chemical synthesis , Histone Deacetylase Inhibitors/chemistry , Hydroxamic Acids/chemistry , Molecular Structure , Procainamide/chemistry , Structure-Activity Relationship
3.
Cell Rep Med ; 1(7): 100125, 2020 10 20.
Article En | MEDLINE | ID: mdl-33205075

Enteroviruses are suspected to contribute to insulin-producing ß cell loss and hyperglycemia-induced diabetes. However, mechanisms are not fully defined. Here, we show that coxsackievirus B type 4 (CVB4) infection in human islet-engrafted mice and in rat insulinoma cells displays loss of unconventional prefoldin RPB5 interactor (URI) and PDX1, affecting ß cell function and identity. Genetic URI ablation in the mouse pancreas causes PDX1 depletion in ß cells. Importantly, diabetic PDX1 heterozygous mice overexpressing URI in ß cells are more glucose tolerant. Mechanistically, URI loss triggers estrogen receptor nuclear translocation leading to DNA methyltransferase 1 (DNMT1) expression, which induces Pdx1 promoter hypermethylation and silencing. Consequently, demethylating agent procainamide-mediated DNMT1 inhibition reinstates PDX1 expression and protects against diabetes in pancreatic URI-depleted mice . Finally, the ß cells of human diabetes patients show correlations between viral protein 1 and URI, PDX1, and DNMT1 levels. URI and DNMT1 expression and PDX1 silencing provide a causal link between enterovirus infection and diabetes.


Capsid Proteins/genetics , Coxsackievirus Infections/genetics , DNA (Cytosine-5-)-Methyltransferase 1/genetics , Diabetes Mellitus, Type 2/genetics , Enterovirus B, Human/genetics , Homeodomain Proteins/genetics , Repressor Proteins/genetics , Trans-Activators/genetics , Animals , Capsid Proteins/metabolism , Coxsackievirus Infections/metabolism , Coxsackievirus Infections/pathology , Coxsackievirus Infections/virology , DNA (Cytosine-5-)-Methyltransferase 1/antagonists & inhibitors , DNA (Cytosine-5-)-Methyltransferase 1/metabolism , Diabetes Mellitus, Type 2/metabolism , Diabetes Mellitus, Type 2/pathology , Diabetes Mellitus, Type 2/virology , Disease Models, Animal , Enterovirus B, Human/metabolism , Enterovirus B, Human/pathogenicity , Female , Gene Expression Regulation , Glucose/metabolism , Glucose/pharmacology , Homeodomain Proteins/metabolism , Humans , Insulin-Secreting Cells/drug effects , Insulin-Secreting Cells/metabolism , Insulin-Secreting Cells/pathology , Insulin-Secreting Cells/transplantation , Male , Mice , Mice, Transgenic , Procainamide/pharmacology , Rats , Repressor Proteins/metabolism , Signal Transduction , Trans-Activators/metabolism , Transplantation, Heterologous
4.
Eur J Med Genet ; 63(2): 103661, 2020 Feb.
Article En | MEDLINE | ID: mdl-31051269

CHARGE syndrome is an autosomal dominant congenital disorder caused primarily by mutations in the CHD7 gene. Using a small molecule screen in a zebrafish model of CHARGE syndrome, we identified 4 compounds that rescue embryos from disease-like phenotypes. Our screen yielded DAPT, a Notch signaling inhibitor that could ameliorate the craniofacial, cranial neuronal and myelination defects in chd7 morphant zebrafish embryos. We discovered that Procainamide, an inhibitor of DNA methyltransferase 1, was able to recover the pattern of expression of isl2a, a cranial neuronal marker while also reducing the effect on craniofacial cartilage and myelination. M344, an inhibitor of Histone deacetylases had a strong recovery effect on craniofacial cartilage defects and could also modestly revert the myelination defects in zebrafish embryos. CHIC-35, a SIRT1 inhibitor partially restored the expression of isl2a in cranial neurons while causing a partial reversion of myelination and craniofacial cartilage defects. Our results suggest that a modular approach to phenotypic rescue in multi-organ syndromes might be a more successful approach to treat these disorders. Our findings also open up the possibility of using these compounds for other disorders with shared phenotypes.


CHARGE Syndrome/drug therapy , CHARGE Syndrome/physiopathology , DNA Helicases/genetics , DNA-Binding Proteins/genetics , Dipeptides/pharmacology , Procainamide/pharmacology , Vorinostat/pharmacology , Zebrafish Proteins/genetics , Zebrafish/embryology , Animals , Animals, Genetically Modified , CHARGE Syndrome/genetics , Cartilage/drug effects , Cartilage/pathology , DNA (Cytosine-5-)-Methyltransferase 1/antagonists & inhibitors , DNA Helicases/metabolism , DNA-Binding Proteins/metabolism , Dipeptides/therapeutic use , Disease Models, Animal , Embryo, Nonmammalian/diagnostic imaging , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/metabolism , Embryo, Nonmammalian/physiopathology , Gene Knockdown Techniques , Histone Deacetylase Inhibitors/pharmacology , Histone Deacetylase Inhibitors/therapeutic use , LIM-Homeodomain Proteins/genetics , LIM-Homeodomain Proteins/metabolism , Nerve Fibers, Myelinated/drug effects , Nerve Fibers, Myelinated/pathology , Neurons/drug effects , Neurons/pathology , Procainamide/therapeutic use , Receptors, Notch/antagonists & inhibitors , Sirtuin 1/antagonists & inhibitors , Transcription Factors/genetics , Transcription Factors/metabolism , Vorinostat/therapeutic use , Zebrafish/genetics , Zebrafish Proteins/metabolism
5.
JACC Clin Electrophysiol ; 5(4): 504-512, 2019 04.
Article En | MEDLINE | ID: mdl-31000106

OBJECTIVES: The authors studied the response rates and relative sensitivity of the most common agents used in the sodium-channel blocker (SCB) challenge. BACKGROUND: A type 1 Brugada electrocardiographic pattern precipitated by an SCB challenge confers a diagnosis of Brugada syndrome. METHODS: Patients undergoing an SCB challenge were prospectively enrolled across Canada and the United Kingdom. Patients with no prior cardiac arrest and family histories of sudden cardiac death or Brugada syndrome were included. RESULTS: Four hundred twenty-five subjects underwent SCB challenge (ajmaline, n = 331 [78%]; procainamide, n = 94 [22%]), with a mean age of 39 ± 15 years (54% men). Baseline non-type 1 Brugada ST-segment elevation was present in 10%. A total of 154 patients (36%) underwent signal-averaged electrocardiography, with 41% having late potentials. Positive results were seen more often with ajmaline than procainamide infusion (26% vs. 4%, p < 0.001). On multivariate analysis, baseline non-type 1 Brugada ST-segment elevation (odds ratio [OR]: 6.92; 95% confidence interval [CI]: 3.15 to 15.2; p < 0.001) and ajmaline use (OR: 8.76; 95% CI: 2.62 to 29.2; p < 0.001) were independent predictors of positive results to SCB challenge. In the subgroup undergoing signal-averaged electrocardiography, non-type 1 Brugada ST-segment elevation (OR: 9.28; 95% CI: 2.22 to 38.8; p = 0.002), late potentials on signal-averaged electrocardiography (OR: 4.32; 95% CI: 1.50 to 12.5; p = 0.007), and ajmaline use (OR: 12.0; 95% CI: 2.45 to 59.1; p = 0.002) were strong predictors of SCB outcome. CONCLUSIONS: The outcome of SCB challenge was significantly affected by the drug used, with ajmaline more likely to provoke a type 1 Brugada electrocardiographic pattern compared with procainamide. Patients undergoing SCB challenge may have contrasting results depending on the drug used, with potential clinical, psychosocial, and socioeconomic implications.


Ajmaline/pharmacology , Brugada Syndrome/diagnosis , Electrocardiography/drug effects , Procainamide/pharmacology , Voltage-Gated Sodium Channel Blockers/pharmacology , Adult , Brugada Syndrome/physiopathology , Cohort Studies , Female , Humans , Male , Middle Aged , Young Adult
6.
JACC Clin Electrophysiol ; 5(2): 212-219, 2019 02.
Article En | MEDLINE | ID: mdl-30784693

OBJECTIVES: This study sought to compare the differences between procainamide and flecainide to stress the His-Purkinje system during electrophysiological study (EPS) in patients with syncope and bundle branch block (BBB). BACKGROUND: Patients with syncope and BBB are at risk of developing atrioventricular block. EPS is recommended including class I drug challenge to unmask His-Purkinje disease in cases with baseline normal His-ventricular interval. There is little data on differences between different class I drugs. METHODS: This was a prospective study of all consecutive patients undergoing EPS for syncope and BBB at a single center (January 1, 2012 to June 30, 2017). Of those patients with negative baseline EPS, 2 cohorts were compared: group A (historical cohort: procainamide) and group B (flecainide). RESULTS: During the study, 271 patients (age 73.9 ± 12.1 years, 64.9% male, QRS duration: 139.4 ± 13.9 ms) underwent EPS. In 166, baseline EPS was negative and class I drug challenge was performed (90 procainamide, 76 flecainide). The final value and percentage increase in the His-ventricular interval (76 ± 16 ms vs. 64 ± 10 ms and 22.5 ± 6.2% vs. 11.8 ± 5.3%; p < 0.001) and diagnostic yield (14.5% vs. 7.8%, p = 0.04) were higher with flecainide. No differences were found in baseline characteristics. During follow-up (25.8 ± 6.3 months), 39 patients (24.8%) with negative EPS (19.2% with flecainide vs. 30.1% with procainamide: relative risk: 5.1; 95% confidence interval: 2.6 to 10.2; p < 0. 001) received a pacemaker. CONCLUSIONS: Flecainide has a higher diagnostic yield than does procainamide in patients with BBB, syncope, and negative baseline EPS due to a greater increase of the His-ventricular interval. Additionally, there is a lesser need for pacemaker implantation in patients in whom the class I drug test using flecainide was negative.


