Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 114
Filter
1.
Eur J Pharmacol ; 925: 175000, 2022 Jun 15.
Article in English | MEDLINE | ID: mdl-35525312

ABSTRACT

Dequalinium chloride has been used primarily as antiseptic compounds, but recently has been investigated for its effects on specific targets, including muscarinic acetylcholine receptors. Here we investigated dequalinium chloride as an antagonist to α7 nicotinic acetylcholine receptors. The pharmacological properties of dequalinium were established using cell lines stably co-transfected with the calcium-permeable human α7 nicotinic acetylcholine receptors and its chaperone NACHO, calcium dye fluorescent measurements or a calcium-sensitive protein reporter, and patch clamp recording of ionic currents. Using calcium dye fluorescence plate reader measurements, we find dequalinium chloride is an antagonist of α7 nicotinic acetylcholine receptors with an IC50 of 672 nM in response to activation with 500 µM acetylcholine chloride and positive allosteric modulator PNU-120596. However, using a membrane-tethered GCAMP7s calcium reporter allowed detection of α7-mediated calcium flux in the absence of PNU-120596. Using this approach revealed an IC50 of 157 nM for dequalinium on 300 µM acetylcholine-evoked currents. Using patch clamp recordings with 300 µM acetylcholine chloride and 10 µM PNU-120596, we find lower concentrations are sufficient to block ionic currents, with IC50 of 120 nM for dequalinium chloride and 54 nM for the related UCL 1684 compound. In summary, we find that dequalinium chloride and UCL1684, which are generally used to block SK-type potassium channels, are also highly effective antagonists of α7 nicotinic acetylcholine receptors. This finding, in combination with previous studies of muscarinic acetylcholine receptors, clearly establishes dequalinium compounds within the class of general anti-cholinergic antagonists.


Subject(s)
Dequalinium , Nicotinic Antagonists , alpha7 Nicotinic Acetylcholine Receptor , Acetylcholine/pharmacology , Calcium/metabolism , Cell Line , Dequalinium/pharmacology , Humans , Nicotinic Antagonists/pharmacology , Phenylurea Compounds/pharmacology , Receptors, Nicotinic/drug effects , alpha7 Nicotinic Acetylcholine Receptor/drug effects , alpha7 Nicotinic Acetylcholine Receptor/metabolism
2.
ChemMedChem ; 16(1): 105-107, 2021 01 08.
Article in English | MEDLINE | ID: mdl-33428329

ABSTRACT

Here we highlight a sound and unique work reported by Chen and co-workers entitled "HIV-1 fusion inhibitors targeting the membrane-proximal external region of Env spikes" (Xiao et al., Nat. Chem. Biol. 2020, 16, 529). In this article, the authors identify, by means of a clever antibody-guided strategy, several small molecules as fusion inhibitors of HIV-1 replication acting at the membrane proximal external region (MPER) of the HIV-1 envelope (Env) spike. MPER, which was previously recognized as a vaccine target, emerges as a novel druggable target for the discovery of HIV-1 fusion inhibitors. The compounds (exemplified by dequalinium and dequalinium-inspired analogues) prevent the conformational changes of Env from the prefusion species to the intermediate states required for membrane fusion. This work not only paves the way to novel, specific and useful anti-HIV-1 inhibitors, but also discloses new therapeutic strategies against other infectious diseases.


Subject(s)
HIV Fusion Inhibitors/chemistry , HIV-1/metabolism , env Gene Products, Human Immunodeficiency Virus/chemistry , Antibodies, Neutralizing/chemistry , Antibodies, Neutralizing/immunology , Binding Sites , Dequalinium/analogs & derivatives , Dequalinium/metabolism , Dequalinium/pharmacology , HIV Fusion Inhibitors/metabolism , HIV Fusion Inhibitors/pharmacology , Humans , Structure-Activity Relationship , Virus Internalization/drug effects , env Gene Products, Human Immunodeficiency Virus/metabolism
3.
Vet Microbiol ; 251: 108913, 2020 Dec.
Article in English | MEDLINE | ID: mdl-33166843

ABSTRACT

As a severe disease characterized by reproductive failure and respiratory distress, porcine reproductive and respiratory syndrome (PRRS) is one of the most leading threats to the swine industry worldwide. Highly evolving porcine reproductive and respiratory syndrome virus (PRRSV) strains with distinct genetic diversity make the current vaccination strategy much less cost-effective and thus urge alternative protective host directed therapeutic approaches. RACK1-PKC-NF-κB signalling axis was suggested as a potential therapeutic target for PRRS control, therefore we tested the inhibitory effect of PKC inhibitor dequalinium chloride (DECA) on the PRRSV infection in vitro. RT-qPCR, western blot, Co-IP and cytopathic effect (CPE) observations revealed that DECA suppressed PRRSV infection and protected Marc-145 cells and porcine alveolar macrophages (PAMs) from severe cytopathic effects, by repressing the PKCα expression, the interaction between RACK1 and PKCα, and subsequently the NF-κB activation. In conclusion, the data presented in this study shed more light on deeper understanding of the molecular pathogenesis upon PRRSV infection and more importantly suggested DECA as a potential promising drug candidate for PRRS control.


