Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 10 de 10
Filter
Add more filters










Publication year range
1.
Biochem Pharmacol ; 186: 114467, 2021 04.
Article in English | MEDLINE | ID: mdl-33577890

ABSTRACT

For more than 60 years dequalinium chloride (DQ) has been used as anti-infective drug, mainly to treat local infections. It is a standard drug to treat bacterial vaginosis and an active ingredient of sore-throat lozenges. As a lipophilic bis-quaternary ammonium molecule, the drug displays membrane effects and selectively targets mitochondria to deplete DNA and to block energy production in cells. But beyond its mitochondriotropic property, DQ can interfere with the correct functioning of diverse proteins. A dozen of DQ protein targets have been identified and their implication in the antibacterial, antiviral, antifungal, antiparasitic and anticancer properties of the drug is discussed here. The anticancer effects of DQ combine a mitochondrial action, a selective inhibition of kinases (PKC-α/ß, Cdc7/Dbf4), and a modulation of Ca2+-activated K+ channels. At the bacterial level, DQ interacts with different multidrug transporters (QacR, AcrB, EmrE) and with the transcriptional regulator RamR. Other proteins implicated in the antiviral (MPER domain of gp41 HIV-1) and antiparasitic (chitinase A from Vibrio harveyi) activities have been identified. DQ also targets α -synuclein oligomers to restrict protofibrils formation implicated in some neurodegenerative disorders. In addition, DQ is a typical bolaamphiphile molecule, well suited to form liposomes and nanoparticules useful for drug entrapment and delivery (DQAsomes and others). Altogether, the review highlights the many pharmacological properties and therapeutic benefits of this old 'multi-talented' drug, which may be exploited further. Its multiple sites of actions in cells should be kept in mind when using DQ in experimental research.


Subject(s)
Anti-Infective Agents, Local/administration & dosage , Cell Cycle Proteins/antagonists & inhibitors , Dequalinium/administration & dosage , Drug Delivery Systems/methods , Mitochondria/drug effects , Animals , Anti-Infective Agents, Local/metabolism , Cell Cycle Proteins/chemistry , Cell Cycle Proteins/metabolism , Dequalinium/metabolism , Humans , Mitochondria/metabolism , Mycoses/drug therapy , Mycoses/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Protein Structure, Secondary , Streptococcal Infections/drug therapy , Streptococcal Infections/metabolism , Vibrio Infections/drug therapy , Vibrio Infections/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.
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
4.
Pharm Res ; 28(11): 2848-62, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21833794

ABSTRACT

PURPOSE: We describe a novel strategy for expression of GFP in mammalian mitochondria. METHODS: The key components of the strategy were an artificially created mitochondrial genome pmtGFP and a DQAsome transfection system. RESULTS: Using immunofluorescence and a combination of immunohistochemical and molecular based techniques, we show that DQAsomes are capable of delivering the pmtGFP construct to the mitochondrial compartment of the mouse macrophage cell line RAW264.7, albeit at low efficiency (1-5%), resulting in the expression of GFP mRNA and protein. Similar transfection efficiencies were also demonstrated in a range of other mammalian cell lines. CONCLUSIONS: The DQAsome-transfection technique was able to deliver the exogenous DNA into the cellular mitochondria and the pmtGFP was functional. Further optimization of this strategy would provide a flexible and rapid way to generate mutant cells and useful animal models of mitochondrial disease.


Subject(s)
Anti-Infective Agents, Local/chemistry , Dequalinium/chemistry , Drug Delivery Systems , Genetic Therapy/methods , Green Fluorescent Proteins/metabolism , Mitochondria/metabolism , Oligonucleotides/chemistry , Animals , Anti-Infective Agents, Local/metabolism , Cell Line , Cell Proliferation/drug effects , DNA, Mitochondrial/genetics , DNA, Mitochondrial/metabolism , Dequalinium/metabolism , Drug Compounding , Genome, Mitochondrial , Green Fluorescent Proteins/chemistry , Green Fluorescent Proteins/genetics , Humans , Liposomes/chemistry , Liposomes/metabolism , Macrophages , Mammals , Mice , Mitochondria/chemistry , Mitochondria/genetics , Oligonucleotides/metabolism , Plasmids/chemistry , Plasmids/metabolism , Transfection
5.
J Mol Biol ; 377(4): 1094-103, 2008 Apr 04.
Article in English | MEDLINE | ID: mdl-18295794

ABSTRACT

EmrE is a Small Multidrug Resistance transporter (SMR) family member that mediates counter transport of protons and hydrophobic cationic drugs such as tetraphenylphosphonium (TPP+), ethidium, propidium and dequalinium. It is thought that the selectivity of the drug binding site in EmrE is defined by two negatively charged glutamate residues within a hydrophobic pocket formed from six of the alpha-helices, three from each monomer of the asymmetric EmrE homodimer. It is not apparent how such a binding pocket accommodates drugs of various sizes and shapes or whether the conformational changes that occur upon drug binding are identical for drugs of diverse chemical nature. Here, using electron cryomicroscopy of EmrE two-dimensional crystals we have determined projection structures of EmrE bound to three structurally different planar drugs, ethidium, propidium and dequalinium. Using image analysis and rigorous comparisons between these density maps and the density maps of the ligand-free and TPP+-bound forms of EmrE, we identify regions within the transporter that adapt differentially depending on the type of ligand bound. We show that all three planar drugs bind at the same pocket within the protein as TPP+. Furthermore, our analysis indicates that, while retaining the overall fold of the protein, binding of the planar drugs is accompanied by small rearrangements of the transmembrane domains that are different to those that occur when TPP+ binds. The regions in the EmrE dimer that are remodelled surround the drug binding site and include transmembrane domains from both monomers.


Subject(s)
Antiporters/chemistry , Binding Sites , Escherichia coli Proteins/chemistry , Anti-Infective Agents, Local/chemistry , Anti-Infective Agents, Local/metabolism , Antiporters/metabolism , Antiviral Agents/chemistry , Antiviral Agents/metabolism , Carrier Proteins/chemistry , Carrier Proteins/metabolism , Cryoelectron Microscopy , Crystallography , Dequalinium/chemistry , Dequalinium/metabolism , Dimerization , Escherichia coli Proteins/metabolism , Ethidium/chemistry , Ethidium/metabolism , Intercalating Agents/chemistry , Intercalating Agents/metabolism , Models, Biological , Propidium/chemistry , Propidium/metabolism , Protein Structure, Tertiary , Structure-Activity Relationship
6.
Science ; 300(5621): 976-80, 2003 May 09.
Article in English | MEDLINE | ID: mdl-12738864

ABSTRACT

Multidrug efflux pumps cause serious problems in cancer chemotherapy and treatment of bacterial infections. Yet high-resolution structures of ligand transporter complexes have previously been unavailable. We obtained x-ray crystallographic structures of the trimeric AcrB pump from Escherichia coli with four structurally diverse ligands. The structures show that three molecules of ligands bind simultaneously to the extremely large central cavity of 5000 cubic angstroms, primarily by hydrophobic, aromatic stacking and van der Waals interactions. Each ligand uses a slightly different subset of AcrB residues for binding. The bound ligand molecules often interact with each other, stabilizing the binding.


Subject(s)
Carrier Proteins/chemistry , Carrier Proteins/metabolism , Escherichia coli Proteins/chemistry , Escherichia coli Proteins/metabolism , Membrane Proteins/chemistry , Membrane Proteins/metabolism , Anti-Infective Agents/chemistry , Anti-Infective Agents/metabolism , Anti-Infective Agents, Local/chemistry , Anti-Infective Agents, Local/metabolism , Binding Sites , Carrier Proteins/isolation & purification , Cell Membrane/chemistry , Chemical Phenomena , Chemistry, Physical , Ciprofloxacin/chemistry , Ciprofloxacin/metabolism , Crystallization , Crystallography, X-Ray , Dequalinium/chemistry , Dequalinium/metabolism , Escherichia coli Proteins/isolation & purification , Ethidium/chemistry , Ethidium/metabolism , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Ligands , Membrane Proteins/isolation & purification , Models, Molecular , Multidrug Resistance-Associated Proteins , Protein Binding , Protein Conformation , Protein Structure, Quaternary , Protein Structure, Secondary , Protein Structure, Tertiary , Rhodamines/chemistry , Rhodamines/metabolism , Static Electricity
7.
Science ; 294(5549): 2158-63, 2001 Dec 07.
Article in English | MEDLINE | ID: mdl-11739955

ABSTRACT

The Staphylococcus aureus multidrug binding protein QacR represses transcription of the qacA multidrug transporter gene and is induced by structurally diverse cationic lipophilic drugs. Here, we report the crystal structures of six QacR-drug complexes. Compared to the DNA bound structure, drug binding elicits a coil-to-helix transition that causes induction and creates an expansive multidrug-binding pocket, containing four glutamates and multiple aromatic and polar residues. These structures indicate the presence of separate but linked drug-binding sites within a single protein. This multisite drug-binding mechanism is consonant with studies on multidrug resistance transporters.


Subject(s)
Gentian Violet/metabolism , Heterocyclic Compounds/metabolism , Repressor Proteins/chemistry , Rosaniline Dyes/metabolism , Bacterial Proteins/chemistry , Bacterial Proteins/metabolism , Berberine/chemistry , Berberine/metabolism , Binding Sites , Crystallization , Crystallography, X-Ray , DNA/metabolism , Dequalinium/chemistry , Dequalinium/metabolism , Dimerization , Drug Resistance, Multiple, Bacterial , Ethidium/chemistry , Ethidium/metabolism , Gentian Violet/chemistry , Glutamates/chemistry , Heterocyclic Compounds/chemistry , Hydrogen Bonding , Hydrophobic and Hydrophilic Interactions , Models, Molecular , Molecular Structure , Protein Conformation , Protein Structure, Secondary , Protein Structure, Tertiary , Repressor Proteins/metabolism , Rhodamines/chemistry , Rhodamines/metabolism , Rosaniline Dyes/chemistry , Staphylococcus aureus
8.
J Biol Chem ; 273(4): 2390-5, 1998 Jan 23.
Article in English | MEDLINE | ID: mdl-9442087

ABSTRACT

1,1'-Decamethylenebis-4-aminoquinaldinium diiodide (DECA; dequalinium) is an anti-tumor agent and protein kinase C (PKC) inhibitor whose mechanism of action with PKC is unknown. This study reports that with human PKC alpha, DECA exhibited competitive inhibition (Ki = 11.5 +/- 5 microM) with respect to RACK-1 (receptor for activated C kinase-1), an adaptor protein that has been proposed to bind activated PKC following translocation (Ron, D., Luo, J., and Mochly-Rosen, D. (1995) J. Biol. Chem. 270, 24180-24187). When exposed to UV light, DECA covalently modified and irreversibly inhibited PKC (alpha or beta), with IC50 = 7-18 microM. UV/DECA treatment of synthetic peptides modeled after the RACK-1-binding site in the C2 region of PKC beta induced modification of Ser218-Leu-Asn-Pro-Glu-Trp-Asn-Glu-Thr226, but not of a control peptide. This modification occurred at a tryptophan residue (Trp223) that is conserved in all conventional PKC isoforms. In overlay assays with native RACK-1 that had been immobilized on nitrocellulose, UV-treated control PKC alpha bound well to RACK-1, whereas UV/DECA-inactivated PKC alpha had reduced binding activity. The significance of these findings is shown with adenocarcinoma cells, which, when pretreated with 10 microM DECA and UV light, exhibited diminished 12-O-tetradecanoylphorbol-13-acetate-induced PKC alpha translocation. Overall, this work identifies DECA as a tool that prevents PKC translocation by inhibiting formation of the PKC.RACK-1 complex.


Subject(s)
Anti-Infective Agents, Local/pharmacology , Dequalinium/pharmacology , Protein Kinase C/antagonists & inhibitors , Receptors, Cell Surface/metabolism , Amino Acid Sequence , Anti-Infective Agents, Local/metabolism , Binding Sites , Dequalinium/metabolism , Humans , Isoenzymes/antagonists & inhibitors , Isoenzymes/metabolism , Molecular Sequence Data , Photochemistry , Protein Kinase C/metabolism , Protein Kinase C-alpha , Proton-Translocating ATPases/metabolism , Receptors for Activated C Kinase , Tetradecanoylphorbol Acetate/pharmacology , Tumor Cells, Cultured , Ultraviolet Rays
9.
Biochemistry ; 33(1): 126-33, 1994 Jan 11.
Article in English | MEDLINE | ID: mdl-8286329

ABSTRACT

The F1-ATPase from Bacillus PS3 (TF1) hydrolyzes 50 microM ATP in three kinetic phases. An initial burst rapidly decelerates to a partially inhibited, intermediate phase, which, in turn, gradually accelerates to an uninhibited, final steady-state rate. Lauryl dimethylamine oxide (LDAO) stimulates the final rate over 4-fold. The stimulatory effect saturates at about 0.1% LDAO. Under these conditions, the intermediate phase is nearly absent. Dequalinium inhibits TF1 reversibly in the dark in the presence or absence of LDAO. The apparent affinity of TF1 for dequalinium increases in the presence of LDAO. Dixon plots of the initial rates of the intermediate phase and the final rates against dequalinium concentration at a series of fixed ATP concentrations in the presence and absence of 0.03% LDAO indicate noncompetitive inhibition in each case. Replots of the slopes of the Dixon plots for the initial rate of the intermediate phase and the final rate against 1/[ATP] reveal apparent Km values of 770 microM and 144 microM, respectively, when obtained in the absence of LDAO. The apparent Km values determined from the data obtained in the presence of LDAO for the same phases are 303 microM and 163 microM, respectively. These results suggest that LDAO stimulates ATPase activity either by increasing the affinity of noncatalytic sites for ATP, which promotes release of inhibitory MgADP from a catalytic site, or by directly promoting release of MgADP from the affected catalytic site. Dequalinium retards this process without affecting the affinity of noncatalytic sites for ATP. When irradiated in the presence of dequalinium, TF1 is rapidly inactivated with an apparent Kd of 12.5 microM in the presence or absence of LDAO.(ABSTRACT TRUNCATED AT 250 WORDS)


Subject(s)
Bacillus/enzymology , Dequalinium/pharmacology , Dimethylamines/pharmacology , Proton-Translocating ATPases/metabolism , Amino Acid Sequence , Carbon Radioisotopes , Cyanogen Bromide , Dequalinium/metabolism , Detergents/pharmacology , Enzyme Activation , Kinetics , Molecular Sequence Data , Peptide Fragments/chemistry , Peptide Fragments/isolation & purification , Peptide Mapping , Proton-Translocating ATPases/antagonists & inhibitors , Proton-Translocating ATPases/chemistry , Sequence Homology, Amino Acid , Trypsin
10.
Cancer Res ; 50(4): 1165-9, 1990 Feb 15.
Article in English | MEDLINE | ID: mdl-2297765

ABSTRACT

Dequalinium belongs to a group of cationic lipophilic drugs believed to be selectively cytotoxic to malignant cells of epithelial origin by virtue of their accumulation in mitochondria. In addition, they are potent inhibitors of calmodulin and, therefore, might sensitize multidrug-resistant cells to chemotherapeutic agents. We compared the responsiveness of multidrug-resistant cells to the effect of dequalinium with that to trifluoperazine, a potent phenothiazine inhibitor of calmodulin. In addition, we studied the effect of these drugs on the responsiveness of multidrug-resistant cell lines to doxorubicin. The effect of drugs on P388 murine leukemic cells was determined by cell counting, [3H]thymidine incorporation into DNA, or soft agar cloning. Drug accumulation was measured by fluorescence spectrophotometry. We found that multidrug-resistant lines were less sensitive than parental cell lines to the intrinsic growth inhibitory effects of dequalinium (IC50, 4.4 versus 0.3 microM in multidrug-resistant and sensitive P388 cells, respectively), whereas they were equally sensitive as the parental line to the effects of trifluoperazine. Following a 3-h exposure of P388/doxorubicin-resistant cells to 0-100 microM doxorubicin with or without either 10 microM dequalinium or 10 microM trifluoperazine, the latter increased the sensitivity to doxorubicin whereas the former had little effect (IC50 values were doxorubicin, 30 microM; doxorubicin plus dequalinium, 25 microM; doxorubicin plus trifluoperazine, 4 microM). Calmodulin prepared from resistant cells were equally sensitive to inhibition by dequalinium and trifluoperazine. P388/doxorubicin-resistant cells accumulated 4.5-fold less dequalinium than P388 cells whereas trifluoperazine was accumulated equally in both. The addition of 4 microM trifluoperazine to resistant cells exposed to 0-100 microM dequalinium completely reversed the alteration in accumulation and resistance to the dequalinium. These studies demonstrate that certain multidrug-resistant lines are cross-resistant to dequalinium and that sensitivity can be completely restored by nontoxic concentrations of trifluoperazine. The resistance appears to be due to changes in drug accumulation and not to be related to an altered sensitivity of calmodulin.


Subject(s)
Dequalinium/metabolism , Leukemia P388/metabolism , Leukemia, Experimental/metabolism , Quinolinium Compounds/metabolism , Trifluoperazine/metabolism , Animals , Calmodulin/antagonists & inhibitors , DNA, Neoplasm/drug effects , DNA, Neoplasm/metabolism , Dequalinium/pharmacology , Doxorubicin/metabolism , Doxorubicin/pharmacology , Drug Resistance , Phenotype , Trifluoperazine/pharmacology , Tumor Cells, Cultured/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...