Electrocardiography/drug effects , Electrophysiologic Techniques, Cardiac/methods , Flecainide/pharmacology , Procainamide/pharmacology , Syncope , Aged , Aged, 80 and over , Bundle-Branch Block/diagnosis , Bundle-Branch Block/physiopathology , Female , Flecainide/therapeutic use , Heart Conduction System/drug effects , Humans , Male , Middle Aged , Procainamide/therapeutic use , Syncope/diagnosis , Syncope/physiopathology
7.
Scand Cardiovasc J ; 52(4): 218-226, 2018 08.
Article En | MEDLINE | ID: mdl-29798684

OBJECTIVES: In normal conditions, sudden heart rate acceleration provokes a rapid reduction in ventricular action potential duration (APD). The protracted APD rate adaptation favors early afterdepolarizations and precipitates arrhythmia. Nevertheless, it is uncertain as to whether the rate-dependent changes of ventricular repolarization can be adversely modified by arrhythmogenic drugs (quinidine and procainamide) and hypokalemia, in comparison to the agents with safe therapeutic profile, such as lidocaine. DESIGN: The rate adaptation of QT interval and monophasic APD obtained from the left ventricular (LV) and the right ventricular (RV) epicardium was examined during rapid cardiac pacing applied in isolated, perfused guinea-pig heart preparations. RESULTS: At baseline, an abrupt increase in cardiac activation rate was associated with a substantial reduction of the QT interval and ventricular APD in the first two cardiac cycles, which was followed by a gradual shortening of repolarization over subsequent pacing intervals. The time constants of the fast (τfast) and slow (τslow) components of the APD dynamics determined from a double exponential fit were longer in RV compared to LV chamber. Quinidine, procainamide, and hypokalemia prolonged ventricular repolarization and delayed the rate adaptation of the QT interval and APD in LV and RV, as evidenced by increased τfast and τslow values. In contrast, lidocaine had no effect on the dynamic changes of ventricular repolarization upon heart rate acceleration. CONCLUSIONS: The rate adaptation of ventricular repolarization is delayed by arrhythmogenic interventions, such as quinidine, procainamide, and hypokalemia, but not changed by lidocaine, a clinically safe antiarrhythmic agent.


Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/drug therapy , Heart Rate/drug effects , Heart Ventricles/drug effects , Hypokalemia/physiopathology , Ventricular Function, Left/drug effects , Ventricular Function, Right/drug effects , Action Potentials , Adaptation, Physiological , Animals , Anti-Arrhythmia Agents/toxicity , Arrhythmias, Cardiac/chemically induced , Arrhythmias, Cardiac/physiopathology , Cardiac Pacing, Artificial , Female , Guinea Pigs , Heart Ventricles/physiopathology , Hypokalemia/complications , Isolated Heart Preparation , Lidocaine/pharmacology , Procainamide/pharmacology , Quinidine/pharmacology , Time Factors
9.
Appl Biochem Biotechnol ; 182(4): 1491-1496, 2017 Aug.
Article En | MEDLINE | ID: mdl-28116573

The problem of gelation of concentrated protein solutions, which poses challenges for both downstream protein processing and liquid formulations of pharmaceutical proteins, is addressed herein by employing previously discovered viscosity-lowering bulky salts. Procainamide-HCl and the salt of camphor-10-sulfonic acid with L-arginine (CSA-Arg) greatly retard gelation upon heating and subsequent cooling of the model proteins gelatin and casein in water: Whereas in the absence of additives the proteins form aqueous gels within several hours at room temperature, procainamide-HCl for both proteins and also CSA-Arg for casein prevent gel formation for months under the same conditions. The inhibition of gelation by CSA-Arg stems exclusively from the CSA moiety: CSA-Na was as effective as CSA-Arg, while Arg-HCl was marginally or not effective. The tested bulky salts did not inhibit (and indeed accelerated) temperature-induced gel formation in aqueous solutions of all examined carbohydrates-starch, agarose, alginate, gellan gum, and carrageenan.