Subject(s)
Dequalinium/pharmacology , Porcine respiratory and reproductive syndrome virus/drug effects , Protein Kinase C-alpha/antagonists & inhibitors , Virus Replication/drug effects , Animals , Cell Line , Cells, Cultured , Cytopathogenic Effect, Viral , Macrophages, Alveolar/drug effects , Macrophages, Alveolar/virology , Signal Transduction , Swine
4.
Int J Mol Sci ; 21(17)2020 Aug 19.
Article in English | MEDLINE | ID: mdl-32825082

ABSTRACT

Structure-based virtual screening is a truly productive repurposing approach provided that reliable target structures are available. Recent progresses in the structural resolution of the G-Protein Coupled Receptors (GPCRs) render these targets amenable for structure-based repurposing studies. Hence, the present study describes structure-based virtual screening campaigns with a view to repurposing known drugs as potential allosteric (and/or orthosteric) ligands for the hM2 muscarinic subtype which was indeed resolved in complex with an allosteric modulator thus allowing a precise identification of this binding cavity. First, a docking protocol was developed and optimized based on binding space concept and enrichment factor optimization algorithm (EFO) consensus approach by using a purposely collected database including known allosteric modulators. The so-developed consensus models were then utilized to virtually screen the DrugBank database. Based on the computational results, six promising molecules were selected and experimentally tested and four of them revealed interesting affinity data; in particular, dequalinium showed a very impressive allosteric modulation for hM2. Based on these results, a second campaign was focused on bis-cationic derivatives and allowed the identification of other two relevant hM2 ligands. Overall, the study enhances the understanding of the factors governing the hM2 allosteric modulation emphasizing the key role of ligand flexibility as well as of arrangement and delocalization of the positively charged moieties.


Subject(s)
Allosteric Site , Anti-Infective Agents, Local/pharmacology , Cholinergic Agents/pharmacology , Dequalinium/pharmacology , Drug Repositioning , Receptors, Muscarinic/chemistry , Allosteric Regulation , Animals , Anti-Infective Agents, Local/chemistry , CHO Cells , Cholinergic Agents/chemistry , Cricetinae , Cricetulus , Dequalinium/chemistry , Humans , Ligands , Molecular Docking Simulation , Protein Binding , Receptors, Muscarinic/metabolism
5.
Acta Neurochir (Wien) ; 162(7): 1683-1690, 2020 07.
Article in English | MEDLINE | ID: mdl-32410120

ABSTRACT

BACKGROUND: Our current understanding of the role of dequalinium chloride (DECA) in the progression of glioma remains very limited. This study was aimed to investigate the effect of DECA on human glioma cell lines in vitro and vivo. METHODS: The underlying molecular mechanism was analyzed for developing potential targeted agents. MTT assay, genomic DNA electrophoresis, DAPI staining, TUNEL staining, and wound scratch assay were performed to evaluate the effect of DECA on human glioma cell lines. Bioinformatics methods were used to screen the possible signaling pathway proteins, and the expression of these proteins and the corresponding mRNA was measured. RESULTS: DECA significantly inhibited the growth and proliferation of human glioma cells. Screening of apoptosis-related proteins showed the mRNA expression level of 6 genes was significantly changed after DECA administration. CONCLUSION: This study shows that DECA effectively inhibits the growth of glioma cells in vitro and vivo. DECA may promote glioma cell apoptosis by affecting the expression of NFKB2, HRAS, NF1, CBL, RAF1, and BCL-2 genes.


