Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 64
Filter
1.
Int J Clin Pharmacol Ther ; 60(6): 253-263, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35445658

ABSTRACT

OBJECTIVE: To develop a physiologically based pharmacokinetic (PBPK) model for amiloride, an acid-sensing ion channel (ASIC) antagonist, and to simulate its pharmacokinetics in plasma and the central nervous system following intranasal administration in a virtual human population. MATERIALS AND METHODS: We first developed a PBPK model of amiloride after oral administration and optimized the model using data from five clinical studies. Next, we added a nasal compartment to the amiloride oral PBPK model and parameterized using data from previous clinical studies. We simulated amiloride's pharmacokinetics in plasma, brain, and cerebrospinal fluid (CSF) after intranasal administration of amiloride at various doses in a virtual human population. RESULTS: The target amiloride concentration in the central nervous system required for maximal ASIC inhibition was achieved with a 75-mg intranasal amiloride dose. However, this finding is based on simulations performed using a mathematical model and needs to be further validated with appropriate clinical data. CONCLUSION: The nasal PBPK model of amiloride could be used to design future clinical studies and allow for successful clinical translation of intranasal amiloride formulation.


Subject(s)
Acid Sensing Ion Channel Blockers , Amiloride , Anxiety Disorders , Acid Sensing Ion Channel Blockers/administration & dosage , Acid Sensing Ion Channel Blockers/pharmacokinetics , Acid Sensing Ion Channels/drug effects , Administration, Intranasal , Administration, Oral , Amiloride/administration & dosage , Amiloride/pharmacokinetics , Anxiety Disorders/drug therapy , Computer Simulation , Humans , Models, Biological
2.
PLoS Pathog ; 17(5): e1009519, 2021 05.
Article in English | MEDLINE | ID: mdl-34003853

ABSTRACT

SARS-CoV-2 is the novel coronavirus that is the causative agent of COVID-19, a sometimes-lethal respiratory infection responsible for a world-wide pandemic. The envelope (E) protein, one of four structural proteins encoded in the viral genome, is a 75-residue integral membrane protein whose transmembrane domain exhibits ion channel activity and whose cytoplasmic domain participates in protein-protein interactions. These activities contribute to several aspects of the viral replication-cycle, including virion assembly, budding, release, and pathogenesis. Here, we describe the structure and dynamics of full-length SARS-CoV-2 E protein in hexadecylphosphocholine micelles by NMR spectroscopy. We also characterized its interactions with four putative ion channel inhibitors. The chemical shift index and dipolar wave plots establish that E protein consists of a long transmembrane helix (residues 8-43) and a short cytoplasmic helix (residues 53-60) connected by a complex linker that exhibits some internal mobility. The conformations of the N-terminal transmembrane domain and the C-terminal cytoplasmic domain are unaffected by truncation from the intact protein. The chemical shift perturbations of E protein spectra induced by the addition of the inhibitors demonstrate that the N-terminal region (residues 6-18) is the principal binding site. The binding affinity of the inhibitors to E protein in micelles correlates with their antiviral potency in Vero E6 cells: HMA ≈ EIPA > DMA >> Amiloride, suggesting that bulky hydrophobic groups in the 5' position of the amiloride pyrazine ring play essential roles in binding to E protein and in antiviral activity. An N15A mutation increased the production of virus-like particles, induced significant chemical shift changes from residues in the inhibitor binding site, and abolished HMA binding, suggesting that Asn15 plays a key role in maintaining the protein conformation near the binding site. These studies provide the foundation for complete structure determination of E protein and for structure-based drug discovery targeting this protein.


Subject(s)
Amiloride/pharmacology , COVID-19 Drug Treatment , Coronavirus Envelope Proteins/metabolism , SARS-CoV-2/drug effects , SARS-CoV-2/metabolism , Amiloride/pharmacokinetics , Animals , Antiviral Agents/pharmacology , Binding Sites/drug effects , COVID-19/virology , Chlorocebus aethiops , Coronavirus Envelope Proteins/chemistry , Humans , Ion Channels/metabolism , Nuclear Magnetic Resonance, Biomolecular , Protein Binding/drug effects , Protein Conformation/drug effects , Protein Domains , Vero Cells , Virus Assembly/drug effects
3.
Bioorg Med Chem ; 37: 116116, 2021 05 01.
Article in English | MEDLINE | ID: mdl-33799173

ABSTRACT

The K+-sparing diuretic amiloride elicits anticancer activities in multiple animal models. During our recent medicinal chemistry campaign aiming to identify amiloride analogs with improved properties for potential use in cancer, we discovered novel 6-(hetero)aryl-substituted amiloride and 5-(N,N-hexamethylene)amiloride (HMA) analogs with up to 100-fold higher potencies than the parent compounds against urokinase plasminogen activator (uPA), one of amiloride's putative anticancer targets, and no diuretic or antikaliuretic effects. Here, we report the systematic evaluation of structure-property relationships (lipophilicity, aqueous solubility and in vitro metabolic stability in human and mouse liver microsomes) in twelve matched pair analogs selected from our 6-substituted amiloride and HMA libraries. Mouse plasma stability, plasma protein binding, Caco-2 cell permeability, cardiac ion channel activity and pharmacokinetics in mice (PO and IV) and rats (IV) are described alongside amiloride and HMA comparators for a subset of the four most promising matched-pair analogs. The findings combined with earlier uPA activity/selectivity and other data ultimately drove selection of two analogs (AA1-39 and AA1-41) that showed efficacy in separate mouse cancer metastasis studies.


