1
|
Sun J, Fang C, Qin X, Si W, Wang F, Li Y, Yan X. Hemozoin: a waste product after heme detoxification? Parasit Vectors 2025; 18:83. [PMID: 40038801 PMCID: PMC11881329 DOI: 10.1186/s13071-025-06699-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2024] [Accepted: 01/29/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Hemozoin is considered a waste byproduct of heme detoxification following hemoglobin digestion; consequently, the biological functions of hemozoin in hemozoin-producing organisms have often been overlooked. However, recent findings indicate that Schistosoma hemozoin facilitates the transfer of iron from erythrocytes to eggs through its formation and degradation, thereby increasing interest in the role of malarial hemozoin. METHODS Using transmission electron microscopy, we compared the formation of Schistosoma hemozoin and malaria hemozoin. Through transcriptome analysis of different stages of P. falciparum 3D7WT and P. falciparum 3D7C580Y,- where the latter serves as a control with reduced hemozoin production, -we analyzed expression patterns of genes related to DNA synthesis, iron, and heme utilization. Using light microscopy, we observed hemozoin aggregation following artemether treatment, and macrophage morphology after ingesting hemozoin in vivo and in vitro. RESULTS Similar to Schistosoma hemozoin, malaria hemozoin consists of heme aggregation and a lipid matrix, likely involved in lipid processing and the utilization of heme and iron. Transcriptome analysis reveals that during the trophozoite stage, the expression levels of these genes in P. falciparum 3D7WT and P. falciparum 3D7C580Y are higher than those during the schizont stage. Correspondingly, less hemozoin was detected at the trophozoite stage, while more was observed during the schizont stage. These results suggest that when more heme and iron are utilized, less heme is available for hemozoin formation. Conversely, when less heme and iron are utilized, they can accumulate for hemozoin formation during the schizont stage, likely benefiting lipid remodeling. Disruption of heme utilization and hemozoin aggregation may lead to parasite death. In addition, the hemozoin released by schizonts can impair macrophage functions, potentially protecting merozoites from phagocytosis. Furthermore, it may be carried by gametocytes into the next host, fulfilling their requirements for iron and heme during their development in mosquitoes. CONCLUSIONS Hemozoin is not a waste byproduct of heme detoxification but instead plays a crucial role in the parasite's life cycle.
Collapse
Affiliation(s)
- Jun Sun
- School of Medicine, Tongji University, 500 Zhennan Road, Shanghai, 200331, People's Republic of China.
| | - Chuantao Fang
- School of Medicine, Tongji University, 500 Zhennan Road, Shanghai, 200331, People's Republic of China
- Shanghai Tenth People's Hospital, Tenth peoples hospital of Tongji university, Shanghai, People's Republic of China, Shanghai, China
| | - Xixi Qin
- School of Medicine, Tongji University, 500 Zhennan Road, Shanghai, 200331, People's Republic of China
| | - Wenwen Si
- School of Medicine, Tongji University, 500 Zhennan Road, Shanghai, 200331, People's Republic of China
| | - Fei Wang
- School of Medicine, Tongji University, 500 Zhennan Road, Shanghai, 200331, People's Republic of China
| | - Yanna Li
- School of Medicine, Tongji University, 500 Zhennan Road, Shanghai, 200331, People's Republic of China
| | - Xiaoli Yan
- School of Medicine, Tongji University, 500 Zhennan Road, Shanghai, 200331, People's Republic of China
| |
Collapse
|
2
|
Adeoye AO, Lobb KA. Malaria parasite cysteine and aspartic proteases as key drug targets for antimalarial therapy. J Mol Model 2025; 31:78. [PMID: 39920505 DOI: 10.1007/s00894-025-06303-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2024] [Accepted: 01/27/2025] [Indexed: 02/09/2025]
Abstract