Subject(s)
Apoptosis/drug effects , Brain Neoplasms/metabolism , Cell Proliferation/drug effects , Dequalinium/pharmacology , Glioma/metabolism , Brain Neoplasms/genetics , Cell Line, Tumor , Glioma/genetics , Humans
6.
Nat Chem Biol ; 16(5): 529-537, 2020 05.
Article in English | MEDLINE | ID: mdl-32152540

ABSTRACT

Combination antiretroviral therapy has transformed HIV-1 infection, once a fatal illness, into a manageable chronic condition. Drug resistance, severe side effects and treatment noncompliance bring challenges to combination antiretroviral therapy implementation in clinical settings and indicate the need for additional molecular targets. Here, we have identified several small-molecule fusion inhibitors, guided by a neutralizing antibody, against an extensively studied vaccine target-the membrane proximal external region (MPER) of the HIV-1 envelope spike. These compounds specifically inhibit the HIV-1 envelope-mediated membrane fusion by blocking CD4-induced conformational changes. An NMR structure of one compound complexed with a trimeric MPER construct reveals that the compound partially inserts into a hydrophobic pocket formed exclusively by the MPER residues, thereby stabilizing its prefusion conformation. These results suggest that the MPER is a potential therapeutic target for developing fusion inhibitors and that strategies employing an antibody-guided search for novel therapeutics may be applied to other human diseases.


Subject(s)
Anti-HIV Agents/chemistry , Anti-HIV Agents/pharmacology , HIV Envelope Protein gp41/chemistry , HIV Envelope Protein gp41/metabolism , Virus Internalization/drug effects , Binding Sites , CD4 Antigens/metabolism , Cell Membrane/metabolism , Dequalinium/chemistry , Dequalinium/pharmacology , Drug Evaluation, Preclinical/methods , Fluorescence Polarization , HEK293 Cells , HIV Envelope Protein gp41/genetics , HIV-1/pathogenicity , Humans , Hydrophobic and Hydrophilic Interactions , Magnetic Resonance Spectroscopy , Molecular Structure , Mutation , Small Molecule Libraries/chemistry , Small Molecule Libraries/pharmacology , Structure-Activity Relationship , Surface Plasmon Resonance
7.
BMC Cancer ; 19(1): 1224, 2019 Dec 16.
Article in English | MEDLINE | ID: mdl-31842863

ABSTRACT

BACKGROUND: Mitochondria are considered a primary intracellular site of reactive oxygen species (ROS) generation. Generally, cancer cells with mitochondrial genetic abnormalities (copy number change and mutations) have escalated ROS levels compared to normal cells. Since high levels of ROS can trigger apoptosis, treating cancer cells with low doses of mitochondria-targeting / ROS-stimulating agents may offer cancer-specific therapy. This study aimed to investigate how baseline ROS levels might influence cancer cells' response to ROS-stimulating therapy. METHODS: Four cancer and one normal cell lines were treated with a conventional drug (cisplatin) and a mitochondria-targeting agent (dequalinium chloride hydrate) separately and jointly. Cell viability was assessed and drug combination synergisms were indicated by the combination index (CI). Mitochondrial DNA copy number (mtDNAcn), ROS and mitochondrial membrane potential (MMP) were measured, and the relative expression levels of the genes and proteins involved in ROS-mediated apoptosis pathways were also investigated. RESULTS: Our data showed a correlation between the baseline ROS level, mtDNAcn and drug sensitivity in the tested cells. Synergistic effect of both drugs was also observed with ROS being the key contributor in cell death. CONCLUSIONS: Our findings suggest that mitochondria-targeting therapy could be more effective compared to conventional treatments. In addition, cancer cells with low levels of ROS may be more sensitive to the treatment, while cells with high levels of ROS may be more resistant. Doubtlessly, further studies employing a wider range of cell lines and in vivo experiments are needed to validate our results. However, this study provides an insight into understanding the influence of intracellular ROS on drug sensitivity, and may lead to the development of new therapeutic strategies to improve efficacy of anticancer therapy.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cisplatin/pharmacology , Dequalinium/pharmacology , Mitochondria/metabolism , Neoplasms/drug therapy , Reactive Oxygen Species/metabolism , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Cell Survival , Cisplatin/therapeutic use , Dequalinium/therapeutic use , Female , Humans , Male , Membrane Potential, Mitochondrial , Mitochondria/drug effects , Neoplasms/metabolism , Prognosis , Treatment Outcome
8.
Int J Nanomedicine ; 13: 4209-4226, 2018.
Article in English | MEDLINE | ID: mdl-30140154

