1
|
Lindblom JR, Zhang X, Lehane AM. A pH Fingerprint Assay to Identify Inhibitors of Multiple Validated and Potential Antimalarial Drug Targets. ACS Infect Dis 2024; 10:1185-1200. [PMID: 38499199 PMCID: PMC11019546 DOI: 10.1021/acsinfecdis.3c00588] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2023] [Revised: 01/22/2024] [Accepted: 02/09/2024] [Indexed: 03/20/2024]
Abstract
New drugs with novel modes of action are needed to safeguard malaria treatment. In recent years, millions of compounds have been tested for their ability to inhibit the growth of asexual blood-stage Plasmodium falciparum parasites, resulting in the identification of thousands of compounds with antiplasmodial activity. Determining the mechanisms of action of antiplasmodial compounds informs their further development, but remains challenging. A relatively high proportion of compounds identified as killing asexual blood-stage parasites show evidence of targeting the parasite's plasma membrane Na+-extruding, H+-importing pump, PfATP4. Inhibitors of PfATP4 give rise to characteristic changes in the parasite's internal [Na+] and pH. Here, we designed a "pH fingerprint" assay that robustly identifies PfATP4 inhibitors while simultaneously allowing the detection of (and discrimination between) inhibitors of the lactate:H+ transporter PfFNT, which is a validated antimalarial drug target, and the V-type H+ ATPase, which was suggested as a possible target of the clinical candidate ZY19489. In our pH fingerprint assays and subsequent secondary assays, ZY19489 did not show evidence for the inhibition of pH regulation by the V-type H+ ATPase, suggesting that it has a different mode of action in the parasite. The pH fingerprint assay also has the potential to identify protonophores, inhibitors of the acid-loading Cl- transporter(s) (for which the molecular identity(ies) remain elusive), and compounds that act through inhibition of either the glucose transporter PfHT or glycolysis. The pH fingerprint assay therefore provides an efficient starting point to match a proportion of antiplasmodial compounds with their mechanisms of action.
Collapse
Affiliation(s)
| | | | - Adele M. Lehane
- Research School of Biology, Australian National University, Canberra, Australian Capital
Territory 2600, Australia
| |
Collapse
|
2
|
Paulikat M, Piccini G, Ippoliti E, Rossetti G, Arnesano F, Carloni P. Physical Chemistry of Chloroquine Permeation through the Cell Membrane with Atomistic Detail. J Chem Inf Model 2023; 63:7124-7132. [PMID: 37947485 PMCID: PMC10685453 DOI: 10.1021/acs.jcim.3c01363] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 10/11/2023] [Accepted: 10/12/2023] [Indexed: 11/12/2023]
Abstract
We provide a molecular-level description of the thermodynamics and mechanistic aspects of drug permeation through the cell membrane. As a case study, we considered the antimalaria FDA approved drug chloroquine. Molecular dynamics simulations of the molecule (in its neutral and protonated form) were performed in the presence of different lipid bilayers, with the aim of uncovering key aspects of the permeation process, a fundamental step for the drug's action. Free energy values obtained by well-tempered metadynamics simulations suggest that the neutral form is the only permeating protomer, consistent with experimental data. H-bond interactions of the drug with water molecules and membrane headgroups play a crucial role for permeation. The presence of the transmembrane potential, investigated here for the first time in a drug permeation study, does not qualitatively affect these conclusions.
Collapse
Affiliation(s)
- Mirko Paulikat
- Computational
Biomedicine, Institute of Advanced Simulations IAS-5/Institute for
Neuroscience and Medicine INM-9, Forschungszentrum
Jülich GmbH, 52428 Jülich, Germany
| | - GiovanniMaria Piccini
- Institute
of Technical and Macromolecular Chemistry, RWTH Aachen University, 52074 Aachen, Germany
| | - Emiliano Ippoliti
- Computational
Biomedicine, Institute of Advanced Simulations IAS-5/Institute for
Neuroscience and Medicine INM-9, Forschungszentrum
Jülich GmbH, 52428 Jülich, Germany
| | - Giulia Rossetti
- Computational
Biomedicine, Institute of Advanced Simulations IAS-5/Institute for
Neuroscience and Medicine INM-9, Forschungszentrum
Jülich GmbH, 52428 Jülich, Germany
- Jülich
Supercomputing Centre (JSC), Forschungszentrum
Jülich GmbH, 52428 Jülich, Germany
- Department
of Neurology, RWTH Aachen University, Aachen 52062, Germany
| | - Fabio Arnesano
- Department
of Chemistry, University of Bari “Aldo
Moro”, Bari 70125, Italy
| | - Paolo Carloni
- Computational
Biomedicine, Institute of Advanced Simulations IAS-5/Institute for
Neuroscience and Medicine INM-9, Forschungszentrum
Jülich GmbH, 52428 Jülich, Germany
- Department
of Physics, RWTH Aachen University, Aachen 52062, Germany
| |
Collapse
|
3
|
Domes R, Frosch T. Molecular Interactions Identified by Two-Dimensional Analysis-Detailed Insight into the Molecular Interactions of the Antimalarial Artesunate with the Target Structure β-Hematin by Means of 2D Raman Correlation Spectroscopy. Anal Chem 2023; 95:12719-12731. [PMID: 37586701 PMCID: PMC10469332 DOI: 10.1021/acs.analchem.3c01415] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Accepted: 07/11/2023] [Indexed: 08/18/2023]
Abstract
A thorough understanding of the interaction of endoperoxide antimalarial agents with their biological target structures is of utmost importance for the tailored design of future efficient antimalarials. Detailed insights into molecular interactions between artesunate and β-hematin were derived with a combination of resonance Raman spectroscopy, two-dimensional correlation analysis, and density functional theory calculations. Resonance Raman spectroscopy with three distinct laser wavelengths enabled the specific excitation of different chromophore parts of β-hematin. The resonance Raman spectra of the artesunate-β-hematin complexes were thoroughly analyzed with the help of high-resolution and highly sensitive two-dimensional correlation spectroscopy. Spectral changes in the peak properties were found with increasing artesunate concentration. Changes in the low-frequency, morphology-sensitive Raman bands indicated a loss in crystallinity of the drug-target complexes. Differences in the high-wavenumber region were assigned to increased distortions of the planarity of the structure of the target molecule due to the appearance of various coexisting alkylation species. Evidence for the appearance of high-valent ferryl-oxo species could be observed with the help of differences in the peak properties of oxidation-state sensitive Raman modes. To support those findings, the relaxed ground-state structures of ten possible covalent mono- and di-meso(Cm)-alkylated hematin-dihydroartemisinyl complexes were calculated using density functional theory. A very good agreement with the experimental peak properties was achieved, and the out-of-plane displacements along the lowest-frequency normal coordinates were investigated by normal coordinate structural decomposition analysis. The strongest changes in all data were observed in vibrations with a high participation of Cm-parts of β-hematin.
Collapse
Affiliation(s)
- Robert Domes
- Leibniz
Institute of Photonic Technology, Albert Einstein Strasse 9, D-07745 Jena, Germany
| | - Torsten Frosch
- Biophotonics and
Biomedical Engineering Group, Technical
University Darmstadt, Merckstraße 25, 64283 Darmstadt, Germany
- Leibniz
Institute of Photonic Technology, Albert Einstein Strasse 9, D-07745 Jena, Germany
| |
Collapse
|
4
|
Kumar S, Kapkoti DS, Mina PR, Gupta M, Kumar R, Kumar P, Pathak P, Bhakuni RS, Rout P, Pal A, Darokar MP. Effect of liquiritigenin on chloroquine accumulation in digestive vacuole leading to apoptosis-like death of chloroquine-resistant P. falciparum. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2023; 114:154738. [PMID: 36940579 DOI: 10.1016/j.phymed.2023.154738] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/30/2022] [Revised: 02/23/2023] [Accepted: 02/28/2023] [Indexed: 06/18/2023]
Abstract
BACKGROUND Malaria remains one of the major health concerns, especially in tropical countries. Although drugs such as artemisinin-based combinations are efficient for treating Plasmodium falciparum, the growing threat from multi-drug resistance has become a major challenge. Thus, there is a constant need to identify and validate new combinations to sustain current disease control strategies to overcome the challenge of drug resistance in the malaria parasites. To meet this demand, liquiritigenin (LTG) has been found to positively interact in combination with the existing clinically used drug chloroquine (CQ), which has become unfunctional due to acquired drug resistance. PURPOSE To evaluate the best interaction between LTG and CQ against CQ- resistant strain of P. falciparum. Furthermore, the in vivo antimalarial efficacy and possible mechanism of action of the best combination was also assessed. METHODS The in vitro anti-plasmodial potential of LTG against CQ- resistant strain K1 of P. falciparum was tested using Giemsa staining method. The behaviour of the combinations was evaluated using the fix ratio method and evaluated the interaction of LTG and CQ by calculating the fractional inhibitory concentration index (FICI). Oral toxicity study was carried out in a mice model. In vivo antimalarial efficacy of LTG alone and in combination with CQ was evaluated using a four-day suppression test in a mouse model. The effect of LTG on CQ accumulation was measured using HPLC and the rate of alkalinization of the digestive vacuole. Cytosolic Ca2+ level, mitochondrial membrane potential, caspase-like activity, terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay, and Annexin V Apoptosis assay to assess anti-plasmodial potential. Proteomics analysis was evaluated by LC-MS/MS analysis. RESULTS LTG possesses anti-plasmodial activity on its own and it showed to be an adjuvant of CQ. In in vitro studies, LTG showed synergy with CQ only in the ratio (CQ: LTG-1:4) against CQ-resistant strain (K1) of P. falciparum. Interestingly, in vivo studies, LTG in combination with CQ showed higher chemo-suppression and enhanced mean survival time at much lower concentrations compared to individual doses of LTG and CQ against CQ- resistant strain (N67) of Plasmodium yoelli nigeriensis. LTG was found to increase the CQ accumulation into digestive vacuole, reducing the rate of alkalinization, in turn increasing cytosolic Ca2+ level, loss of mitochondrial potential, caspase-3 activity, DNA damage and externalization of phosphatidylserine of the membrane (in vitro). These observations indicate the involvement of apoptosis-like death of P. falciparum that might be due to the accumulation of CQ. CONCLUSION LTG showed synergy with CQ in the ratio LTG: CQ, 4:1) in vitro and was able to curtail the IC50 of CQ and LTG. Interestingly, in vivo in combination with CQ, LTG showed higher chemo-suppression as well as enhanced mean survival time at a much lower concentrations of both the partners as compared to an individual dose of CQ and LTG. Thus, synergistic drug combination offers the possibility to enhance CQ efficacy in chemotherapy.
Collapse
Affiliation(s)
- Saurabh Kumar
- Bioprospection and Product Development Division, CSIR- Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Deepak Singh Kapkoti
- Phytochemistry Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Pooja Rani Mina
- Bioprospection and Product Development Division, CSIR- Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Madhuri Gupta
- Phytochemistry Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Ravi Kumar
- Bioprospection and Product Development Division, CSIR- Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Parmanand Kumar
- Bioprospection and Product Development Division, CSIR- Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Priyanka Pathak
- Bioprospection and Product Development Division, CSIR- Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - R S Bhakuni
- Phytochemistry Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Prasant Rout
- Phytochemistry Division, CSIR-Central Institute of Medicinal and Aromatic Plants, Lucknow, India
| | - Anirban Pal
- Bioprospection and Product Development Division, CSIR- Central Institute of Medicinal and Aromatic Plants, Lucknow, India.
| | - Mahendra P Darokar
- Bioprospection and Product Development Division, CSIR- Central Institute of Medicinal and Aromatic Plants, Lucknow, India.
| |
Collapse
|
5
|
Domes R, Frosch T. Investigations on the Novel Antimalarial Ferroquine in Biomimetic Solutions Using Deep UV Resonance Raman Spectroscopy and Density Functional Theory. Anal Chem 2023; 95:7630-7639. [PMID: 37141178 DOI: 10.1021/acs.analchem.3c00539] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/05/2023]
Abstract
Deep ultraviolet (DUV) resonance Raman experiments are performed, investigating the novel, promising antimalarial ferroquine (FQ). Two buffered aqueous solutions with pH values of 5.13 and 7.00 are used, simulating the acidic and neutral conditions inside a parasite's digestive vacuole and cytosol, respectively. To imitate the different polarities of the membranes and interior, the buffer's 1,4-dioxane content was increased. These experimental conditions should mimic the transport of the drug inside malaria-infected erythrocytes through parasitophorous membranes. Supporting density functional theory (DFT) calculations on the drug's micro-speciation were performed, which could be nicely assigned to shifts in the peak positions of resonantly enhanced high-wavenumber Raman signals at λexc = 257 nm. FQ is fully protonated in polar mixtures like the host interior and the parasite's cytoplasm or digestive vacuole (DV) and is only present as a free base in nonpolar ones, such as the host's and parasitophorous membranes. Additionally, the limit of detection (LoD) of FQ at vacuolic pH values was determined using DUV excitation wavelengths at 244 and 257 nm. By applying the resonant laser line at λexc = 257 nm, a minimal FQ concentration of 3.1 μM was detected, whereas the pre-resonant excitation wavelength 244 nm provides an LoD of 6.9 μM. These values were all up to one order of magnitude lower than the concentration found for the food vacuole of a parasitized erythrocyte.
