Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Int J Biol Macromol ; 164: 45-52, 2020 Dec 01.
Article in English | MEDLINE | ID: mdl-32679335

ABSTRACT

Nanotechnology based antimicrobial drugs are developed to enhance their properties to combat multidrug resistant microbes. Carbenoxolone (CBX) is a semi-synthetic derivate of a natural substance from the licorice plant, with anti- (inflammatory, fungal, viral, microbial, fibrotic and cancer) properties. Though used to treat gastric ulcers, its low aqueous stability, low bioavailability and toxicity limited the drug's utility. To enhance its antimicrobial activity and reduce cytotoxicity, a controlled release nanoformulation was developed using natural biodegradable polymer chitosan (CS) as a carrier which is biocompatible, nontoxic with placid antimicrobial property. UV-visible spectroscopy, electron microscopy, and Fourier transform infrared spectroscopy were used for characterization of the resultant CS-CBX nanoparticles (NPs). They were spherical with uniform dispersion, ~200 nm in size with surface charge of +18.6 mV and drug encapsulation of >80%. Drug release kinetics exhibited a controlled release of 86% over 36 h following zero order kinetics. The anti-microbial activity against common pathogenic Gram -ve and +ve bacteria and yeast increased ~2-fold with a concomitant 4-fold reduction in cytotoxicity assessed using human lung adeno carcinoma (A549) cells. This study demonstrates the affirmative aspects of CS-CBX NPs as a promising antibacterial agent and may facilitate repositioning of the drug for diverse applications.


Subject(s)
Carbenoxolone/chemistry , Chitosan/chemistry , Nanoparticles/chemistry , Anti-Infective Agents/pharmacology , Candida albicans/drug effects , Carbenoxolone/pharmacology , Carbenoxolone/toxicity , Cell Line, Tumor , Chitosan/pharmacology , Chitosan/toxicity , Drug Carriers , Drug Liberation , Gram-Negative Bacteria/drug effects , Gram-Positive Bacteria/drug effects , Humans , Hydrogen-Ion Concentration , Materials Testing , Microbial Sensitivity Tests , Nanoparticles/toxicity , Spectroscopy, Fourier Transform Infrared , Static Electricity
2.
Neuropharmacology ; 170: 108041, 2020 06 15.
Article in English | MEDLINE | ID: mdl-32213357

ABSTRACT

Our previous studies have shown that ginsenoside Rg1 (Rg1) exerts antidepressant-like effects in animal models of depression, accompanied by an improvement of astrocytic gap junction functions. However, whether connexin 43 (Cx43), the major connexin forming gap junctions between astrocytes, is the key regulator of Rg1-induced antidepressant-like effects is still unknown. In this study, we examine in vitro and in vivo the involvement of Cx43 in the antidepressant effects of Rg1. Corticosterone was used to establish an in vitro rat model of depression. Treatment with Rg1 1 h prior to corticosterone significantly improved the cell viability of astrocytes, which was significantly inhibited by carbenoxolone, a widely used gap junction inhibitor. Moreover, Rg1 treatment significantly ameliorated antidepressant-sensitive behaviours induced by infusion of carbenoxolone or Gap26, a selective inhibitor of Cx43, into the prefrontal cortex of the animals. Rg1 treatment increased the expression of Cx43 compared with Gap26 group. According to these results, the antidepressant-like effects of Rg1 were mainly mediated by Cx43-formed gap junctions.