ABSTRACT

INTRODUCTION: Multidrug resistance (MDR) of breast cancer is the major challenge to successful chemotherapy while mitochondria-targeting therapy was a promising strategy to overcome MDR. MATERIALS AND METHODS: In this study, HER-2 peptide-PEG2000-Schiff base-cholesterol (HPSC) derivate was synthesized successfully and incorporated it on the surface of the doxorubicin (DOX)-loaded dequalinium (DQA) chloride vesicle (HPS-DQAsomes) to treat drug-resistant breast cancer. Evaluations were performed using human breast cancer cell and DOX-resistant breast cancer cell lines (MCF-7 and MCF-7/ADR). RESULTS: The particle size of HPS-DQAsomes was ~110 nm with spherical shape. In vitro cytotoxicity assay indicated that HPS-DQAsomes could increase the cytotoxicity against MCF-7/ADR cell line. Cellular uptake and mitochondria-targeting assay demonstrated that HPS-DQAsomes could target delivering therapeutical agent to mitochondria and inducing mitochondria-driven apoptosis process. In vivo antitumor assay suggested that HPS-DQAsomes could reach favorable antitumor activity due to both tumor targetability and sub-organelles' targetability. Histological assay also indicated that HPS-DQAsomes showed a strong apoptosis-inducing effect. No obvious systematic toxicity of HPS-DQAsomes could be observed. CONCLUSION: In summary, multifunctional HPS-DQAsomes provide a novel and versatile approach for overcoming MDR via mitochondrial pathway in cancer treatment.


Subject(s)
Antineoplastic Agents/pharmacology , Dequalinium/pharmacology , Doxorubicin/pharmacology , Drug Delivery Systems , Mitochondria/metabolism , Peptides/pharmacology , Receptor, ErbB-2/metabolism , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Cholesterol/chemistry , Cytochromes c/metabolism , Dequalinium/therapeutic use , Doxorubicin/therapeutic use , Drug Liberation , Drug Resistance, Neoplasm/drug effects , Endocytosis/drug effects , Female , Humans , Hydrogen-Ion Concentration , Liposomes , MCF-7 Cells , Membrane Potential, Mitochondrial/drug effects , Mice, Nude , Mitochondria/drug effects , Particle Size , Polyethylene Glycols/chemistry , Schiff Bases/chemistry , Toxicity Tests
9.
Eur J Pharm Biopharm ; 124: 104-115, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29305141

ABSTRACT

Mitochondria are targets with great potential for therapeutics for many human disorders. However, drug delivery systems for such therapeutics remain in need of more efficient mitochondrial-targeting carriers. In this study, we report that nanosomes composed of Dequalinium/DOTAP (1,2-dioleoyl-3-trimethylammonium-propane)/DOPE (1,2-dioleoyl-sn-glycero-3-phosphoethanolamine), called DQA80s, can act in the dual role of mitochondrial-targeting carrier and anticancer agent for therapeutic interventions against mitochondrial diseases. In cytotoxicity assays, DQA80s were shown to be more toxic than DQAsomes. The DQA80s showed significantly increased cellular uptake as compared to that of DQAsomes, and DQA80s also showed more efficient escape from the endolysosome to the cytosol. We observed the efficient targeting of DQA80s to mitochondria in living cells using flow cytometry, confocal microscopy, and TEM imaging. We also found evidence of anticancer potential that mitochondrial-targeted DQA80s induced apoptosis by production of reactive oxygen species (ROS) via MAPK signaling pathways, loss of mitochondrial membrane potential, and the caspase-3 activation. The present study demonstrates that DQA80s have excellent dual potential both as a carrier and as an anticancer therapeutic for mitochondria-related disease therapy in vivo.


Subject(s)
Antineoplastic Agents/pharmacology , Dequalinium/pharmacology , Drug Carriers , Mitochondria/drug effects , Nanoparticles , Neoplasms/drug therapy , Antineoplastic Agents/chemistry , Antineoplastic Agents/metabolism , Apoptosis/drug effects , Caspase 3/metabolism , Cell Cycle/drug effects , Dequalinium/chemistry , Dequalinium/metabolism , Dose-Response Relationship, Drug , Drug Compounding , Fatty Acids, Monounsaturated/chemistry , Flow Cytometry , HeLa Cells , Humans , Membrane Potential, Mitochondrial/drug effects , Microscopy, Confocal , Microscopy, Electron, Transmission , Mitochondria/metabolism , Mitochondria/pathology , Nanomedicine/methods , Neoplasms/metabolism , Neoplasms/pathology , Phosphatidylethanolamines/chemistry , Quaternary Ammonium Compounds/chemistry , Reactive Oxygen Species/metabolism , Technology, Pharmaceutical/methods
10.
Arch Gynecol Obstet ; 293(3): 469-84, 2016 Mar.
Article in English | MEDLINE | ID: mdl-26506926

ABSTRACT

BACKGROUND: Vaginal infections are responsible for a large proportion of gynaecological outpatient visits. Those are bacterial vaginosis (BV), vulvovaginal candidosis (VVC), aerobic vaginitis (AV) associated with aerobic bacteria, and mixed infections. Usual treatments show similar acceptable short-term efficacy, but frequent recurrences and increasing microbial resistance are unsolved issues. Furthermore, vaginal infections are associated with a variety of serious adverse outcomes in pregnancy and generally have a major impact on quality of life. Identifying the correct therapy can be challenging for the clinician, particularly in mixed infections. FINDINGS: Dequalinium chloride (DQC) is an anti-microbial antiseptic agent with a broad bactericidal and fungicidal activity. Systemic absorption after vaginal application of DQC is very low and systemic effects negligible. Vaginal DQC (Fluomizin vaginal tablets) has been shown to have equal clinical efficacy as clindamycin in the treatment of BV. Its broad antimicrobial activity makes it appropriate for the treatment of mixed vaginal infections and in case of uncertain diagnosis. Moreover, resistance of pathogens is unlikely due to its multiple mode of action, and vaginal DQC provides also a reduced risk for post-treatment vaginal infections. CONCLUSIONS: Vaginal DQC (10 mg) as 6-day therapy offers a safe and effective option for empiric therapy of different vaginal infections in daily practice. This review summarizes the available and relevant pharmacological and clinical data for the therapy of vaginal infections with vaginal DQC and provides the rationale for its use in daily gynaecologic practice.


Subject(s)
Anti-Bacterial Agents/therapeutic use , Anti-Infective Agents, Local/pharmacology , Bacteria/drug effects , Dequalinium/pharmacology , Vaginal Creams, Foams, and Jellies/therapeutic use , Vaginal Diseases/microbiology , Anti-Infective Agents, Local/therapeutic use , Antifungal Agents/pharmacology , Bacteria, Aerobic , Candidiasis, Vulvovaginal/diagnosis , Female , Humans , Pregnancy , Quality of Life , Vaginal Diseases/drug therapy , Vaginosis, Bacterial/microbiology
12.
Ann Hematol ; 94(3): 379-92, 2015 Mar.
Article in English | MEDLINE | ID: mdl-25322811

ABSTRACT

Current frontline therapies have improved overall survival in acute promyelocytic leukemia (APL) patients to exceptional rates; however, relapse is still a problem among high-risk and old patients. Therefore, the development of better and safer therapies continues to be a goal in the treatment of this disease. In the present work, we examined three different pathways that hinder cell death in the APL cell line NB4, shedding light on the mechanisms that underlie resistance to apoptosis in these cells and that might help provide them with a proliferative advantage. We found that the proteasome inhibitor MG-132 specifically induces in NB4 cells an Nrf2-mediated antioxidant response which counteracts mitochondria-dependent apoptosis induced by the lipophilic cation dequalinium. More importantly, we also demonstrated that high basal autophagy levels and the gain-of-function of mutant p53 are intrinsic mechanisms of resistance to apoptosis in this cell line. According to our results, the pharmacological inhibition of autophagy and p53 mutants are useful tools to explore resistance to apoptosis in APL and other types of cancer and could be the bases of new therapeutic approaches that improve the efficiency and allow dose reduction of the current treatments.


Subject(s)
Apoptosis/genetics , Drug Resistance, Neoplasm/genetics , Leukemia, Promyelocytic, Acute/genetics , Leukemia, Promyelocytic, Acute/pathology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Antioxidants/metabolism , Apoptosis/drug effects , Cell Line, Tumor , Cell Nucleus/drug effects , Cell Nucleus/genetics , Cell Nucleus/metabolism , Dequalinium/administration & dosage , Dequalinium/pharmacology , Dose-Response Relationship, Drug , Gene Expression Regulation, Leukemic/drug effects , HL-60 Cells , Humans , Leukemia, Promyelocytic, Acute/drug therapy , Leupeptins/administration & dosage , Leupeptins/pharmacology , Protein Transport/drug effects , Tumor Suppressor Protein p53/metabolism
13.
Proc Natl Acad Sci U S A ; 111(40): 14406-11, 2014 Oct 07.
Article in English | MEDLINE | ID: mdl-25237136

