1
|
Lemke J, Gollasch M, Tsvetkov D, Schulig L. Advances in the design and development of chemical modulators of the voltage-gated potassium channels K V7.4 and K V7.5. Expert Opin Drug Discov 2025; 20:47-62. [PMID: 39627683 DOI: 10.1080/17460441.2024.2438226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 10/20/2024] [Accepted: 12/02/2024] [Indexed: 12/10/2024]
Abstract
INTRODUCTION Hypertension remains a major public health concern, with significant morbidity and mortality worldwide. Despite the availability of various antihypertensive medications, blood pressure control remains suboptimal in many individuals. During the last decades, KV7.4 and KV7.5, which were already known from the view of neuronal regulation, emerged as possible important players in the regulation of vascular tone and blood pressure. AREAS COVERED This review covers physiological functions and current advancements in the development of KV7.4 and KV7.5 channel modulators. The authors highlight the structural elements likely to be important for the future design of KV7 subtype-selective modulators, underscoring their potential as an innovative hypertension treatment. EXPERT OPINION Extensive research has been focused on targeting neuronal KV7.2 and KV7.3 channels, while KV7.4 and KV7.5 attracted less attention. Many of the developed compounds represent derivatives of flupirtine or retigabine, whereby subtype channel selectivity has only been demonstrated for a handful of individual compounds. Novel substances address additional sites within the binding pocket by incorporating new functional groups. A comprehensive and systematic evaluation of a compound set with significant subtype selectivity should be performed. The discovery of new highly active, less toxic, and selective compounds, therefore, remains the goal of further research in the coming years.
Collapse
Affiliation(s)
- Jana Lemke
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, University of Greifswald, Greifswald, Germany
| | - Maik Gollasch
- Department of Internal Medicine and Geriatrics, University Medicine, Greifswald, Germany
| | - Dmitry Tsvetkov
- Department of Internal Medicine and Geriatrics, University Medicine, Greifswald, Germany
| | - Lukas Schulig
- Department of Pharmaceutical and Medicinal Chemistry, Institute of Pharmacy, University of Greifswald, Greifswald, Germany
| |
Collapse
|
2
|
Rithanyaa R, Radhakrishnan N. In silico Analysis of Selected Four-Leaf Water Clover ( Marsilea species) Constituents as Human Acetyl Cholinesterase (hAchE), Carbonic Anhydrase II (hCA-II), and Protein Tyrosine Phosphatase 1B (PTP1B) Inhibitory Agents. JOURNAL OF PHARMACY AND BIOALLIED SCIENCES 2024; 16:S1237-S1241. [PMID: 38882880 PMCID: PMC11174185 DOI: 10.4103/jpbs.jpbs_549_23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2023] [Revised: 11/03/2023] [Accepted: 11/05/2023] [Indexed: 06/18/2024] Open
Abstract
A four-leaf water clover (Marsilea species) has been reported to exhibit various biological activities. In the present study, we aimed to evaluate 23 selected constituents of a four-leaf water clover (Marsilea species) as potent inhibitory agents of human acetyl cholinesterase (hAchE), carbonic anhydrase II (hCA-II), and protein tyrosine phosphatase 1B (hPTP-1B) using an in silico method. The 23 selected constituents of the four-leaf water clover (Marsilea species) were studied on the docking behavior of hAchE, hCA-II, and hPTP-1B by using the Webina docking method. In addition to docking, toxicity analysis was also performed using the pkCSM web server. Toxicity analysis has shown that 10 ligands (44%) of the four-leaf water clover (Marsilea species) were predicted to have hERG II (human ether-a-go-go-related gene) inhibition activity. The docking analysis showed that marsilin has exhibited the maximum binding energy (-11.3 kcal/mol) with the hAchE, whereas it fails to dock with both the target enzymes (hCA-II and hPTP-1B). Thus, the present find provides a new understanding about the 23 selected ligands of the four-leaf water clover (Marsilea species) as potent inhibitory agents of human acetyl cholinesterase (hAchE), carbonic anhydrase II (hCA-II), and protein tyrosine phosphatase 1B (hPTP-1B).
Collapse
Affiliation(s)
- Ramesh Rithanyaa
- Department of Biochemistry, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (Deemed to be University), Thandalam, Chennai, Tamil Nadu, India
| | - Narayanaswamy Radhakrishnan
- Department of Biochemistry, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (Deemed to be University), Thandalam, Chennai, Tamil Nadu, India
| |
Collapse
|
3
|
Tamilarasu N, Narayanaswamy R, Prabhakaran VS. In Silico Analysis of Selected Glycyrrhiza glabra (Licorice) Constituents: Exploring Their Modulatory Effects on Human Superoxide Dismutase, Human Phosphodiesterase-9 and Human Dipeptidyl Peptidase-4. Cureus 2024; 16:e53494. [PMID: 38440026 PMCID: PMC10911055 DOI: 10.7759/cureus.53494] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Accepted: 02/02/2024] [Indexed: 03/06/2024] Open
Abstract
Background Glycyrrhiza glabra (Licorice) has been known for its various biological activities. In the current investigation, we aimed to evaluate 11 (10 natural and one synthetic) selected constituents of G. glabra as potent modulatory agents of human superoxide dismutase (hSOD), human phosphodiesterase-9 (hPDE 9) and human dipeptidyl peptidase-4 (hDPP 4) using in silico method. Methodology The 11 selected constituents of G. glabra (Licorice) were investigated on the docking behaviour of hSOD, hPDE 9 and hDPP 4 by using the PatchDock method. In addition to docking, toxicity analysis was also carried out using the pkCSM free online server (University of Melbourne, Melbourne, AUS). Results Toxicity analysis has shown that four ligands (36%) of G. glabra (Licorice) are predicted to have human ether-a-go-go-related gene-2 (hERG 2) inhibition activity. The docking analysis showed that glabridin (-224.13 kcal/mol) has shown the highest atomic contact binding energy with the hSOD enzyme, whereas carbenoxolone has shown the maximum atomic contact binding energy with both the hPDE 9 and hDPP 4 enzymes (-239.57 and -173.50 kcal/mol) respectively. Conclusion Thus the present finding provides new information about 11 selected ligands of G. glabra (Licorice) as potent modulatory agents of hSOD, hPDE 9 and hDPP 4.