Subject(s)
Connexin 43/biosynthesis , Depression/drug therapy , Depression/metabolism , Disease Models, Animal , Ginsenosides/administration & dosage , Animals , Animals, Newborn , Astrocytes/drug effects , Astrocytes/metabolism , Carbenoxolone/administration & dosage , Carbenoxolone/toxicity , Cells, Cultured , Central Nervous System Agents/administration & dosage , Connexin 43/antagonists & inhibitors , Depression/chemically induced , Dose-Response Relationship, Drug , Male , Peptides/administration & dosage , Peptides/toxicity , Prefrontal Cortex/drug effects , Prefrontal Cortex/metabolism , Rats , Rats, Sprague-Dawley
3.
J Toxicol Sci ; 43(8): 493-506, 2018.
Article in English | MEDLINE | ID: mdl-30078835

ABSTRACT

We used motion field imaging to characterize the conduction and contraction of a sheet of cardiomyocytes derived from human induced pluripotent stem cells (hiPS-CMs). A hiPS-CMs sheet of 2.8 mm × 2.8 mm allowed us to simultaneously measure the conduction and the contraction properties in the same cells. Pharmacological responses in the hiPS-CMs of four typical cardiac functional modulators, Na+ channel blocker (lidocaine), Ca2+ channel blocker (diltiazem), gap-junction inhibitor (carbenoxolone), and ß-adrenergic stimulator (isoproterenol), were investigated, and the results were compared to those found using the isolated guinea-pig heart model perfused by the Langendorff method. The conduction speed of excitation waves in hiPS-CMs was decreased by lidocaine, diltiazem, and carbenoxolone, and increased by isoproterenol, and these results were in accordance with the changes in the conduction parameters of electrocardiogram (QRS duration, PR interval, and P duration) in the Langendorff guinea-pig heart model. The maximum speeds for contraction and relaxation, which respectively represent the contraction and relaxation kinetics of hiPS-CMs, were decreased by lidocaine and diltiazem, and increased by isoproterenol. These results also corresponded to alterations in the contractile and relaxation parameters found by measuring left ventricular pressure (LVdP/dtmax and LVdP/dtmin) in the Langendorff guinea-pig heart model. From these lines of evidence, it was suggested that hiPS-CMs enable us to evaluate the cardiac toxicities associated with conduction disturbance or contractile dysfunction, and thereby would be useful as an integrated assessment of cardiac function.


Subject(s)
Heart Rate/drug effects , Heart Rate/physiology , Heart/diagnostic imaging , Heart/physiology , Induced Pluripotent Stem Cells , Isolated Heart Preparation , Myocardial Contraction/drug effects , Myocardial Contraction/physiology , Myocytes, Cardiac/physiology , Animals , Carbenoxolone/toxicity , Diltiazem/toxicity , Guinea Pigs , Humans , Isoproterenol/toxicity , Lidocaine/toxicity
4.
Neuroscience ; 194: 262-71, 2011 Oct 27.
Article in English | MEDLINE | ID: mdl-21839806

ABSTRACT

Cortical spreading depression (CSD) has been shown to cause secondary cell loss in experimental models of brain injury and in patients, and blocking of CSD is a potential neuroprotective strategy. Here we tested the hypothesis that gap junctions affect CSD under physiological conditions as well as infarct development in a rat two-vein occlusion model suited to study pathophysiology of the penumbra (n = 71). We applied the gap junction blocker carbenoxolone (CBX) or saline intra-ventricularly. Interestingly, CBX temporarily increased systemic blood pressure and cortical blood flow (41% and 53%, 15 min after 250 µg CBX). We induced CSD with cortical microinjection of potassium chloride (KCl), counted how many spontaneous CSDs after CSD induction were elicited and measured the propagation velocity. After 250 µg CBX administration, significant 37.5 ± 6.5 additional CSDs were seen. CSD velocity increased significantly after 50 µg and 250 µg CBX. Occlusion of two adjacent cortical veins using Rose Bengal dye and fiberoptic illumination followed by 250 µg CBX or saline showed a significant more than doubling of infarct volumes 7 days after CBX. The current experiments provide evidence that CBX can accelerate the initiation and propagation of CSD suggesting opening of gap junctions is not required for CSD propagation. Blocking gap junctions worsens outcome from focal cerebral ischemia. Hence, measures intended to improve spatial buffering via astroglial gap junctions could have therapeutic potential in disease processes involving CSD.