Carbohydrates/chemistry , Caseins/chemistry , Excipients/chemistry , Excipients/pharmacology , Gelatin/chemistry , Salts/chemistry , Salts/pharmacology , Animals , Arginine/chemistry , Camphor/analogs & derivatives , Camphor/pharmacology , Cattle , Gels , Procainamide/pharmacology , Solutions , Sulfones/pharmacology , Temperature , Viscosity
10.
J Emerg Med ; 52(4): 484-492, 2017 Apr.
Article En | MEDLINE | ID: mdl-27751700

BACKGROUND: Ventricular tachycardia (VT) and ventricular fibrillation are the causes of approximately 300,000 deaths per year in the United States. VT is classified based on hemodynamic status and appearance. Stable, monomorphic VT treatment is controversial. OBJECTIVE: Our aim was to provide emergency physicians with an evidence-based review of the medical management of stable, monomorphic VT. DISCUSSION: Stable, monomorphic VT is part of a larger class of ventricular dysrhythmias defined by a rate of at least 120 beats/min with QRS > 120 ms without regularly occurring P:QRS association. Little controversy exists for the treatment of hemodynamically unstable VT. The medical management of hemodynamically stable monomorphic VT is surrounded by controversy. Direct current cardioversion is most efficacious. Guidelines for the treatment of stable VT from the American Heart Association provide a IIa recommendation for procainamide, compared with a IIb recommendation for both amiodarone and sotalol. Studies evaluating procainamide, lidocaine, amiodarone, and sotalol suffer from poor design, difference in inclusion and exclusion criteria, small sample size, and outcome determination. Procainamide demonstrates the greatest efficacy. If procainamide is selected, a maximum dose of 10 mg/kg at 50-100 mg/min intravenous (IV) over 10-20 min should be provided with monitoring of blood pressure and electrocardiogram. Monomorphic VT with acute myocardial ischemia requires further study. CONCLUSIONS: Optimal management of stable, monomorphic VT includes direct current cardioversion. If medical management is chosen, procainamide is most efficacious, though current literature suffers from poor design.


Anti-Arrhythmia Agents/therapeutic use , Electric Countershock/standards , Review Literature as Topic , Tachycardia, Ventricular/drug therapy , Amiodarone/pharmacology , Amiodarone/therapeutic use , Anti-Arrhythmia Agents/pharmacology , Electric Countershock/methods , Electrocardiography/methods , Emergency Service, Hospital/organization & administration , Evidence-Based Medicine/methods , Humans , Lidocaine/pharmacology , Lidocaine/therapeutic use , Procainamide/administration & dosage , Procainamide/pharmacology , Procainamide/therapeutic use , Pyrimidinones/pharmacology , Pyrimidinones/therapeutic use , Sotalol/pharmacology , Sotalol/therapeutic use , Tachycardia, Ventricular/mortality
11.
Chem Res Toxicol ; 29(9): 1393-5, 2016 09 19.
Article En | MEDLINE | ID: mdl-27501273

We have previously reported the enhancement of the antiproliferative and apoptotic activities of cis-diamminedichloroplatinum(II) (DDP) when it is coadministered with a class I antiarrhythmic drug procainamide hydrochloride (PA). Here, we determined the antiproliferative activity of DDP, either in solution or loaded in liposomes, in the presence of PA, in the bulk solution, or directly embedded in liposomes together with DDP. Our results show that PA potentiates the activity of DDP-liposomes and that this effect is maintained at least in some of the investigated cell types when both drugs were mixed and loaded together into liposomes.


Cisplatin/pharmacology , Liposomes/chemistry , Procainamide/pharmacology , A549 Cells , Animals , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Synergism , Inhibitory Concentration 50 , Liposomes/chemical synthesis
12.
Toxicol Appl Pharmacol ; 305: 55-65, 2016 08 15.
Article En | MEDLINE | ID: mdl-27288732

Cation trapping in acidic cell compartments determines an antiproliferative effect that has a potential interest in oncology, as shown by clinical data and trials involving chloroquine and hydroxychloroquine. To further characterize the mechanism of this effect, we studied a series of 6 substituted triethylamine (s-Et3N) drugs that encompasses a wide range of liposolubility (amiodarone, quinacrine, chloroquine, hydroxychloroquine, lidocaine, and procainamide). Three tumor cell lines and primary human endothelial cells were exploited in proliferation assays (48h, cell counts). Accumulation of the autophagic effector LC3 II and the apoptotic marker cleaved PARP1 (immunoblots), cytotoxicity, cell cycle analysis and endocytic function were further tested in the p53-null histiocytic lymphoma U937 line. A profound and desynchronized antiproliferative effect was observed in response to all s-Et3Ns with essentially no cell type specificity. Predictors of s-Et3N potency were liposolubility and the acute accumulation of the autophagic effector LC3 II (6h-treatments). For each s-Et3N, there was an antiproliferative concentration range where cytotoxicity and apoptosis were not triggered in U937 cells (24-48h-treatments). Quinacrine was the most potent cytostatic drug (1-5µM). Co-treatment of cells with inhibitors of cholesterol, ß-cyclodextrin or lovastatin, partially reversed the antiproliferative effect of each s-Et3N. The cytopathology induced by cationic drug accumulation includes a cytostatic effect. Its intensity is cell type- and p53-independent, but predicted by the inhibition of autophagic flux and by the liposolubility of individual drugs and alleviated by cholesterol ablation. The superiority of quinacrine, biomarker value of LC3 II and antagonism by a statin may be clinically relevant.


Anticholesteremic Agents/pharmacology , Autophagy/drug effects , Cholesterol/metabolism , Cytostatic Agents/pharmacology , Cytotoxins/pharmacology , Lovastatin/pharmacology , Amiodarone/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cells, Cultured , Chloroquine/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Hydroxychloroquine/pharmacology , Lidocaine/pharmacology , Lysosomes , Microtubule-Associated Proteins/metabolism , Poly (ADP-Ribose) Polymerase-1/metabolism , Procainamide/pharmacology , Quinacrine/pharmacology , beta-Cyclodextrins/pharmacology
13.
Mol Pharm ; 13(6): 2126-35, 2016 06 06.
Article En | MEDLINE | ID: mdl-27112518

To improve the anticolitic efficacy of 5-aminosalicylic acid (5-ASA), a colon-specific mutual prodrug of 5-ASA was designed. 5-ASA was coupled to procainamide (PA), a local anesthetic, via an azo bond to prepare 5-(4-{[2-(diethylamino)ethyl]carbamoyl}phenylazo)salicylic acid (5-ASA-azo-PA). 5-ASA-azo-PA was cleaved to 5-ASA and PA up to about 76% at 10 h in the cecal contents while remaining stable in the small intestinal contents. Oral gavage of 5-ASA-azo-PA and sulfasalazine, a colon-specific prodrug currently used in clinic, to rats showed similar efficiency in delivery of 5-ASA to the large intestine, and PA was not detectable in the blood after 5-ASA-azo-PA administration. Oral gavage of 5-ASA-azo-PA alleviated 2,4,6-trinitrobenzenesulfonic acid-induced rat colitis. Moreover, combined intracolonic treatment with 5-ASA and PA elicited an additive ameliorative effect. Furthermore, combined treatment with 5-ASA and PA additively inhibited nuclear factor-kappaB (NFκB) activity in human colon carcinoma cells and inflamed colonic tissues. Finally, 5-ASA-azo-PA administered orally was able to reduce inflammatory mediators, NFκB target gene products, in the inflamed colon. 5-ASA-azo-PA may be a colon-specific mutual prodrug acting against colitis, and the mutual anticolitic effects occurred at least partly through the cooperative inhibition of NFκB activity.


Azo Compounds/pharmacology , Colitis/drug therapy , Mesalamine/pharmacology , NF-kappa B/metabolism , Procainamide/pharmacology , Prodrugs/pharmacology , Animals , Azo Compounds/chemistry , Colon/drug effects , Male , Mesalamine/chemistry , Procainamide/chemistry , Prodrugs/chemistry , Rats , Rats, Sprague-Dawley , Trinitrobenzenesulfonic Acid/chemistry , Trinitrobenzenesulfonic Acid/pharmacology
14.
Pharmacol Rep ; 68(3): 654-61, 2016 Jun.
Article En | MEDLINE | ID: mdl-27026293

BACKGROUND: We describe the potentiation of antiproliferative and apoptotic activities triggered by cis-diamminedichloroplatinum(II) (DDP), and obtained in vitro by the co-administration of procainamide hydrochloride (PdHCl) in murine P388, and human A2780 and A549 cells. METHODS: We determined the antiproliferative and apoptotic activities of DDP and PdHCl combinations by different techniques. Moreover, cell cycle analysis, restriction enzyme inhibition followed by agarose gel electrophoresis, and TUNEL analysis of tumour cells in vivo were also used to strengthen our hypothesis. RESULTS: Our results show that PdHCl may significantly increase the inhibition of cell proliferation and apoptosis. Experiments in vivo showed that the co-administration of DDP and PdHCl increased the percentage of apoptotic cells compared to DDP alone treatment, both in subcutaneous (sc) and intraperitoneal (ip) P388 tumours. We finally demonstrated that the co-administration of PdHCl prevents DNA digestion accounting for a restriction enzyme inhibition that in some cases was greater than that obtained by DDP alone. Moreover, when PdHCl was mixed with the reaction products (RP) of DDP (RP-PdHCl) we obtained a restriction enzyme inhibition greater for some enzymes (Bsp1407I, Hin1II, and Psp1406I) than that obtained by the DDP-PdHCl solution. CONCLUSIONS: On the whole our data demonstrate that the class I antiarrhythmic drug PdHCl may increase the antiproliferative activity of DDP by improving its triggering of apoptosis, and that this phenomenon may be likely linked to the formation of a new Pt compound.