Collapse
Affiliation(s)
- Robert Domes
- Leibniz Institute of Photonic Technology, Albert-Einstein Strasse 9, 07751 Jena, Germany
| | - Torsten Frosch
- Leibniz Institute of Photonic Technology, Albert-Einstein Strasse 9, 07751 Jena, Germany
- Biophotonics and Biomedical Engineering Group, Technical University Darmstadt, Merckstrasse 25, 64283 Darmstadt, Germany
| |
Collapse
|
6
|
Paulikat M, Vitone D, Schackert FK, Schuth N, Barbanente A, Piccini G, Ippoliti E, Rossetti G, Clark AH, Nachtegaal M, Haumann M, Dau H, Carloni P, Geremia S, De Zorzi R, Quintanar L, Arnesano F. Molecular Dynamics and Structural Studies of Zinc Chloroquine Complexes. J Chem Inf Model 2023; 63:161-172. [PMID: 36468829 DOI: 10.1021/acs.jcim.2c01164] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/10/2022]
Abstract
Chloroquine (CQ) is a first-choice drug against malaria and autoimmune diseases. It has been co-administered with zinc against SARS-CoV-2 and soon dismissed because of safety issues. The structural features of Zn-CQ complexes and the effect of CQ on zinc distribution in cells are poorly known. In this study, state-of-the-art computations combined with experiments were leveraged to solve the structural determinants of zinc-CQ interactions in solution and the solid state. NMR, ESI-MS, and X-ray absorption and diffraction methods were combined with ab initio molecular dynamics calculations to address the kinetic lability of this complex. Within the physiological pH range, CQ binds Zn2+ through the quinoline ring nitrogen, forming [Zn(CQH)Clx(H2O)3-x](3+)-x (x = 0, 1, 2, and 3) tetrahedral complexes. The Zn(CQH)Cl3 species is stable at neutral pH and at high chloride concentrations typical of the extracellular medium, but metal coordination is lost at a moderately low pH as in the lysosomal lumen. The pentacoordinate complex [Zn(CQH)(H2O)4]3+ may exist in the absence of chloride. This in vitro/in silico approach can be extended to other metal-targeting drugs and bioinorganic systems.
Collapse
Affiliation(s)
- Mirko Paulikat
- Computational Biomedicine (IAS-5/INM-9), Forschungszentrum Jülich GmbH, 52428Jülich, Germany
| | - Daniele Vitone
- Department of Chemistry, University of Bari "Aldo Moro", 70125Bari, Italy
| | - Florian K Schackert
- Computational Biomedicine (IAS-5/INM-9), Forschungszentrum Jülich GmbH, 52428Jülich, Germany.,Department of Physics, RWTH Aachen University, 52062Aachen, Germany
| | - Nils Schuth
- Department of Chemistry, Center for Research and Advanced Studies (Cinvestav), 07360Mexico City, Mexico
| | | | | | - Emiliano Ippoliti
- Computational Biomedicine (IAS-5/INM-9), Forschungszentrum Jülich GmbH, 52428Jülich, Germany
| | - Giulia Rossetti
- Computational Biomedicine (IAS-5/INM-9), Forschungszentrum Jülich GmbH, 52428Jülich, Germany.,Jülich Supercomputing Centre (JSC), Forschungszentrum Jülich GmbH, 52428Jülich, Germany.,Department of Neurology, RWTH Aachen University, 52062Aachen, Germany
| | - Adam H Clark
- Paul Scherrer Institute, 5232Villigen, Switzerland
| | | | - Michael Haumann
- Department of Physics, Freie Universität Berlin, 14195Berlin, Germany
| | - Holger Dau
- Department of Physics, Freie Universität Berlin, 14195Berlin, Germany
| | - Paolo Carloni
- Computational Biomedicine (IAS-5/INM-9), Forschungszentrum Jülich GmbH, 52428Jülich, Germany.,Department of Physics, RWTH Aachen University, 52062Aachen, Germany
| | - Silvano Geremia
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, 34127Trieste, Italy
| | - Rita De Zorzi
- Department of Chemical and Pharmaceutical Sciences, University of Trieste, 34127Trieste, Italy
| | - Liliana Quintanar
- Department of Chemistry, Center for Research and Advanced Studies (Cinvestav), 07360Mexico City, Mexico
| | - Fabio Arnesano
- Department of Chemistry, University of Bari "Aldo Moro", 70125Bari, Italy
| |
Collapse
|
7
|
Molecular Dynamic, Hirshfeld Surface, Molecular Docking and Drug likeness Studies of a Potent Anti-oxidant, Anti-malaria and Anti-Inflammatory medicine: Pyrogallol. RESULTS IN CHEMISTRY 2023. [DOI: 10.1016/j.rechem.2023.100763] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
|
8
|
Agyapong J, Rohrbach P. Quantifying pH in Malaria Using pHluorin and Flow Cytometry. Methods Mol Biol 2023; 2644:211-224. [PMID: 37142924 DOI: 10.1007/978-1-0716-3052-5_13] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/06/2023]
Abstract
Intracellular pH (pHi) plays a critical role in the regulation of numerous biological functions where specific pH ranges are required for optimal operation within cells. Slight pH changes can impact the regulation of diverse molecular processes, including enzymatic activities, ion channels, and transporters, which all play a role in cell functions. Methods for quantifying pHi continue to evolve and include various optical methods using fluorescent pH indicators. Here, we provide a protocol to measure pHi in the cytosol of Plasmodium falciparum blood stage parasites by means of flow cytometry and using pHluorin2, a pH-sensitive fluorescent protein that has been introduced into the genome of the parasite.
Collapse
Affiliation(s)
- Jeffrey Agyapong
- Institute of Parasitology, McGill University, Montreal, QC, Canada
| | - Petra Rohrbach
- Institute of Parasitology, McGill University, Montreal, QC, Canada.
| |
Collapse
|
9
|
Vásquez-Ocmín PG, Gallard JF, Van Baelen AC, Leblanc K, Cojean S, Mouray E, Grellier P, Guerra CAA, Beniddir MA, Evanno L, Figadère B, Maciuk A. Biodereplication of Antiplasmodial Extracts: Application of the Amazonian Medicinal Plant Piper coruscans Kunth. Molecules 2022; 27:7638. [PMID: 36364460 PMCID: PMC9656727 DOI: 10.3390/molecules27217638] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/01/2022] [Accepted: 11/05/2022] [Indexed: 09/08/2024] Open
Abstract
Improved methodological tools to hasten antimalarial drug discovery remain of interest, especially when considering natural products as a source of drug candidates. We propose a biodereplication method combining the classical dereplication approach with the early detection of potential antiplasmodial compounds in crude extracts. Heme binding is used as a surrogate of the antiplasmodial activity and is monitored by mass spectrometry in a biomimetic assay. Molecular networking and automated annotation of targeted mass through data mining were followed by mass-guided compound isolation by taking advantage of the versatility and finely tunable selectivity offered by centrifugal partition chromatography. This biodereplication workflow was applied to an ethanolic extract of the Amazonian medicinal plant Piper coruscans Kunth (Piperaceae) showing an IC50 of 1.36 µg/mL on the 3D7 Plasmodium falciparum strain. It resulted in the isolation of twelve compounds designated as potential antiplasmodial compounds by the biodereplication workflow. Two chalcones, aurentiacin (1) and cardamonin (3), with IC50 values of 2.25 and 5.5 µM, respectively, can be considered to bear the antiplasmodial activity of the extract, with the latter not relying on a heme-binding mechanism. This biodereplication method constitutes a rapid, efficient, and robust technique to identify potential antimalarial compounds in complex extracts such as plant extracts.
Collapse
Affiliation(s)
| | - Jean-François Gallard
- Institut de Chimie des Substances Naturelles CNRS UPR 2301, Université Paris-Saclay, 1 Avenue de la Terrasse, 91198 Gif-sur-Yvette, France
| | - Anne-Cécile Van Baelen
- Université Paris-Saclay, CNRS, BioCIS, 91400 Orsay, France
- Département Médicaments et Technologies pour la Santé (DMTS), CEA, SIMoS, Université Paris-Saclay, F-91191 Gif-sur-Yvette, France
| | - Karine Leblanc
- Université Paris-Saclay, CNRS, BioCIS, 91400 Orsay, France
| | | | - Elisabeth Mouray
- Unité Molécules de Communication et Adaptation des Microorganismes (MCAM, UMR 7245), Muséum National d’Histoire Naturelle, CNRS, Sorbonne Universités, CP52, 57 Rue Cuvier, 75005 Paris, France
| | - Philippe Grellier
- Unité Molécules de Communication et Adaptation des Microorganismes (MCAM, UMR 7245), Muséum National d’Histoire Naturelle, CNRS, Sorbonne Universités, CP52, 57 Rue Cuvier, 75005 Paris, France
| | - Carlos A. Amasifuén Guerra
- Dirección de Recursos Genéticos y Biotecnología (DRGB), Instituto Nacional de Innovación Agraria (INIA), Avenida La Molina N° 1981, La Molina, Lima 15024, Peru
| | | | - Laurent Evanno
- Université Paris-Saclay, CNRS, BioCIS, 91400 Orsay, France
| | - Bruno Figadère
- Université Paris-Saclay, CNRS, BioCIS, 91400 Orsay, France
| | | |
Collapse
|
10
|
Olivier T, Loots L, Kok M, de Villiers M, Reader J, Birkholtz LM, Arnott GE, de Villiers KA. Adsorption to the Surface of Hemozoin Crystals: Structure-Based Design and Synthesis of Amino-Phenoxazine β-Hematin Inhibitors. ChemMedChem 2022; 17:e202200139. [PMID: 35385211 PMCID: PMC9119941 DOI: 10.1002/cmdc.202200139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2022] [Revised: 04/05/2022] [Indexed: 11/07/2022]
Abstract
In silico adsorption of eight antimalarials that inhibit β-hematin (synthetic hemozoin) formation identified a primary binding site on the (001) face, which accommodates inhibitors via formation of predominantly π-π interactions. A good correlation (r2 =0.64, P=0.017) between adsorption energies and the logarithm of β-hematin inhibitory activity was found for this face. Of 53 monocyclic, bicyclic and tricyclic scaffolds, the latter yielded the most favorable adsorption energies. Five new amino-phenoxazine compounds were pursued as β-hematin inhibitors based on adsorption behaviour. The 2-substituted phenoxazines show good to moderate β-hematin inhibitory activity (<100 μM) and Plasmodium falciparum blood stage activity against the 3D7 strain. N1 ,N1 -diethyl-N4 -(10H-phenoxazin-2-yl)pentane-1,4-diamine (P2a) is the most promising hit with IC50 values of 4.7±0.6 and 0.64±0.05 μM, respectively. Adsorption energies are predictive of β-hematin inhibitory activity, and thus the in silico approach is a beneficial tool for structure-based development of new non-quinoline inhibitors.
Collapse
Affiliation(s)
- Tania Olivier
- Department of Chemistry and Polymer Science, Stellenbosch University, Private BagX1, Matieland, 7602, South Africa
| | - Leigh Loots
- Department of Chemistry and Polymer Science, Stellenbosch University, Private BagX1, Matieland, 7602, South Africa
| | - Michélle Kok
- Department of Biochemistry, Stellenbosch University, Private BagX1, Matieland, 7602, South Africa
| | - Marianne de Villiers
- Department of Biochemistry, Stellenbosch University, Private BagX1, Matieland, 7602, South Africa
| | - Janette Reader
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Pretoria, 0028, South Africa
| | - Lyn-Marié Birkholtz
- Department of Biochemistry, Genetics and Microbiology, Institute for Sustainable Malaria Control, University of Pretoria, Pretoria, 0028, South Africa
| | - Gareth E Arnott
- Department of Chemistry and Polymer Science, Stellenbosch University, Private BagX1, Matieland, 7602, South Africa
| | - Katherine A de Villiers
- Department of Chemistry and Polymer Science, Stellenbosch University, Private BagX1, Matieland, 7602, South Africa
| |
Collapse
|
11
|
Mechanistic basis for multidrug resistance and collateral drug sensitivity conferred to the malaria parasite by polymorphisms in PfMDR1 and PfCRT. PLoS Biol 2022; 20:e3001616. [PMID: 35507548 PMCID: PMC9067703 DOI: 10.1371/journal.pbio.3001616] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2021] [Accepted: 03/31/2022] [Indexed: 01/16/2023] Open
Abstract
Polymorphisms in the Plasmodium falciparum multidrug resistance protein 1 (pfmdr1) gene and the Plasmodium falciparum chloroquine resistance transporter (pfcrt) gene alter the malaria parasite’s susceptibility to most of the current antimalarial drugs. However, the precise mechanisms by which PfMDR1 contributes to multidrug resistance have not yet been fully elucidated, nor is it understood why polymorphisms in pfmdr1 and pfcrt that cause chloroquine resistance simultaneously increase the parasite’s susceptibility to lumefantrine and mefloquine—a phenomenon known as collateral drug sensitivity. Here, we present a robust expression system for PfMDR1 in Xenopus oocytes that enables direct and high-resolution biochemical characterizations of the protein. We show that wild-type PfMDR1 transports diverse pharmacons, including lumefantrine, mefloquine, dihydroartemisinin, piperaquine, amodiaquine, methylene blue, and chloroquine (but not the antiviral drug amantadine). Field-derived mutant isoforms of PfMDR1 differ from the wild-type protein, and each other, in their capacities to transport these drugs, indicating that PfMDR1-induced changes in the distribution of drugs between the parasite’s digestive vacuole (DV) and the cytosol are a key driver of both antimalarial resistance and the variability between multidrug resistance phenotypes. Of note, the PfMDR1 isoforms prevalent in chloroquine-resistant isolates exhibit reduced capacities for chloroquine, lumefantrine, and mefloquine transport. We observe the opposite relationship between chloroquine resistance-conferring mutations in PfCRT and drug transport activity. Using our established assays for characterizing PfCRT in the Xenopus oocyte system and in live parasite assays, we demonstrate that these PfCRT isoforms transport all 3 drugs, whereas wild-type PfCRT does not. We present a mechanistic model for collateral drug sensitivity in which mutant isoforms of PfMDR1 and PfCRT cause chloroquine, lumefantrine, and mefloquine to remain in the cytosol instead of sequestering within the DV. This change in drug distribution increases the access of lumefantrine and mefloquine to their primary targets (thought to be located outside of the DV), while simultaneously decreasing chloroquine’s access to its target within the DV. The mechanistic insights presented here provide a basis for developing approaches that extend the useful life span of antimalarials by exploiting the opposing selection forces they exert upon PfCRT and PfMDR1.
Collapse
|
12
|
Henderson JA, Shen J. Exploring the pH- and Ligand-Dependent Flap Dynamics of Malarial Plasmepsin II. J Chem Inf Model 2021; 62:150-158. [PMID: 34964641 DOI: 10.1021/acs.jcim.1c01180] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Malaria remains a global health threat─over 400,000 deaths occurred in 2019. Plasmepsins are promising targets of antimalarial therapeutics; however, no inhibitors have reached the clinic. To fuel the progress, a detailed understanding of the pH- and ligand-dependent conformational dynamics of plasmepsins is needed. Here we present the continuous constant pH molecular dynamics study of the prototypical plasmepsin II and its complexed form with a substrate analogue. The simulations revealed that the catalytic dyads D34 and D214 are highly coupled in the apo protein and that the pepstatin binding enhances the difference in proton affinity, making D34 the general base and D214 the general acid. The simulations showed that the flap adopts an open state regardless of pH; however, upon pepstatin binding the flap can close or open depending on the protonation state of D214. These and other data are discussed and compared with the off-targets human cathepsin D and renin. This study lays the groundwork for a systematic investigation of pH- and ligand-modulated dynamics of the entire family of plasmepsins to help design more potent and selective inhibitors.
Collapse
Affiliation(s)
- Jack A Henderson
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| | - Jana Shen
- Department of Pharmaceutical Sciences, University of Maryland School of Pharmacy, Baltimore, Maryland 21201, United States
| |
Collapse
|
13
|
Broichhagen J, Kilian N. Chemical Biology Tools To Investigate Malaria Parasites. Chembiochem 2021; 22:2219-2236. [PMID: 33570245 PMCID: PMC8360121 DOI: 10.1002/cbic.202000882] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2020] [Revised: 02/10/2021] [Indexed: 02/06/2023]
Abstract
Parasitic diseases like malaria tropica have been shaping human evolution and history since the beginning of mankind. After infection, the response of the human host ranges from asymptomatic to severe and may culminate in death. Therefore, proper examination of the parasite's biology is pivotal to deciphering unique molecular, biochemical and cell biological processes, which in turn ensure the identification of treatment strategies, such as potent drug targets and vaccine candidates. However, implementing molecular biology methods for genetic manipulation proves to be difficult for many parasite model organisms. The development of fast and straightforward applicable alternatives, for instance small-molecule probes from the field of chemical biology, is essential. In this review, we will recapitulate the highlights of previous molecular and chemical biology approaches that have already created insight and understanding of the malaria parasite Plasmodium falciparum. We discuss current developments from the field of chemical biology and explore how their application could advance research into this parasite in the future. We anticipate that the described approaches will help to close knowledge gaps in the biology of P. falciparum and we hope that researchers will be inspired to use these methods to gain knowledge - with the aim of ending this devastating disease.
Collapse
Affiliation(s)
- Johannes Broichhagen
- Leibniz-Forschungsinstitut für Molekulare Pharmakologie (FMP)Robert-Roessle-Strasse 1013125BerlinGermany
| | - Nicole Kilian
- Centre for Infectious DiseasesParasitologyHeidelberg University HospitalIm Neuenheimer Feld 32469120HeidelbergGermany
| |
Collapse
|
14
|
Mills B, Isaac RE, Foster R. Metalloaminopeptidases of the Protozoan Parasite Plasmodium falciparum as Targets for the Discovery of Novel Antimalarial Drugs. J Med Chem 2021; 64:1763-1785. [PMID: 33534577 DOI: 10.1021/acs.jmedchem.0c01721] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Malaria poses a significant threat to approximately half of the world's population with an annual death toll close to half a million. The emergence of resistance to front-line antimalarials in the most lethal human parasite species, Plasmodium falciparum (Pf), threatens progress made in malaria control. The prospect of losing the efficacy of antimalarial drugs is driving the search for small molecules with new modes of action. Asexual reproduction of the parasite is critically dependent on the recycling of amino acids through catabolism of hemoglobin (Hb), which makes metalloaminopeptidases (MAPs) attractive targets for the development of new drugs. The Pf genome encodes eight MAPs, some of which have been found to be essential for parasite survival. In this article, we discuss the biological structure and function of each MAP within the Pf genome, along with the drug discovery efforts that have been undertaken to identify novel antimalarial candidates of therapeutic value.
Collapse
Affiliation(s)
- Belinda Mills
- School of Chemistry, University of Leeds, Leeds, U.K., LS2 9JT
| | - R Elwyn Isaac
- School of Biology, University of Leeds, Leeds, U.K., LS2 9JT
| | - Richard Foster
- School of Chemistry, University of Leeds, Leeds, U.K., LS2 9JT
| |
Collapse
|
15
|
Shafik SH, Cobbold SA, Barkat K, Richards SN, Lancaster NS, Llinás M, Hogg SJ, Summers RL, McConville MJ, Martin RE. The natural function of the malaria parasite's chloroquine resistance transporter. Nat Commun 2020; 11:3922. [PMID: 32764664 PMCID: PMC7413254 DOI: 10.1038/s41467-020-17781-6] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2019] [Accepted: 07/15/2020] [Indexed: 01/27/2023] Open
Abstract
The Plasmodium falciparum chloroquine resistance transporter (PfCRT) is a key contributor to multidrug resistance and is also essential for the survival of the malaria parasite, yet its natural function remains unresolved. We identify host-derived peptides of 4-11 residues, varying in both charge and composition, as the substrates of PfCRT in vitro and in situ, and show that PfCRT does not mediate the non-specific transport of other metabolites and/or ions. We find that drug-resistance-conferring mutations reduce both the peptide transport capacity and substrate range of PfCRT, explaining the impaired fitness of drug-resistant parasites. Our results indicate that PfCRT transports peptides from the lumen of the parasite's digestive vacuole to the cytosol, thereby providing a source of amino acids for parasite metabolism and preventing osmotic stress of this organelle. The resolution of PfCRT's native substrates will aid the development of drugs that target PfCRT and/or restore the efficacy of existing antimalarials.
Collapse
Affiliation(s)
- Sarah H Shafik
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| | - Simon A Cobbold
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Kawthar Barkat
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| | - Sashika N Richards
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| | - Nicole S Lancaster
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| | - Manuel Llinás
- Department of Biochemistry and Molecular Biology, Department of Chemistry, and Huck Center for Malaria Research, The Pennsylvania State University, University Park, PA, 16802, USA
| | - Simon J Hogg
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| | - Robert L Summers
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia
| | - Malcolm J McConville
- Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, VIC, 3052, Australia
| | - Rowena E Martin
- Research School of Biology, The Australian National University, Canberra, ACT, 2601, Australia.
| |
Collapse
|
16
|
Mode of action of quinoline antimalarial drugs in red blood cells infected by Plasmodium falciparum revealed in vivo. Proc Natl Acad Sci U S A 2019; 116:22946-22952. [PMID: 31659055 PMCID: PMC6859308 DOI: 10.1073/pnas.1910123116] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The most widely used antimalarial drugs belong to the quinoline family. The question of their mode of action has been open for centuries. It has been recently narrowed down to whether these drugs interfere with the process of crystallization of heme in the malaria parasite. To date, all studies of the drug action on heme crystals have been done either on model systems or on dried parasites, which yielded limited data and ambiguity. This study was done in actual parasites in their near-native environment, revealing the mode of action of these drugs in vivo. The approach adopted in this study can be extended to other families of antimalarial drugs, such as artemisinins, provided appropriate derivatives can be synthesized. The most widely used antimalarial drugs belong to the quinoline family. Their mode of action has not been characterized at the molecular level in vivo. We report the in vivo mode of action of a bromo analog of the drug chloroquine in rapidly frozen Plasmodium falciparum-infected red blood cells. The Plasmodium parasite digests hemoglobin, liberating the heme as a byproduct, toxic to the parasite. It is detoxified by crystallization into inert hemozoin within the parasitic digestive vacuole. By mapping such infected red blood cells with nondestructive X-ray microscopy, we observe that bromoquine caps hemozoin crystals. The measured crystal surface coverage is sufficient to inhibit further hemozoin crystal growth, thereby sabotaging heme detoxification. Moreover, we find that bromoquine accumulates in the digestive vacuole, reaching submillimolar concentration, 1,000-fold more than that of the drug in the culture medium. Such a dramatic increase in bromoquine concentration enhances the drug’s efficiency in depriving heme from docking onto the hemozoin crystal surface. Based on direct observation of bromoquine distribution in the digestive vacuole and at its membrane surface, we deduce that the excess bromoquine forms a complex with the remaining heme deprived from crystallization. This complex is driven toward the digestive vacuole membrane, increasing the chances of membrane puncture and spillage of heme into the interior of the parasite.
Collapse
|
17
|
Sayers EJ, Peel SE, Schantz A, England RM, Beano M, Bates SM, Desai AS, Puri S, Ashford MB, Jones AT. Endocytic Profiling of Cancer Cell Models Reveals Critical Factors Influencing LNP-Mediated mRNA Delivery and Protein Expression. Mol Ther 2019; 27:1950-1962. [PMID: 31427168 DOI: 10.1016/j.ymthe.2019.07.018] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2019] [Revised: 07/18/2019] [Accepted: 07/26/2019] [Indexed: 12/26/2022] Open
Abstract
Lipid nanoparticles have great potential for delivering nucleic-acid-based therapeutics, but low efficiency limits their broad clinical translation. Differences in transfection capacity between in vitro models used for nanoparticle pre-clinical testing are poorly understood. To address this, using a clinically relevant lipid nanoparticle (LNP) delivering mRNA, we highlight specific endosomal characteristics in in vitro tumor models that impact protein expression. A 30-cell line LNP-mRNA transfection screen identified three cell lines having low, medium, and high transfection that correlated with protein expression when they were analyzed in tumor models. Endocytic profiling of these cell lines identified major differences in endolysosomal morphology, localization, endocytic uptake, trafficking, recycling, and endolysosomal pH, identified using a novel pH probe. High-transfecting cells showed rapid LNP uptake and trafficking through an organized endocytic pathway to lysosomes or rapid exocytosis. Low-transfecting cells demonstrated slower endosomal LNP trafficking to lysosomes and defective endocytic organization and acidification. Our data establish that efficient LNP-mRNA transfection relies on an early and narrow endosomal escape window prior to lysosomal sequestration and/or exocytosis. Endocytic profiling should form an important pre-clinical evaluation step for nucleic acid delivery systems to inform model selection and guide delivery-system design for improved clinical translation.
Collapse
Affiliation(s)
- Edward J Sayers
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff CF10 3NB, Wales
| | - Samantha E Peel
- Discovery Biology, Discovery Sciences, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Anna Schantz
- Advanced Drug Delivery, Pharmaceutical Sciences, IMED Biotech Unit, AstraZeneca, Gothenburg, Sweden
| | - Richard M England
- Early Chemical Development, Pharmaceutical Sciences, IMED Biotech Unit, AstraZeneca, Macclesfield, UK
| | - Maya Beano
- Pathology, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Stephanie M Bates
- Pathology, Drug Safety and Metabolism, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Arpan S Desai
- Advanced Drug Delivery, Pharmaceutical Sciences, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Sanyogitta Puri
- Advanced Drug Delivery, Pharmaceutical Sciences, IMED Biotech Unit, AstraZeneca, Cambridge, UK
| | - Marianne B Ashford
- Advanced Drug Delivery, Pharmaceutical Sciences, IMED Biotech Unit, AstraZeneca, Cambridge, UK.
| | - Arwyn T Jones
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, Redwood Building, King Edward VII Avenue, Cardiff CF10 3NB, Wales.
| |
Collapse
|
18
|
Ismail M, Du Y, Ling L, Li X. Artesunate-heparin conjugate based nanocapsules with improved pharmacokinetics to combat malaria. Int J Pharm 2019; 562:162-171. [DOI: 10.1016/j.ijpharm.2019.03.031] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 03/13/2019] [Accepted: 03/14/2019] [Indexed: 12/17/2022]
|
19
|
Farkaš R, Beňová-Liszeková D, Mentelová L, Beňo M, Babišová K, Trusinová-Pečeňová L, Raška O, Chase BA, Raška I. Endosomal vacuoles of the prepupal salivary glands of Drosophila play an essential role in the metabolic reallocation of iron. Dev Growth Differ 2018; 60:411-430. [PMID: 30123964 DOI: 10.1111/dgd.12562] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Revised: 06/30/2018] [Accepted: 07/02/2018] [Indexed: 12/17/2022]
Abstract
In the recent past, we demonstrated that a great deal is going on in the salivary glands of Drosophila in the interval after they release their glycoprotein-rich secretory glue during pupariation. The early-to-mid prepupal salivary glands undergo extensive endocytosis with widespread vacuolation of the cytoplasm followed by massive apocrine secretion. Here, we describe additional novel properties of these endosomes. The use of vital pH-sensitive probes provided confirmatory evidence that these endosomes have acidic contents and that there are two types of endocytosis seen in the prepupal glands. The salivary glands simultaneously generate mildly acidic, small, basally-derived endosomes and strongly acidic, large and apical endosomes. Staining of the large vacuoles with vital acidic probes is possible only after there is ambipolar fusion of both basal and apical endosomes, since only basally-derived endosomes can bring fluorescent probes into the vesicular system. We obtained multiple lines of evidence that the small basally-derived endosomes are chiefly involved in the uptake of dietary Fe3+ iron. The fusion of basal endosomes with the larger and strongly acidic apical endosomes appears to facilitate optimal conditions for ferrireductase activity inside the vacuoles to release metabolic Fe2+ iron. While iron was not detectable directly due to limited staining sensitivity, we found increasing fluorescence of the glutathione-sensitive probe CellTracker Blue CMAC in large vacuoles, which appeared to depend on the amount of iron released by ferrireductase. Moreover, heterologous fluorescently-labeled mammalian iron-bound transferrin is actively taken up, providing direct evidence for active iron uptake by basal endocytosis. In addition, we serendipitously found that small (basal) endosomes were uniquely recognized by PNA lectin, whereas large (apical) vacuoles bound DBA lectin.
Collapse
Affiliation(s)
- Robert Farkaš
- Laboratory of Developmental Genetics, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Denisa Beňová-Liszeková
- Laboratory of Developmental Genetics, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Lucia Mentelová
- Laboratory of Developmental Genetics, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia.,Department of Genetics, Comenius University, Bratislava, Slovakia
| | - Milan Beňo
- Laboratory of Developmental Genetics, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Klaudia Babišová
- Laboratory of Developmental Genetics, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia.,Department of Genetics, Comenius University, Bratislava, Slovakia
| | - Ludmila Trusinová-Pečeňová
- Laboratory of Developmental Genetics, Institute of Experimental Endocrinology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Otakar Raška
- Institute of Biology and Medical Genetics, 1st Faculty of Medicine, Charles University, Prague, Czech Republic.,Department of Normal, Pathological and Clinical Physiology, Third Faculty of Medicine, Charles University, Prague, Czech Republic
| | - Bruce A Chase
- Department of Biology, University of Nebraska, Omaha, Nebraska
| | - Ivan Raška
- Institute of Biology and Medical Genetics, 1st Faculty of Medicine, Charles University, Prague, Czech Republic
| |
Collapse
|
20
|
Lawrence N, Dennis ASM, Lehane AM, Ehmann A, Harvey PJ, Benfield AH, Cheneval O, Henriques ST, Craik DJ, McMorran BJ. Defense Peptides Engineered from Human Platelet Factor 4 Kill Plasmodium by Selective Membrane Disruption. Cell Chem Biol 2018; 25:1140-1150.e5. [PMID: 30033131 DOI: 10.1016/j.chembiol.2018.06.009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2018] [Revised: 06/01/2018] [Accepted: 06/25/2018] [Indexed: 11/29/2022]
Abstract
Malaria is a serious threat to human health and additional classes of antimalarial drugs are greatly needed. The human defense protein, platelet factor 4 (PF4), has intrinsic antiplasmodial activity but also undesirable chemokine properties. We engineered a peptide containing the isolated PF4 antiplasmodial domain, which through cyclization, retained the critical structure of the parent protein. The peptide, cPF4PD, killed cultured blood-stage Plasmodium falciparum with low micromolar potency by specific disruption of the parasite digestive vacuole. Its mechanism of action involved selective penetration and accumulation inside the intraerythrocytic parasite without damaging the host cell or parasite membranes; it did not accumulate in uninfected cells. This selective activity was accounted for by observations of the peptide's specific binding and penetration of membranes with exposed negatively charged phospholipid headgroups. Our findings highlight the tremendous potential of the cPF4PD scaffold for developing antimalarial peptide drugs with a distinct and selective mechanism of action.
Collapse
Affiliation(s)
- Nicole Lawrence
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Adelaide S M Dennis
- Research School of Biology, The Australian National University, Canberra, ACT 2600, Australia
| | - Adele M Lehane
- Research School of Biology, The Australian National University, Canberra, ACT 2600, Australia
| | - Anna Ehmann
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2600, Australia
| | - Peta J Harvey
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Aurélie H Benfield
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Olivier Cheneval
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia
| | - Sónia Troeira Henriques
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - David J Craik
- Institute for Molecular Bioscience, The University of Queensland, Brisbane, QLD 4072, Australia.
| | - Brendan J McMorran
- The John Curtin School of Medical Research, The Australian National University, Canberra, ACT 2600, Australia.
| |
Collapse
|
21
|
Ismail M, Ling L, Du Y, Yao C, Li X. Liposomes of dimeric artesunate phospholipid: A combination of dimerization and self-assembly to combat malaria. Biomaterials 2018; 163:76-87. [DOI: 10.1016/j.biomaterials.2018.02.026] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2017] [Revised: 02/03/2018] [Accepted: 02/09/2018] [Indexed: 10/18/2022]
|
22
|
Adamantane amine-linked chloroquinoline derivatives as chloroquine resistance modulating agents in Plasmodium falciparum. Bioorg Med Chem Lett 2018; 28:1287-1291. [DOI: 10.1016/j.bmcl.2018.03.026] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2018] [Revised: 03/09/2018] [Accepted: 03/10/2018] [Indexed: 12/27/2022]
|
23
|
Fitzroy SM, Gildenhuys J, Olivier T, Tshililo NO, Kuter D, de Villiers KA. The Effects of Quinoline and Non-Quinoline Inhibitors on the Kinetics of Lipid-Mediated β-Hematin Crystallization. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2017; 33:7529-7537. [PMID: 28689414 PMCID: PMC5709178 DOI: 10.1021/acs.langmuir.7b01132] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/07/2023]
Abstract
The throughput of a biomimetic lipid-mediated assay used to investigate the effects of inhibitors on the kinetics of β-hematin formation has been optimized through the use of 24-well microplates. The rate constant for β-hematin formation mediated by monopalmitoyl-rac-glycerol was reduced from 0.17 ± 0.04 min-1 previously measured in Falcon tubes to 0.019 ± 0.002 min-1 in the optimized assay. While this necessitated longer incubation times, transferring aliquots from multiple 24-well plates to a single 96-well plate for final absorbance measurements actually improved the overall turnaround time per inhibitor. This assay has been applied to investigate the effects of four clinically relevant antimalarial drugs (chloroquine, amodiaquine, quinidine, and quinine) as well as several short-chain 4-aminoquinoline derivatives and non-quinoline (benzamide) compounds on the kinetics of β-hematin formation. The adsorption strength of these inhibitors to crystalline β-hematin (Kads) was quantified using a theoretical kinetic model that is based on the Avrami equation and the Langmuir isotherm. Statistically significant linear correlations between lipid-mediated β-hematin inhibitory activity and Kads values for quinoline (r2 = 0.76, P-value = 0.0046) and non-quinoline compounds (r2 = 0.99, P-stat = 0.0006), as well as between parasite inhibitory activity (D10) and Kads values for quinoline antimalarial drugs and short-chain chloroquine derivatives (r2 = 0.64, P-value = 0.0098), provide a strong indication that drug action involves adsorption to the surface of β-hematin crystals. Independent support in this regard is provided by experiments that spectrophotometrically monitor the direct adsorption of antimalarial drugs to preformed β-hematin.
Collapse
|
24
|
A Variant PfCRT Isoform Can Contribute to Plasmodium falciparum Resistance to the First-Line Partner Drug Piperaquine. mBio 2017; 8:mBio.00303-17. [PMID: 28487425 PMCID: PMC5424201 DOI: 10.1128/mbio.00303-17] [Citation(s) in RCA: 76] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Current efforts to reduce the global burden of malaria are threatened by the rapid spread throughout Asia of Plasmodium falciparum resistance to artemisinin-based combination therapies, which includes increasing rates of clinical failure with dihydroartemisinin plus piperaquine (PPQ) in Cambodia. Using zinc finger nuclease-based gene editing, we report that addition of the C101F mutation to the chloroquine (CQ) resistance-conferring PfCRT Dd2 isoform common to Asia can confer PPQ resistance to cultured parasites. Resistance was demonstrated as significantly higher PPQ concentrations causing 90% inhibition of parasite growth (IC90) or 50% parasite killing (50% lethal dose [LD50]). This mutation also reversed Dd2-mediated CQ resistance, sensitized parasites to amodiaquine, quinine, and artemisinin, and conferred amantadine and blasticidin resistance. Using heme fractionation assays, we demonstrate that PPQ causes a buildup of reactive free heme and inhibits the formation of chemically inert hemozoin crystals. Our data evoke inhibition of heme detoxification in the parasite’s acidic digestive vacuole as the primary mode of both the bis-aminoquinoline PPQ and the related 4-aminoquinoline CQ. Both drugs also inhibit hemoglobin proteolysis at elevated concentrations, suggesting an additional mode of action. Isogenic lines differing in their pfmdr1 copy number showed equivalent PPQ susceptibilities. We propose that mutations in PfCRT could contribute to a multifactorial basis of PPQ resistance in field isolates. The global agenda to eliminate malaria depends on the continued success of artemisinin-based combination therapies (ACTs), which target the asexual blood stages of the intracellular parasite Plasmodium. Partial resistance to artemisinin, however, is now established in Southeast Asia, exposing the partner drugs to increased selective pressure. Plasmodium falciparum resistance to the first-line partner piperaquine (PPQ) is now spreading rapidly in Cambodia, resulting in clinical treatment failures. Here, we report that a variant form of the Plasmodium falciparum chloroquine resistance transporter, harboring a C101F mutation edited into the chloroquine (CQ)-resistant Dd2 isoform prevalent in Asia, can confer PPQ resistance in cultured parasites. This was accompanied by a loss of CQ resistance. Biochemical assays showed that PPQ, like CQ, inhibits the detoxification of reactive heme that is formed by parasite-mediated catabolism of host hemoglobin. We propose that novel PfCRT variants emerging in the field could contribute to a multigenic basis of PPQ resistance.
Collapse
|
25
|
Shin S, Shardt O, Warren PB, Stone HA. Membraneless water filtration using CO 2. Nat Commun 2017; 8:15181. [PMID: 28462929 PMCID: PMC5418569 DOI: 10.1038/ncomms15181] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 03/07/2017] [Indexed: 11/13/2022] Open
Abstract
Water purification technologies such as microfiltration/ultrafiltration and reverse osmosis utilize porous membranes to remove suspended particles and solutes. These membranes, however, cause many drawbacks such as a high pumping cost and a need for periodic replacement due to fouling. Here we show an alternative membraneless method for separating suspended particles by exposing the colloidal suspension to CO2. Dissolution of CO2 into the suspension creates solute gradients that drive phoretic motion of particles. Due to the large diffusion potential generated by the dissociation of carbonic acid, colloidal particles move either away from or towards the gas–liquid interface depending on their surface charge. Using the directed motion of particles induced by exposure to CO2, we demonstrate a scalable, continuous flow, membraneless particle filtration process that exhibits low energy consumption, three orders of magnitude lower than conventional microfiltration/ultrafiltration processes, and is essentially free from fouling. Water treatment processes mostly rely on the use of membranes and filters, which have high pumping costs and require periodic replacement. Here, the authors describe an efficient membraneless method that induces directed motion of suspended colloidal particles by exposing the suspension to CO2.
Collapse
Affiliation(s)
- Sangwoo Shin
- Department of Mechanical and Aerospace Engineering, Princeton University, Princeton, New Jersey 08544, USA
| | - Orest Shardt
- Department of Mechanical and Aerospace Engineering, Princeton University, Princeton, New Jersey 08544, USA
| | | | - Howard A Stone
- Department of Mechanical and Aerospace Engineering, Princeton University, Princeton, New Jersey 08544, USA
| |
Collapse
|
26
|
Jida M, Sanchez CP, Urgin K, Ehrhardt K, Mounien S, Geyer A, Elhabiri M, Lanzer M, Davioud-Charvet E. A Redox-Active Fluorescent pH Indicator for Detecting Plasmodium falciparum Strains with Reduced Responsiveness to Quinoline Antimalarial Drugs. ACS Infect Dis 2017; 3:119-131. [PMID: 28183182 DOI: 10.1021/acsinfecdis.5b00141] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mutational changes in the Plasmodium falciparum chloroquine resistance transporter (PfCRT) have been associated with differential responses to a wide spectrum of biologically active compounds including current and former quinoline and quinoline-like antimalarial drugs. PfCRT confers altered drug responsiveness by acting as a transport system, expelling drugs from the parasite's digestive vacuole where these drugs exert, at least part of, their antiplasmodial activity. To preserve the efficacy of these invaluable drugs, novel functional tools are required for epidemiological surveys of parasite strains carrying mutant PfCRT variants and for drug development programs aimed at inhibiting or circumventing the action of PfCRT. Here we report the synthesis and characterization of a pH-sensitive fluorescent chloroquine analogue consisting of 7-chloro-N-{2-[(propan-2-yl)amino]ethyl}quinolin-4-amine functionalized with the fluorochrome 7-nitrobenzofurazan (NBD) (henceforth termed Fluo-CQ). In the parasite, Fluo-CQ accumulates in the digestive vacuole, giving rise to a strong fluorescence signal but only in parasites carrying the wild type PfCRT. In parasites carrying the mutant PfCRT, Fluo-CQ does not accumulate. The differential handling of the fluorescent probe, combined with live cell imaging, provides a diagnostic tool for quick detection of those P. falciparum strains that carry a PfCRT variant associated with altered responsiveness to quinoline and quinoline-like antimalarial drugs. In contrast to the accumulation studies, chloroquine (CQ)-resistant parasites were observed cross-resistant to Fluo-CQ when the chemical probe was tested in various CQ-sensitive and -resistant parasite strains. NBD derivatives were found to act as redox cyclers of two essential targets, using a coupled assay based on methemoglobin and the NADPH-dependent glutathione reductase (GRs) from P. falciparum. This redox activity is proposed to contribute to the dual action of Fluo-CQ on redox equilibrium and methemoglobin reduction via PfCRT-mediated drug efflux in the cytosol and then continuous redox-dependent shuttling between food vacuole and cytosol. Taking into account these physicochemical characteristics, a model was proposed to explain Fluo-CQ antimalarial effects involving the contribution of PfCRT-mediated transport, methemoglobin reduction, hematin binding, and NBD reduction activity catalyzed by PfGR in CQ-resistant versus CQ-sensitive parasites. Therefore, introduction of NBD fluorophore in drugs is not inert and should be taken into account in drug transport and imaging studies.
Collapse
Affiliation(s)
- Mouhamad Jida
- UMR 7509 Centre National de la Recherche Scientifique and University of Strasbourg, European School of Chemistry, Polymers and Materials (ECPM), 25 rue Becquerel, F-67087 Strasbourg, France
| | - Cecilia P. Sanchez
- Zentrum
für Infektiologie, Parasitologie, Universität Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Karène Urgin
- UMR 7509 Centre National de la Recherche Scientifique and University of Strasbourg, European School of Chemistry, Polymers and Materials (ECPM), 25 rue Becquerel, F-67087 Strasbourg, France
| | - Katharina Ehrhardt
- UMR 7509 Centre National de la Recherche Scientifique and University of Strasbourg, European School of Chemistry, Polymers and Materials (ECPM), 25 rue Becquerel, F-67087 Strasbourg, France
- Zentrum
für Infektiologie, Parasitologie, Universität Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Saravanan Mounien
- UMR 7509 Centre National de la Recherche Scientifique and University of Strasbourg, European School of Chemistry, Polymers and Materials (ECPM), 25 rue Becquerel, F-67087 Strasbourg, France
| | - Aurelia Geyer
- UMR 7509 Centre National de la Recherche Scientifique and University of Strasbourg, European School of Chemistry, Polymers and Materials (ECPM), 25 rue Becquerel, F-67087 Strasbourg, France
| | - Mourad Elhabiri
- UMR 7509 Centre National de la Recherche Scientifique and University of Strasbourg, European School of Chemistry, Polymers and Materials (ECPM), 25 rue Becquerel, F-67087 Strasbourg, France
| | - Michael Lanzer
- Zentrum
für Infektiologie, Parasitologie, Universität Heidelberg, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Elisabeth Davioud-Charvet
- UMR 7509 Centre National de la Recherche Scientifique and University of Strasbourg, European School of Chemistry, Polymers and Materials (ECPM), 25 rue Becquerel, F-67087 Strasbourg, France
| |
Collapse
|
27
|
Hapuarachchi SV, Cobbold SA, Shafik SH, Dennis ASM, McConville MJ, Martin RE, Kirk K, Lehane AM. The Malaria Parasite's Lactate Transporter PfFNT Is the Target of Antiplasmodial Compounds Identified in Whole Cell Phenotypic Screens. PLoS Pathog 2017; 13:e1006180. [PMID: 28178359 PMCID: PMC5298231 DOI: 10.1371/journal.ppat.1006180] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2016] [Accepted: 01/11/2017] [Indexed: 11/19/2022] Open
Abstract
In this study the ‘Malaria Box’ chemical library comprising 400 compounds with antiplasmodial activity was screened for compounds that perturb the internal pH of the malaria parasite, Plasmodium falciparum. Fifteen compounds induced an acidification of the parasite cytosol. Two of these did so by inhibiting the parasite’s formate nitrite transporter (PfFNT), which mediates the H+-coupled efflux from the parasite of lactate generated by glycolysis. Both compounds were shown to inhibit lactate transport across the parasite plasma membrane, and the transport of lactate by PfFNT expressed in Xenopus laevis oocytes. PfFNT inhibition caused accumulation of lactate in parasitised erythrocytes, and swelling of both the parasite and parasitised erythrocyte. Long-term exposure of parasites to one of the inhibitors gave rise to resistant parasites with a mutant form of PfFNT that showed reduced inhibitor sensitivity. This study provides the first evidence that PfFNT is a druggable antimalarial target. The emergence and spread of Plasmodium falciparum strains resistant to leading antimalarial drugs has intensified the need to discover and develop drugs that kill the parasite via new mechanisms. Here we screened compounds that are known to inhibit P. falciparum growth for their effects on the pH inside the parasite. We identified fifteen compounds that decrease the pH inside the parasite, and determined the mechanism by which two of these, MMV007839 and MMV000972, disrupt pH and kill the parasite. The two compounds were found to inhibit the P. falciparum formate nitrite transporter (PfFNT), a transport protein that is located on the parasite surface and that serves to remove the waste product lactic acid from the parasite. The compounds inhibited both the H+-coupled transport of lactate across the parasite plasma membrane and the transport of lactate by PfFNT expressed in Xenopus oocytes. In addition to disrupting pH, PfFNT inhibition led to a build-up of lactate in the parasite-infected red blood cell and the swelling of both the parasite and the infected red blood cell. Exposing parasites to MMV007839 over a prolonged time period gave rise to resistant parasites with a mutant form of PfFNT that was less sensitive to the compound. This study validates PfFNT as a novel antimalarial drug target.
Collapse
Affiliation(s)
| | - Simon A Cobbold
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, VIC, Australia
| | - Sarah H Shafik
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Adelaide S M Dennis
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Malcolm J McConville
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Melbourne, VIC, Australia
| | - Rowena E Martin
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Kiaran Kirk
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| | - Adele M Lehane
- Research School of Biology, Australian National University, Canberra, ACT, Australia
| |
Collapse
|
28
|
Goodman CD, Austarheim I, Mollard V, Mikolo B, Malterud KE, McFadden GI, Wangensteen H. Natural products from Zanthoxylum heitzii with potent activity against the malaria parasite. Malar J 2016; 15:481. [PMID: 27649682 PMCID: PMC5029023 DOI: 10.1186/s12936-016-1533-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2016] [Accepted: 09/09/2016] [Indexed: 01/07/2023] Open
Abstract
Background Zanthoxylum heitzii (Rutaceae) (olon) is used in traditional medicine in Central and West Africa to treat malaria. To identify novel compounds with anti-parasitic activity and validate medicinal usage, extracts and compounds isolated from this tree were tested against the erythrocytic stages of the human malaria parasite Plasmodium falciparum and for inhibition of transmission in rodent malaria parasite Plasmodium berghei. Results Hexane bark extract showed activity against P. falciparum (IC50 0.050 μg/ml), while leaf and seed extracts were inactive. Fractionation of the hexane bark extract led to the identification of three active constituents; dihydronitidine, pellitories and heitziquinone. Dihydronitidine was the most active compound with an IC50 value of 0.0089 µg/ml (25 nM). This compound was slow acting, requiring 50 % longer exposure time than standard anti-malarials to reach full efficacy. Heitziquinone and pellitorine were less potent, with IC50 values of 3.55 μg/ml and 1.96 µg/ml, but were fast-acting. Plasmodium berghei ookinete conversion was also inhibited by the hexane extract (IC50 1.75 µg/ml), dihydronitidine (0.59 µg/ml) and heitziquinone (6.2 µg/ml). Water extracts of Z. heitzii bark contain only low levels of dihydronitidine and show modest anti-parasitic activity. Conclusions Three compounds with anti-parasitic activity were identified in Z. heitzii bark extract. The alkaloid dihydronitidine is the most effective of these, accounting for the bulk of activity in both erythrocytic and transmission-blocking assays. These compounds may present good leads for development of novel anti-malarials and add to the understanding of the chemical basis of the anti-parasitic activity in these classes of natural product. Electronic supplementary material The online version of this article (doi:10.1186/s12936-016-1533-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | - Ingvild Austarheim
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Oslo, P. O. Box 1068, Blindern, 0316, Oslo, Norway
| | - Vanessa Mollard
- School of BioSciences, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Bertin Mikolo
- National Polytechnic High School, Marien Ngouabi University, BP 69, Brazzaville, Republic of Congo
| | - Karl Egil Malterud
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Oslo, P. O. Box 1068, Blindern, 0316, Oslo, Norway
| | - Geoffrey I McFadden
- School of BioSciences, University of Melbourne, Parkville, VIC, 3010, Australia
| | - Helle Wangensteen
- Department of Pharmaceutical Chemistry, School of Pharmacy, University of Oslo, P. O. Box 1068, Blindern, 0316, Oslo, Norway.
| |
Collapse
|
29
|
Richards SN, Nash MN, Baker ES, Webster MW, Lehane AM, Shafik SH, Martin RE. Molecular Mechanisms for Drug Hypersensitivity Induced by the Malaria Parasite's Chloroquine Resistance Transporter. PLoS Pathog 2016; 12:e1005725. [PMID: 27441371 PMCID: PMC4956231 DOI: 10.1371/journal.ppat.1005725] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2016] [Accepted: 06/03/2016] [Indexed: 01/23/2023] Open
Abstract
Mutations in the Plasmodium falciparum ‘chloroquine resistance transporter’ (PfCRT) confer resistance to chloroquine (CQ) and related antimalarials by enabling the protein to transport these drugs away from their targets within the parasite’s digestive vacuole (DV). However, CQ resistance-conferring isoforms of PfCRT (PfCRTCQR) also render the parasite hypersensitive to a subset of structurally-diverse pharmacons. Moreover, mutations in PfCRTCQR that suppress the parasite’s hypersensitivity to these molecules simultaneously reinstate its sensitivity to CQ and related drugs. We sought to understand these phenomena by characterizing the functions of PfCRTCQR isoforms that cause the parasite to become hypersensitive to the antimalarial quinine or the antiviral amantadine. We achieved this by measuring the abilities of these proteins to transport CQ, quinine, and amantadine when expressed in Xenopus oocytes and complemented this work with assays that detect the drug transport activity of PfCRT in its native environment within the parasite. Here we describe two mechanistic explanations for PfCRT-induced drug hypersensitivity. First, we show that quinine, which normally accumulates inside the DV and therewithin exerts its antimalarial effect, binds extremely tightly to the substrate-binding site of certain isoforms of PfCRTCQR. By doing so it likely blocks the normal physiological function of the protein, which is essential for the parasite’s survival, and the drug thereby gains an additional killing effect. In the second scenario, we show that although amantadine also sequesters within the DV, the parasite’s hypersensitivity to this drug arises from the PfCRTCQR-mediated transport of amantadine from the DV into the cytosol, where it can better access its antimalarial target. In both cases, the mutations that suppress hypersensitivity also abrogate the ability of PfCRTCQR to transport CQ, thus explaining why rescue from hypersensitivity restores the parasite’s sensitivity to this antimalarial. These insights provide a foundation for understanding clinically-relevant observations of inverse drug susceptibilities in the malaria parasite. In acquiring resistance to one drug, many pathogens and cancer cells become hypersensitive to other drugs. This phenomenon could be exploited to combat existing drug resistance and to delay the emergence of resistance to new drugs. However, much remains to be understood about the mechanisms that underlie drug hypersensitivity in otherwise drug-resistant microbes. Here, we describe two mechanisms by which the Plasmodium falciparum ‘chloroquine resistance transporter’ (PfCRT) causes the malaria parasite to become hypersensitive to structurally-diverse drugs. First, we show that an antimalarial drug that normally exerts its killing effect within the parasite’s digestive vacuole is also able to bind extremely tightly to certain forms of PfCRT. This activity will block the natural, essential function of the protein and thereby provide the drug with an additional killing effect. The second mechanism arises when a cytosolic-acting drug that normally sequesters within the digestive vacuole is leaked back into the cytosol via PfCRT. In both cases, mutations that suppress hypersensitivity also abrogate the ability of PfCRT to transport chloroquine, thus explaining why rescue from hypersensitivity restores the parasite’s sensitivity to this antimalarial. These insights provide a foundation for understanding and exploiting the hypersensitivity of chloroquine-resistant parasites to several of the current antimalarials.
Collapse
Affiliation(s)
- Sashika N. Richards
- Research School of Biology, Australian National University, Canberra, Australia
| | - Megan N. Nash
- Research School of Biology, Australian National University, Canberra, Australia
| | - Eileen S. Baker
- Research School of Biology, Australian National University, Canberra, Australia
| | - Michael W. Webster
- Research School of Biology, Australian National University, Canberra, Australia
| | - Adele M. Lehane
- Research School of Biology, Australian National University, Canberra, Australia
| | - Sarah H. Shafik
- Research School of Biology, Australian National University, Canberra, Australia
| | - Rowena E. Martin
- Research School of Biology, Australian National University, Canberra, Australia
- * E-mail:
| |
Collapse
|
30
|
Abstract
Some hours after invading the erythrocytes of its human host, the malaria parasite Plasmodium falciparum induces an increase in the permeability of the erythrocyte membrane to monovalent ions. The resulting net influx of Na(+) and net efflux of K(+), down their respective concentration gradients, converts the erythrocyte cytosol from an initially high-K(+), low-Na(+) solution to a high-Na(+), low-K(+) solution. The intraerythrocytic parasite itself exerts tight control over its internal Na(+), K(+), Cl(-), and Ca(2+) concentrations and its intracellular pH through the combined actions of a range of membrane transport proteins. The molecular mechanisms underpinning ion regulation in the parasite are receiving increasing attention, not least because PfATP4, a P-type ATPase postulated to be involved in Na(+) regulation, has emerged as a potential antimalarial drug target, susceptible to inhibition by a wide range of chemically unrelated compounds.
Collapse
Affiliation(s)
- Kiaran Kirk
- Research School of Biology, The Australian National University, Canberra, ACT 2601, Australia;
| |
Collapse
|
31
|
Šegan S, Opsenica I, Zlatović M, Milojković-Opsenica D, Šolaja B. Quantitative structure retention/activity relationships of biologically relevant 4-amino-7-chloroquinoline based compounds. J Chromatogr B Analyt Technol Biomed Life Sci 2016; 1012-1013:144-52. [DOI: 10.1016/j.jchromb.2016.01.033] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Revised: 01/15/2016] [Accepted: 01/19/2016] [Indexed: 12/31/2022]
|
32
|
van Schalkwyk DA, Nash MN, Shafik SH, Summers RL, Lehane AM, Smith PJ, Martin RE. Verapamil-Sensitive Transport of Quinacrine and Methylene Blue via the Plasmodium falciparum Chloroquine Resistance Transporter Reduces the Parasite's Susceptibility to these Tricyclic Drugs. J Infect Dis 2015; 213:800-10. [PMID: 26503982 DOI: 10.1093/infdis/jiv509] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2015] [Accepted: 10/15/2015] [Indexed: 12/16/2022] Open
Abstract
BACKGROUND It is becoming increasingly apparent that certain mutations in the Plasmodium falciparum chloroquine resistance transporter (PfCRT) alter the parasite's susceptibility to diverse compounds. Here we investigated the interaction of PfCRT with 3 tricyclic compounds that have been used to treat malaria (quinacrine [QC] and methylene blue [MB]) or to study P. falciparum (acridine orange [AO]). METHODS We measured the antiplasmodial activities of QC, MB, and AO against chloroquine-resistant and chloroquine-sensitive P. falciparum and determined whether QC and AO affect the accumulation and activity of chloroquine in these parasites. We also assessed the ability of mutant (PfCRT(Dd2)) and wild-type (PfCRT(D10)) variants of the protein to transport QC, MB, and AO when expressed at the surface of Xenopus laevis oocytes. RESULTS Chloroquine resistance-conferring isoforms of PfCRT reduced the susceptibility of the parasite to QC, MB, and AO. In chloroquine-resistant (but not chloroquine-sensitive) parasites, AO and QC increased the parasite's accumulation of, and susceptibility to, chloroquine. All 3 compounds were shown to bind to PfCRT(Dd2), and the transport of QC and MB via this protein was saturable and inhibited by the chloroquine resistance-reverser verapamil. CONCLUSIONS Our findings reveal that the PfCRT(Dd2)-mediated transport of tricyclic antimalarials reduces the parasite's susceptibility to these drugs.
Collapse
Affiliation(s)
| | - Megan N Nash
- Research School of Biology, Australian National University, Canberra, Australia
| | - Sarah H Shafik
- Research School of Biology, Australian National University, Canberra, Australia
| | - Robert L Summers
- Research School of Biology, Australian National University, Canberra, Australia
| | - Adele M Lehane
- Research School of Biology, Australian National University, Canberra, Australia
| | - Peter J Smith
- Division of Pharmacology, Department of Medicine, University of Cape Town, Rondebosch, South Africa
| | - Rowena E Martin
- Research School of Biology, Australian National University, Canberra, Australia
| |
Collapse
|
33
|
Fong KY, Sandlin RD, Wright DW. Identification of β-hematin inhibitors in the MMV Malaria Box. INTERNATIONAL JOURNAL FOR PARASITOLOGY-DRUGS AND DRUG RESISTANCE 2015; 5:84-91. [PMID: 26150923 PMCID: PMC4486462 DOI: 10.1016/j.ijpddr.2015.05.003] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/18/2015] [Revised: 05/15/2015] [Accepted: 05/19/2015] [Indexed: 11/22/2022]
Abstract
The Malaria Box, assembled by the Medicines for Malaria Venture, is a set of 400 structurally diverse, commercially available compounds with demonstrated activity against blood-stage Plasmodium falciparum. The compounds are a representative subset of the 20,000 in vitro antimalarials identified from the high-throughput screening efforts of St. Jude Children's Research Hospital (TN, USA), Novartis and GlaxoSmithKline. In addition, a small set of active compounds from commercially available libraries was added to this group, but it has not previously been published. Elucidation of the biochemical pathways on which these compounds act is a major challenge; therefore, access to these compounds has been made available free of charge to the investigator community. Here, the Malaria Box compounds were tested for activity against the formation of β-hematin, a synthetic form of the heme detoxification biomineral, hemozoin. Further, the mechanism of action of these compounds within the malaria parasite was explored. Ten of the Malaria Box compounds demonstrated significant inhibition of β-hematin formation. In this assay, dose–response data revealed IC50 values ranging from 8.7 to 22.7 μM for these hits, each of which is more potent than chloroquine (a known inhibitor of hemozoin formation). The in vitro antimalarial activity of these ten hits was confirmed in cultures of the chloroquine sensitive D6 strain of the parasite resulting in IC50 values of 135–2165 nM, followed by testing in the multidrug resistant strain, C235. Cultures of P. falciparum (D6) were then examined for their heme distribution following treatment with nine of the commercially available confirmed compounds, seven of which disrupted the hemozoin pathway. Ten of 400 Malaria Box compounds were found to be potent β-hematin inhibitors. We confirmed similar in vitro antimalarial activity to results from previous screens. 7 of the 9 commercially available hits were validated hemozoin inhibitors in culture.
Collapse
Affiliation(s)
- Kim Y Fong
- Department of Chemistry, Vanderbilt University, Station B 351822, Nashville, TN 37235, USA
| | - Rebecca D Sandlin
- Department of Chemistry, Vanderbilt University, Station B 351822, Nashville, TN 37235, USA
| | - David W Wright
- Department of Chemistry, Vanderbilt University, Station B 351822, Nashville, TN 37235, USA
| |
Collapse
|
34
|
Siwo GH, Tan A, Button-Simons KA, Samarakoon U, Checkley LA, Pinapati RS, Ferdig MT. Predicting functional and regulatory divergence of a drug resistance transporter gene in the human malaria parasite. BMC Genomics 2015; 16:115. [PMID: 25765049 PMCID: PMC4352545 DOI: 10.1186/s12864-015-1261-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2014] [Accepted: 01/22/2015] [Indexed: 12/05/2022] Open
Abstract
Background The paradigm of resistance evolution to chemotherapeutic agents is that a key coding mutation in a specific gene drives resistance to a particular drug. In the case of resistance to the anti-malarial drug chloroquine (CQ), a specific mutation in the transporter pfcrt is associated with resistance. Here, we apply a series of analytical steps to gene expression data from our lab and leverage 3 independent datasets to identify pfcrt-interacting genes. Resulting networks provide insights into pfcrt’s biological functions and regulation, as well as the divergent phenotypic effects of its allelic variants in different genetic backgrounds. Results To identify pfcrt-interacting genes, we analyze pfcrt co-expression networks in 2 phenotypic states - CQ-resistant (CQR) and CQ-sensitive (CQS) recombinant progeny clones - using a computational approach that prioritizes gene interactions into functional and regulatory relationships. For both phenotypic states, pfcrt co-expressed gene sets are associated with hemoglobin metabolism, consistent with CQ’s expected mode of action. To predict the drivers of co-expression divergence, we integrate topological relationships in the co-expression networks with available high confidence protein-protein interaction data. This analysis identifies 3 transcriptional regulators from the ApiAP2 family and histone acetylation as potential mediators of these divergences. We validate the predicted divergences in DNA mismatch repair and histone acetylation by measuring the effects of small molecule inhibitors in recombinant progeny clones combined with quantitative trait locus (QTL) mapping. Conclusions This work demonstrates the utility of differential co-expression viewed in a network framework to uncover functional and regulatory divergence in phenotypically distinct parasites. pfcrt-associated co-expression in the CQ resistant progeny highlights CQR-specific gene relationships and possible targeted intervention strategies. The approaches outlined here can be readily generalized to other parasite populations and drug resistances. Electronic supplementary material The online version of this article (doi:10.1186/s12864-015-1261-6) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Geoffrey H Siwo
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN, USA. .,Geisel School of Medicine, Dartmouth College, Hanover, NH, USA.
| | - Asako Tan
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN, USA. .,Epicentre, Madison, WI, USA.
| | - Katrina A Button-Simons
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN, USA.
| | - Upeka Samarakoon
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN, USA. .,Harvard Medical School, Boston, MA, USA.
| | - Lisa A Checkley
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN, USA.
| | - Richard S Pinapati
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN, USA.
| | - Michael T Ferdig
- Department of Biological Sciences, Eck Institute for Global Health, University of Notre Dame, Notre Dame, IN, USA.
| |
Collapse
|
35
|
Vekilov PG, Rimer JD, Olafson KN, Ketchum MA. Lipid or aqueous medium for hematin crystallization? CrystEngComm 2015. [DOI: 10.1039/c5ce01178g] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Hematin crystallization, the primary heme detoxification mechanism of malaria parasites infecting human erythrocytes, most likely requires the participation of lipid structures.
Collapse
Affiliation(s)
- Peter G. Vekilov
- Department of Chemical and Biomolecular Engineering
- University of Houston
- Houston, USA
- Department of Chemistry
- University of Houston
| | - Jeffrey D. Rimer
- Department of Chemical and Biomolecular Engineering
- University of Houston
- Houston, USA
| | - Katy N. Olafson
- Department of Chemical and Biomolecular Engineering
- University of Houston
- Houston, USA
| | - Megan A. Ketchum
- Department of Chemical and Biomolecular Engineering
- University of Houston
- Houston, USA
| |
Collapse
|
36
|
An atomic scale mechanism for the antimalarial action of chloroquine from density functional theory calculations. TRANSIT METAL CHEM 2014. [DOI: 10.1007/s11243-014-9868-z] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
37
|
Abo M, Minakami R, Miyano K, Kamiya M, Nagano T, Urano Y, Sumimoto H. Visualization of phagosomal hydrogen peroxide production by a novel fluorescent probe that is localized via SNAP-tag labeling. Anal Chem 2014; 86:5983-90. [PMID: 24862209 DOI: 10.1021/ac501041w] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Hydrogen peroxide (H2O2), a member of reactive oxygen species (ROS), plays diverse physiological roles including host defense and cellular signal transduction. During ingestion of invading microorganisms, professional phagocytes such as macrophages release H2O2 specifically into the phagosome to direct toxic ROS toward engulfed microbes. Although H2O2 is considered to exert discrete effects in living systems depending on location of its production, accumulation, and consumption, there have been limitations of techniques for probing this oxygen metabolite with high molecular specificity at the subcellular resolution. Here we describe the development of an O(6)-benzylguanine derivative of 5-(4-nitrobenzoyl)carbonylfluorescein (NBzF-BG), a novel H2O2-specific fluorescent probe; NBzF-BG is covalently and selectively conjugated with the SNAP-tag protein, leading to formation of the fluorophore-protein conjugate (SNAP-NBzF). SNAP-NBzF rapidly reacts with H2O2 and thereby shows a 9-fold enhancement in fluorescence. When SNAP-tag is expressed in HEK293T cells and RAW264.7 macrophages as a protein C-terminally fused to the transmembrane domain of platelet-derived growth factor receptor (PDGFR), the tag is presented on the outside of the plasma membrane; conjugation of NBzF-BG with the cell surface SNAP-tag enables detection of H2O2 added exogenously. We also demonstrate molecular imaging of H2O2 that is endogenously produced in phagosomes of macrophages ingesting IgG-coated latex beads. Thus, NBzF-BG, combined with the SNAP-tag technology, should be useful as a tool to measure local production of H2O2 in living cells.
Collapse
Affiliation(s)
- Masahiro Abo
- Departments of Biochemistry and ‡Health Sciences, Kyushu University Graduate School of Medical Sciences , Fukuoka 812-8582, Japan
| | | | | | | | | | | | | |
Collapse
|
38
|
Olafson KN, Rimer JD, Vekilov PG. Growth of Large Hematin Crystals in Biomimetic Solutions. CRYSTAL GROWTH & DESIGN 2014; 14:2123-2127. [PMID: 24839403 PMCID: PMC4018177 DOI: 10.1021/cg5002682] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/23/2014] [Revised: 04/07/2014] [Indexed: 06/03/2023]
Abstract
Hematin crystallization is an essential component of the physiology of malaria parasites. Several antimalarial drugs are believed to inhibit crystallization and expose the parasites to toxic soluble hematin. Hence, understanding the mechanisms of hematin crystal growth and inhibition is crucial for the design of new drugs. A major obstacle to microscopic, spectroscopic, and crystallographic studies of hematin crystallization has been the unavailability of large hematin crystals grown under conditions representative of the parasite anatomy. We have developed a biomimetic method to reproducibly grow large hematin crystals reaching 50 μm in length. We imitate the digestive vacuole of Plasmodium falciparum and employ a two-phase solution of octanol and citric buffer. The nucleation of seeds is enhanced at the interface between the aqueous and organic phases, where an ordered layer of octanol molecules is known to serve as substrate for nucleation. The seeds are transferred to hematin-saturated octanol in contact with citric buffer. We show that the crystals grow in the octanol layer, while the buffer supplies hydrogen ions needed for bonds that link the hematin molecules in the crystal. The availability of large hematin crystals opens new avenues for studies of hematin detoxification of malaria parasites in host erythrocytes.
Collapse
Affiliation(s)
- Katy N. Olafson
- Department
of Chemical and Biomolecular Engineering,
and Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Jeffrey D. Rimer
- Department
of Chemical and Biomolecular Engineering,
and Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| | - Peter G. Vekilov
- Department
of Chemical and Biomolecular Engineering,
and Department of Chemistry, University of Houston, Houston, Texas 77204, United States
| |
Collapse
|
39
|
Diverse mutational pathways converge on saturable chloroquine transport via the malaria parasite's chloroquine resistance transporter. Proc Natl Acad Sci U S A 2014; 111:E1759-67. [PMID: 24728833 DOI: 10.1073/pnas.1322965111] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Mutations in the chloroquine resistance transporter (PfCRT) are the primary determinant of chloroquine (CQ) resistance in the malaria parasite Plasmodium falciparum. A number of distinct PfCRT haplotypes, containing between 4 and 10 mutations, have given rise to CQ resistance in different parts of the world. Here we present a detailed molecular analysis of the number of mutations (and the order of addition) required to confer CQ transport activity upon the PfCRT as well as a kinetic characterization of diverse forms of PfCRT. We measured the ability of more than 100 variants of PfCRT to transport CQ when expressed at the surface of Xenopus laevis oocytes. Multiple mutational pathways led to saturable CQ transport via PfCRT, but these could be separated into two main lineages. Moreover, the attainment of full activity followed a rigid process in which mutations had to be added in a specific order to avoid reductions in CQ transport activity. A minimum of two mutations sufficed for (low) CQ transport activity, and as few as four conferred full activity. The finding that diverse PfCRT variants are all limited in their capacity to transport CQ suggests that resistance could be overcome by reoptimizing the CQ dosage.
Collapse
|
40
|
Abstract
As it grows and replicates within the erythrocytes of its host the malaria parasite takes up nutrients from the extracellular medium, exports metabolites and maintains a tight control over its internal ionic composition. These functions are achieved via membrane transport proteins, integral membrane proteins that mediate the passage of solutes across the various membranes that separate the biochemical machinery of the parasite from the extracellular environment. Proteins of this type play a key role in antimalarial drug resistance, as well as being candidate drug targets in their own right. This review provides an overview of recent work on the membrane transport biology of the malaria parasite-infected erythrocyte, encompassing both the parasite-induced changes in the membrane transport properties of the host erythrocyte and the cell physiology of the intracellular parasite itself.
Collapse
|
41
|
Han Y, Shchukin D, Schneider J, Möhwald H. Fluorescence indicative pH drop in sonication. Colloids Surf A Physicochem Eng Asp 2014. [DOI: 10.1016/j.colsurfa.2013.12.061] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
|
42
|
Kuter D, Benjamin SJ, Egan TJ. Multiple spectroscopic and magnetic techniques show that chloroquine induces formation of the μ-oxo dimer of ferriprotoporphyrin IX. J Inorg Biochem 2014; 133:40-9. [PMID: 24480793 DOI: 10.1016/j.jinorgbio.2014.01.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2013] [Revised: 01/03/2014] [Accepted: 01/07/2014] [Indexed: 11/28/2022]
Abstract
Interaction of the antimalarial chloroquine (CQ) with ferriprotoporphyrin IX, Fe(III)PPIX, was investigated in aqueous solution (pH7.4) and as a precipitate from aqueous medium at pH5.0. In solution, spectrophotometric titrations indicated strong association (logKobs 13.3±0.2) and a Job plot gave a stoichiometry of 1:2 CQ:Fe(III)PPIX. UV-visible absorbance and magnetic circular dichroism spectra of the complex were compared to various Fe(III)PPIX species. Close similarity to the spectra of the μ-oxo dimer, μ-[Fe(III)PPIX]2O, was revealed. The induction of this species by CQ was confirmed by magnetic susceptibility measurements using the Evans NMR method. The observed low-magnetic moment (2.25±0.02 μB) could only be attributed to antiferromagnetically coupled Fe(III) centers. The value was comparable to that of μ-[Fe(III)PPIX]2O (2.0±0.1 μB). In the solid-state, mass spectrometry confirmed the presence of CQ in the complex. Dissolution of this solid in aqueous solution (pH7.4) resulted in a solution with a UV-visible spectrum consistent with the same 1:2 stoichiometry observed in the Job plot. Magnetic susceptibility measurements made on the solid using an Evans balance produced a magnetic moment (2.3±0.1 μB) consistent with that in solution. Diffusion coefficients of CQ and its complex with Fe(III)PPIX were measured in aqueous solution (3.3±0.3 and 0.6±0.2×10(-10) m(2)·s(-1), respectively). The latter was used in conjunction with an empirical relationship between diffusion coefficient and molar volume to estimate the degree of aggregation. The findings suggest the formation of a 2:4 CQ:Fe(III)PPIX complex in aqueous solution at pH7.4.
Collapse
Affiliation(s)
- David Kuter
- Department of Chemistry, University of Cape Town, Private Bag, Rondebosch 7701, South Africa
| | - Stefan J Benjamin
- Department of Chemistry, University of Cape Town, Private Bag, Rondebosch 7701, South Africa
| | - Timothy J Egan
- Department of Chemistry, University of Cape Town, Private Bag, Rondebosch 7701, South Africa.
| |
Collapse
|
43
|
Egan TJ, Kuter D. Dual-functioning antimalarials that inhibit the chloroquine-resistance transporter. Future Microbiol 2013; 8:475-89. [PMID: 23534360 PMCID: PMC7099626 DOI: 10.2217/fmb.13.18] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Malaria remains a major international health challenge. Resistance to a number of existing drugs and evidence of the emergence of artemisinin resistance has emphasized the need for new antimalarials. A new approach has been the preparation of dual-function compounds that include a chloroquine-like antimalarial group and a group that resembles a chloroquine chemosensitizer. This article reviews the recent discovery of such dual-function antimalarials that are proposed to target both hemozoin formation and the chloroquine resistance transporter, PfCRT. These are discussed in relation to the mechanism of action of 4-aminoquinolines, chloroquine resistance and resistance reversal.
Collapse
Affiliation(s)
- Timothy J Egan
- Department of Chemistry, University of Cape Town, Private Bag, Rondebosch 7701, South Africa.
| | | |
Collapse
|
44
|
Wolfe DM, Lee JH, Kumar A, Lee S, Orenstein SJ, Nixon RA. Autophagy failure in Alzheimer's disease and the role of defective lysosomal acidification. Eur J Neurosci 2013; 37:1949-61. [PMID: 23773064 PMCID: PMC3694736 DOI: 10.1111/ejn.12169] [Citation(s) in RCA: 260] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 01/22/2013] [Accepted: 01/28/2013] [Indexed: 12/12/2022]
Abstract
Autophagy is a lysosomal degradative process which recycles cellular waste and eliminates potentially toxic damaged organelles and protein aggregates. The important cytoprotective functions of autophagy are demonstrated by the diverse pathogenic consequences that may stem from autophagy dysregulation in a growing number of neurodegenerative disorders. In many of the diseases associated with autophagy anomalies, it is the final stage of autophagy-lysosomal degradation that is disrupted. In several disorders, including Alzheimer's disease (AD), defective lysosomal acidification contributes to this proteolytic failure. The complex regulation of lysosomal pH makes this process vulnerable to disruption by many factors, and reliable lysosomal pH measurements have become increasingly important in investigations of disease mechanisms. Although various reagents for pH quantification have been developed over several decades, they are not all equally well suited for measuring the pH of lysosomes. Here, we evaluate the most commonly used pH probes for sensitivity and localisation, and identify LysoSensor yellow/blue-dextran, among currently used probes, as having the optimal profile of properties for measuring lysosomal pH. In addition, we review evidence that lysosomal acidification is defective in AD and extend our original findings, of elevated lysosomal pH in presenilin 1 (PS1)-deficient blastocysts and neurons, to additional cell models of PS1 and PS1/2 deficiency, to fibroblasts from AD patients with PS1 mutations, and to neurons in the PS/APP mouse model of AD.
Collapse
Affiliation(s)
- Devin M. Wolfe
- Center for Dementia Research, Nathan S. Kline Institute, 140 Old Orangeburg Road, Orangeburg, NY, USA, 10962
| | - Ju-hyun Lee
- Center for Dementia Research, Nathan S. Kline Institute, 140 Old Orangeburg Road, Orangeburg, NY, USA, 10962
- Department of Psychiatry, New York University, 550 First Ave, New York, NY, USA 10016
| | - Asok Kumar
- Center for Dementia Research, Nathan S. Kline Institute, 140 Old Orangeburg Road, Orangeburg, NY, USA, 10962
- Department of Pathology, New York University, 550 First Ave, New York, NY, USA 10016
| | - Sooyeon Lee
- Center for Dementia Research, Nathan S. Kline Institute, 140 Old Orangeburg Road, Orangeburg, NY, USA, 10962
| | - Samantha J. Orenstein
- Department of Developmental and Molecular Biology, Marion Bessin Liver Research Center, Albert Einstein College of Medicine, 1300 Morris Park Ave, Bronx, NY, USA 10461
| | - Ralph A. Nixon
- Center for Dementia Research, Nathan S. Kline Institute, 140 Old Orangeburg Road, Orangeburg, NY, USA, 10962
- Department of Psychiatry, New York University, 550 First Ave, New York, NY, USA 10016
- Department of Cell Biology, New York University, 550 First Ave, New York, NY, USA 10016
| |
Collapse
|
45
|
van Schalkwyk DA, Saliba KJ, Biagini GA, Bray PG, Kirk K. Loss of pH control in Plasmodium falciparum parasites subjected to oxidative stress. PLoS One 2013; 8:e58933. [PMID: 23536836 PMCID: PMC3594203 DOI: 10.1371/journal.pone.0058933] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2012] [Accepted: 02/08/2013] [Indexed: 11/29/2022] Open
Abstract
The intraerythrocytic malaria parasite is susceptible to oxidative stress and this may play a role in the mechanism of action of some antimalarial agents. Here we show that exposure of the intraerythrocytic malaria parasite to the oxidising agent hydrogen peroxide results in a fall in the intracellular ATP level and inhibition of the parasite's V-type H+-ATPase, causing a loss of pH control in both the parasite cytosol and the internal digestive vacuole. In contrast to the V-type H+-ATPase, the parasite's digestive vacuole H+-pyrophosphatase is insensitive to hydrogen peroxide-induced oxidative stress. This work provides insights into the effects of oxidative stress on the intraerythrocytic parasite, as well as providing an alternative possible explanation for a previous report that light-induced oxidative stress causes selective lysis of the parasite's digestive vacuole.
Collapse
Affiliation(s)
- Donelly A van Schalkwyk
- Research School of Biology, The Australian National University, Canberra, Australian Capital Territory, Australia
| | | | | | | | | |
Collapse
|
46
|
Affiliation(s)
- Paloma F. Salas
- Medicinal Inorganic Chemistry
Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia
V6T 1Z1, Canada
| | - Christoph Herrmann
- Medicinal Inorganic Chemistry
Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia
V6T 1Z1, Canada
- Advanced
Applied Physics Solutions, TRIUMF, 4004
Wesbrook Mall, Vancouver, British Columbia
V6T 2A3, Canada
| | - Chris Orvig
- Medicinal Inorganic Chemistry
Group, Department of Chemistry, University of British Columbia, 2036 Main Mall, Vancouver, British Columbia
V6T 1Z1, Canada
| |
Collapse
|
47
|
Clark M, Fisher NC, Kasthuri R, Cerami Hand C. Parasite maturation and host serum iron influence the labile iron pool of erythrocyte stage Plasmodium falciparum. Br J Haematol 2013; 161:262-9. [PMID: 23398516 DOI: 10.1111/bjh.12234] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2012] [Accepted: 12/20/2012] [Indexed: 12/30/2022]
Abstract
Iron is a critical and tightly regulated nutrient for both the malaria parasite and its human host. The importance of the relationship between host iron and the parasite has been underscored recently by studies showing that host iron supplementation may increase the risk of falciparum malaria. It is unclear what host iron sources the parasite is able to access. We developed a flow cytometry-based method for measuring the labile iron pool (LIP) of parasitized erythrocytes using the nucleic acid dye STYO 61 and the iron sensitive dye, calcein acetoxymethyl ester (CA-AM). This new approach enabled us to measure the LIP of P. falciparum through the course of its erythrocytic life cycle and in response to the addition of host serum iron sources. We found that the LIP increases as the malaria parasite develops from early ring to late schizont stage, and that the addition of either transferrin or ferric citrate to culture media increases the LIP of trophozoites. Our method for detecting the LIP within malaria parasitized RBCs provides evidence that the parasite is able to access serum iron sources as part of the host vs. parasite arms race for iron.
Collapse
Affiliation(s)
- Martha Clark
- Department of Microbiology and Immunology, University of North Carolina-Chapel Hill, Chapel Hill, USA, United States
| | | | | | | |
Collapse
|
48
|
Griffin CE, Hoke JM, Samarakoon U, Duan J, Mu J, Ferdig MT, Warhurst DC, Cooper RA. Mutation in the Plasmodium falciparum CRT protein determines the stereospecific activity of antimalarial cinchona alkaloids. Antimicrob Agents Chemother 2012; 56:5356-64. [PMID: 22869567 PMCID: PMC3457399 DOI: 10.1128/aac.05667-11] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2011] [Accepted: 07/30/2012] [Indexed: 11/20/2022] Open
Abstract
The Cinchona alkaloids are quinoline aminoalcohols that occur as diastereomer pairs, typified by (-)-quinine and (+)-quinidine. The potency of (+)-isomers is greater than the (-)-isomers in vitro and in vivo against Plasmodium falciparum malaria parasites. They may act by the inhibition of heme crystallization within the parasite digestive vacuole in a manner similar to chloroquine. Earlier studies showed that a K76I mutation in the digestive vacuole-associated protein, PfCRT (P. falciparum chloroquine resistance transporter), reversed the normal potency order of quinine and quinidine toward P. falciparum. To further explore PfCRT-alkaloid interactions in the malaria parasite, we measured the in vitro susceptibility of eight clonal lines of P. falciparum derived from the 106/1 strain, each containing a unique pfcrt allele, to four Cinchona stereoisomer pairs: quinine and quinidine; cinchonidine and cinchonine; hydroquinine and hydroquinidine; 9-epiquinine and 9-epiquinidine. Stereospecific potency of the Cinchona alkaloids was associated with changes in charge and hydrophobicity of mutable PfCRT amino acids. In isogenic chloroquine-resistant lines, the IC(50) ratio of (-)/(+) CA pairs correlated with side chain hydrophobicity of the position 76 residue. Second-site PfCRT mutations negated the K76I stereospecific effects: charge-change mutations C72R or Q352K/R restored potency patterns similar to the parent K76 line, while V369F increased susceptibility to the alkaloids and nullified stereospecific differences between alkaloid pairs. Interactions between key residues of the PfCRT channel/transporter with (-) and (+) alkaloids are stereospecifically determined, suggesting that PfCRT binding plays an important role in the antimalarial activity of quinine and other Cinchona alkaloids.
Collapse
Affiliation(s)
- Carol E. Griffin
- Department of Biological Sciences, Old Dominion University, Norfolk, Virginia, USA
| | - Jonathan M. Hoke
- Department of Biological Sciences, Old Dominion University, Norfolk, Virginia, USA
| | - Upeka Samarakoon
- Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, South Bend, Indiana, USA
| | - Junhui Duan
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Jianbing Mu
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, USA
| | - Michael T. Ferdig
- Eck Institute for Global Health, Department of Biological Sciences, University of Notre Dame, South Bend, Indiana, USA
| | - David C. Warhurst
- Department of Pathogen Molecular Biology, Faculty of Infectious and Tropical Disease, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Roland A. Cooper
- Department of Biological Sciences, Old Dominion University, Norfolk, Virginia, USA
- Department of Natural Sciences and Mathematics, Dominican University of California, San Rafael, California, USA
| |
Collapse
|
49
|
PfCRT and its role in antimalarial drug resistance. Trends Parasitol 2012; 28:504-14. [PMID: 23020971 DOI: 10.1016/j.pt.2012.08.002] [Citation(s) in RCA: 185] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2012] [Revised: 08/09/2012] [Accepted: 08/13/2012] [Indexed: 12/15/2022]
Abstract
Plasmodium falciparum resistance to chloroquine, the former gold standard antimalarial drug, is mediated primarily by mutant forms of the chloroquine resistance transporter (PfCRT). These mutations impart upon PfCRT the ability to efflux chloroquine from the intracellular digestive vacuole, the site of drug action. Recent studies reveal that PfCRT variants can also affect parasite fitness, protect immature gametocytes against chloroquine action, and alter P. falciparum susceptibility to current first-line therapies. These results highlight the need to be vigilant in screening for the appearance of novel pfcrt alleles that could contribute to new multi-drug resistance phenotypes.
Collapse
|
50
|
Orjih AU, Mathew TC, Cherian PT. Erythrocyte membranes convert monomeric ferriprotoporphyrin IX to β-hematin in acidic environment at malarial fever temperature. Exp Biol Med (Maywood) 2012; 237:884-93. [PMID: 22890028 DOI: 10.1258/ebm.2012.012013] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Hemozoin production makes it possible for intraerythrocytic malaria parasites to digest massive quantities of hemoglobin but still avoid potential ferriprotoporphyrin IX (FP) toxicity, which they cannot decompose further. Some antimalarial drugs, such as chloroquine, work by inhibiting this production, forcing the parasite to starve to death. As part of the efforts to identify possible biological mechanisms of FP polymerization, we have used normal human erythrocyte membranes as a model, to promote β-hematin (β-h) synthesis. Hemin in 35% aqueous dimethyl sulfoxide (DMSO) was reacted with isolated erythrocyte membranes and incubated overnight in sodium acetate buffer, pH 4.8, at 41°C. Infrared spectroscopy and electron microscopy showed that β-h was produced. Hemin in 10% was less effective as the substrate than when it was in 35% DMSO. A high malarial temperature seemed to be necessary, because FP polymerization was less at 37°C than at 41°C. Production was partially inhibited by chloroquine. These observations are of interest because other investigators have reported that membrane lipids mediated FP polymerization, but whole membranes were ineffective. On the other hand, our hypothesis is that the transport vesicles (TV) in malaria parasites could provide the receptor for FP and the lipids that promote hemozoin formation. Erythrocyte membranes may not be directly involved, but Plasmodium species transport hemoglobin in membrane-bound TV into food vacuoles, where hemoglobin catabolism is completed and hemozoin crystals are stored.
Collapse
Affiliation(s)
- Augustine U Orjih
- Department of Medical Laboratory Sciences, Kuwait University, Kuwait, Arabian Gulf.
| | | | | |
Collapse
|