Subject(s)
Brain Infarction/physiopathology , Brain Ischemia/physiopathology , Carbenoxolone/toxicity , Cortical Spreading Depression/drug effects , Gap Junctions/drug effects , Animals , Anti-Ulcer Agents/toxicity , Brain Infarction/chemically induced , Brain Infarction/metabolism , Brain Ischemia/chemically induced , Brain Ischemia/metabolism , Cerebrovascular Circulation/drug effects , Cerebrovascular Circulation/physiology , Cortical Spreading Depression/physiology , Disease Models, Animal , Gap Junctions/physiology , Male , Rats , Rats, Wistar
5.
Pak J Pharm Sci ; 22(4): 349-54, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19783510

ABSTRACT

Previous experimental studies have shown the protective effects of CBX on brain ischemic injures in global and in vitro models of ischemia. However, effects of CBX in temporary model of focal cerebral ischemia are not clear. Hence, the aim of this study was to investigate the effects of central microinjection of CBX on post-ischemic reperfusion injuries in a temporary model of focal cerebral ischemia. Transient focal cerebral ischemia was induced in rats by 60 min middle cerebral artery occlusion (MCAO), followed by 23 h reperfusion. CBX was administered into the right ventricle at doses of 1, 12, 25, 50 and/or 100 microg/kg at the beginning of MCAO. Cortical and striatal infarct volumes and motor dysfunctions were assessed 24 h after MCAO. Administration of CBX at doses of 1, 12, 25 and/or 50 microg/kg significantly reduced cortical infarct volumes by 35%, 49%, 41% and 43%, respectively (P<0.001). In addition, CBX only at dose of 25 microg/kg significantly reduced striatal infarct volume and improved neurological dysfunctions (P<0.01). Our findings indicated that central microinjection of CBX has protective effect on against ischemic reperfusion injuries in a transient model of focal cerebral ischemia.


Subject(s)
Carbenoxolone/administration & dosage , Carbenoxolone/toxicity , Cerebral Cortex , Ischemic Attack, Transient/chemically induced , Neostriatum , Animals , Disease Models, Animal , Dyskinesia, Drug-Induced/psychology , Infarction, Middle Cerebral Artery/chemically induced , Infarction, Middle Cerebral Artery/pathology , Ischemic Attack, Transient/pathology , Ischemic Attack, Transient/psychology , Male , Microinjections , Rats , Rats, Wistar
6.
J Neurochem ; 102(2): 508-21, 2007 Jul.
Article in English | MEDLINE | ID: mdl-17403140

ABSTRACT

The beneficial or detrimental role of gap junction communication in the pathophysiology of brain injury is still controversial. We used co-cultures of hippocampal astrocytes and neurons, where we identified homocellular astrocyte-astrocyte and heterocellular astrocyte-neuron coupling by fluorescence recovery after photobleaching, which was decreased by the gap junction blocker carbenoxolone (CBX). In these cultures, we determined the cell type-specific effects of CBX on the excitotoxic damage caused by N-methyl-D-aspartate (NMDA). We determined in both astrocytes and neurons the influence of CBX, alone or together with NMDA challenge, on cytotoxicity using propidium iodide labeling. CBX alone was not cytotoxic, but CBX treatment differentially accelerated the NMDA-induced cell death in both astrocytes and neurons. In addition, we measured mitochondrial potential using rhodamine 123, membrane potential using the oxonol dye bis(1,3-diethylthiobarbituric acid)trimethine oxonol, cytosolic Ca(2+) level using fura-2, and formation of reactive oxygen species (ROS) using dihydroethidium. CBX alone induced neither an intracellular Ca(2+) rise nor a membrane depolarization. However, CBX elicited a mitochondrial depolarization in both astrocytes and neurons and increased the ROS formation in neurons. In contrast, NMDA caused a membrane depolarization in neurons, coinciding with intracellular Ca(2+) rise, but neither mitochondrial depolarization nor ROS production seem to be involved in NMDA-mediated cytotoxicity. Pre-treatment with CBX accelerated the NMDA-induced membrane depolarization and prevented the repolarization of neurons after the NMDA challenge. We hypothesize that these effects are possibly mediated via blockage of gap junctions, and might be involved in the mechanism of CBX-induced acceleration of excitotoxic cell death, whereas the CBX-induced mitochondrial depolarization and ROS formation are not responsible for the increase in cytotoxicity. We conclude that both in astrocytes and neurons gap junctions provide protection against NMDA-induced cytotoxicity.