ABSTRACT

Primary hyperoxaluria 1 (PH1; Online Mendelian Inheritance in Man no. 259900), a typically lethal biochemical disorder, may be caused by the AGT(P11LG170R) allele in which the alanine:glyoxylate aminotransferase (AGT) enzyme is mistargeted from peroxisomes to mitochondria. AGT contains a C-terminal peroxisomal targeting sequence, but mutations generate an N-terminal mitochondrial targeting sequence that directs AGT from peroxisomes to mitochondria. Although AGT(P11LG170R) is functional, the enzyme must be in the peroxisome to detoxify glyoxylate by conversion to alanine; in disease, amassed glyoxylate in the peroxisome is transported to the cytosol and converted to oxalate by lactate dehydrogenase, leading to kidney failure. From a chemical genetic screen, we have identified small molecules that inhibit mitochondrial protein import. We tested whether one promising candidate, Food and Drug Administration (FDA)-approved dequalinium chloride (DECA), could restore proper peroxisomal trafficking of AGT(P11LG170R). Indeed, treatment with DECA inhibited AGT(P11LG170R) translocation into mitochondria and subsequently restored trafficking to peroxisomes. Previous studies have suggested that a mitochondrial uncoupler might work in a similar manner. Although the uncoupler carbonyl cyanide m-chlorophenyl hydrazone inhibited AGT(P11LG170R) import into mitochondria, AGT(P11LG170R) aggregated in the cytosol, and cells subsequently died. In a cellular model system that recapitulated oxalate accumulation, exposure to DECA reduced oxalate accumulation, similar to pyridoxine treatment that works in a small subset of PH1 patients. Moreover, treatment with both DECA and pyridoxine was additive in reducing oxalate levels. Thus, repurposing the FDA-approved DECA may be a pharmacologic strategy to treat PH1 patients with mutations in AGT because an additional 75 missense mutations in AGT may also result in mistrafficking.


Subject(s)
Dequalinium/pharmacology , Hyperoxaluria, Primary/metabolism , Transaminases/metabolism , Animals , Anti-Infective Agents, Local/pharmacology , CHO Cells , Cricetinae , Cricetulus , Drug Evaluation, Preclinical/methods , Embryo, Nonmammalian/drug effects , Embryo, Nonmammalian/embryology , Humans , Hyperoxaluria, Primary/genetics , Hyperoxaluria, Primary/prevention & control , Immunoblotting , Microscopy, Fluorescence , Mitochondria/metabolism , Mutation , Oxalates/metabolism , Peroxisomes/metabolism , Protein Transport/drug effects , Protein Transport/genetics , Pyridoxine/pharmacology , Transaminases/genetics , Zebrafish/embryology
14.
Leuk Res ; 38(7): 795-803, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24811390

ABSTRACT

Delocalized lipophilic cation dequalinium (DQA) selectively accumulates in mitochondria and displays anticancer activity in different malignancies. Our previous studies indicate a DQA-induced cytotoxicity in human acute promyelocytic leukemia NB4 cells by early disturbance in mitochondrial function and oxidative stress. This study shows the ability of DQA to downregulate Raf/MEK/ERK1/2 and PI3K/Akt signaling pathways in NB4 cells which leads to cell death by apoptosis and/or necrosis. Moreover, DQA potentiates the action of specific inhibitors of these pathways. These DQA effects could be mediated by redox regulation of Akt. Our results contribute to a better understanding of the cytotoxic DQA mechanism on leukemia cells and encourage the performance of further studies in combination with other agents such as kinase inhibitors for improving the efficacy of therapies against acute promyelocytic leukemia.


Subject(s)
Dequalinium/pharmacology , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Leukemia/drug therapy , Mitogen-Activated Protein Kinase Kinases/antagonists & inhibitors , Phosphoinositide-3 Kinase Inhibitors , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , Signal Transduction/drug effects , raf Kinases/antagonists & inhibitors , Apoptosis/drug effects , Down-Regulation , Extracellular Signal-Regulated MAP Kinases/physiology , Glutathione/metabolism , Humans , Leukemia/pathology , Mitogen-Activated Protein Kinase Kinases/physiology , Phosphatidylinositol 3-Kinases/physiology , Proto-Oncogene Proteins c-akt/physiology , raf Kinases/physiology
15.
Biomaterials ; 35(21): 5591-604, 2014 Jul.
Article in English | MEDLINE | ID: mdl-24726749

ABSTRACT

Invasive brain glioma is the most lethal type of cancer and is highly infiltrating. This leads to an extremely poor prognosis and makes complete surgical removal of the tumor virtually impossible. Non-penetration of therapeutic drugs across the blood-brain barrier (BBB), brain cancer stem cells (CSCs), and brain cancer vasculogenic mimicry (VM) results in relapse after surgical and radio therapy. We developed a functional targeting chemotherapy for transporting drugs across the BBB, destroying VM channels, and eliminating CSCs and cancer cells in the brain. The studies were undertaken on brain glioma cells in vitro and in brain glioma-bearing rats. Using paclitaxel as the anticancer drug and artemether as the regulator of apoptosis and inhibitor of VM channels, a kind of functional targeting paclitaxel plus artemether liposomes was developed by modifying two new functional materials: a mannose-vitamin E derivative conjugate (MAN-TPGS1000) and a dequalinium-lipid derivative conjugate (DQA-PEG2000-DSPE). The transport mechanism across the BBB was associated with receptor-mediated endocytosis by MAN-TPGS1000 conjugate via glucose transporters and adsorptive-mediated endocytosis by DQA-PEG2000-DSPE conjugate via electric charge-based interactions. The efficacy was related to the destruction of VM channels by regulating VM indicators, as well as the induction of apoptosis in brain cancer cells and CSCs by activating apoptotic enzymes and pro-apoptotic proteins and inhibiting anti-apoptotic proteins. These data suggest that the chemotherapy using functional targeting paclitaxel plus artemether liposomes could provide a new strategy for treating invasive brain glioma.


Subject(s)
Artemisinins/pharmacology , Brain Neoplasms/drug therapy , Glioma/drug therapy , Liposomes/pharmacology , Paclitaxel/pharmacology , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Artemether , Blood-Brain Barrier/drug effects , Blood-Brain Barrier/metabolism , Brain/drug effects , Brain/metabolism , Brain/pathology , Cell Line, Tumor , Dequalinium/pharmacology , Drug Delivery Systems/methods , Male , Mannose/pharmacology , Mice , Mice, Inbred ICR , Phosphatidylethanolamines/pharmacology , Polyethylene Glycols/pharmacology , Rats , Rats, Sprague-Dawley , Vitamin E/pharmacology
16.
Mol Biol (Mosk) ; 48(3): 416-28, 2014.
Article in Russian | MEDLINE | ID: mdl-25831891

ABSTRACT

Mitochondria play central roles in diverse physiological and pathological conditions associated with cell survival and death. Delocalized lipophilic cations, such as dequalinium (DQA), are accumulated in cancer cells attracted by the highly negative mitochondrial transmembrane potential of these cells. DQA showed a potent anticancer activity in cells from different malignancies. Here, we report the effect of DQA on PC-3 prostate cancer cells. Incubation with DQA at concentrations between 1.5 and 100 microM from 24 to 48 h decreases cell viability. The decrease in cell viability together with a loss of mitochondrial transmembrane potential induced an increase in reactive oxygen species production and cell death via caspase-3 dependent apoptotic pathway. QA was shown to cause moderate to strong cell death in a time and concentration dependent manner, causing a most advantageous effect at a concentration of 10 microM applied for a long 48 h time period, which might be a consequence of the kinetics of intracellular DQA accumulation in mitochondria, but also of the mechanisms of DQA-induced cell death. This data shows DQA as a promising agent against the human prostate cancer PC-3 cell line, activating the caspase-3 dependent apoptotic pathway. This fact might be beneficial for possible future applications in cancer therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Dequalinium/pharmacology , Membrane Potential, Mitochondrial , Mitochondria/metabolism , Oxidative Stress , Prostatic Neoplasms/metabolism , Apoptosis , Caspase 3/metabolism , Cell Line, Tumor , Cell Survival , Dose-Response Relationship, Drug , Humans , Male , Mitochondria/drug effects , Reactive Oxygen Species/metabolism
17.
J Steroid Biochem Mol Biol ; 138: 267-72, 2013 Nov.
Article in English | MEDLINE | ID: mdl-23831356

ABSTRACT

High doses of anabolic androgenic steroids (AAS) impair the cardioprotective effects of exercise against ischemia/reperfusion (I/R) insult, possibly through cellular redox imbalance. Here, the effect of nandrolone decanoate (DECA) treatment on heart redox metabolism was investigated during I/R in sedentary and exercised rats. DECA treatment significantly reduced superoxide dismutase and glutathione reductase activities in exercised rats after heart reperfusion. Catalase and glutathione peroxidase activities were not affected by DECA in both sedentary and trained rats, regardless the I/R period. DECA also induced myocardial oxidative stress, as evidenced by the reduced levels of total reduced thiols after heart reperfusion in exercised rats treated with the anabolic steroid. These results indicate that cardiotoxic effects of supraphysiological doses of AAS involve reduced heart antioxidant capacity.


Subject(s)
Anabolic Agents/adverse effects , Heart/drug effects , Physical Conditioning, Animal/physiology , Animals , Dequalinium/analogs & derivatives , Dequalinium/pharmacology , Glutathione Reductase/metabolism , Heart/physiology , Male , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Rats , Rats, Wistar , Superoxide Dismutase/metabolism
18.
BMC Res Notes ; 5: 151, 2012 Mar 19.
Article in English | MEDLINE | ID: mdl-22429611

ABSTRACT

BACKGROUND: Atopobium vaginae and Gardnerella vaginalis are major markers for bacterial vaginosis. We aimed to determine the MIC and MBC range of the broad-spectrum anti-infective and antiseptic dequalinium chloride for 28 strains, belonging to 4 species of the genus Atopobium, i.e. A. vaginae, A. minutum, A. rimae and A. parvulum. METHODS: The MIC was determined with a broth microdilution assay. RESULTS: The MIC and MBC for Atopobium spp. for dequalinium chloride ranged between < 0.0625 and 2 µg/ml. CONCLUSIONS: This study demonstrated that dequalinium chloride inhibits and kills clinical isolates of A. vaginae at concentrations similar to those of clindamycin and lower than those of metronidazole.