Collapse
Affiliation(s)
- Naveena Tamilarasu
- Biochemistry, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, IND
| | - Radhakrishnan Narayanaswamy
- Biochemistry, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences, Chennai, IND
| | | |
Collapse
|
4
|
Arulselvan AJ, Manimuthu MS, Narayanaswamy R. Molecular Docking Analysis of Selected Urtica dioica Constituents As Human Carbonic Anhydrase II (hCA-II), Human 11 Beta-Hydroxysteroid Dehydrogenases Type 1 (h11beta-HSD1), and Human Dual Specificity Phosphatase (hCDC25B) Inhibitory Agents. Cureus 2024; 16:e53886. [PMID: 38465091 PMCID: PMC10925072 DOI: 10.7759/cureus.53886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Accepted: 02/08/2024] [Indexed: 03/12/2024] Open
Abstract
Background Urtica dioica (Stinging nettle)has been reported to exhibit various pharmacological activities. In the present study, we aimed to evaluate 24 selected constituents of U. dioica as potent inhibitory agents of human carbonic anhydrase II (hCA-II), human 11 beta-hydroxysteroid dehydrogenases type 1 (h11beta-HSD1), and human dual specificity phosphatase (hCDC25B) using in silico method. Methodology The 24 selected constituents of U. dioica (Stinging nettle) were studied on the docking behavior of hCA-II, h11beta-HSD1, and hCDC25B by using the Webina docking method. In addition to docking, toxicity analysis was also performed using the pkCSM free web server, respectively. Results Toxicity analysis has shown that six ligands (25%) of U. dioica (Stinging nettle) are predicted to have hERG II (Human ether-a-go-go-related gene) inhibition activity. The docking analysis showed that afzelin, stigmastane-3, 6-diol, and astragalin of U. dioica have shown the maximum binding energy (-7.2, -9.5, and -8.5 kcal/mol) with the hCA-II, h11beta-HSD1 and hCDC25B, respectively. Conclusions Thus, the current finding provides new knowledge about the 24 selected ligands of U. dioica (Stinging nettle) as potent inhibitory agents of human carbonic anhydrase II (hCA-II), human 11 beta-hydroxysteroid dehydrogenases type 1 (h11beta-HSD1) and human dual specificity phosphatase (hCDC25B).
Collapse
Affiliation(s)
- Angel Jenifer Arulselvan
- Biochemistry, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (Deemed to be University), Chennai, IND
| | - Mani Sankar Manimuthu
- Biochemistry, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (Deemed to be University), Chennai, IND
| | - Radhakrishnan Narayanaswamy
- Biochemistry, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (Deemed to be University), Chennai, IND
| |
Collapse
|
5
|
Surya Prakash V, Radhakrishnan N, Vasantha-Srinivasan P, Veeramani C, El Newehy AS, Alsaif MA, Al-Numair KS. In silico analysis of selected nutrition rich fruit of Bunch berry ( Lantana camara) constituents as human acetylcholinesterase (hAchE), carbonic anhydrase II (hCA-II) and carboxylesterase 1 (hCES-1) inhibitory agents. Saudi J Biol Sci 2023; 30:103847. [PMID: 37961045 PMCID: PMC10638019 DOI: 10.1016/j.sjbs.2023.103847] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2023] [Revised: 10/11/2023] [Accepted: 10/19/2023] [Indexed: 11/15/2023] Open
Abstract
Background Bunch berry (Lantana camara) is primarily composed of flavonoids and vitamin C; therefore, it has been shown to possess various medical characteristics, including the ability to relieve fever, inflammation, and urinary tract infections. Objective In this study, we intended to assess twenty chosen constituents of Bunch berry as potent inhibitory agents of human acetylcholinesterase (hAchE), carbonic anhydrase II (hCA-II) and carboxylesterase 1 (hCES-1) employing in silico techniques. Methods The twenty chosen Bunch berry components were examined about docking behaviour of hAchE, hCA-II and hCES-I by using the Swissdock method. Apart from to docking, Molecular physico-chemical, drug-likeness, ADME (ingesting, dispersing, metabolising, and excreting), and toxicity assessments were also performed utilising the Molinspiration, Swiss ADME, pkCSM, and STITCH web sites, correspondingly. Results Eight ligands (40 %) have exhibited strict adherence to Lipinski's rule of five (Ro5), according to molecular physico-chemical study. Drug-likeness property analysis has shown that five ligands (25 %) of Bunch berry predicted to exhibit moderate bioactivity score against all the descriptors. ADME analysis has shown that five ligands (25 %) of Bunch berry are predicted to possess high gastrointestinal absorption property Toxicity analysis has shown that six ligands (30 %) of Bunch berry are predicted to have hERG II (Human ether-a-go-go-related gene) inhibition activity. According to the docking analysis, lantic acid has the lowest atomic binding energy for all three target enzymes, hAchE (-6.23 kcal/mol), hCA-II (-4.46 kcal/mol), and hCES-I (-5.99 kcal/mol), respectively. Conclusions Thus the current find provides an advanced understanding the twenty selected ligands of Bunch berry as potent inhibitory agents of human acetylcholinesterase (hAchE), carbonic anhydrase II (hCA-II) and carboxylesterase 1 (hCES-1).
Collapse
Affiliation(s)
- V. Surya Prakash
- Department of Biochemistry, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai, Tamil Nadu 602 105, India
| | - N. Radhakrishnan
- Department of Biochemistry, Saveetha Medical College and Hospital, Saveetha Institute of Medical and Technical Sciences (SIMATS), Thandalam, Chennai, Tamil Nadu 602 105, India
| | - P. Vasantha-Srinivasan
- Department of Bio-Informatics, Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences (Deemed to be University), Chennai 602105, India
| | - Chinnadurai Veeramani
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh 11433, Saudi Arabia
| | - Ahmed S. El Newehy
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh 11433, Saudi Arabia
| | - Mohammed A. Alsaif
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh 11433, Saudi Arabia
| | - Khalid S. Al-Numair
- Department of Community Health Sciences, College of Applied Medical Sciences, King Saud University, P.O. Box 10219, Riyadh 11433, Saudi Arabia
| |
Collapse
|
6
|
Wang Y, Su R, Chen J, Liu X, Luo J, Lao Y, Huang P, Shi J, Jiang C, Liao L, Zhang J. Synthesis of 1,3,5-triphenyl-1,2,4-triazole derivatives and their neuroprotection by anti-oxidative stress and anti-inflammation and protecting BBB. Eur J Med Chem 2023; 260:115742. [PMID: 37651874 DOI: 10.1016/j.ejmech.2023.115742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2023] [Revised: 08/12/2023] [Accepted: 08/16/2023] [Indexed: 09/02/2023]
Abstract
Acute ischemic stroke (AIS) is a serious cardiovascular and cerebrovascular disease; Oxidative stress and neuroinflammation are important factors which destroy blood-brain barrier (BBB) in AIS. In the study, a series of 1,3,5-triphenyl-1,2,4-triazole derivatives were designed and synthesized; the optimal compound 9 was obtained by screening their anti-oxidant and anti-inflammatory effects; the neuroprotection effect of compound 9 was evaluated with a rat middle cerebral artery occlusion (MCAO) model. Subsequently, the mechanism of neuroprotection were explored via Western blot. The results prompt compound 9 maybe exert anti-AIS neuroprotection by inhibiting oxidative stress and neuroinflammation inhibition by inhibiting Keap1, COX-2 and iNOS. At the same time, it can protect BBB by reducing glycocalyx degradation and matrix metallopeptidase-9 levels. Its LD50 > 1000 mg/kg on mice and hERG channel inhibition IC50 > 30 μM, which lower acute toxicity and hERG channel inhibition would make compound 9 a promising stroke treatment candidate.
Collapse
Affiliation(s)
- Yang Wang
- Department of Medicinal Chemistry, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Ruiqi Su
- Department of Medicinal Chemistry, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Jianwen Chen
- Department of Medicinal Chemistry, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Xuan Liu
- Department of Medicinal Chemistry, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Jingning Luo
- Department of Medicinal Chemistry, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Yaoqiang Lao
- Department of Medicinal Chemistry, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Ping Huang
- Department of Medicinal Chemistry, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Jinguo Shi
- Department of Medicinal Chemistry, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Caibao Jiang
- Department of Medicinal Chemistry, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Liping Liao
- Department of Medicinal Chemistry, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, PR China
| | - Jingxia Zhang
- Department of Medicinal Chemistry, School of Pharmaceutical Science, Sun Yat-sen University, Guangzhou, 510006, PR China.
| |
Collapse
|
7
|
Reynolds T, Blagg BSJ. Synthesis and Validation of the First Cell-Impermeable Hsp90α-Selective Inhibitors. ACS Med Chem Lett 2023; 14:1250-1256. [PMID: 37736193 PMCID: PMC10510499 DOI: 10.1021/acsmedchemlett.3c00265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Accepted: 08/02/2023] [Indexed: 09/23/2023] Open
Abstract
Hsp90α is an isoform of the heat shock protein 90 (Hsp90) family of molecular chaperones and mediates the folding and activation of ∼400 client proteins. However, inhibition of intracellular Hsp90α has caused detrimental side effects and significantly hindered the clinical development of Hsp90 inhibitors. As an alternative strategy, 14 Hsp90α-selective inhibitors were synthesized to introduce permanently charged moieties onto the solvent-exposed portion of the Hsp90α binding site to produce cell-impermeable extracellular Hsp90α-selective inhibitors. The resulting lead compounds were cell-permeable dimethylamine 14 (NDNA3), with an affinity of 0.51 μM for Hsp90α and >196-fold selectivity over the other Hsp90 isoforms, and cell-impermeable quaternary ammonium 17 (NDNA4), with an affinity of 0.34 μM for Hsp90α and >294-fold selectivity. The permanently charged analogs were determined to have low membrane permeability, to be nontoxic against Ovcar-8 and MCF-10A cells, to avoid disruption of hERG channel maturation, and not to induce the heat shock response or Hsp90α-dependent client degradation.
Collapse
Affiliation(s)
- Tyelor
S. Reynolds
- Department of Chemistry and
Biochemistry, The University of Notre Dame, 305 McCourtney Hall, Notre Dame, Indiana 46556, United States
| | - Brian S. J. Blagg
- Department of Chemistry and
Biochemistry, The University of Notre Dame, 305 McCourtney Hall, Notre Dame, Indiana 46556, United States
| |
Collapse
|
8
|
Sanchez-Sandoval AL, Hernández-Plata E, Gomora JC. Voltage-gated sodium channels: from roles and mechanisms in the metastatic cell behavior to clinical potential as therapeutic targets. Front Pharmacol 2023; 14:1206136. [PMID: 37456756 PMCID: PMC10348687 DOI: 10.3389/fphar.2023.1206136] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 06/21/2023] [Indexed: 07/18/2023] Open
Abstract
During the second half of the last century, the prevalent knowledge recognized the voltage-gated sodium channels (VGSCs) as the proteins responsible for the generation and propagation of action potentials in excitable cells. However, over the last 25 years, new non-canonical roles of VGSCs in cancer hallmarks have been uncovered. Their dysregulated expression and activity have been associated with aggressive features and cancer progression towards metastatic stages, suggesting the potential use of VGSCs as cancer markers and prognostic factors. Recent work has elicited essential information about the signalling pathways modulated by these channels: coupling membrane activity to transcriptional regulation pathways, intracellular and extracellular pH regulation, invadopodia maturation, and proteolytic activity. In a promising scenario, the inhibition of VGSCs with FDA-approved drugs as well as with new synthetic compounds, reduces cancer cell invasion in vitro and cancer progression in vivo. The purpose of this review is to present an update regarding recent advances and ongoing efforts to have a better understanding of molecular and cellular mechanisms on the involvement of both pore-forming α and auxiliary β subunits of VGSCs in the metastatic processes, with the aim at proposing VGSCs as new oncological markers and targets for anticancer treatments.
Collapse
Affiliation(s)
- Ana Laura Sanchez-Sandoval
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
- Medicina Genómica, Hospital General de México “Dr Eduardo Liceaga”, Mexico City, Mexico
| | - Everardo Hernández-Plata
- Consejo Nacional de Humanidades, Ciencias y Tecnologías and Instituto Nacional de Medicina Genómica, Mexico City, Mexico
| | - Juan Carlos Gomora
- Departamento de Neuropatología Molecular, Instituto de Fisiología Celular, Universidad Nacional Autónoma de México, Mexico City, Mexico
| |
Collapse
|
9
|
Chen L, Cho HY, Chuang TH, Ke TL, Wu SN. The Effectiveness of Isoplumbagin and Plumbagin in Regulating Amplitude, Gating Kinetics, and Voltage-Dependent Hysteresis of erg-mediated K + Currents. Biomedicines 2022; 10:780. [PMID: 35453530 PMCID: PMC9029050 DOI: 10.3390/biomedicines10040780] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2022] [Revised: 03/22/2022] [Accepted: 03/25/2022] [Indexed: 02/04/2023] Open
Abstract
Isoplumbagin (isoPLB, 5-hydroxy-3-methyl-1,4-naphthoquinone), a naturally occurring quinone, has been observed to exercise anti-inflammatory, antimicrobial, and antineoplastic activities. Notably, whether and how isoPLB, plumbagin (PLB), or other related compounds impact transmembrane ionic currents is not entirely clear. In this study, during GH3-cell exposure to isoPLB, the peak and sustained components of an erg (ether-à-go-go related gene)-mediated K+ current (IK(erg)) evoked with long-lasting-step hyperpolarization were concentration-dependently decreased, with a concomitant increase in the decaying time constant of the deactivating current. The presence of isoPLB led to a differential reduction in the peak and sustained components of deactivating IK(erg) with effective IC50 values of 18.3 and 2.4 μM, respectively, while the KD value according to the minimum binding scheme was estimated to be 2.58 μM. Inhibition by isoPLB of IK(erg) was not reversed by diazoxide; however, further addition of isoPLB, during the continued exposure to 4,4'-dithiopyridine, did not suppress IK(erg) further. The recovery of IK(erg) by a two-step voltage pulse with a geometric progression was slowed in the presence of isoPLB, and the decaying rate of IK(erg) activated by the envelope-of-tail method was increased in its presence. The strength of the IK(erg) hysteresis in response to an inverted isosceles-triangular ramp pulse was diminished by adding isoPLB. A mild inhibition of the delayed-rectifier K+ current (IK(DR)) produced by the presence of isoPLB was seen in GH3 cells, while minimal changes in the magnitude of the voltage-gated Na+ current were demonstrated in its presence. Moreover, the IK(erg) identified in MA-10 Leydig tumor cells was blocked by adding isoPLB. Therefore, the effects of isoPLB or PLB on ionic currents (e.g., IK(erg) and IK(DR)) demonstrated herein would be upstream of our previously reported perturbations on mitochondrial morphogenesis or respiration. Taken together, the perturbations of ionic currents by isoPLB or PLB demonstrated herein are likely to contribute to the underlying mechanism through which they, or other structurally similar compounds, result in adjustments in the functional activities of different neoplastic cells (e.g., GH3 and MA-10 cells), presuming that similar in vivo observations occur.
Collapse
Affiliation(s)
- Linyi Chen
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan; (L.C.); (T.-L.K.)
- Department of Medical Science, National Tsing Hua University, Hsinchu 30013, Taiwan
| | - Hsin-Yen Cho
- Department of Physiology, National Cheng Kung University Medical College, Tainan City 70101, Taiwan; (H.-Y.C.); (T.-H.C.)
| | - Tzu-Hsien Chuang
- Department of Physiology, National Cheng Kung University Medical College, Tainan City 70101, Taiwan; (H.-Y.C.); (T.-H.C.)
| | - Ting-Ling Ke
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu 30013, Taiwan; (L.C.); (T.-L.K.)
| | - Sheng-Nan Wu
- Department of Physiology, National Cheng Kung University Medical College, Tainan City 70101, Taiwan; (H.-Y.C.); (T.-H.C.)
- Institute of Basic Medical Sciences, National Cheng Kung University Medical College, Tainan City 70101, Taiwan
| |
Collapse
|
10
|
Meléndez TA, Huanosta-Gutiérrez A, Barriga-Montoya C, González-Andrade M, Gómez-Lagunas F. Dronedarone blockage of the tumor-related Kv10.1 channel: a comparison with amiodarone. Pflugers Arch 2020; 472:75-87. [DOI: 10.1007/s00424-019-02342-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 12/12/2019] [Accepted: 12/17/2019] [Indexed: 12/24/2022]
|
11
|
Kim S, Lee H, Ko JW, Kim JR. Effects of Celecoxib on the QTc Interval: A Thorough QT/QTc Study. Clin Ther 2019; 41:2204-2218. [PMID: 31564512 DOI: 10.1016/j.clinthera.2019.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2019] [Revised: 09/01/2019] [Accepted: 09/03/2019] [Indexed: 11/29/2022]
Abstract
PURPOSE Celecoxib is a selective cyclooxygenase-2 inhibitor widely used in patients with osteoarthritis and rheumatoid arthritis. Recently, nonclinical data on the inhibition of human ether-à-go-go-related gene potassium channels by celecoxib were reported, but there is no compelling evidence for this finding in humans. The aim of this study was to assess the potential effects of celecoxib on cardiac repolarization by conducting a thorough QT study, which was designed in compliance with the related guidelines. METHODS This randomized, open-label, positive- and negative-controlled, crossover clinical study was conducted in healthy male and female subjects. Each subject received, in 1 of 4 randomly assigned sequences, all of the following 3 interventions: celecoxib 400 mg once daily for 6 days; a single dose of moxifloxacin 400 mg, which served as a positive control to assess the assay sensitivity; and water without any drug, which served as a negative control. Serial 12-lead ECG and blood samples for pharmacokinetic analysis were collected periodically over 24 h. Individually RR-corrected QT intervals (QTcI) and Fridericia method-corrected QT intervals (QTcF) were calculated and evaluated. FINDINGS Twenty-eight subjects were allocated to 1 of the 4 intervention sequences. The largest time-matched mean effects of celecoxib on the QTcI and QTcF were <5 ms, and the upper bounds of the 1-sided 95% CIs of those values did not exceed 10 ms. Moreover, none of the subjects had an absolute QTcI value of >450 ms or a change from baseline in QTcI of >60 ms after multiple administrations of celecoxib. The QTcI did not show a positive correlation with celecoxib concentrations in the range up to ~2700 μg/L. The overall effects of moxifloxacin on the QTcI and QTcF were enough to establish assay sensitivity. No serious adverse events were reported, with a total of 11 AEs reported in 8 subjects. IMPLICATIONS Celecoxib caused no clinically relevant increase in the QT/QTc interval at the maximum dose level used in current practice settings. ClinicalTrials.gov identifier: NCT03822520.
Collapse
Affiliation(s)
- Seokuee Kim
- Department of Clinical Pharmacology and Therapeutics, Samsung Medical Center, Seoul, Republic of Korea
| | - Hyeryeon Lee
- Department of Clinical Pharmacology and Therapeutics, Samsung Medical Center, Seoul, Republic of Korea
| | - Jae-Wook Ko
- Department of Clinical Pharmacology and Therapeutics, Samsung Medical Center, Seoul, Republic of Korea
| | - Jung-Ryul Kim
- Department of Clinical Pharmacology and Therapeutics, Samsung Medical Center, Seoul, Republic of Korea; Department of Clinical Research Design and Evaluation, SAIHST, Sungkyunkwan University, Seoul, Republic of Korea.
| |
Collapse
|
12
|
Bakhriansyah M, Souverein PC, Klungel OH, de Boer A, Blom MT, Tan HL. Non-steroidal anti-inflammatory drugs and the risk of out-of-hospital cardiac arrest: a case-control study. Europace 2019; 21:99-105. [PMID: 30107407 PMCID: PMC6321960 DOI: 10.1093/europace/euy180] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 07/17/2018] [Indexed: 11/18/2022] Open
Abstract
AIMS Non-steroidal anti-inflammatory drugs (NSAIDs), particularly selective COX-2 inhibitors, are associated with an increased risk of cardiovascular adverse events. However, the association between these drugs and out-of-hospital cardiac arrest with electrocardiogram-documented ventricular tachycardia/ventricular fibrillation (VT/VF-OHCA) has not been studied yet. This study was aimed to evaluate the association between the use of selective COX-2 inhibitors or conventional NSAIDs and VT/VF-OHCA compared with non-use. METHODS AND RESULTS A case-control study was conducted among 2483 cases with VT/VF-OHCA from the AmsteRdam REsuscitation STudies (ARREST) registry, an ongoing Dutch registry of OHCA, and 10 441 non-VT/VF-OHCA-controls from the Dutch PHARMO Database Network, containing drug dispensing records of community pharmacies, over the period July 2005-December 2011. Up to five controls were matched for age and sex to one case at the date of VT/VF-OHCA (index date). Odds ratios (ORs) and 95% confidence intervals (CIs) were calculated by conditional logistic regression analysis. Of the cases, 0.5% was currently exposed at the index date to selective COX-2 inhibitors and 2.5% to conventional NSAIDs. Neither current use of selective COX-2 inhibitors nor conventional NSAIDs were associated with an increased risk of VT/VF-OHCA (adjusted OR 1.11, 95% CI: 0.79-1.56 and adjusted OR 0.97, 95% CI: 0.86-1.10, respectively) compared with non-use. Stratification for VT/VF-OHCA with presence/absence of acute myocardial infarction did not change these results. CONCLUSION Exposure to selective COX-2 inhibitors or conventional NSAIDs was not associated with an increased risk of VT/VF-OHCA compared with non-use.
Collapse
Affiliation(s)
- Mohammad Bakhriansyah
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3508 TB, the Netherlands
- Department of Pharmacology, Faculty of Medicine, Lambung Mangkurat University, Banjarmasin, Indonesia
| | - Patrick C Souverein
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3508 TB, the Netherlands
| | - Olaf H Klungel
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3508 TB, the Netherlands
| | - Anthonius de Boer
- Division of Pharmacoepidemiology and Clinical Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht 3508 TB, the Netherlands
| | - Marieke T Blom
- Department of Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, DD, the Netherlands
| | - Hanno L Tan
- Department of Cardiology, Academic Medical Center, University of Amsterdam, Amsterdam, DD, the Netherlands
| |
Collapse
|
13
|
Ruscica M, Ferri N, Macchi C, Corsini A, Sirtori CR. Lipid lowering drugs and inflammatory changes: an impact on cardiovascular outcomes? Ann Med 2018; 50:461-484. [PMID: 29976096 DOI: 10.1080/07853890.2018.1498118] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Inflammatory changes are responsible for maintenance of the atherosclerotic process and may underlie some of the most feared vascular complications. Among the multiple mechanisms of inflammation, the arterial deposition of lipids and particularly of cholesterol crystals is the one responsible for the activation of inflammasome NLRP3, followed by the rise of circulating markers, mainly C-reactive protein (CRP). Elevation of lipoproteins, LDL but also VLDL and remnants, associates with increased inflammatory changes and coronary risk. Lipid lowering medications can reduce cholesterolemia and CRP: patients with elevations of both are at greatest cardiovascular (CV) risk and receive maximum benefit from therapy. Evaluation of the major drug series indicates that statins exert the largest LDL and CRP reduction, accompanied by reduced CV events. Other drugs, mainly active on the triglyceride/HDL axis, for example, PPAR agonists, may improve CRP and the lipid pattern, especially in patients with metabolic syndrome. PCSK9 antagonists, the newest most potent medications, do not induce significant changes in inflammatory markers, but patients with the highest baseline CRP levels show the best CV risk reduction. Parallel evaluation of lipids and inflammatory changes clearly indicates a significant link, both guiding to patients at highest risk, and to the best pharmacological approach. Key messages Lipid lowering agents with "pleiotropic" effects provide a more effective approach to CV prevention In CANTOS study, patients achieving on-treatment hsCRP concentrations ≤2 mg/L had a higher benefit in terms of reduction in major CV events The anti-inflammatory activity of PCSK9 antagonists appears to be of a minimal extent.
Collapse
Affiliation(s)
- M Ruscica
- a Dipartimento di Scienze Farmacologiche e Biomolecolari , Università degli Studi di Milano , Milan , Italy
| | - N Ferri
- b Dipartimento di Scienze del Farmaco , Università degli Studi di Padova , Padova , Italy
| | - C Macchi
- a Dipartimento di Scienze Farmacologiche e Biomolecolari , Università degli Studi di Milano , Milan , Italy
| | - A Corsini
- a Dipartimento di Scienze Farmacologiche e Biomolecolari , Università degli Studi di Milano , Milan , Italy
| | - C R Sirtori
- c Centro Dislipidemie , A.S.S.T. Grande Ospedale Metropolitano Niguarda , Milan , Italy
| |
Collapse
|
14
|
Klivinyi C, Bornemann-Cimenti H. Pain medication and long QT syndrome. Korean J Pain 2018; 31:3-9. [PMID: 29372020 PMCID: PMC5780213 DOI: 10.3344/kjp.2018.31.1.3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2017] [Revised: 10/03/2017] [Accepted: 11/06/2017] [Indexed: 02/02/2023] Open
Abstract
Long QT syndrome is a cardiac repolarization disorder and is associated with an increased risk of torsades de pointes. The acquired form is most often attributable to administration of specific medications and/or electrolyte imbalance. This review provides insights into the risk for QT prolongation associated with drugs frequently used in the treatment of chronic pain. In the field of pain medicine all the major drug classes (i.e. NSAIDs, opioids, anticonvulsive and antidepressant drugs, cannabinoids, muscle relaxants) contain agents that increase the risk of QT prolongation. Other substances, not used in the treatment of pain, such as proton pump inhibitors, antiemetics, and diuretics are also associated with long QT syndrome. When the possible benefits of therapy outweigh the associated risks, slow dose titration and electrocardiography monitoring are recommended.
Collapse
Affiliation(s)
- Christoph Klivinyi
- Department of Anesthesiology and Intensive Care Medicine, Medical University of Graz, Graz, Austria
| | - Helmar Bornemann-Cimenti
- Department of Anesthesiology and Intensive Care Medicine, Medical University of Graz, Graz, Austria
| |
Collapse
|
15
|
Ahmad S, Panda BP, Kohli K, Fahim M, Dubey K. Folic acid ameliorates celecoxib cardiotoxicity in a doxorubicin heart failure rat model. PHARMACEUTICAL BIOLOGY 2017; 55:1295-1303. [PMID: 28274156 PMCID: PMC6130581 DOI: 10.1080/13880209.2017.1299768] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2016] [Revised: 02/19/2017] [Accepted: 02/21/2017] [Indexed: 05/30/2023]
Abstract
CONTEXT The cardiotoxic effect of selective cyclo-oxygenase-2 inhibitors is well known. While rofecoxib and valdecoxib have been withdrawn, celecoxib remains on the market. Folic acid, a naturally occurring vitamin, has been shown to reduce myocardial ischemia and post-reperfusion injury in rats. OBJECTIVE This study examined the cardiac effects of celecoxib and folic acid on doxorubicin-induced cardiomyopathy in rats. MATERIALS AND METHODS Cardiomyopathy was induced in male Wistar rats with six intraperitoneal injections of 2.5 mg/kg doxorubicin over a period of two weeks. The effect of 28 days of celecoxib (100 mg/kg/day) and its combination with folic acid (10 mg/kg/day) was studied on doxorubicin-induced cardiomyopathy according to serum lactate dehydrogenase (LDH), creatine kinase (CK-MB), troponin-T (Tn-T), tumor necrosis factor alpha (TNF-α), cardiac thiobarbituric acid reactive substance (TBARS), and glutathione (GSH) levels as well as systolic blood pressure (SBP), heart rate (HR) and ultrastructural studies. RESULTS Celecoxib cardiotoxicity was manifested by significant increases in the LDH, Tn-T, TNF-α, CK-MB, SBP, HR (p < 0.001) and TBARS (p < 0.01) levels and a significant decrease in the GSH (p < 0.05) level when used alone or administered with doxorubicin. However, the combination of folic acid with celecoxib caused a significant reversal of these parameters and reduced the cardiotoxicity of celecoxib that was aggravated by doxorubicin. The ultrastructural study also revealed myocardial protection with this combination. DISCUSSION AND CONCLUSION Folic acid protects against the cardiotoxic effects of celecoxib, which are aggravated in the presence of doxorubicin. Folic acid may act as a useful adjunct in patients who are taking celecoxib.
Collapse
Affiliation(s)
- Shafique Ahmad
- Department of Pharmacology, Faculty of Pharmacy, Jamia Hamdard, New Delhi, India
| | - Bibhu Prasad Panda
- Department of Pharmacognosy and Phytochemistry, Faculty of Pharmacy, Jamia Hamdard, New Delhi, India
| | - Kanchan Kohli
- Department of Pharmaceutics, Faculty of Pharmacy, Jamia Hamdard, New Delhi, India
| | - Mohammad Fahim
- Department of Physiology, Hamdard Institute of Medical Sciences and Research, Jamia Hamdard, New Delhi, India
| | - Kiran Dubey
- Department of Pharmacology, Faculty of Pharmacy, Jamia Hamdard, New Delhi, India
| |
Collapse
|
16
|
Schneider P, Schneider G. A Computational Method for Unveiling the Target Promiscuity of Pharmacologically Active Compounds. Angew Chem Int Ed Engl 2017; 56:11520-11524. [DOI: 10.1002/anie.201706376] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 07/07/2017] [Indexed: 12/17/2022]
Affiliation(s)
| | - Gisbert Schneider
- Department of Chemistry and Applied Biosciences; Swiss Federal Institute of Technology (ETH); Vladimir-Prelog-Weg 4 8093 Zurich Switzerland
- inSili.com LLC; Segantinisteig 3 8049 Zurich Switzerland
| |
Collapse
|
17
|
Schneider P, Schneider G. A Computational Method for Unveiling the Target Promiscuity of Pharmacologically Active Compounds. Angew Chem Int Ed Engl 2017. [DOI: 10.1002/ange.201706376] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
| | - Gisbert Schneider
- Department of Chemistry and Applied Biosciences; Swiss Federal Institute of Technology (ETH); Vladimir-Prelog-Weg 4 8093 Zurich Switzerland
- inSili.com LLC; Segantinisteig 3 8049 Zurich Switzerland
| |
Collapse
|
18
|
Spyra S, Meisner A, Schaefer M, Hill K. COX-2-selective inhibitors celecoxib and deracoxib modulate transient receptor potential vanilloid 3 channels. Br J Pharmacol 2017; 174:2696-2705. [PMID: 28567799 DOI: 10.1111/bph.13893] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2016] [Revised: 05/11/2017] [Accepted: 05/11/2017] [Indexed: 01/03/2023] Open
Abstract
BACKGROUND AND PURPOSE The transient receptor potential vanilloid 3 (TRPV3) channel is a heat-sensitive ion channel, which is predominantly expressed in keratinocytes. TRPV3 channels are involved in numerous physiological and pathophysiological processes within the skin, including cutaneous nociception, temperature sensation and development of itch. The role of TRPV3 channels in such processes is poorly understood; therefore, the establishment of selective modulators of TRPV3 channels is highly desirable. EXPERIMENTAL APPROACH Novel TRPV3-modulating compounds were identified using fluorometric intracellular Ca2+ assays and further evaluated with electrophysiological techniques. KEY RESULTS TRPV3 activity, elicited by 2-aminoethoxydiphenyl borate (2-APB), was efficaciously enhanced by deracoxib and celecoxib, two COX-2-selective inhibitors. They exerted their potentiating effect via a direct interaction with TRPV3 as evident from excised inside-out recordings. Structurally-related COX-2 inhibitors affected TRPV3 channel gating to a much lesser degree. Similar results were obtained in HEK293 cells stably expressing cyan fluorescent protein-tagged mouse TRPV3 channels and in a mouse keratinocyte cell line, endogenously expressing TRPV3. The effects of celecoxib and deracoxib on TRPV3 were dependent on the stimulus used to activate TRPV3. While 2-APB and heat-activated TRPV3 channels were potentiated by celecoxib, carvacrol-activated channels were inhibited by celecoxib. CONCLUSIONS AND IMPLICATIONS We identified a new class of drugs that modulate TRPV3 channels. The most potent compound celecoxib is an approved analgesic and anti-inflammatory drug, which is currently being investigated for its topical application in the treatment of skin cancer. As TRPV3 is highly expressed in skin, celecoxib might affect TRPV3 activity in vivo when used at high local concentrations.
Collapse
Affiliation(s)
- Stefan Spyra
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universität Leipzig, Leipzig, Germany
| | - Anne Meisner
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universität Leipzig, Leipzig, Germany
| | - Michael Schaefer
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universität Leipzig, Leipzig, Germany
| | - Kerstin Hill
- Rudolf-Boehm-Institut für Pharmakologie und Toxikologie, Universität Leipzig, Leipzig, Germany
| |
Collapse
|
19
|
Puratchikody A, Sriram D, Umamaheswari A, Irfan N. 3-D structural interactions and quantitative structural toxicity studies of tyrosine derivatives intended for safe potent inflammation treatment. Chem Cent J 2016; 10:24. [PMID: 27141229 PMCID: PMC4852405 DOI: 10.1186/s13065-016-0169-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2015] [Accepted: 04/11/2016] [Indexed: 12/24/2022] Open
Abstract
Background Drugs that inhibit cyclooxygenase-2 (COX-2) while sparing cyclooxygenase-1 (COX-1) represent a new attractive therapeutic development and offer new perspective for further use of COX-2 inhibitors. Intention of this work is to develop safer, selective COX-2 inhibitors that do not produce harmful effects. Results A series of 55 tyrosine derivatives were designed for evaluation as selective COX-2 inhibitors and investigated by in silico for their anti-inflammatory activities using C-Docker. The results of docking study showed that 35 molecules were found to selectively inhibit the enzyme COX-2. These molecules formed stable π hydrophobic and additional van der Waals interactions in the active site side pocket of COX-2. The molecules selected from docking studies were examined through ADMET descriptors and Osiris property explorer to find its safety profile as well. The tyrosine derivatives containing toxic fragments were eliminated. Conclusion The results conclude that out of 55, 19 molecules possessed best binding energy (< −3.333 kcal/mol) and these molecules had more selective and safer COX-2 inhibitor profile compared to the standard celecoxib.3-D structural interactions of COX-2 inhibiting tyrosine derivatives. ![]()
Collapse
Affiliation(s)
- Ayarivan Puratchikody
- Drug Discovery and Development Research Group, Department of Pharmaceutical Technology, Anna University Chennai, BIT Campus, Tiruchirappalli, 620024 India
| | - Dharmaraj Sriram
- Pharmacy Group, Birla Institute of Technology and Sciences, Pilani, Hyderabad Campus, Jawahar Nagar, Secunderabad, Telangana 500 078 India
| | - Appavoo Umamaheswari
- Drug Discovery and Development Research Group, Department of Pharmaceutical Technology, Anna University Chennai, BIT Campus, Tiruchirappalli, 620024 India
| | - Navabshan Irfan
- Drug Discovery and Development Research Group, Department of Pharmaceutical Technology, Anna University Chennai, BIT Campus, Tiruchirappalli, 620024 India
| |
Collapse
|
20
|
Papageorgiou N, Zacharia E, Briasoulis A, Charakida M, Tousoulis D. Celecoxib for the treatment of atherosclerosis. Expert Opin Investig Drugs 2016; 25:619-633. [PMID: 26940257 DOI: 10.1517/13543784.2016.1161756] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION It is widely accepted that inflammation plays a pivotal role in the progression of atherosclerosis. Anti-inflammatory drugs and especially selective cyclooxygenase-2 (COX-2) inhibitors have attracted a keen interest. AREAS COVERED In the present drug evaluation article, the authors elucidate the role of celecoxib, a selective COX-2 inhibitor, in the treatment of atherosclerosis. They discuss the atherogenic properties of the COX-2 enzyme. In addition, they address the studies that support an atheroprotective role of celecoxib. Moreover, they provide a review of the literature on the role of COX-2 inhibitors in increasing the rate of major adverse cardiovascular events. Finally, they discuss the emerging evidence that supports celecoxib as an adjuvant or neo-adjuvant therapy to percutaneous coronary intervention (PCI). EXPERT OPINION Several studies have demonstrated a beneficial effect of celecoxib on the progression of atherosclerosis. Nevertheless, this evidence is mainly derived from preliminary data, while a substantial number of clinical studies have raised concerns regarding the cardiovascular safety of COX-2 inhibitors. Interestingly, recent clinical studies have supported the advantages of short-term celecoxib administration in patients undergoing PCI. However, many more large scale clinical trials are required to assess the long-term safety and efficacy of celecoxib administration in patients with cardiovascular disease.
Collapse
Affiliation(s)
| | - Effimia Zacharia
- b 1st Department of Cardiology, Hippokration Hospital , University of Athens , Athens , Greece
| | - Alexandros Briasoulis
- c Division of Cardiology , Wayne State University/Detroit Medical Center , Detroit , MI , USA
| | - Marietta Charakida
- d Vascular Physiology Unit, Institute of Cardiovascular Science , University College London , London , UK
| | - Dimitris Tousoulis
- b 1st Department of Cardiology, Hippokration Hospital , University of Athens , Athens , Greece
| |
Collapse
|
21
|
Acute and chronic toxicity, cytochrome p450 enzyme inhibition, and HERG channel blockade studies with a polyherbal, ayurvedic formulation for inflammation. BIOMED RESEARCH INTERNATIONAL 2015; 2015:971982. [PMID: 25893199 PMCID: PMC4381553 DOI: 10.1155/2015/971982] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2014] [Revised: 02/20/2015] [Accepted: 02/20/2015] [Indexed: 12/13/2022]
Abstract
Ayurvedic plants are known for thousands of years to have anti-inflammatory and antiarthritic effect. We have recently shown that BV-9238, a proprietary formulation of Withania somnifera, Boswellia serrata, Zingiber officinale, and Curcuma longa, inhibits LPS-induced TNF-alpha and nitric oxide production from mouse macrophage and reduces inflammation in different animal models. To evaluate the safety parameters of BV-9238, we conducted a cytotoxicity study in RAW 264.7 cells (0.005–1 mg/mL) by MTT/formazan method, an acute single dose (2–10 g/kg bodyweight) toxicity study and a 180-day chronic study with 1 g and 2 g/kg bodyweight in Sprague Dawley rats. Some sedation, ptosis, and ataxia were observed for first 15–20 min in very high acute doses and hence not used for further chronic studies. At the end of 180 days, gross and histopathology, blood cell counts, liver and renal functions were all at normal levels. Further, a modest attempt was made to assess the effects of BV-9238 (0.5 µg/mL) on six major human cytochrome P450 enzymes and 3H radioligand binding assay with human hERG receptors. BV-9238 did not show any significant inhibition of these enzymes at the tested dose. All these suggest that BV-9238 has potential as a safe and well tolerated anti-inflammatory formulation for future use.
Collapse
|
22
|
Frolov RV, Singh S. Evidence of more ion channels inhibited by celecoxib: KV1.3 and L-type Ca(2+) channels. BMC Res Notes 2015; 8:62. [PMID: 25889233 PMCID: PMC4348164 DOI: 10.1186/s13104-015-1023-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 02/19/2015] [Indexed: 01/01/2023] Open
Abstract
Background Celecoxib, a selective inhibitor of cyclooxygenase-2, can directly modulate many voltage-activated potassium, sodium and calcium channels and alter functioning of excitable cells. The inhibitory and facilitating effects of celecoxib on ion channels occur at low micromolar concentrations, bordering on therapeutic concentrations achievable in the clinical setting. The experiments described here were performed with the goals (1) to increase the range of ion channels tested, and (2) to examine possible differences in celecoxib’s effects on channels from different species. Findings The channels examined in this study using patch-clamp and intracellular recording methods were human KV1.3 channels expressed in CHO cells, L-type Ca2+ channels (LTCC) from guinea pig cardiomyocytes, and LTCCs from Drosophila larval body-wall muscles. Celecoxib inhibited KV1.3 currents with IC50 of 5.0 μM at the end of 200 ms pulses to +20 mV. Celecoxib inhibited peak currents through guinea pig and Drosophila LTCCs with IC50s of 10.6 and 76.0 μM, respectively. Conclusions As blockade of KV1.3 channels is associated with suppression of inflammatory immune reactions, the finding that celecoxib can inhibit these channels raises a question of possible contribution of KV1.3 inhibition to the anti-inflammatory effects of celecoxib. On the other hand, the Ca2+ channel results are consistent with previous observations indicating that, in contrast to K+ channels, strength of celecoxib effects on LTCCs strongly varies from species to species.
Collapse
Affiliation(s)
- Roman V Frolov
- Department of Physical Sciences, Division of Biophysics, University of Oulu, P.O. Box 3000, Oulun Yliopisto, 90014, Finland.
| | - Satpal Singh
- Department of Pharmacology and Toxicology, State University of New York at Buffalo, Buffalo, NY, 14214, USA.
| |
Collapse
|
23
|
Frolov RV, Singh S. Celecoxib and ion channels: a story of unexpected discoveries. Eur J Pharmacol 2014; 730:61-71. [PMID: 24630832 DOI: 10.1016/j.ejphar.2014.02.032] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2013] [Revised: 01/28/2014] [Accepted: 02/18/2014] [Indexed: 10/25/2022]
Abstract
Celecoxib (Celebrex), a highly popular selective inhibitor of cyclooxygenase-2, can modulate ion channels and alter functioning of neurons and myocytes at clinically relevant concentrations independently of cyclooxygenase inhibition. In experimental systems varying from Drosophila to primary mammalian and human cell lines, celecoxib inhibits many voltage-activated Na(+), Ca(2+), and K(+) channels, including NaV1.5, L- and T-type Ca(2+) channels, KV1.5, KV2.1, KV4.3, KV7.1, KV11.1 (hERG), while stimulating other K(+) channels-KV7.2-5 and, possibly, KV11.1 (hERG) channels under certain conditions. In this review, we summarize the information currently available on the effects of celecoxib on ion channels, examine mechanistic aspects of drug action and the concomitant changes at the cellular and organ levels, and discuss these findings in the therapeutic context.
Collapse
Affiliation(s)
- Roman V Frolov
- Department of Physical Sciences, Division of Biophysics, University of Oulu, PO Box 3000, 90014 Oulun Yliopisto, Finland.
| | - Satpal Singh
- Department of Pharmacology and Toxicology, State University of New York at Buffalo, Buffalo, NY 14214, USA
| |
Collapse
|
24
|
Choi SW, Kim KS, Shin DH, Yoo HY, Choe H, Ko TH, Youm JB, Kim WK, Zhang YH, Kim SJ. Class 3 inhibition of hERG K+ channel by caffeic acid phenethyl ester (CAPE) and curcumin. Pflugers Arch 2013; 465:1121-34. [PMID: 23440458 DOI: 10.1007/s00424-013-1239-7] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2013] [Revised: 02/03/2013] [Accepted: 02/04/2013] [Indexed: 12/30/2022]
Abstract
Human ether-á-go-go-related gene (hERG) K(+) channel current (I hERG ) is inhibited by various compounds and genetic mutations, potentially resulting in cardiac arrhythmia. Here, we investigated effects of caffeic acid phenethyl ester (CAPE) and curcumin, two natural anti-inflammatory polyphenols, on I hERG in HEK-293 cells overexpressed with hERG. CAPE dose-dependently decreased repolarization tail current of hERG (I hERG,tail; IC50, 10.6 ± 0.5 μM). CAPE also shifted half-activation voltage (V 1/2) to the left (from -17.5 to -26.5 mV) and accelerated activation and inactivation kinetics. The CAPE inhibition of I hERG,tail was not attenuated in the pore-blocker site mutants of hERG (Y652A and F656A). A point mutation of Cys723 (C723S) mimicked the effects of CAPE and caused a left shift of V 1/2 and acceleration of I hERG,tail deactivation. However, I hERG,tail inhibition by CAPE was still observed in C723S. Taken together, CAPE inhibits hERG channel by class 3 mechanism, i.e., modification of gating, not by blocking the pore. Curcumin induced changes of I hERG similar to those of CAPE, while additional interaction with pore-blocking sites was suggested from attenuated I hERG,tail inhibition in Y652A and F656A. Interestingly, I hERG induced by human action potential voltage clamp was increased by CAPE while decreased by curcumin. Mathematical simulation of action potential derived from the experimental results of CAPE and curcumin supports that CAPE, but not curcumin, would induce shortening of AP duration by facilitation of I hERG . The above results revealed intriguing roles of Cys723 in hERG kinetics and suggested that conventional drug screening by using step pulse protocol for I hERG,tail would overlook the hERG kinetic modulations that could compensate the decrease of I hERG,tail.
Collapse
Affiliation(s)
- Seong Woo Choi
- Department of Physiology, Seoul National University College of Medicine, 103 Daehakro, Chongro-gu, Seoul, 110-799, Republic of Korea
| | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
The role of potassium channel activation in celecoxib-induced analgesic action. PLoS One 2013; 8:e54797. [PMID: 23358696 PMCID: PMC3554616 DOI: 10.1371/journal.pone.0054797] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2012] [Accepted: 12/14/2012] [Indexed: 01/17/2023] Open
Abstract
BACKGROUND AND PURPOSE Celecoxib (CXB) is a widely prescribed COX-2 inhibitor used clinically to treat pain and inflammation. Recently, COX-2 independent mechanisms have been described to be the targets of CXB. For instance, ion channels such as the voltage-gated sodium channel, L-type calcium channel, Kv2.1, Kv1.5, Kv4.3 and HERG potassium channel were all reported to be inhibited by CXB. Our recent study revealed that CXB is a potent activator of Kv7/M channels. M currents expressed in dorsal root ganglia play an important role in nociception. Our study was aimed at establishing the role of COX-2 independent M current activation in the analgesic action of CXB. METHODS AND RESULTS We compared the effects of CXB and its two structural analogues, unmethylated CXB (UMC) and 2,5-dimethyl-CXB (DMC), on Kv7/M currents and pain behavior in animal models. UMC is a more potent inhibitor of COX-2 than CXB while DMC has no COX-2 inhibiting activity. We found that CXB, UMC and DMC concentration-dependently activated Kv7.2/7.3 channels expressed in HEK293 cells and the M-type current in dorsal root ganglia neurons, negatively shifted I-V curve of Kv7.2/7.3 channels, with a potency and efficiency inverse to their COX-2 inhibitory potential. Furthermore, CXB, UMC and DMC greatly reduced inflammatory pain behavior induced by bradykinin, mechanical pain behavior induced by stimulation with von Frey filaments and thermal pain behavior in the Hargreaves test. CXB and DMC also significantly attenuated hyperalgesia in chronic constriction injury neuropathic pain. CONCLUSION CXB, DMC and UMC are openers of Kv7/M K(+) channels with effects independent of COX-2 inhibition. The analgesic effects of CXBs on pain behaviors, especially those of DMC, suggest that activation of Kv7/M K(+) channels may play an important role in the analgesic action of CXB. This study strengthens the notion that Kv7/M K(+) channels are a potential target for pain treatment.
Collapse
|
26
|
Inhibition of ion channels and heart beat in Drosophila by selective COX-2 inhibitor SC-791. PLoS One 2012; 7:e38759. [PMID: 22701705 PMCID: PMC3368905 DOI: 10.1371/journal.pone.0038759] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 05/12/2012] [Indexed: 11/19/2022] Open
Abstract
Recent findings suggest that modulation of ion channels might be implicated in some of the clinical effects of coxibs, selective inhibitors of cyclooxygenase-2 (COX-2). Celecoxib and its inactive analog 2,5-dimethyl-celecoxib, but not rofecoxib, can suppress or augment ionic currents and alter functioning of neurons and myocytes. To better understand these unexpected effects, we have recently investigated the mechanism of inhibition of human Kv2.1 channels by a highly selective COX-2 inhibitor SC-791. In this study we have further explored the SC-791 action on ion channels and heartbeat in Drosophila, which lacks cyclooxygenases and thus can serve as a convenient model to study COX-2-independent mechanisms of coxibs. Using intracellular recordings in combination with a pharmacological approach and utilizing available Drosophila mutants, we found that SC-791 inhibited voltage-activated K+ and L-type Ca2+ channels in larval body-wall muscles and reduced heart rate in a concentration-dependent manner. Unlike celecoxib and several other K+ channel blockers, SC-791 did not induce arrhythmia. Instead, application of SC-791 resulted in a dramatic slowing of contractions and, at higher concentrations, in progressively weaker contractions with gradual cessation of heartbeat. Isradipine, a selective blocker of L-type Ca2+ channels, showed a similar pattern of heart arrest, though no prolongation of contractions was observed. Ryanodine was the only channel modulating compound of those tested additionally that was capable of slowing contractions. Like SC-791, ryanodine reduced heart rate without arrhythmia. However, it could not stop heartbeat completely even at 500 µM, the highest concentration used. The magnitude of heart rate reduction, when SC-791 and ryanodine were applied together, was smaller than expected for independent mechanisms, raising the possibility that SC-791 might be interfering with excitation-contraction coupling in Drosophila heart.
Collapse
|