Apoptosis/drug effects , Cell Proliferation/drug effects , Cisplatin/pharmacology , Procainamide/pharmacology , Animals , Anti-Arrhythmia Agents/pharmacology , Cell Cycle/drug effects , Cells, Cultured , Drug Synergism , Humans , Mice , Restriction Mapping
15.
PLoS One ; 11(2): e0150319, 2016.
Article En | MEDLINE | ID: mdl-26918767

Overt systemic inflammatory response is a predisposing mechanism for infection-induced skeletal muscle damage and rhabdomyolysis. Aberrant DNA methylation plays a crucial role in the pathophysiology of excessive inflammatory response. The antiarrhythmic drug procainamide is a non-nucleoside inhibitor of DNA methyltransferase 1 (DNMT1) used to alleviate DNA hypermethylation. Therefore, we evaluated the effects of procainamide on the syndromes and complications of rhabdomyolysis rats induced by lipopolysaccharide (LPS). Rhabdomyolysis animal model was established by intravenous infusion of LPS (5 mg/kg) accompanied by procainamide therapy (50 mg/kg). During the experimental period, the changes of hemodynamics, muscle injury index, kidney function, blood gas, blood electrolytes, blood glucose, and plasma interleukin-6 (IL-6) levels were examined. Kidneys and lungs were exercised to analyze superoxide production, neutrophil infiltration, and DNMTs expression. The rats in this model showed similar clinical syndromes and complications of rhabdomyolysis including high levels of plasma creatine kinase, acute kidney injury, hyperkalemia, hypocalcemia, metabolic acidosis, hypotension, tachycardia, and hypoglycemia. The increases of lung DNMT1 expression and plasma IL-6 concentration were also observed in rhabdomyolysis animals induced by LPS. Treatment with procainamide not only inhibited the overexpression of DNMT1 but also diminished the overproduction of IL-6 in rhabdomyolysis rats. In addition, procainamide improved muscle damage, renal dysfunction, electrolytes disturbance, metabolic acidosis, hypotension, and hypoglycemia in the rats with rhabdomyolysis. Moreover, another DNMT inhibitor hydralazine mitigated hypoglycemia, muscle damage, and renal dysfunction in rhabdomyolysis rats. These findings reveal that therapeutic effects of procainamide could be based on the suppression of DNMT1 and pro-inflammatory cytokine in endotoxin-induced rhabdomyolysis.


DNA (Cytosine-5-)-Methyltransferases/antagonists & inhibitors , Endotoxins/toxicity , Procainamide/therapeutic use , Rhabdomyolysis/drug therapy , Acidosis/drug therapy , Acidosis/etiology , Animals , Bicarbonates/blood , Biomarkers , Creatinine/blood , DNA (Cytosine-5-)-Methyltransferase 1 , DNA (Cytosine-5-)-Methyltransferases/biosynthesis , DNA Methylation/drug effects , DNA Methyltransferase 3A , Drug Evaluation, Preclinical , Electrolytes/blood , Endotoxemia/complications , Hydralazine/pharmacology , Hydralazine/therapeutic use , Hypertension/drug therapy , Hypertension/etiology , Interleukin-6/blood , Kidney/immunology , Kidney/pathology , Kidney/physiopathology , Lung/enzymology , Lung/pathology , Male , Muscle, Skeletal/pathology , Neutrophils/pathology , Procainamide/pharmacology , Random Allocation , Rats , Rats, Wistar , Rhabdomyolysis/blood , Rhabdomyolysis/chemically induced , Rhabdomyolysis/complications , Superoxides/analysis , Tachycardia/drug therapy , Tachycardia/etiology , DNA Methyltransferase 3B
16.
Actas dermo-sifiliogr. (Ed. impr.) ; 105(1): 18-30, ene.-feb. 2014. ilus, tab
Article Es | IBECS | ID: ibc-129401

El término lupus eritematoso inducido por fármacos (LEIF) hace referencia a una entidad caracterizada por la aparición de manifestaciones clínicas, histopatológicas e inmunológicas similares a aquellas que aparecen en el lupus eritematoso idiopático, pero que cronológicamente coinciden con la toma de ciertos fármacos y que se resuelven tras la retirada de los mismos. Más de 90 fármacos se han asociado con la aparición de LEIF. Esta lista de fármacos implicados sigue aumentando. Al igual que el lupus eritematoso idiopático, el LEIF se puede subclasificar en lupus eritematoso sistémico inducido por fármacos, lupus eritematoso cutáneo subagudo inducido por fármacos y lupus eritematoso cutáneo crónico inducido por fármacos. Reconocer estas entidades es de gran interés, ya que este cuadro suele revertir tras la retirada del fármaco implicado


Drug-induced lupus erythematosus (DILE) refers to a condition whose clinical, histological, and immunological features are similar to those seen in idiopathic lupus erythematosus but that occurs when certain drugs are taken and resolves after their withdrawal. Over 90 drugs have been linked to DILE to date and the list is growing. Like idiopathic lupus erythematosus, DILE has systemic, subacute cutaneous, and chronic cutaneous forms. A correct diagnosis is very important, as this condition usually resolves after withdrawal of the offending drug


Humans , Male , Female , Lupus Erythematosus, Cutaneous/chemically induced , Lupus Erythematosus, Cutaneous/immunology , Lupus Erythematosus, Cutaneous/pathology , Vasculitis/pathology , Vasculitis/therapy , Thalidomide/therapeutic use , Procainamide/pharmacology , Arthralgia/complications , Hydralazine/adverse effects , Hydralazine/pharmacology , Isoniazid/toxicity
17.
Fundam Clin Pharmacol ; 28(4): 382-93, 2014 Aug.
Article En | MEDLINE | ID: mdl-23952942

Procainamide is class Ia Na(+) channel blocker that may prolong ventricular repolarization secondary to inhibition of IK r , the rapid component of the delayed rectifier K(+) current. In contrast to selective IN a blockers such as lidocaine, procainamide was shown to produce arrhythmogenic effects in the clinical setting. This study examined whether pro-arrhythmic responses to procainamide may be accounted for by drug-induced repolarization abnormalities including impaired electrical restitution kinetics, spatial gradients in action potential duration (APD), and activation-to-repolarization coupling. In perfused guinea-pig hearts, procainamide was found to prolong the QT interval on ECG and left ventricular (LV) epicardial monophasic APD, increased the maximum slope of electrical restitution, enhanced transepicardial APD variability, and eliminated the inverse correlation between the local APD and activation time values determined at distinct epicardial recording sites prior to drug infusion. In contrast, lidocaine had no effect on electrical restitution, the degree of transepicardial repolarization heterogeneities, and activation-to-repolarization coupling. Spontaneous episodes of monomorphic ventricular tachycardia were observed in 57% of procainamide-treated heart preparations. No arrhythmia was induced by lidocaine. In summary, this study suggests that abnormal changes in repolarization may contribute to pro-arrhythmic effects of procainamide.


Anti-Arrhythmia Agents/toxicity , Arrhythmias, Cardiac/chemically induced , Lidocaine/toxicity , Procainamide/toxicity , Action Potentials/drug effects , Animals , Anti-Arrhythmia Agents/pharmacology , Arrhythmias, Cardiac/physiopathology , Electrocardiography , Female , Guinea Pigs , Lidocaine/pharmacology , Long QT Syndrome/chemically induced , Long QT Syndrome/physiopathology , Procainamide/pharmacology , Tachycardia, Ventricular/chemically induced , Tachycardia, Ventricular/physiopathology , Voltage-Gated Sodium Channel Blockers/pharmacology , Voltage-Gated Sodium Channel Blockers/toxicity
19.
Antimicrob Agents Chemother ; 57(3): 1347-51, 2013 Mar.
Article En | MEDLINE | ID: mdl-23274657

Aggregatibacter actinomycetemcomitans invades periodontal pocket epithelium and is therefore difficult to eliminate by periodontal scaling and root planing. It is susceptible to azithromycin, which is taken up by many types of mammalian cells. This led us to hypothesize that azithromycin accumulation by gingival epithelium could enhance the killing of intraepithelial A. actinomycetemcomitans. [(3)H]azithromycin transport by Smulow-Glickman gingival epithelial cells and SCC-25 oral epithelial cells was characterized. To test our hypothesis, we infected cultured Smulow-Glickman cell monolayers with A. actinomycetemcomitans (Y4 or SUNY 465 strain) for 2 h, treated them with gentamicin to eliminate extracellular bacteria, and then incubated them with azithromycin for 1 to 4 h. Viable intracellular bacteria were released, plated, and enumerated. Azithromycin transport by both cell lines exhibited Michaelis-Menten kinetics and was competitively inhibited by l-carnitine and several other organic cations. Cell incubation in medium containing 5 µg/ml azithromycin yielded steady-state intracellular concentrations of 144 µg/ml in SCC-25 cells and 118 µg/ml in Smulow-Glickman cells. Azithromycin induced dose- and time-dependent intraepithelial killing of both A. actinomycetemcomitans strains. Treatment of infected Smulow-Glickman cells with 0.125 µg/ml azithromycin killed approximately 29% of the intraepithelial CFU of both strains within 4 h, while treatment with 8 µg/ml azithromycin killed ≥82% of the CFU of both strains (P < 0.05). Addition of carnitine inhibited the killing of intracellular bacteria by azithromycin (P < 0.05). Thus, human gingival epithelial cells actively accumulate azithromycin through a transport system that facilitates the killing of intraepithelial A. actinomycetemcomitans and is shared with organic cations.


Aggregatibacter actinomycetemcomitans/drug effects , Anti-Bacterial Agents/pharmacology , Azithromycin/pharmacology , Epithelial Cells/drug effects , Gingiva/drug effects , Aggregatibacter actinomycetemcomitans/growth & development , Biological Transport/drug effects , Carnitine/pharmacology , Cell Line , Dose-Response Relationship, Drug , Epithelial Cells/cytology , Epithelial Cells/microbiology , Gingiva/cytology , Gingiva/microbiology , Humans , Kinetics , Probenecid/pharmacology , Procainamide/pharmacology , Pyrilamine/pharmacology , Quinidine/pharmacology , Time Factors
20.
Circ Arrhythm Electrophysiol ; 4(2): 128-35, 2011 Apr.
Article En | MEDLINE | ID: mdl-21270101

BACKGROUND: Catecholaminergic polymorphic ventricular tachycardia (CPVT) is caused by mutations in the cardiac ryanodine receptor (RyR2) or calsequestrin (Casq2) and can be difficult to treat. The class Ic antiarrhythmic drug flecainide blocks RyR2 channels and prevents CPVT in mice and humans. It is not known whether other class I antiarrhythmic drugs also block RyR2 channels and to what extent RyR2 channel inhibition contributes to antiarrhythmic efficacy in CPVT. METHODS AND RESULTS: We first measured the effect of all class I antiarrhythmic drugs marketed in the United States (quinidine, procainamide, disopyramide, lidocaine, mexiletine, flecainide, and propafenone) on single RyR2 channels incorporated into lipid bilayers. Only flecainide and propafenone inhibited RyR2 channels, with the S-enantiomer of propafenone having a significantly lower potency than R-propafenone or flecainide. In Casq2(-/-) myocytes, the propafenone enantiomers and flecainide significantly reduced arrhythmogenic Ca(2+) waves at clinically relevant concentrations, whereas Na(+) channel inhibitors without RyR2 blocking properties did not. In Casq2(-/-) mice, 5 mg/kg R-propafenone or 20 mg/kg S-propafenone prevented exercise-induced CPVT, whereas procainamide (20 mg/kg) or lidocaine (20 mg/kg) were ineffective (n=5 to 9 mice, P<0.05). QRS duration was not significantly different, indicating a similar degree of Na(+) channel inhibition. Clinically, propafenone (900 mg/d) prevented ICD shocks in a 22-year-old CPVT patient who had been refractory to maximal standard drug therapy and bilateral stellate ganglionectomy. CONCLUSIONS: RyR2 cardiac Ca(2+) release channel inhibition appears to determine efficacy of class I drugs for the prevention of CPVT in Casq2(-/-) mice. Propafenone may be an alternative to flecainide for CPVT patients symptomatic on ß-blockers.


Anti-Arrhythmia Agents/pharmacology , Calcium Channel Blockers/pharmacology , Flecainide/pharmacology , Myocytes, Cardiac/drug effects , Propafenone/pharmacology , Ryanodine Receptor Calcium Release Channel/drug effects , Analysis of Variance , Animals , Calsequestrin/deficiency , Calsequestrin/genetics , Defibrillators, Implantable , Disease Models, Animal , Dose-Response Relationship, Drug , Electric Countershock/instrumentation , Electrocardiography , Humans , Ion Channel Gating/drug effects , Lidocaine/pharmacology , Male , Mice , Mice, Knockout , Mutation, Missense , Myocytes, Cardiac/metabolism , Procainamide/pharmacology , Ryanodine Receptor Calcium Release Channel/genetics , Ryanodine Receptor Calcium Release Channel/metabolism , Sodium Channel Blockers/pharmacology , Tachycardia, Ventricular/drug therapy , Tachycardia, Ventricular/genetics , Tachycardia, Ventricular/metabolism , Tachycardia, Ventricular/physiopathology , Time Factors , Young Adult
...