Subject(s)
Amiloride/analogs & derivatives , Amiloride/pharmacology , Antineoplastic Agents/pharmacology , Amiloride/pharmacokinetics , Animals , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacokinetics , Caco-2 Cells , Drug Screening Assays, Antitumor , Female , Humans , Male , Mice, Inbred BALB C , Microsomes, Liver/drug effects , Molecular Structure , Rats, Sprague-Dawley , Structure-Activity Relationship
4.
J Renin Angiotensin Aldosterone Syst ; 21(4): 1470320320975893, 2020.
Article in English | MEDLINE | ID: mdl-33234024

ABSTRACT

Amiloride is a potassium retaining diuretic and natriuretic which acts by reversibly blocking luminal epithelial sodium channels (ENaCs) in the late distal tubule and collecting duct. Amiloride is indicated in oedematous states, and for potassium conservation adjunctive to thiazide or loop diuretics for hypertension, congestive heart failure and hepatic cirrhosis with ascites. Historical studies on its use in hypertension were poorly controlled and there is insufficient data on dose-response. It is clearly highly effective in combination with thiazide diuretics where it counteracts the adverse metabolic effects of the thiazides and its use in the Medical Research Council Trial of Older Hypertensive Patients, demonstrated convincing outcome benefits on stroke and coronary events. Recently it has been shown to be as effective as spironolactone in resistant hypertension but there is a real need to establish its potential role in the much larger number of patients with mild to moderate hypertension in whom there is a paucity of information with amiloride particularly across an extended dose range.


Subject(s)
Amiloride/therapeutic use , Amiloride/adverse effects , Amiloride/chemistry , Amiloride/pharmacokinetics , Animals , Clinical Trials as Topic , Diuretics/adverse effects , Diuretics/chemistry , Diuretics/pharmacokinetics , Diuretics/therapeutic use , Humans , Hypertension/drug therapy
5.
J Tissue Eng Regen Med ; 14(3): 464-474, 2020 03.
Article in English | MEDLINE | ID: mdl-31840422

ABSTRACT

Large bone defects represent a significant unmet medical challenge. Cost effectiveness and better stability make small molecule organic compounds a more promising alternative compared with biomacromolecules, for example, growth factors/hormones, in regenerative medicine. However, one common challenge for the application of these small compounds is their side-effect issue. Phenamil is emerging as an intriguing small molecule to promote bone repair by strongly activating bone morphogenetic protein signaling pathway. In addition to osteogenesis, phenamil also induces significant adipogenesis based on some in vitro studies, which is a concern that impedes it from potential clinical applications. Besides the soluble chemical signals, cellular differentiation is heavily dependent on the microenvironments provided by the 3D scaffolds. Therefore, we developed a 3D nanofibrous biomimetic scaffold-based strategy to harness the phenamil-induced stem cell lineage differentiation. Based on the gene expression, alkaline phosphatase activity, and mineralization data, we indicated that bone-matrix mimicking mineralized-gelatin nanofibrous scaffold effectively improved phenamil-induced osteoblastic differentiation, while mitigating the adipogenic differentiation in vitro. In addition to normal culture conditions, we also indicated that mineralized matrix can significantly improve phenamil-induced osteoblastic differentiation in simulated inflammatory condition. In viewing of the crucial role of mineralized matrix, we developed an innovative and facile mineral deposition-based strategy to sustain release of phenamil from 3D scaffolds for efficient local bone regeneration. Overall, our study demonstrated that biomaterials played a crucial role in modulating small molecule drug phenamil-induced osteoblastic differentiation by providing a bone-matrix mimicking mineralized gelatin nanofibrous scaffolds.


Subject(s)
Adipogenesis/drug effects , Amiloride/analogs & derivatives , Cell Differentiation/drug effects , Nanofibers/chemistry , Osteoblasts/metabolism , Tissue Scaffolds/chemistry , Amiloride/chemistry , Amiloride/pharmacokinetics , Amiloride/pharmacology , Animals , Cell Line , Mice , Osteoblasts/cytology
6.
Artif Cells Nanomed Biotechnol ; 46(sup3): S192-S207, 2018.
Article in English | MEDLINE | ID: mdl-30032652

ABSTRACT

AIM: To develop a nanoemulsion-nanoformulation in order to enhance brain bioavailability for Amiloride (Amilo) via intranasal (i.n.) drug delivery in the brain. MATERIAL AND METHODS: Oleic Acid, Tween-20 and Carbitol were selected as oil, surfactant and co-surfactant, respectively. For nanoemulsion preparation, an aqueous micro titration method followed by a high energy ultra-sonication method was used whereas three-factor three-level central composite design was employed to get the best formulation. The independent variables selected for the optimization were %oil, % Surfactant and co-surfactant (Smix) and sonication time (seconds). RESULTS: Based on the constraints applied for independent and dependent variables, the optimized formulation was selected with 2.5% oil, 10% Smix and a sonication time of 45 s. The experimental values observed for dependent variables such as hydrodynamic diameter (nm), % transmittance and % cumulative drug release were found to be 89.36 ± 11.18 nm, 99.23 ± 0.84% and 80.36 ± 5.48%, respectively. Results showed; a spherical shape (transmission electron microscopy and scanning electron microscopy - assisted morphological characterization), polydispersity index (0.231 ± 0.018), zeta potential (-9.83 ± 0.12 mV), refractive index (1.38 ± 0.042), viscosity (41 ± 5 cp), pH (6.4 ± 0.18) and drug content of 98.28 ± 0.29%, for optimized Amiloride-loaded-Nanoemulsion (Amilo-NE). For bioavailability evaluation, ultra-performance liquid chromatography-mass spectroscopy based bioanalytical method was developed and validated for pharmacokinetics, biodistribution, brain-targeting efficiency (1992.67 ± 45.63%) and nose-to-brain transport (586.18 ± 11.63%) whereby an enhanced Amilo-brain bioavailability was observed as compared to intravenous administration (i.v.). Furthermore, Amilo-NE enhanced the treatment in seizure threshold i.e. both rodent models of epilepsy (increasing current electroshock and pentylenetetrazole) induced seizures in mice. CONCLUSION: A significant role of Amilo-NE as observed after high targeting potential and efficiency of the formulation supports the easy brain targeting for Amilo-NE.


Subject(s)
Amiloride , Epilepsy , Nanoparticles , Ultrasonic Waves , Administration, Intranasal , Amiloride/chemistry , Amiloride/pharmacokinetics , Amiloride/pharmacology , Animals , Emulsions , Epilepsy/drug therapy , Epilepsy/metabolism , Epilepsy/pathology , Epilepsy/physiopathology , Ethylene Glycols/chemistry , Humans , Male , Mice , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Particle Size , Polysorbates/chemistry , Tissue Distribution
7.
Acta Biomater ; 60: 93-108, 2017 09 15.
Article in English | MEDLINE | ID: mdl-28713017

ABSTRACT

Inducing differentiation and maturation of resident multipotent stem cells (MSCs) is an important strategy to regenerate hard tissues in mal-calcification conditions. Here we explore a co-delivery approach of therapeutic molecules comprised of ion and drug through a mesoporous bioglass nanoparticle (MBN) for this purpose. Recently, MBN has offered unique potential as a nanocarrier for hard tissues, in terms of high mesoporosity, bone bioactivity (and possibly degradability), tunable delivery of biomolecules, and ionic modification. Herein Sr ion is structurally doped to MBN while drug Phenamil is externally loaded as a small molecule activator of BMP signaling, for the stimulation of osteo/odontogenesis and mineralization of human MSCs derived from dental pulp. The Sr-doped MBN (85Si:10Ca:5Sr) sol-gel processed presents a high mesoporosity with a pore size of ∼6nm. In particular, Sr ion is released slowly at a daily rate of ∼3ppm per mg nanoparticles for up to 7days, a level therapeutically effective for cellular stimulation. The Sr-MBN is internalized to most MSCs via an ATP dependent macropinocytosis within hours, increasing the intracellular levels of Sr, Ca and Si ions. Phenamil is loaded maximally ∼30% into Sr-MBN and then released slowly for up to 7days. The co-delivered molecules (Sr ion and Phenamil drug) have profound effects on the differentiation and maturation of cells, i.e., significantly enhancing expression of osteo/odontogenic genes, alkaline phosphatase activity, and mineralization of cells. Of note, the stimulation is a result of a synergism of Sr and Phenamil, through a Trb3-dependent BMP signaling pathway. This biological synergism is further evidenced in vivo in a mal-calcification condition involving an extracted tooth implantation in dorsal subcutaneous tissues of rats. Six weeks post operation evidences the osseous-dentinal hard tissue formation, which is significantly stimulated by the Sr/Phenamil delivery, based on histomorphometric and micro-computed tomographic analyses. The bioactive nanoparticles releasing both Sr ion and Phenamil drug are considered to be a promising therapeutic nanocarrier platform for hard tissue regeneration. Furthermore, this novel ion/drug co-delivery concept through nanoparticles can be extensively used for other tissues that require different therapeutic treatment. STATEMENT OF SIGNIFICANCE: This study reports a novel design concept in inorganic nanoparticle delivery system for hard tissues - the co-delivery of therapeutic molecules comprised of ion (Sr) and drug (Phenamil) through a unique nanoparticle of mesoporous bioactive glass (MBN). The physico-chemical and biological properties of MBN enabled an effective loading of both therapeutic molecules and a subsequently sustained/controlled release. The co-delivered Sr and Phenamil demonstrated significant stimulation of adult stem cell differentiation in vitro and osseous/dentinal regeneration in vivo, through BMP signaling pathways. We consider the current combination of Sr ion with Phenamil is suited for the osteo/odontogenesis of stem cells for hard tissue regeneration, and further, this ion/drug co-delivery concept can extend the applications to other areas that require specific cellular and tissue functions.