CONTEXT Cysteine and aspartic proteases are enzyme families that play crucial roles in the life cycle of Plasmodium, the parasite responsible for malaria. These proteases are involved in vital biological processes, such as hemoglobin degradation within the host's red blood cells, protein turnover, and regulation of parasite development. Inhibiting these proteases with small molecule drugs can block the parasite's growth and survival. Chemically, these enzymes have specific active sites where inhibitors can bind, preventing the breakdown of key proteins, making them attractive targets for the design of novel antimalarial compounds. Understanding the structure and catalytic mechanisms of these proteases is critical for developing selective and potent inhibitors. The degradation of hemoglobin occurs in the parasite's digestive vacuole, and disruption of this process by targeting these proteases can inhibit parasite development, leading to the death of the parasite. Hence, these proteases are critical for maintaining the parasite's metabolic functions, and inhibiting them can disrupt the parasite's life cycle. Malaria remains a major global health problem, particularly in tropical and subtropical regions, where resistance to existing antimalarial drugs, such as chloroquine and artemisinin-based therapies, is an escalating issue. The emergence of drug-resistant Plasmodium strains highlights the urgent need for new therapeutic strategies. Targeting cysteine and aspartic proteases offers a novel approach to antimalarial drug development, as these enzymes are crucial for parasite survival and have not been widely exploited in current therapies. By inhibiting these proteases, researchers aim to develop new antimalarial treatments that could overcome resistance mechanisms and provide more effective options for malaria control and eradication. METHODS The application of computational methods such as molecular docking, dynamics simulations, and quantum mechanical calculations, combined with powerful molecular modeling tools, provides a comprehensive framework for discovering and optimizing inhibitors targeting Plasmodium cysteine and aspartic proteases. These methods facilitate the rational design of novel antimalarial drugs, offering a pathway to overcome drug resistance and improve therapeutic outcomes.
Collapse
Affiliation(s)
- Akinwunmi O Adeoye
- Biomembrane and Toxicology Unit, Department of Biochemistry, Federal University Oye-Ekiti, Ekiti State, Nigeria.
- Department of Chemistry, Rhodes University, Grahamstown, South Africa.
| | - Kevin A Lobb
- Department of Chemistry, Rhodes University, Grahamstown, South Africa
| |
Collapse
|
3
|
Li Q, Vetter L, Veith Y, Christ E, Végvári Á, Sahin C, Ribacke U, Wahlgren M, Ankarklev J, Larsson O, Chun-Leung Chan S. tRNA regulation and amino acid usage bias reflect a coordinated metabolic adaptation in Plasmodium falciparum. iScience 2024; 27:111167. [PMID: 39524331 PMCID: PMC11544085 DOI: 10.1016/j.isci.2024.111167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 08/20/2024] [Accepted: 10/10/2024] [Indexed: 11/16/2024] Open
Abstract
An adaptive feature of malaria-causing parasites is the digestion of host hemoglobin (HB) to acquire amino acids (AAs). Here, we describe a link between nutrient availability and translation dependent regulation of gene expression as an adaptive strategy. We show that tRNA expression in Plasmodium falciparum does not match the decoding need expected for optimal translation. A subset of tRNAs decoding AAs that are insufficiently provided by HB are lowly expressed, wherein the abundance of a protein-coding transcript is negatively correlated with the decoding requirement of these tRNAs. Proliferation-related genes have evolved a high requirement of these tRNAs, thereby proliferation can be modulated by repressing protein synthesis of these genes during nutrient stress. We conclude that the parasite modulates translation elongation by maintaining a discordant tRNA profile to exploit variations in AA-composition among genes as an adaptation strategy. This study exemplifies metabolic adaptation as an important driving force for protein evolution.
Collapse
Affiliation(s)
- Qian Li
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Leonie Vetter
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Ylva Veith
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Elena Christ
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Ákos Végvári
- Division of Chemistry I, Department of Medical Biochemistry and Biophysics (MBB), Karolinska Institutet, Stockholm, Sweden
| | - Cagla Sahin
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Ulf Ribacke
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Mats Wahlgren
- Department of Microbiology, Tumor and Cell Biology (MTC), Karolinska Institutet, Stockholm, Sweden
| | - Johan Ankarklev
- Department of Molecular Biosciences, The Wenner-Gren Institute, Stockholm University, SE-106 91 Stockholm, Sweden
| | - Ola Larsson
- Department of Oncology-Pathology, Science for Life Laboratories, Karolinska Institutet, Stockholm, Sweden
| | - Sherwin Chun-Leung Chan
- Department of Oncology-Pathology, Science for Life Laboratories, Karolinska Institutet, Stockholm, Sweden
| |
Collapse
|
4
|
Shadija N, Dass S, Xu W, Wang L, Ke H. Functionality of the V-type ATPase during asexual growth and development of Plasmodium falciparum. J Biol Chem 2024; 300:107608. [PMID: 39084459 PMCID: PMC11387698 DOI: 10.1016/j.jbc.2024.107608] [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: 12/24/2023] [Revised: 07/02/2024] [Accepted: 07/03/2024] [Indexed: 08/02/2024] Open
Abstract
Vacuolar type ATPases (V-type ATPases) are highly conserved hetero-multisubunit proton pumping machineries found in all eukaryotes. They utilize ATP hydrolysis to pump protons, acidifying intracellular or extracellular compartments, and are thus crucial for various biological processes. Despite their evolutionary conservation in malaria parasites, this proton pump remains understudied. To understand the localization and biological functions of Plasmodium falciparum V-type ATPase, we employed CRISPR/Cas9 to endogenously tag the subunit A of the V1 domain. V1A (PF3D7_1311900) was tagged with a triple hemagglutinin epitope and the TetR-DOZI-aptamer system for conditional expression under the regulation of anhydrotetracycline. Via immunofluorescence assays, we identified that V-type ATPase is expressed throughout the intraerythrocytic developmental cycle and is mainly localized to the digestive vacuole and parasite plasma membrane. Immuno-electron microscopy further revealed that V-type ATPase is also localized on secretory organelles in merozoites. Knockdown of V1A led to cytosolic pH imbalance and blockage of hemoglobin digestion in the digestive vacuole, resulting in an arrest of parasite development in the trophozoite-stage and, ultimately, parasite demise. Using bafilomycin A1, a specific inhibitor of V-type ATPases, we found that the P. falciparum V-type ATPase is likely involved in parasite invasion but is not critical for ring-stage development. Further, we detected a large molecular weight complex in blue native-PAGE (∼1.0 MDa), corresponding to the total molecular weights of V1 and Vo domains. Together, we show that V-type ATPase is localized to multiple subcellular compartments in P. falciparum, and its functionality throughout the asexual cycle varies depending on the parasite developmental stages.
Collapse
Affiliation(s)
- Neeta Shadija
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Swati Dass
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Wei Xu
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Liying Wang
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA
| | - Hangjun Ke
- Center for Molecular Parasitology, Department of Microbiology and Immunology, Drexel University College of Medicine, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
5
|
Wendt C, Miranda K. Endocytosis in malaria parasites: An ultrastructural perspective of membrane interplay in a unique infection model. CURRENT TOPICS IN MEMBRANES 2024; 93:27-49. [PMID: 39181577 DOI: 10.1016/bs.ctm.2024.05.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Malaria remains a major global threat, representing a severe public health problem worldwide. Annually, it is responsible for a high rate of morbidity and mortality in many tropical developing countries where the disease is endemic. The causative agent of malaria, Plasmodium spp., exhibits a complex life cycle, alternating between an invertebrate vector, which transmits the disease, and the vertebrate host. The disease pathology observed in the vertebrate host is attributed to the asexual development of Plasmodium spp. inside the erythrocyte. Once inside the red blood cell, malaria parasites cause extensive changes in the host cell, increasing membrane rigidity and altering its normal discoid shape. Additionally, during their intraerythrocytic development, malaria parasites incorporate and degrade up to 70 % of host cell hemoglobin. This mechanism is essential for parasite development and represents an important drug target. Blocking the steps related to hemoglobin endocytosis or degradation impairs parasite development and can lead to its death. The ultrastructural analysis of hemoglobin endocytosis on Plasmodium spp. has been broadly explored along the years. However, it is only recently that the proteins involved in this process have started to emerge. Here, we will review the most important features related to hemoglobin endocytosis and catabolism on malaria parasites. A special focus will be given to the recent analysis obtained through 3D visualization approaches and to the molecules involved in these mechanisms.
Collapse
Affiliation(s)
- Camila Wendt
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho and Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil; Laboratório de Biomineralização, Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| | - Kildare Miranda
- Laboratório de Ultraestrutura Celular Hertha Meyer, Instituto de Biofísica Carlos Chagas Filho and Centro Nacional de Biologia Estrutural e Bioimagem, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil.
| |
Collapse
|
6
|
Giannangelo C, Challis MP, Siddiqui G, Edgar R, Malcolm TR, Webb CT, Drinkwater N, Vinh N, Macraild C, Counihan N, Duffy S, Wittlin S, Devine SM, Avery VM, De Koning-Ward T, Scammells P, McGowan S, Creek DJ. Chemoproteomics validates selective targeting of Plasmodium M1 alanyl aminopeptidase as an antimalarial strategy. eLife 2024; 13:RP92990. [PMID: 38976500 PMCID: PMC11230628 DOI: 10.7554/elife.92990] [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] [Indexed: 07/10/2024] Open
Abstract
New antimalarial drug candidates that act via novel mechanisms are urgently needed to combat malaria drug resistance. Here, we describe the multi-omic chemical validation of Plasmodium M1 alanyl metalloaminopeptidase as an attractive drug target using the selective inhibitor, MIPS2673. MIPS2673 demonstrated potent inhibition of recombinant Plasmodium falciparum (PfA-M1) and Plasmodium vivax (PvA-M1) M1 metalloaminopeptidases, with selectivity over other Plasmodium and human aminopeptidases, and displayed excellent in vitro antimalarial activity with no significant host cytotoxicity. Orthogonal label-free chemoproteomic methods based on thermal stability and limited proteolysis of whole parasite lysates revealed that MIPS2673 solely targets PfA-M1 in parasites, with limited proteolysis also enabling estimation of the binding site on PfA-M1 to within ~5 Å of that determined by X-ray crystallography. Finally, functional investigation by untargeted metabolomics demonstrated that MIPS2673 inhibits the key role of PfA-M1 in haemoglobin digestion. Combined, our unbiased multi-omic target deconvolution methods confirmed the on-target activity of MIPS2673, and validated selective inhibition of M1 alanyl metalloaminopeptidase as a promising antimalarial strategy.
Collapse
Affiliation(s)
- Carlo Giannangelo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Matthew P Challis
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Ghizal Siddiqui
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Rebecca Edgar
- School of Medicine, Deakin UniversityGeelongAustralia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin UniversityGeelongAustralia
| | - Tess R Malcolm
- Monash Biomedicine Discovery Institute and Department of Microbiology, Monash UniversityClaytonAustralia
- Centre to Impact AMR, Monash UniversityClaytonAustralia
| | - Chaille T Webb
- Monash Biomedicine Discovery Institute and Department of Microbiology, Monash UniversityClaytonAustralia
- Centre to Impact AMR, Monash UniversityClaytonAustralia
| | - Nyssa Drinkwater
- Monash Biomedicine Discovery Institute and Department of Microbiology, Monash UniversityClaytonAustralia
- Centre to Impact AMR, Monash UniversityClaytonAustralia
| | - Natalie Vinh
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Christopher Macraild
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Natalie Counihan
- School of Medicine, Deakin UniversityGeelongAustralia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin UniversityGeelongAustralia
| | - Sandra Duffy
- Discovery Biology, Centre for Cellular Phenomics, Griffith UniversityNathanAustralia
| | - Sergio Wittlin
- Swiss Tropical and Public Health InstituteAllschwilSwitzerland
- University of BaselBaselSwitzerland
| | - Shane M Devine
- The Walter and Eliza Hall Institute of Medical ResearchParkvilleAustralia
- Department of Medical Biology, The University of MelbourneParkvilleAustralia
| | - Vicky M Avery
- Discovery Biology, Centre for Cellular Phenomics, Griffith UniversityNathanAustralia
- School of Environment and Science, Griffith UniversityNathanAustralia
| | - Tania De Koning-Ward
- School of Medicine, Deakin UniversityGeelongAustralia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin UniversityGeelongAustralia
| | - Peter Scammells
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Sheena McGowan
- Monash Biomedicine Discovery Institute and Department of Microbiology, Monash UniversityClaytonAustralia
- Centre to Impact AMR, Monash UniversityClaytonAustralia
| | - