Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 20 de 473
1.
Int J Pharm ; 640: 123024, 2023 Jun 10.
Article En | MEDLINE | ID: mdl-37156309

Bilosomes are innovative vesicular carriers containing bile salt with a non-ionic surfactant. Being highly flexible, bilosomes can squeeze themselves through the skin carrying the drug to the action site and improving its skin penetration. The objective of this research was to encapsulate niflumic acid (NA), a non-steroidal anti-inflammatory drug into Brij® integrated bilosomes (BIBs) for effective treatment of osteoarthritis through transdermal delivery. BIBs were formulated using 100 mg of Span 20 with different amounts of sodium cholate (NaC), sodium taurocholate (NaTC), or sodium glycocholate (NaGC) as bile salt, with the addition of 5 mg of Brij-93 or Brij-35. BIBs were prepared utilizing ethanol injection method with the application of (31 × 22) complete factorial design using Design-Expert® software. The optimal BIBs formulation determined was (B5) which contains 5 mg of NaTC used as bile salt and 5 mg of Brij-93. B5 exhibited entrapment efficiency% = 95.21 ± 0.00%, particle size = 373.05 ± 0.07 nm, polydispersity index = 0.27 ± 0.01, and zeta potential = -32.00 ± 0.00 mV. It also had a high elasticity with a spherical shape. B5 gel displayed a sustained release profile with a significantly 2.3 folds' higher drug permeation percent across rat skin than that permeated from NA gel. Moreover, in vivo anti-osteoarthritic and histopathological studies assured the efficacy and safety of B5 gel and its superiority over NA gel. Generally, the outcomes confirmed the great efficacy of NA loaded BIBs for the topical treatment of osteoarthritis.


Liposomes , Niflumic Acid , Rats , Animals , Niflumic Acid/pharmacology , Liposomes/pharmacology , Administration, Cutaneous , Skin , Bile Acids and Salts , Permeability , Particle Size , Drug Delivery Systems
2.
Med Chem ; 19(5): 445-459, 2023.
Article En | MEDLINE | ID: mdl-36537605

BACKGROUND: 1,3,4-oxadizole and pyrazole derivatives are very important scaffolds for medicinal chemistry. A literature survey revealed that they possess a wide spectrum of biological activities including anti-inflammatory and antitumor effects. OBJECTIVES: To describe the synthesis and evaluation of two classes of new niflumic acid (NF) derivatives, the 1,3,4-oxadizole derivatives (compounds 3 and (4A-E) and pyrazole derivatives (compounds 5 and 6), as EGFR tyrosine kinase inhibitors in silico and in vitro. METHODS: The designed compounds were synthesized using conventional organic synthesis methods. The antitumor activities of the new NF derivatives against HepG2 hepatocellular carcinoma and A549 non-small cell lung cancer cell lines were assessed in vitro via MTT assay, flow cytometry, RT-PCR, as well as via molecular docking studies. RESULTS: The cytotoxicity results indicated that the newly synthesized NF derivatives were cytotoxic against the two cancer cell lines, with compound 6 being the most cytotoxic, achieving the lowest IC50 concentration. Furthermore, compound 6 targeted EGFR tyrosine kinase leading to cell cycle arrest at the G2/M cell cycle phase and induction of apoptosis. The in vitro biological investigation results matched those of the molecular docking analysis. In conclusion, the new NF derivatives, specifically compound 6, exhibited favorable pharmacokinetic features and are promising EGFR tyrosine kinase inhibitors. CONCLUSION: A series of niflumic acid derivatives (3, 4A-E, 5, and 6) were successfully created, and FT-IR, 1H, 13CNMR, and HRMS were used to confirm their chemical structures. According to molecular docking studies, compounds 3, 5, and 6 have the highest docking scores (ΔG), and most tested compounds have a good pharmacokinetic profile. Results of compound 6 in vitro antitumor activities showed that it is a promising EGFR tyrosine kinase inhibitor.


Antineoplastic Agents , Carcinoma, Non-Small-Cell Lung , Lung Neoplasms , Humans , Structure-Activity Relationship , Niflumic Acid/pharmacology , Cell Proliferation , Molecular Docking Simulation , Cell Line, Tumor , Spectroscopy, Fourier Transform Infrared , Drug Screening Assays, Antitumor , Protein Kinase Inhibitors/chemistry , Antineoplastic Agents/chemistry , ErbB Receptors , Pyrazoles/pharmacology , Molecular Structure , Apoptosis
3.
Dalton Trans ; 52(1): 147-158, 2022 Dec 20.
Article En | MEDLINE | ID: mdl-36472127

To develop new chemotherapeutics with anti-metastasis properties, a series of multi-specific niflumic acid (NFA) platinum(IV) complexes with DNA damage, inflammation inhibition, immunity activation, and angiogenesis suppression mechanisms were designed, synthesized and evaluated as novel antitumor agents. The dual NFA platinum(IV) complex with a cisplatin core showed promising antitumor activities both in vitro and in vivo with lower toxicity than platinum(II) drugs and displayed attractive anti-metastasis performance. It caused serious DNA damage and further elevated the expression of γ-H2AX. Furthermore, it promoted apoptosis by activating the mitochondrial apoptotic pathway and autophagy of tumor cells. Moreover, immune response in tumors was significantly improved by increasing CD3+, CD4+ and CD8+ T infiltrating cells. Subsequently, the pathway ERK/HIF-1α/VEGFA associated with angiogenesis was suppressed by the reduced inflammation and elevated immune response, and the density of microvessels marked by CD34 was significantly reduced in tumors. Accordingly, the multi-specific NFA platinum(IV) complexes have great potential to be developed as novel anti-proliferative and anti-metastatic drugs.


Antineoplastic Agents , Neoplasms , Humans , Platinum/pharmacology , Niflumic Acid/pharmacology , Organoplatinum Compounds/pharmacology , Antineoplastic Agents/pharmacology , Cisplatin/pharmacology , DNA Damage , Apoptosis , Inflammation , Cell Line, Tumor
4.
Article En | MEDLINE | ID: mdl-35988877

The effect of acute hypoosmotic stress on the neural response was investigated using the neurons identified in the abdominal ganglion of the amphibious mollusk Onchidium. The membrane potential of an identified neuron (Ip-1/2) was not significantly altered in 50% hypoosmotic artificial sea water. In isotonic 50% artificial seawater (ASW) with osmolarity that was compensated for using glycerol or urea, the membrane potentials of Ip-1/2 were also not altered compared to those in 50% hypoosmotic ASW. However, hyperpolarization was induced in isotonic 50% ASW when osmolarity was compensated for using sucrose or mannose. In the presence of volume-regulated anion channel (VRAC) inhibitors (niflumic acid and glibenclamide), the Ip-1/2 membrane potentials were hyperpolarized in 50% hypoosmotic ASW. These results suggest that there is a compensatory mechanism involving aquaglyceroporin and VRAC-like channels that maintains membrane potential under hypoosmotic conditions. Here, we detected the expression of aquaglyceroporin mRNA in neural tissues of Onchidium.


Aquaglyceroporins , Gastropoda , Animals , Anions/metabolism , Anions/pharmacology , Aquaglyceroporins/metabolism , Aquaglyceroporins/pharmacology , Gastropoda/metabolism , Glyburide/metabolism , Glyburide/pharmacology , Glycerol/metabolism , Mannose/metabolism , Mannose/pharmacology , Membrane Potentials/physiology , Neurons/metabolism , Niflumic Acid/metabolism , Niflumic Acid/pharmacology , RNA, Messenger/metabolism , Sucrose/metabolism
5.
Chem Biol Drug Des ; 98(6): 1025-1037, 2021 12.
Article En | MEDLINE | ID: mdl-34587361

The Hippo signaling pathway extorts several signals that concomitantly target the activity of transcriptional cofactor yes associated protein (YAP). YAP is a key regulator that elicits signature gene expression by coupling with transcriptional enhanced associate domain (TEAD) family of transcriptional factors. The YAP-TEAD complex via target gene expression gets associated with the development, proliferation, and progression of cancerous cells. Moreover, YAP adorns cells with several oncogenic traits such as inhibition of apoptosis, enhanced proliferation, drug resistance, and immune response suppression, which later became associated with various diseases, particularly cancer. Therefore, inhibition of the YAP activity is an appealing and viable therapeutic target for cancer treatment. This review highlights the recent advances in existing and novel synthetic therapeutics targeting YAP inhibition and regulation. The synthetically produced YAPD93A belonging to cyclic peptides and DC-TEADin02 and vinyl sulfonamide class of compounds are the most potent compounds to inhibit the YAP-TEAD expression by targeting protein-protein interaction (IC50  = 25 nM) and palmitate binding central pocket of TEAD (IC50  = 197 nM), respectively. On the other hand, Chlorpromazine belonging to phenothiazines class has the least potential to suppress YAP via proteasomal degradation (cell viability value of <20% at 40 µM).


Antineoplastic Agents/pharmacology , Molecular Targeted Therapy/methods , YAP-Signaling Proteins/metabolism , Animals , Antineoplastic Agents/chemistry , Curcumin/analogs & derivatives , Curcumin/pharmacology , Humans , Niflumic Acid/pharmacology , Oxadiazoles/chemistry , Oxadiazoles/pharmacology , Pyrones/chemistry , TEA Domain Transcription Factors/metabolism , Triazines/chemistry , Triazines/pharmacology , Verteporfin/pharmacology , YAP-Signaling Proteins/chemistry , Zoledronic Acid/pharmacology
6.
Sci Rep ; 10(1): 13999, 2020 08 19.
Article En | MEDLINE | ID: mdl-32814817

Glycine is one of the major neurotransmitters in the brainstem and the spinal cord. Glycine binds to and activates glycine receptors (GlyRs), increasing Cl- conductance at postsynaptic sites. This glycinergic synaptic transmission contributes to the generation of respiratory rhythm and motor patterns. Strychnine inhibits GlyR by binding to glycine-binding site, while picrotoxin blocks GlyR by binding to the channel pore. We have previously reported that bath application of strychnine to zebrafish embryos causes bilateral muscle contractions in response to tactile stimulation. To explore the drug-mediated inhibition of GlyRs, we screened a chemical library of ~ 1,000 approved drugs and pharmacologically active molecules by observing touch-evoked response of zebrafish embryos in the presence of drugs. We found that exposure of zebrafish embryos to nifedipine (an inhibitor of voltage-gated calcium channel) or niflumic acid (an inhibitor of cyclooxygenase 2) caused bilateral muscle contractions just like strychnine-treated embryos showed. We then assayed strychnine, picrotoxin, nifedipine, and niflumic acid for concentration-dependent inhibition of glycine-mediated currents of GlyRs in oocytes and calculated IC50s. The results indicate that all of them concentration-dependently inhibit GlyR in the order of strychnine > picrotoxin > nifedipine > niflumic acid.


Niflumic Acid/pharmacology , Receptors, Glycine/antagonists & inhibitors , Synaptic Transmission/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Convulsants/pharmacology , Embryo, Nonmammalian/cytology , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/physiology , Female , Glycine/pharmacology , Membrane Potentials/drug effects , Nifedipine/pharmacology , Oocytes/drug effects , Oocytes/metabolism , Oocytes/physiology , Picrotoxin/pharmacology , Receptors, Glycine/agonists , Receptors, Glycine/metabolism , Strychnine/pharmacology , Synaptic Transmission/physiology , Vasodilator Agents/pharmacology , Xenopus laevis , Zebrafish/embryology , Zebrafish/metabolism
7.
Mol Pharmacol ; 98(1): 61-71, 2020 07.
Article En | MEDLINE | ID: mdl-32358165

The blood-brain barrier (BBB) is essential for the maintenance of homeostasis in the brain. Brain capillary endothelial cells (BCECs) comprise the BBB, and thus a delicate balance between their proliferation and death is required. Although the activity of ion channels in BCECs is involved in BBB functions, the underlying molecular mechanisms remain unclear. In the present study, the molecular components of Ca2+-activated Cl- (ClCa) channels and their physiological roles were examined using mouse BCECs (mBCECs) and a cell line derived from bovine BCECs, t-BBEC117. Expression analyses revealed that TMEM16A was strongly expressed in mBCECs and t-BBEC117 cells. In t-BBEC117 cells, whole-cell Cl- currents were sensitive to the ClCa channel blockers, 100 µM niflumic acid and 10 µM T16Ainh-A01, and were also reduced markedly by small-interfering RNA (siRNA) knockdown of TMEM16A. Importantly, block of ClCa currents with ClCa channel blockers or TMEM16A siRNA induced membrane hyperpolarization. Moreover, treatment with TMEM16A siRNA caused an increase in resting cytosolic Ca2+ concentration ([Ca2+]cyt). T16Ainh-A01 reduced cell viability in a concentration-dependent manner. Either ClCa channel blockers or TMEM16A siRNA also curtailed cell proliferation and migration. Furthermore, ClCa channel blockers attenuated the trans-endothelial permeability. In combination, these results strongly suggest that TMEM16A contributes to ClCa channel conductance and can regulate both the resting membrane potential and [Ca2+]cyt in BCECs. Our data also reveal how these BCECs may be involved in the maintenance of BBB functions, as both the proliferation and migration are altered following changes in channel activity. SIGNIFICANCE STATEMENT: In brain capillary endothelial cells (BCECs) of the blood-brain barrier (BBB), TMEM16A is responsible for Ca2+-activated Cl- channels and can regulate both the resting membrane potential and cytosolic Ca2+ concentration, contributing to the proliferation and migration of BCECs. The present study provides novel information on the molecular mechanisms underlying the physiological functions of BCECs in the BBB and a novel target for therapeutic drugs for disorders associated with dysfunctions in the BBB.


Anoctamin-1/metabolism , Blood-Brain Barrier/metabolism , Brain/cytology , Calcium/metabolism , Chloride Channels/metabolism , Animals , Anoctamin-1/antagonists & inhibitors , Blood-Brain Barrier/cytology , Blood-Brain Barrier/diagnostic imaging , Brain/drug effects , Brain/metabolism , Cattle , Cell Line , Cell Movement/drug effects , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Endothelial Cells/cytology , Endothelial Cells/drug effects , Endothelial Cells/metabolism , HEK293 Cells , Humans , Male , Membrane Potentials/drug effects , Mice , Niflumic Acid/pharmacology , Pyrimidines/pharmacology , Thiazoles/pharmacology
8.
Assay Drug Dev Technol ; 18(3): 134-147, 2020 04.
Article En | MEDLINE | ID: mdl-32319819

The calcium-activated chloride channel, TMEM16A, is involved in airway hydration and bronchoconstriction and is a promising target for respiratory disease. Drug development efforts around channels require an electrophysiology-based assay for identifying inhibitors or activators. TMEM16A has proven to be a difficult channel to record on automated electrophysiology platforms due to its propensity for rundown. We developed an automated, whole-cell, electrophysiology assay on the QPatch-48 to evaluate small-molecule inhibitors of TMEM16A. In this assay, currents remained stable for a duration of roughly 11 min, allowing for the cumulative addition of five concentrations of compounds and resulted in reproducible IC50s. The absence of rundown was likely due to a low internal free-calcium level of 250 nM, which was high enough to produce large currents, but also maintained the voltage dependence of the channel. Current amplitude averaged 6 nA using the single-hole QPlate and the channel maintained outward rectification throughout the recording. Known TMEM16A inhibitors were tested and their IC50s aligned with those reported in the literature using manual patch-clamp. Once established, this assay was used to validate novel TMEM16A inhibitors that were identified in our high-throughput fluorescent-based assay, as well as to assist in structure-activity relationship efforts by the chemists. Overall, we demonstrate an easy to operate, reproducible, automated electrophysiology assay using the QPatch-48 for TMEM16A drug development efforts.


Automation , Benzbromarone/analysis , Drug Development , High-Throughput Screening Assays , Niflumic Acid/analysis , Small Molecule Libraries/analysis , Anoctamin-1/antagonists & inhibitors , Benzbromarone/pharmacology , Electrophysiological Phenomena/drug effects , Fluorescence , HEK293 Cells , Humans , Neoplasm Proteins/antagonists & inhibitors , Niflumic Acid/pharmacology , Small Molecule Libraries/pharmacology , Software
9.
Neuropharmacology ; 160: 107795, 2019 12 01.
Article En | MEDLINE | ID: mdl-31560908

Fenamates mefanamic and niflumic acids (MFA and NFA) induced dual potentiating and inhibitory effects on GABA currents recorded in isolated cerebellar Purkinje cells using the whole-cell patch-clamp and fast-application techniques. Regardless of the concentration, both drugs induced a pronounced prolongation of the current response. We demonstrated that the same concentration of drugs can produce both potentiating and inhibitory effects, depending on the GABA concentration, which indicates that both processes take place simultaneously and the net effect depends on the concentrations of both the agonist and fenamate. We found that the NFA-induced block is strongly voltage-dependent. The Woodhull analysis of the block suggests that NFA has two binding sites in the pore - shallow and deep. We built a homology model of the open GABAAR based on the cryo-EM structure of the open α1 GlyR and applied Monte-Carlo energy minimization to optimize the ligand-receptor complexes. A systematic search for MFA/NFA binding sites in the GABAAR pore revealed the existence of two sites, the location of which coincides well with predictions of the Woodhull model. In silico docking suggests that two fenamate molecules are necessary to occlude the pore. We showed that MFA, acting as a PAM, competes with an intravenous anesthetic etomidate for a common binding site. We built structural models of MFA and NFA binding at the transmembrane ß(+)/α(-) intersubunit interface. We suggested a hypothesis on the molecular mechanism underlying the prolongation of the receptor lifetime in open state after MFA/NFA binding and ß subunit specificity of the fenamate potentiation.


Anti-Inflammatory Agents, Non-Steroidal/pharmacology , GABA-A Receptor Antagonists/pharmacology , Mefenamic Acid/pharmacology , Niflumic Acid/pharmacology , Receptors, GABA-A/metabolism , Anesthetics, Intravenous/pharmacology , Animals , Anti-Inflammatory Agents, Non-Steroidal/metabolism , Binding Sites/drug effects , Cells, Cultured , Drug Synergism , Etomidate/pharmacology , GABA-A Receptor Antagonists/metabolism , Mefenamic Acid/metabolism , Niflumic Acid/metabolism , Purkinje Cells/drug effects , Rats
10.
Cancer Res ; 79(17): 4399-4411, 2019 09 01.
Article En | MEDLINE | ID: mdl-31289134

The Hippo pathway plays a critical role in cell growth and tumorigenesis. The activity of TEA domain transcription factor 4 (TEAD4) determines the output of Hippo signaling; however, the regulation and function of TEAD4 has not been explored extensively. Here, we identified glucocorticoids (GC) as novel activators of TEAD4. GC treatment facilitated glucocorticoid receptor (GR)-dependent nuclear accumulation and transcriptional activation of TEAD4. TEAD4 positively correlated with GR expression in human breast cancer, and high expression of TEAD4 predicted poor survival of patients with breast cancer. Mechanistically, GC activation promoted GR interaction with TEAD4, forming a complex that was recruited to the TEAD4 promoter to boost its own expression. Functionally, the activation of TEAD4 by GC promoted breast cancer stem cells maintenance, cell survival, metastasis, and chemoresistance both in vitro and in vivo. Pharmacologic inhibition of TEAD4 inhibited GC-induced breast cancer chemoresistance. In conclusion, our study reveals a novel regulation and functional role of TEAD4 in breast cancer and proposes a potential new strategy for breast cancer therapy. SIGNIFICANCE: This study provides new insight into the role of glucocorticoid signaling in breast cancer, with potential for clinical translation.


Breast Neoplasms/pathology , DNA-Binding Proteins/metabolism , Drug Resistance, Neoplasm , Muscle Proteins/metabolism , Receptors, Glucocorticoid/metabolism , Transcription Factors/metabolism , Adaptor Proteins, Signal Transducing/genetics , Adaptor Proteins, Signal Transducing/metabolism , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Breast Neoplasms/mortality , Cell Line, Tumor , Cell Nucleus/metabolism , DNA-Binding Proteins/genetics , Dexamethasone/pharmacology , Drug Resistance, Neoplasm/drug effects , Female , Glucocorticoids/metabolism , Glucocorticoids/pharmacology , Humans , Mice, Nude , Muscle Proteins/genetics , Niflumic Acid/pharmacology , Receptors, Glucocorticoid/genetics , Signal Transduction , TEA Domain Transcription Factors , Trans-Activators/genetics , Trans-Activators/metabolism , Transcription Factors/genetics , Transcriptional Coactivator with PDZ-Binding Motif Proteins , Xenograft Model Antitumor Assays , YAP-Signaling Proteins
11.
Am J Physiol Cell Physiol ; 317(3): C466-C480, 2019 09 01.
Article En | MEDLINE | ID: mdl-31242393

The swelling-activated chloride current (ICl,swell) is induced when a cell swells and plays a central role in maintaining cell volume in response to osmotic stress. The major contributor of ICl,swell is the volume-regulated anion channel (VRAC). Leucine-rich repeat containing 8A (LRRC8A; SWELL1) was recently identified as an essential component of VRAC, but the mechanisms of VRAC activation are still largely unknown; moreover, other Cl- channels, such as anoctamin 1 (ANO1), were also suggested to contribute to ICl,swell. In this present study, we investigated the roles of LRRC8A and ANO1 in activation of ICl,swell; we also explored the role of intracellular Ca2+ in ICl,swell activation. We used a CRISPR/Cas9 gene editing approach, electrophysiology, live fluorescent imaging, selective pharmacology, and other approaches to show that both LRRC8A and ANO1 can be activated by cell swelling in HEK293 cells. Yet, both channels contribute biophysically and pharmacologically distinct components to ICl,swell, with LRRC8A being the major component. Cell swelling induced oscillatory Ca2+ transients, and these Ca2+ signals were required to activate both the LRRC8A- and ANO1-dependent components of ICl,swell. Both ICl,swell components required localized rather than global Ca2+ for activation. Interestingly, while intracellular Ca2+ was necessary and sufficient to activate ANO1, it was necessary but not sufficient to activate LRRC8A-mediated currents. Finally, Ca2+ transients linked to the ICl,swell activation were mediated by the G protein-coupled receptor-independent PLC isoforms.


Calcium Signaling/physiology , Cell Size , Chloride Channels/physiology , Animals , Anoctamin-1/antagonists & inhibitors , Anoctamin-1/physiology , CHO Cells , Calcium Signaling/drug effects , Cell Size/drug effects , Chloride Channels/antagonists & inhibitors , Cricetinae , Cricetulus , Cyclooxygenase Inhibitors/pharmacology , Enzyme Inhibitors/pharmacology , HEK293 Cells , Humans , Membrane Proteins/antagonists & inhibitors , Membrane Proteins/physiology , Neoplasm Proteins/antagonists & inhibitors , Neoplasm Proteins/physiology , Niflumic Acid/pharmacology , Thapsigargin/pharmacology
12.
Neurotox Res ; 35(1): 244-254, 2019 Jan.
Article En | MEDLINE | ID: mdl-30178287

Significant body of evidence suggests that abnormal kynurenic acid (KYNA) level is involved in the pathophysiology of central nervous system disorders. In the brain, KYNA is synthesized from kynurenine (KYN) by kynurenine aminotransferases (KATs), predominantly by KAT II isoenzyme. Blockage of ionotropic glutamate (GLU) receptors is a main cellular effect of KYNA. High KYNA levels have been linked with psychotic symptoms and cognitive dysfunction in animals and humans. As immunological imbalance and impaired glutamatergic neurotransmission are one of the crucial processes in neurological pathologies, we aimed to analyze the effect of anti-inflammatory agents, inhibitors of cyclooxygenase-2 (COX-2): celecoxib, niflumic acid, and parecoxib, on KYNA synthesis and KAT II activity in rat brain in vitro. The influence of COX-2 inhibitors was examined in rat brain cortical slices and on isolated KAT II enzyme. Niflumic acid and parecoxib decreased in a dose-dependent manner KYNA production and KAT II activity in rat brain cortex in vitro, whereas celecoxib was ineffective. Molecular docking results suggested that niflumic acid and parecoxib interact with an active site of KAT II. In conclusion, niflumic acid and parecoxib are dual COX-2 and KAT II inhibitors.


Brain/drug effects , Brain/metabolism , Cyclooxygenase 2 Inhibitors/pharmacology , Kynurenic Acid/metabolism , Animals , Cyclooxygenase 2/metabolism , Cyclooxygenase 2 Inhibitors/chemistry , Dose-Response Relationship, Drug , Humans , Isoxazoles/pharmacology , Male , Molecular Docking Simulation , Niflumic Acid/pharmacology , Rats, Wistar , Tissue Culture Techniques , Transaminases/metabolism
13.
Am J Physiol Cell Physiol ; 316(5): C698-C710, 2019 05 01.
Article En | MEDLINE | ID: mdl-30566392

Multiple types of Cl- channels regulate smooth muscle excitability and contractility in vascular, gastrointestinal, and airway smooth muscle cells. However, little is known about Cl- channels in detrusor smooth muscle (DSM) cells. Here, we used inside-out single channel and whole cell patch-clamp recordings for detailed biophysical and pharmacological characterizations of Cl- channels in freshly isolated guinea pig DSM cells. The recorded single Cl- channels displayed unique gating with multiple subconductive states, a fully opened single-channel conductance of 164 pS, and a reversal potential of -41.5 mV, which is close to the ECl of -65 mV, confirming preferential permeability to Cl-. The Cl- channel demonstrated strong voltage dependence of activation (half-maximum of mean open probability, V0.5, ~-20 mV) and robust prolonged openings at depolarizing voltages. The channel displayed similar gating when exposed intracellularly to solutions containing Ca2+-free or 1 mM Ca2+. In whole cell patch-clamp recordings, macroscopic current demonstrated outward rectification, inhibitions by 4,4'-diisothiocyano-2,2'-stilbenedisulfonic acid (DIDS) and niflumic acid, and insensitivity to chlorotoxin. The outward current was reversibly reduced by 94% replacement of extracellular Cl- with I-, Br-, or methanesulfonate (MsO-), resulting in anionic permeability sequence: Cl->Br->I->MsO-. While intracellular Ca2+ levels (0, 300 nM, and 1 mM) did not affect the amplitude of Cl- current and outward rectification, high Ca2+ slowed voltage-step current activation at depolarizing voltages. In conclusion, our data reveal for the first time the presence of a Ca2+-independent DIDS and niflumic acid-sensitive, voltage-dependent Cl- channel in the plasma membrane of DSM cells. This channel may be a key regulator of DSM excitability.


Cell Membrane/metabolism , Chloride Channels/metabolism , Myocytes, Smooth Muscle/metabolism , Urinary Bladder/cytology , Urinary Bladder/metabolism , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Cell Membrane/drug effects , Cells, Cultured , Chloride Channels/antagonists & inhibitors , Guinea Pigs , Male , Myocytes, Smooth Muscle/drug effects , Niflumic Acid/pharmacology , Urinary Bladder/drug effects
14.
Dig Dis Sci ; 63(12): 3329-3338, 2018 12.
Article En | MEDLINE | ID: mdl-30155840

BACKGROUND: Irritable bowel syndrome (IBS) is a common disease with intestinal dysmotility, whose mechanism remains elusive. TMEM16A is a calcium-activated chloride channel (CaCC) involved in intestinal slow-wave generation. AIMS: To investigate whether TMEM16A is involved in colonic dysmotility in IBS. METHODS: A rat model of IBS was established by chronic water avoidance stress (WAS). Colonic pathological alterations were evaluated histologically, and intestinal motility was assessed by intestinal transit time (ITT) and fecal water content (FWC). Visceral sensitivity was determined by visceromotor response (VMR) to colorectal distension (CRD). TMEM16A expression was evaluated by RT-PCR, Western blot, and immunofluorescence. Colonic muscle strip contractility was measured by isometric transducers, and the effect of niflumic acid (NFA), a CaCC antagonist, on colonic motility was examined. RESULTS: After 10 days of WAS exposure, ITT was decreased and FWC was elevated. Furthermore, VMR magnitude of WAS rats in response to CRD was significantly enhanced. Protein and mRNA levels of TMEM16A in colon were considerably increased after WAS. The percentage of TMEM16A-positive neurons in myenteric plexus (MP) of WAS rats was significantly higher than controls. Pharmacological blockade of TMEM16A activity by NFA considerably enhanced ITT, with concentration-dependent declines in FWC and VMR magnitude in NFA-treated rats. Further, spontaneous contraction of colonic strips of NFA-treated rats was significantly ameliorated in a concentration-dependent manner in vitro. CONCLUSIONS: Upregulation of TMEM16A in MP neurons may play an important role in chronic stress-induced colonic hypermotility, making CaCC-blocking drugs a putatively effective treatment method for colonic hypermotility in IBS.


Anoctamin-1/metabolism , Colon , Gastrointestinal Motility , Irritable Bowel Syndrome , Niflumic Acid/pharmacology , Animals , Anoctamin-1/antagonists & inhibitors , Colon/drug effects , Colon/innervation , Colon/physiopathology , Cyclooxygenase Inhibitors/pharmacology , Disease Models, Animal , Gastrointestinal Motility/drug effects , Gastrointestinal Motility/physiology , Irritable Bowel Syndrome/drug therapy , Irritable Bowel Syndrome/metabolism , Irritable Bowel Syndrome/physiopathology , Myenteric Plexus , Rats , Treatment Outcome , Up-Regulation
15.
Br J Pharmacol ; 175(10): 1770-1780, 2018 05.
Article En | MEDLINE | ID: mdl-29500929

BACKGROUND AND PURPOSE: Although chloride channels are involved in several physiological processes and acquired diseases, the availability of compounds selectively targeting CLC proteins is limited. ClC-1 channels are responsible for sarcolemma repolarization after an action potential in skeletal muscle and have been associated with myotonia congenita and myotonic dystrophy as well as with other muscular physiopathological conditions. To date only a few ClC-1 blockers have been discovered, such as anthracene-9-carboxylic acid (9-AC) and niflumic acid (NFA), whereas no activator exists. The absence of a ClC-1 structure and the limited information regarding the binding pockets in CLC channels hamper the identification of improved modulators. EXPERIMENTAL APPROACH: Here we provide an in-depth characterization of drug binding pockets in ClC-1 through an integrated in silico and experimental approach. We first searched putative cavities in a homology model of ClC-1 built upon an eukaryotic CLC crystal structure, and then validated in silico data by measuring the blocking ability of 9-AC and NFA on mutant ClC-1 channels expressed in HEK 293 cells. KEY RESULTS: We identified four putative binding cavities in ClC-1. 9-AC appears to interact with residues K231, R421 and F484 within the channel pore. We also identified one preferential binding cavity for NFA and propose R421 and F484 as critical residues. CONCLUSIONS AND IMPLICATIONS: This study represents the first effort to delineate the binding sites of ClC-1. This information is fundamental to discover compounds useful in the treatment of ClC-1-associated dysfunctions and might represent a starting point for specifically targeting other CLC proteins.


Algorithms , Anthracenes/pharmacology , Chloride Channels/antagonists & inhibitors , Molecular Docking Simulation , Niflumic Acid/pharmacology , Anthracenes/chemistry , Binding Sites/drug effects , Chloride Channels/genetics , Chloride Channels/metabolism , HEK293 Cells , Humans , Ligands , Mutation , Niflumic Acid/chemistry
16.
J Cell Biochem ; 119(7): 5944-5959, 2018 07.
Article En | MEDLINE | ID: mdl-29600587

Ca2+ -activated Cl- channels (CaCCs) are anionic channels that regulate many important physiological functions associated with chloride and calcium flux in some somatic cells. The molecular identity of CaCCs was revealed to be TMEM16A and TMEM16B (also known as Anoctamin or ANO1 and ANO2, respectively) in all eukaryotes. A recent study suggests the presence of TMEM16A in human sperm and a relationship with the rhZP-induced acrosome reaction. However, to the best of our knowledge, little is known about the role of TMEM16A in other spermatic processes such as capacitation or motility. In this study, we evaluated the effects of two TMEM16A antagonists on capacitation, acrosome reaction, and motility in guinea pig sperm; these antagonists were T16Ainh-A01, belonging to a second generation of potent antagonists of TMEM16A, and niflumic acid (NFA), a well-known antagonist of TMEM16A (CaCCs). First of all, we confirmed that the absence of Cl- in the capacitation medium changes motility parameters, capacitation, and the progesterone-induced acrosome reaction. Using a specific antibody, TMEM16A was found as a protein band of ∼120 kDa, which localization was in the apical crest of the acrosome and the middle piece of the flagellum. Inhibition of TMEM16A by T16Ainh-A01 affected sperm physiology by reducing capacitation, blocking the progesterone-induced acrosome reaction under optimal capacitation conditions, inhibiting progressive motility, and the acquisition of hyperactivated motility, diminishing [Ca2+ ]i, and increasing [Cl- ]i. These changes in sperm kinematic parameters provide new evidence of the important role played by TMEM16A in the production of sperm capable of fertilizing oocytes.


Anoctamin-1/antagonists & inhibitors , Pyrimidines/pharmacology , Sperm Capacitation/drug effects , Sperm Motility/drug effects , Thiazoles/pharmacology , Acrosome Reaction/drug effects , Animals , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Calcium/metabolism , Chloride Channels/antagonists & inhibitors , Chlorides/metabolism , Guinea Pigs , Male , Niflumic Acid/pharmacology
17.
Cell Physiol Biochem ; 45(3): 867-882, 2018.
Article En | MEDLINE | ID: mdl-29421809

BACKGROUND/AIMS: In the human genome, more than 400 genes encode ion channels, which are ubiquitously expressed and often coexist and participate in almost all physiological processes. Therefore, ion channel blockers represent fundamental tools in discriminating the contribution of individual channel types to a physiological phenomenon. However, unspecific effects of these compounds may represent a confounding factor. Three commonly used chloride channel inhibitors, i.e. 4,4'-diisothiocyano-2,2'-stilbene-disulfonic acid (DIDS), 5-nitro-2-[(3-phenylpropyl) amino]benzoic acid (NPPB) and the anti-inflammatory drug niflumic acid were tested to identify the lowest concentration effective on Cl- channels and ineffective on K+ channels. METHODS: The activity of the above mentioned compounds was tested by whole cell patch-clamp on the swelling-activated Cl- current ICl,swell and on the endogenous voltage-dependent, outwardly rectifying K+ selective current in human kidney cell lines (HEK 293/HEK 293 Phoenix). RESULTS: Micromolar (1-10 µM) concentrations of DIDS and NPPB could not discriminate between the Cl- and K+ selective currents. Specifically, 1 µM DIDS only affected the K+ current and 10 µM NPPB equally affected the Cl- and K+ currents. Only relatively high (0.1-1 mM) concentrations of DIDS and prolonged (5 minutes) exposure to 0.1-1 mM NPPB preferentially suppressed the Cl- current. Niflumic acid preferentially inhibited the Cl- current, but also significantly affected the K+ current. The endogenous voltage-dependent, outwardly rectifying K+ selective current in HEK 293/HEK 293 Phoenix cells was shown to arise from the Kv 3.1 channel, which is extensively expressed in brain and is involved in neurological diseases. CONCLUSION: The results of the present study underscore that sensitivity of a given physiological phenomenon to the Cl- channel inhibitors NPPB, DIDS and niflumic acid may actually arise from an inhibition of Cl- channels but can also result from an inhibition of voltage-dependent K+ channels, including the Kv 3.1 channel. The use of niflumic acid as anti-inflammatory drug in patients with concomitant Kv 3.1 dysfunction may result contraindicated.


Action Potentials/drug effects , Kv1.3 Potassium Channel/metabolism , Potassium Channel Blockers/pharmacology , Potassium/metabolism , Animals , Chlorides/metabolism , Epithelial Cells/cytology , HEK293 Cells , Humans , Kidney Tubules, Proximal/cytology , Kv1.3 Potassium Channel/antagonists & inhibitors , Kv1.3 Potassium Channel/genetics , Mice , NIH 3T3 Cells , Niflumic Acid/chemistry , Niflumic Acid/pharmacology , Nitrobenzoates/chemistry , Nitrobenzoates/pharmacology , Patch-Clamp Techniques , Potassium Channel Blockers/chemistry , RNA Interference , RNA, Small Interfering/metabolism
18.
Biopharm Drug Dispos ; 39(3): 135-142, 2018 Mar.
Article En | MEDLINE | ID: mdl-29319909

Canagliflozin, used to treat type 2 diabetes mellitus (T2DM), is commonly co-administered with sulfonylureas. The objective of the present study was to evaluate the possible inhibitory effect of sulfonylureas and non-steroidal anti-inflammatory drugs (NSAIDs) on canagliflozin metabolism in vitro. Three sulfonylurea derivatives were evaluated as inhibitors: chlorpropamide, glimepiride and gliclazide. Two other NSAIDs were used as positive control inhibitors: niflumic acid and diclofenac. The rate of formation of canagliflozin metabolites was determined by HPLC analysis of in vitro incubations of canagliflozin as a substrate with and without inhibitors, using human liver microsomes (HLMs). Among sulfonylureas, glimepiride showed the most potent inhibitory effect against canagliflozin M7 metabolite formation, with an IC50 value of 88 µm, compared to chlorpropamide and gliclazide with IC50 values of more than 500 µm. Diclofenac inhibited M5 metabolite formation more than M7, with IC50 values of 32 µm for M5 and 80 µm for M7. Niflumic acid showed no inhibition activity against M5 formation, but had relatively selective inhibitory potency against M7 formation, which is catalysed by UGT1A9, with an IC50 value of 1.9 µm and an inhibition constant value of 0.8 µm. A clinical pharmacokinetic interaction between canagliflozin and sulfonylureas is unlikely. However, a possible clinically important drug interaction between niflumic acid and canagliflozin has been identified.


Canagliflozin/metabolism , Chlorpropamide/pharmacology , Diclofenac/pharmacology , Gliclazide/pharmacology , Microsomes, Liver/drug effects , Microsomes, Liver/metabolism , Niflumic Acid/pharmacology , Sulfonylurea Compounds/pharmacology , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Canagliflozin/pharmacokinetics , Dose-Response Relationship, Drug , Drug Interactions , Humans , Hypoglycemic Agents/pharmacology
19.
Eur J Med Chem ; 153: 131-139, 2018 Jun 10.
Article En | MEDLINE | ID: mdl-28502586

Three novel Zn(II) complexes of NSAID niflumic acid (Hnif) were prepared and studied, namely; [Zn(MeOH)4(nif)2] (1), [Zn(cyclam)(nif)2] (2) and [Zn(nif)2(tmen)] (3), where nif is deprotonated niflumic acid, cyclam is 1,4,8,11-Tetraazacyclotetradecane and tmen is N,N,N',N'-Tetramethylethylenediamine. The complexes have been characterized by infrared spectroscopy, elemental and thermal analysis and single-crystal X-ray structure analysis. All three complexes contain two deprotonated niflumato anions monodentately coordinated via carboxylato groups. Furthermore, fluorescence binding studies of the prepared compounds with human genomic DNA-EB (ethidium bromide) were carried out, which suggest that all complexes are able to bind to DNA via intercalation. Moreover, from the obtained results it followed that complexes 2 and 3 bind to DNA from the tissue with aortic aneurysm (aDNA) and control (cDNA) with a different strength. Additionally, complexes 1-3 exhibit good binding affinity to human serum albumin with high binding constant.


Anti-Inflammatory Agents, Non-Steroidal/chemistry , Coordination Complexes/chemistry , DNA/metabolism , Intercalating Agents/chemistry , Niflumic Acid/analogs & derivatives , Serum Albumin/metabolism , Zinc/chemistry , Anti-Inflammatory Agents, Non-Steroidal/pharmacology , Coordination Complexes/pharmacology , Crystallography, X-Ray , Humans , Intercalating Agents/pharmacology , Models, Molecular , Niflumic Acid/pharmacology , Zinc/pharmacology
20.
Clin Exp Hypertens ; 39(4): 295-305, 2017.
Article En | MEDLINE | ID: mdl-28513236

The aim of the present study is to investigate the effects of hypertension on the gap junctions between vascular smooth muscle cells (VSMCs) in the cerebral arteries (CAs) of spontaneously hypertensive rats (SHRs). The functions of gap junctions in the CAs of VSMCs in SHRs and control normotensive Wistar-Kyoto (WKY) rats were studied using whole-cell patch clamp recordings and pressure myography, and the expression levels of connexins were analyzed using reverse transcription-quantitative polymerase chain reaction and Western blot analyses. Whole-cell patch clamp measurements revealed that the membrane capacitance and conductance of in situ VSMCs in the CAs were significantly greater in SHRs than in WKY rats, suggesting that gap junction coupling is enhanced between VSMCs in the CAs of SHRs. Application of the endothelium-independent vasoconstrictors KCl or phenylephrine (PE) stimulated a greater vasoconstriction in the CAs of SHRs than in those of WKY rats. The EC50 value of KCl was 24.9 mM (n = 14) and 36.9 mM (n=12) for SHRs and WKY rats, respectively. The EC50 value of PE was 0.9 µM (n = 7) and 2.2 µM (n = 7) for SHRs and WKY rats, respectively. Gap junction inhibitors 18ß-glycyrrhetinic acid (18ß-GA), niflumic acid (NFA), and 2-aminoethoxydiphenyl borate (2-APB) attenuated KCl-induced vasoconstriction in SHRs and WKY rats. The mRNA and protein expression levels of the gap junction protein connexin 45 (Cx45) were significantly higher in the CAs of SHRs than in those of WKY rats. Phosphorylated Cx43 protein expression was significantly higher in the CAs of SHRs than in those of WKY rats, despite the total Cx43 mRNA and protein expression levels in the cerebral artery (CA) exhibiting no significant difference between SHRs and WKY rats. Increases in the expression of Cx45 and phosphorylation of Cx43 may promote gap junction communication among VSMCs in the CAs of SHRs, which may enhance the contractile response of the CA to vasoconstrictors.


Cerebral Arteries/physiopathology , Gap Junctions/drug effects , Gap Junctions/physiology , Hypertension/physiopathology , Muscle, Smooth, Vascular/physiopathology , Animals , Boron Compounds/pharmacology , Cerebral Arteries/drug effects , Cerebral Arteries/metabolism , Connexin 43/genetics , Connexin 43/metabolism , Connexins/genetics , Connexins/metabolism , Electric Capacitance , Electrophysiological Phenomena , Glycyrrhetinic Acid/analogs & derivatives , Glycyrrhetinic Acid/pharmacology , Hypertension/metabolism , Male , Muscle, Smooth, Vascular/drug effects , Muscle, Smooth, Vascular/metabolism , Niflumic Acid/pharmacology , Phenylephrine/pharmacology , Phosphorylation , Potassium Chloride/pharmacology , RNA, Messenger/metabolism , Rats , Rats, Inbred SHR , Rats, Inbred WKY , Vasoconstriction/drug effects , Vasoconstrictor Agents/pharmacology
...