Subject(s)
Bone Morphogenetic Proteins , Calcification, Physiologic/drug effects , Drug Carriers , Mesenchymal Stem Cells/metabolism , Nanoparticles , Signal Transduction/drug effects , Strontium , Amiloride/analogs & derivatives , Amiloride/chemistry , Amiloride/pharmacokinetics , Amiloride/pharmacology , Cell Culture Techniques , Cells, Cultured , Drug Carriers/chemistry , Drug Carriers/pharmacokinetics , Drug Carriers/pharmacology , Humans , Mesenchymal Stem Cells/cytology , Nanoparticles/chemistry , Nanoparticles/therapeutic use , Odontogenesis/drug effects , Osteogenesis/drug effects , Porosity , Strontium/chemistry , Strontium/pharmacokinetics , Strontium/pharmacology
8.
Tissue Eng Part A ; 23(5-6): 195-207, 2017 03.
Article in English | MEDLINE | ID: mdl-27771997

ABSTRACT

Growth factor-based therapeutics using bone morphogenetic protein 2 (BMP-2) presents a promising strategy to reconstruct craniofacial bone defects such as mandible. However, clinical applications require supraphysiological BMP doses that often increase inappropriate adipogenesis, resulting in well-documented, cyst-like bone formation. Here we reported a novel complementary strategy to enhance osteogenesis and mandibular bone repair by using small-molecule phenamil that has been shown to be a strong activator of BMP signaling. Phenamil synergistically induced osteogenic differentiation of human bone marrow mesenchymal stem cells with BMP-2 while suppressing their adipogenic differentiation induced by BMP-2 in vitro. The observed pro-osteogenic and antiadipogenic activity of phenamil was mediated by expression of tribbles homolog 3 (Trb3) that enhanced BMP-smad signaling and inhibited expression of peroxisome proliferator-activated receptor gamma (PPARγ), a master regulator of adipogenesis. The synergistic effect of BMP-2+phenamil on bone regeneration was further confirmed in a critical-sized rat mandibular bone defect by implanting polymer scaffolds designed to slowly release the therapeutic molecules. These findings indicate a new complementary osteoinductive strategy to improve clinical efficacy and safety of current BMP-based therapeutics.


Subject(s)
Amiloride/analogs & derivatives , Bone Morphogenetic Protein 2 , Mandible/metabolism , Mandibular Injuries/drug therapy , Amiloride/pharmacokinetics , Amiloride/pharmacology , Animals , Bone Morphogenetic Protein 2/pharmacokinetics , Bone Morphogenetic Protein 2/pharmacology , Drug Implants/pharmacokinetics , Drug Implants/pharmacology , Humans , Mandible/pathology , Mandibular Injuries/metabolism , Mandibular Injuries/pathology , Mesenchymal Stem Cells/metabolism , Mesenchymal Stem Cells/pathology , Rats, Sprague-Dawley
9.
J Cyst Fibros ; 15(3): 295-301, 2016 05.
Article in English | MEDLINE | ID: mdl-26677762

ABSTRACT

BACKGROUND: In CF patients, the defective ion transport causes a simultaneous reduction of fluid, Cl(-) and HCO3(-) secretion. We aimed to demonstrate that the resulting altered properties of mucus can be recovered using lumacaftor, a CFTR corrector. METHODS: The micro-rheology of non-CF and CF mucus was analysed using Multiple Particle Tracking. RESULTS: The diffusion coefficient of nano-beads imbedded in mucus from CF human bronchial epithelium was lower than in non-CF mucus, and the elastic and viscous moduli were higher. We found that 25% correction of F508del-CFTR mutation with lumacaftor was enough to improve significantly CF mucus properties. Surprisingly, also incubation with amiloride, a compound that reduces fluid absorption but might not change the secretion of HCO3(-) towards the airway surface fluid, improved CF mucus properties. CONCLUSION: CF mucus properties can be recovered by either improving the hydration of the airways or recovering Cl(-) and HCO3(-) secretion across the mutated protein treated with a corrector compound.


Subject(s)
Amiloride , Aminopyridines , Benzodioxoles , Cystic Fibrosis Transmembrane Conductance Regulator/genetics , Cystic Fibrosis , Mucus , Respiratory Mucosa , Amiloride/administration & dosage , Amiloride/pharmacokinetics , Aminopyridines/administration & dosage , Aminopyridines/pharmacokinetics , Benzodioxoles/administration & dosage , Benzodioxoles/pharmacokinetics , Biological Availability , Cell Culture Techniques , Cystic Fibrosis/drug therapy , Cystic Fibrosis/genetics , Epithelial Sodium Channel Blockers/administration & dosage , Epithelial Sodium Channel Blockers/pharmacokinetics , Humans , Microfluidics/methods , Models, Theoretical , Mucus/drug effects , Mucus/metabolism , Mutant Proteins/genetics , Respiratory Mucosa/drug effects , Respiratory Mucosa/metabolism
10.
Rev. esp. cardiol. (Ed. impr.) ; 68(12): 1101-1110, dic. 2015. ilus, tab, graf
Article in Spanish | IBECS | ID: ibc-145616