Subject(s)
Astrocytes/metabolism , Carbenoxolone/toxicity , Gap Junctions/metabolism , Hippocampus/metabolism , N-Methylaspartate/toxicity , Neurons/metabolism , Animals , Animals, Newborn , Anti-Ulcer Agents/toxicity , Astrocytes/drug effects , Cell Death/drug effects , Cell Death/physiology , Cell Survival/drug effects , Cell Survival/physiology , Cells, Cultured , Coculture Techniques , Cytoprotection/drug effects , Cytoprotection/physiology , Drug Synergism , Gap Junctions/drug effects , Hippocampus/cytology , Hippocampus/drug effects , Indicators and Reagents , Membrane Potential, Mitochondrial/drug effects , Membrane Potential, Mitochondrial/physiology , Neurons/drug effects , Neurotoxins/toxicity , Oxidative Stress/drug effects , Oxidative Stress/physiology , Rats , Rats, Wistar , Reactive Oxygen Species/metabolism
7.
Respir Physiol Neurobiol ; 152(1): 51-60, 2006 May.
Article in English | MEDLINE | ID: mdl-16159713

ABSTRACT

The role of gap junctions in the brainstem respiratory control system is ambiguous. In the present study, we used juvenile rats to determine whether blocking gap junctions altered eupnea or gasping in the in situ, arterially perfused rat preparation. Blockade of gap junctions with 100 microM carbenoxolone or 300 microM octanol did not produce any consistent changes in the timing or amplitude of integrated phrenic discharge or in the peak frequency in the power spectrum of phrenic nerve discharge during eupnea or ischemic gasping beyond those changes seen in time-control animals. These findings do not rule out a role for gap junctions in the expression of eupnea or gasping, but they do demonstrate that these intermembrane channels are not obligatory for either rhythm to occur.


Subject(s)
Gap Junctions/physiology , Pulmonary Ventilation/physiology , Respiratory Mechanics/physiology , Aldehydes/toxicity , Animals , Brain Stem/cytology , Carbenoxolone/toxicity , Gap Junctions/drug effects , Male , Phrenic Nerve/drug effects , Phrenic Nerve/physiology , Pulmonary Ventilation/drug effects , Rats , Rats, Sprague-Dawley , Reflex/physiology
8.
Antiviral Res ; 36(3): 191-5, 1997 Dec.
Article in English | MEDLINE | ID: mdl-9477119

ABSTRACT

Triterpenic compounds, such as glycyrrhizic acid (GRa) and carbenoxolone (CBX), have a synergistic effect with prostaglandin A1 on the inhibition of vaccinia virus (VV) replication in L929 cells. The fractional inhibitory concentration (FIC) values for GRa and CBX were 0.5 and 0.25, respectively. In the supernatant of triterpene treated cells, increased production of some prostaglandins was shown, whilst cell-associated prostaglandins and prostaglandins of the A series were only slightly influenced by the presence of triterpenes. From these findings there is no evidence that prostaglandin production and metabolism could be involved in the antiviral activity of triterpenes.


Subject(s)
Carbenoxolone/pharmacology , Glycyrrhizic Acid/pharmacology , Prostaglandins A/pharmacology , Vaccinia virus/drug effects , Animals , Carbenoxolone/toxicity , Cell Line , Chlorocebus aethiops , Drug Synergism , Glycyrrhizic Acid/toxicity , Mice , Molecular Structure , Prostaglandins/biosynthesis , Vero Cells
SELECTION OF CITATIONS
SEARCH DETAIL
...