Subject(s)
Actinobacteria/drug effects , Anti-Bacterial Agents/pharmacology , Bacteroides fragilis/drug effects , Dequalinium/pharmacology , Actinobacteria/growth & development , Actinobacteria/isolation & purification , Bacteroides fragilis/growth & development , Bacteroides fragilis/isolation & purification , Clindamycin/pharmacology , Culture Media , Female , Humans , Metronidazole/pharmacology , Microbial Sensitivity Tests , Vagina/microbiology , Vaginosis, Bacterial/microbiology
19.
J Pharmacol Sci ; 118(2): 156-60, 2012.
Article in English | MEDLINE | ID: mdl-22343363

ABSTRACT

Although the antinociceptive effects of N-palmitoyl-ethanolamine (PEA) were first characterized nearly 50 years ago, the identity of the mechanism that mediates these actions has not been elucidated. The present study investigated the contribution of K(+) channels on peripheral antinociception induced by the CB(2) agonist PEA. Nociceptive thresholds to mechanical paw stimulation of Wistar rats treated with intraplantar prostaglandin E(2) to induce hyperalgesia were measured, and other agents were also given by local injection. PEA (5, 10, and 20 µg/paw) elicited a local peripheral antinociceptive effect. This effect was antagonized by glibenclamide, a selective blocker of ATP-sensitive K(+) channels (20, 40, and 80 µg/paw). In addition, neither the voltage-dependent K(+) channel-specific blocker tetraethylammonium (30 µg/paw) nor the small and large conductance blockers of Ca(2+)-activated K(+) channels, dequalinium (50 µg/paw) and paxilline (20 µg/paw), respectively, were able to block the local antinociceptive effect of PEA. These results indicate that the activation of ATP-sensitive K(+) channels could be the mechanism that induces peripheral antinociception by PEA and that voltage-dependent K(+) channels and small and large conductance Ca(2+)-activated K(+) channels do not appear to be involved in this mechanism.


Subject(s)
Analgesics/pharmacology , Hyperalgesia/drug therapy , KATP Channels/drug effects , Palmitic Acids/pharmacology , Amides , Analgesics/administration & dosage , Animals , Dequalinium/pharmacology , Dinoprostone/pharmacology , Disease Models, Animal , Dose-Response Relationship, Drug , Endocannabinoids , Ethanolamines , Glyburide/administration & dosage , Glyburide/pharmacology , Hyperalgesia/physiopathology , Indoles/pharmacology , KATP Channels/metabolism , Male , Palmitic Acids/administration & dosage , Rats , Rats, Wistar
20.
Leuk Res ; 35(10): 1395-401, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21477862

ABSTRACT

Dequalinium, an amphiphilic quinolinium derivative, selectively accumulates in mitochondria and displays anticancer activity in cells from different malignancies. Previous studies indicate a differential DQA-induced cytotoxicity in NB4 and K562 human leukemia cells as a consequence of an early disturbance in mitochondrial function. Results in this paper show that DQA induces a concentration-dependent oxidative stress by decreasing GSH level and increasing ROS in a cell type specific way. Inhibitors of the JNK and p38 stress regulated kinases potentiate DQA-induced NB4 cell death suggesting a protective function for these enzymes. K562 cells with relatively high GSH levels remained resistant to DQA action.


Subject(s)
Apoptosis/drug effects , Dequalinium , Leukemia/drug therapy , Mitochondria/metabolism , Blotting, Western , Cell Line, Tumor , Dequalinium/pharmacology , Drug Synergism , Glutathione/analysis , Glutathione/biosynthesis , Humans , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , JNK Mitogen-Activated Protein Kinases/metabolism , Leukemia/enzymology , Leukemia/pathology , Mitochondria/drug effects , Organ Specificity , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Protein Kinase Inhibitors/pharmacology , Reactive Oxygen Species/metabolism , Superoxides/analysis , Surface-Active Agents/pharmacology , p38 Mitogen-Activated Protein Kinases/antagonists & inhibitors , p38 Mitogen-Activated Protein Kinases/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...