ABSTRACT

Introducción y objetivos: Se han implicado diversos mecanismos en la respuesta mecánica al estiramiento miocárdico, que incluyen la activación del intercambiador Na+/H+ por acciones autocrinas y paracrinas. Se estudia la participación de estos mecanismos en las respuestas electrofisiológicas al estiramiento agudo miocárdico mediante el análisis de los cambios inducidos con fármacos. Métodos: Se analizan las modificaciones de la fibrilación ventricular inducidas por el estiramiento agudo miocárdico en corazones de conejo aislados y perfundidos utilizando electrodos múltiples epicárdicos y técnicas cartográficas. Se estudian 4 series: control (n = 9); durante la perfusión del antagonista de los receptores de la angiotensina II, losartán (1 miM, n = 8); durante la perfusión del bloqueador del receptor de la endotelina A, BQ-123 (0,1 miM, n = 9), y durante la perfusión del inhibidor del intercambiador Na+/H+, EIPA (5-[N-ethyl-N-isopropyl]-amiloride) (1 miM, n = 9). Resultados: EIPA atenuó el aumento de la frecuencia dominante de la fibrilación producido por el estiramiento (control = 40,4%; losartán = 36% [no significativo]; BQ-123 = 46% [no significativo], y EIPA = 22% [p < 0,001]). Durante el estiramiento, la complejidad de los mapas de activación fue menor en la serie con EIPA (p < 0,0001) y también en esta serie fue mayor la concentración espectral de la arritmia (mayor regularidad): control = 18 ± 3%; EIPA = 26 ± 9% (p < 0,02); losartán = 18 ± 5% (no significativo), y BQ-123 = 18 ± 4% (no significativo). Conclusiones: El inhibidor del intercambiador Na+/H+ EIPA atenúa los efectos electrofisiológicos responsables de la aceleración y del aumento de la complejidad de la fibrilación ventricular producidos por el estiramiento agudo miocárdico. Por el contrario, el antagonista de los receptores de la angiotensina II, losartán, y el del receptor A de la endotelina, BQ-123, no modifican estos efectos (AU)


Introduction and objectives: Mechanical response to myocardial stretch has been explained by various mechanisms, which include Na+ /H+ exchanger activation by autocrine-paracrine system activity. Drug-induced changes were analyzed to investigate the role of these mechanisms in the electrophysiological responses to acute myocardial stretch. Methods: Multiple epicardial electrodes and mapping techniques were used to analyze changes in ventricular fibrillation induced by acute myocardial stretch in isolated perfused rabbit hearts. Four series were studied: control (n = 9); during perfusion with the angiotensin receptor blocker losartan (1 mM, n = 8); during perfusion with the endothelin A receptor blocker BQ-123 (0.1 mM, n = 9), and during perfusion with the Na+ /H+ exchanger inhibitor EIPA (5-[N-ethyl-N-isopropyl]-amiloride) (1 mM, n = 9). Results: EIPA attenuated the increase in the dominant frequency of stretch-induced fibrillation (control = 40.4%; losartan = 36% [not significant]; BQ-123 = 46% [not significant]; and EIPA = 22% [P < .001]). During stretch, the activation maps were less complex (P < .0001) and the spectral concentration of the arrhythmia was greater (greater regularity) in the EIPA series: control = 18 (3%); EIPA = 26 (9%) (P < .02); losartan = 18 (5%) (not significant); and BQ-123 = 18 (4%) (not significant). Conclusions: The Na+ /H+ exchanger inhibitor EIPA attenuated the electrophysiological effects responsible for the acceleration and increased complexity of ventricular fibrillation induced by acute myocardial stretch. The angiotensin II receptor antagonist losartan and the endothelin A receptor blocker BQ-123 did not modify these effects (AU)


Subject(s)
Humans , Losartan/pharmacokinetics , Amiloride/pharmacokinetics , /pharmacokinetics , Arrhythmias, Cardiac/drug therapy , Ventricular Fibrillation/drug therapy , 28573 , Endothelin Receptor Antagonists/pharmacokinetics , Endoplasmic Reticulum Stress , Myocardial Revascularization , Cardiac Electrophysiology/methods
11.
Br J Pharmacol ; 171(22): 5076-92, 2014 Nov.
Article in English | MEDLINE | ID: mdl-24962208

ABSTRACT

BACKGROUND AND PURPOSE: Previously, a systems pharmacology model was developed characterizing drug effects on the interrelationship between mean arterial pressure (MAP), cardiac output (CO) and total peripheral resistance (TPR). The present investigation aims to (i) extend the previously developed model by parsing CO into heart rate (HR) and stroke volume (SV) and (ii) evaluate if the mechanism of action (MoA) of new compounds can be elucidated using only HR and MAP measurements. EXPERIMENTAL APPROACH: Cardiovascular effects of eight drugs with diverse MoAs (amiloride, amlodipine, atropine, enalapril, fasudil, hydrochlorothiazide, prazosin and propranolol) were characterized in spontaneously hypertensive rats (SHR) and normotensive Wistar-Kyoto (WKY) rats following single administrations of a range of doses. Rats were instrumented with ascending aortic flow probes and aortic catheters/radiotransmitters for continuous recording of MAP, HR and CO throughout the experiments. Data were analysed in conjunction with independent information on the time course of the drug concentration following a mechanism-based pharmacokinetic-pharmacodynamic modelling approach. KEY RESULTS: The extended model, which quantified changes in TPR, HR and SV with negative feedback through MAP, adequately described the cardiovascular effects of the drugs while accounting for circadian variations and handling effects. CONCLUSIONS AND IMPLICATIONS: A systems pharmacology model characterizing the interrelationship between MAP, CO, HR, SV and TPR was obtained in hypertensive and normotensive rats. This extended model can quantify dynamic changes in the CVS and elucidate the MoA for novel compounds, with one site of action, using only HR and MAP measurements. Whether the model can be applied for compounds with a more complex MoA remains to be established.


Subject(s)
Hemodynamics/drug effects , Hypertension/metabolism , Models, Biological , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/analogs & derivatives , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacokinetics , 1-(5-Isoquinolinesulfonyl)-2-Methylpiperazine/pharmacology , Amiloride/pharmacokinetics , Amiloride/pharmacology , Amlodipine/pharmacokinetics , Amlodipine/pharmacology , Animals , Atropine/pharmacokinetics , Atropine/pharmacology , Enalapril/pharmacokinetics , Enalapril/pharmacology , Hydrochlorothiazide/pharmacokinetics , Hydrochlorothiazide/pharmacology , Male , Prazosin/pharmacokinetics , Prazosin/pharmacology , Propranolol/pharmacokinetics , Propranolol/pharmacology , Rats, Inbred SHR , Rats, Inbred WKY
12.
Pulm Pharmacol Ther ; 27(1): 76-82, 2014 Feb.
Article in English | MEDLINE | ID: mdl-23932897

ABSTRACT

Although the airway surface is the anatomic target for many lung disease therapies, measuring drug concentrations and activities on these surfaces poses considerable challenges. We tested whether mass spectrometric analysis of exhaled breath condensate (EBC) could be utilized to non-invasively measure airway drug pharmacokinetics and predicted pharmacological activities. Mass spectrometric methods were developed to detect a novel epithelial sodium channel blocker (GS-9411/P-680), two metabolites, a chemically related internal standard, plus naturally occurring solutes including urea as a dilution marker. These methods were then applied to EBC and serum collected from four (Floridian) sheep before, during and after inhalation of nebulized GS-9411/P-680. Electrolyte content of EBC and serum was also assessed as a potential pharmacodynamic marker of drug activity. Airway surface concentrations of drug, metabolites, and electrolytes were calculated from EBC measures using EBC:serum urea based dilution factors. GS-9411/P-680 and its metabolites were quantifiable in the sheep EBC, with peak airway concentrations between 1.9 and 3.4 µM measured 1 h after inhalation. In serum, only Metabolite #1 was quantifiable, with peak concentrations ∼60-fold lower than those in the airway (45 nM at 1 h). EBC electrolyte concentrations suggested a pharmacological effect; but this effect was not statistical significant. Analysis of EBC collected during an inhalation drug study provided a method for quantification of airway drug and metabolites via mass spectrometry. Application of this methodology could provide an important tool in development and testing of drugs for airways diseases.


Subject(s)
Amiloride/analogs & derivatives , Mass Spectrometry/methods , Sodium Channel Blockers/pharmacokinetics , Urea/metabolism , Administration, Inhalation , Amiloride/administration & dosage , Amiloride/pharmacokinetics , Amiloride/pharmacology , Animals , Biomarkers/metabolism , Breath Tests , Female , Sheep , Sodium Channel Blockers/administration & dosage , Sodium Channel Blockers/pharmacology , Tissue Distribution
13.
Eur Respir J ; 42(2): 389-93, 2013 Aug.
Article in English | MEDLINE | ID: mdl-23100510

ABSTRACT

Nasal potential difference (NPD) quantifies abnormal ion transport in cystic fibrosis. It has gained acceptance as an outcome measure for the investigation of new therapies. To quantify the effect of solution temperature on NPD, we first examined the effect of switching from room temperature (20-25°C) to warmed (32-37°C) solutions and vice versa during each perfusion step. Secondly, standard protocols were repeated at both temperatures in the same subjects. Changing solution temperature did not alter NPD during perfusion with Ringer's solution (<1 mV) (p>0.1). During perfusion with zero chloride solution, changing from room temperature to warmed solutions tended to decrease absolute NPD (i.e. it became less negative) by 0.9 mV (p>0.1); changing from warmed to room temperature increased NPD by 2.1 mV (p<0.05). During isoprenaline perfusion, changing from room temperature to warmed solutions increased NPD by 1.5 mV (p<0.01) and from warmed to room temperature decreased NPD by 1.4 mV (p<0.05). For full protocols at room temperature or warmed in the same subjects, mean values were similar (n = 24). During warmed perfusion, group results for total chloride response had a larger standard deviation. As this increased variability will probably decrease the power of trials, this study suggests that solutions at room temperature should be recommended for the measurement of NPD.


Subject(s)
Chloride Channels/drug effects , Chlorides/pharmacokinetics , Nasal Mucosa/physiology , Adolescent , Adult , Amiloride/pharmacokinetics , Cystic Fibrosis/pathology , Healthy Volunteers , Humans , Ion Transport , Ions , Isoproterenol/pharmacokinetics , Middle Aged , Perfusion , Temperature , Time Factors , Young Adult
15.
Small ; 8(6): 943-8, 2012 Mar 26.
Article in English | MEDLINE | ID: mdl-22315201

ABSTRACT

The concept of a long-term sensor for ion changes in the lysosome is presented. The sensor is made by layer-by-layer assembly of oppositely charged polyelectrolytes around ion-sensitive fluorophores, in this case for protons. The sensor is spontaneously incorporated by cells and resides over days in the lysosome. Intracellular changes of the concentration of protons upon cellular stimulation with pH-active agents are monitored by read-out of the sensor fluorescence at real time. With help of this sensor concept it is demonstrated that the different agents used (Monensin, Chloroquine, Bafilomycin A1, Amiloride) possessed different kinetics and mechanisms of action in affecting the intracellular pH values.


Subject(s)
Hydrogen-Ion Concentration , Lysosomes/metabolism , Amiloride/pharmacokinetics , Chloroquine/pharmacokinetics , Fluorescence , Macrolides/pharmacokinetics , Monensin/pharmacokinetics
16.
Clin Pharmacol Ther ; 89(4): 587-94, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21368750

ABSTRACT

Thiazolidinediones (TZDs) are associated with fluid retention that has been suggested to be resistant to treatment with loop diuretics. This resistance is thought to be caused by upregulation of renal epithelial sodium channels (ENaCs). In this study, we tested whether these mechanisms are of clinical significance. We conducted a well-controlled study in 12 insulin-resistant nondiabetic participants, who received treatment for 9 weeks with either rosiglitazone at a dosage of 4 mg b.i.d. or placebo. The aim of the study was to investigate whether upregulation of ENaCs by rosiglitazone reduces furosemide's natriuretic response and enhances the response to the ENaC inhibitor amiloride. The natriuretic response to furosemide and amiloride and the amount of α-ENaC in urinary exosomes were quantified. Rosiglitazone neither reduced furosemide-induced natriuresis nor changed furosemide's concentration-effect curve. Furthermore, rosiglitazone did not change either amiloride-induced natriuresis nor the amount of urinary α-ENaC. This study challenges previous findings regarding TZD-related ENaC upregulation and suggests that TZD-induced fluid retention should respond normally to loop diuretics.


Subject(s)
Diuretics/pharmacology , Epithelial Sodium Channels/drug effects , Hypoglycemic Agents/pharmacology , Insulin Resistance , Thiazolidinediones/pharmacology , Adult , Amiloride/pharmacokinetics , Amiloride/pharmacology , Cross-Over Studies , Double-Blind Method , Drug Interactions , Epithelial Sodium Channels/metabolism , Exosomes/metabolism , Female , Furosemide/pharmacokinetics , Furosemide/pharmacology , Humans , Kidney/drug effects , Kidney/metabolism , Male , Middle Aged , Natriuresis/drug effects , Rosiglitazone , Up-Regulation/drug effects
17.
Gen Comp Endocrinol ; 169(3): 197-202, 2010 Dec 01.
Article in English | MEDLINE | ID: mdl-20800063

ABSTRACT

In Rhinella arenarum spermiation occurs as a consequence of LH/FSH increase during the amplexus or by a single dose of hCG, among other gonadotropins. The present study employs an in vitro system to study the mechanism of action of hCG in the spermiation of R. arenarum. Testicular fragments were incubated for 2h at 28°C in the presence or absence of 20IU hCG with or without different PKA/PKC inhibitors and activators as well as ouabain and amiloride as Na(+)/K(+) ATPase and transcellular Na(+) transport inhibitors, respectively. Ouabain did not induce spermiation in absence of hCG and inhibited hCG-induced spermiation in a dose-dependent manner, reaching 90% inhibition with the higher concentration. In contrast, amiloride neither affected spermiation nor steroidogenesis. Activation of PKA with 8Br-cAMP induced spermiation in the absence of hCG while its inhibition with H89 blocked hCG action. On the other hand, PKC inhibition with Bi or STP did not affect hCG-induced spermiation although PKC activation significantly decreased hCG-dependent sperm release. These results suggest that PKC inhibits spermiation but also that the inhibition exerted by the kinase could be blocked by hCG. Taken together, these observations could indicate that PKA is involved in the mechanism of the gonadotropin action, mechanism also requiring the activation of a non-pumping Na(+)/K(+) ATPase pathway.


Subject(s)
Bufo arenarum/physiology , Chorionic Gonadotropin/pharmacology , Ouabain/pharmacology , Spermatozoa/metabolism , 8-Bromo Cyclic Adenosine Monophosphate/pharmacology , Amiloride/pharmacokinetics , Animals , Cyclic AMP-Dependent Protein Kinases/metabolism , Indoles/pharmacology , Isoquinolines/pharmacology , Male , Maleimides/pharmacology , Protein Kinase C/metabolism , Sertoli Cells/drug effects , Sodium-Potassium-Exchanging ATPase/antagonists & inhibitors , Spermatozoa/drug effects , Staurosporine/pharmacology , Sulfonamides/pharmacology , Tetradecanoylphorbol Acetate/pharmacology
18.
Rapid Commun Mass Spectrom ; 21(21): 3427-34, 2007.
Article in English | MEDLINE | ID: mdl-17902196

ABSTRACT

A new method for simultaneous determination of amiloride and hydrochlorothiazide by liquid chromatography/electrospray tandem mass spectrometry (LC/MS/MS) operated in positive and negative ionization switching mode was developed and validated. Protein precipitation with acetonitrile was selected for sample preparation. The analytes were separated on a Phenomenex Curosil-PFP (250x4.6 mm, 5 microm) column by a gradient elution with a mobile phase consisting of 0.15% formic acid solution containing 0.23% ammonium acetate and methanol pumped at a flow rate of 1.0 mL.min(-1). Rizatriptan was used as the internal standard (IS) for quantification. The determination was carried out on a Waters Quattro-micro triple-quadrupole mass spectrometer operated in multiple reaction monitoring (MRM) mode using the following transitions monitored simultaneously: positive m/z 230-->171 for amiloride, m/z 270-->158 for rizatriptan, and negative m/z 296-->205 for hydrochlorothiazide. The lower limits of quantification (LLOQs) were 0.1 and 1.0 ng.mL(-1) for amiloride and hydrochlorothiazide, respectively, which were lower than other published methods by using ultraviolet (UV), fluorimetric or mass spectrometric detection. The intra- and inter-day precision and accuracy were studied at three different concentration levels and were always better than 15% (n=5). This simple and robust LC/MS/MS method was successfully applied to the pharmacokinetic study of compound amiloride and hydrochlorothiazide tablets in healthy male Chinese volunteers.


Subject(s)
Amiloride/blood , Diuretics/blood , Hydrochlorothiazide/blood , Spectrometry, Mass, Electrospray Ionization/methods , Tandem Mass Spectrometry/methods , Administration, Oral , Amiloride/pharmacokinetics , Chromatography, High Pressure Liquid , Diuretics/pharmacokinetics , Drug Therapy, Combination , Humans , Hydrochlorothiazide/pharmacokinetics , Male , Reproducibility of Results , Serotonin Receptor Agonists/blood , Triazoles/blood , Tryptamines/blood
19.
J Pharmacokinet Pharmacodyn ; 34(5): 711-26, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17653836

ABSTRACT

PURPOSE: To compare the performance of the standard lag time model (LAG model) with the performance of an analytical solution of the transit compartment model (TRANSIT model) in the evaluation of four pharmacokinetic studies with four different compounds. METHODS: The population pharmacokinetic analyses were performed using NONMEM on concentration-time data of glibenclamide, furosemide, amiloride, and moxonidine. In the TRANSIT model, the optimal number of transit compartments was estimated from the data. This was based on an analytical solution for the change in drug concentration arising from a series of transit compartments with the same first-order transfer rate between each compartment. Goodness-of-fit was assessed by the decrease in objective function value (OFV) and by inspection of diagnostic graphs. RESULTS: With the TRANSIT model, the OFV was significantly lower and the goodness-of-fit was markedly improved in the absorption phase compared with the LAG model for all drugs. The parameter estimates related to the absorption differed between the two models while the estimates of the pharmacokinetic disposition parameters were similar. CONCLUSION: Based on these results, the TRANSIT model is an attractive alternative for modeling drug absorption delay, especially when a LAG model poorly describes the drug absorption phase or is numerically unstable.


Subject(s)
Models, Biological , Pharmacokinetics , Absorption , Adult , Aged , Aged, 80 and over , Amiloride/pharmacokinetics , Female , Furosemide/pharmacokinetics , Glyburide/pharmacokinetics , Humans , Imidazoles/pharmacokinetics , Male , Middle Aged , Nonlinear Dynamics
20.
J Nanosci Nanotechnol ; 6(9-10): 3001-9, 2006.
Article in English | MEDLINE | ID: mdl-17048511

ABSTRACT

The purpose of this study was to encapsulate Amiloride Hydrochloride into nano-liposomes, incorporate it into dry powder inhaler, and to provide prolonged effective concentration in airways to enhance mucociliary clearance and prevent secondary infection in cystic fibrosis. Liposomes were prepared by thin film hydration technique and then dispersion was passed through high pressure homogenizer to achieve size of nanometer range. Nano-liposomes were separated by centrifugation and were characterized. They were dispersed in phosphate buffer saline pH 7.4 containing carriers (lactose/sucrose/mannitol), and glycine as anti-adherent. The resultant dispersion was spray dried. The spray dried powders were characterized and in vitro drug release studies were performed using phosphate buffer saline pH 7.4. In vitro and in vivo drug pulmonary deposition was carried out using Andersen Cascade Impactor and by estimating drug in bronchial alveolar lavage and lung homogenate after intratracheal instillation in rats respectively. Nano-liposomes were found to have mean volume diameter of 198 +/- 15 nm, and 57% +/- 1.9% of drug entrapment. Mannitol based formulation was found to have low density, good flowability, particle size of 6.7 +/- 0.6 microm determined by Malvern MasterSizer, maximum fine particle fraction of 67.6 +/- 0.6%, mean mass aerodynamic diameter 2.3 +/- 0.1 microm, and geometric standard deviation 2.4 +/- 0.1. Developed formulations were found to have prolonged drug release following Higuchi's Controlled Release model and in vivo studies showed maximal retention time of drug of 12 hrs within the lungs and slow clearance from the lungs. This study provides a practical approach for direct lung delivery of Amiloride Hydrochloride encapsulated in liposomes for controlled and prolonged retention at the site of action from dry powder inhaler. It can provide a promising alternative to the presently available nebulizers in terms of prolonged pharmacological effect, reducing systemic side effects such as potassium retention due to rapid clearance of the drug from lungs in patients suffering from cystic fibrosis.


Subject(s)
Amiloride/pharmacokinetics , Delayed-Action Preparations/chemistry , Drug Carriers/chemistry , Liposomes/chemistry , Lung/metabolism , Nanostructures/chemistry , Nebulizers and Vaporizers , Administration, Inhalation , Aerosols/administration & dosage , Aerosols/chemistry , Amiloride/administration & dosage , Amiloride/chemistry , Animals , Delayed-Action Preparations/administration & dosage , Delayed-Action Preparations/pharmacokinetics , Desiccation , Metabolic Clearance Rate , Nanostructures/ultrastructure , Particle Size , Powders , Rats , Sodium Channel Blockers/administration & dosage , Sodium Channel Blockers/chemistry , Sodium Channel Blockers/pharmacokinetics , Tissue Distribution
SELECTION OF CITATIONS
SEARCH DETAIL
...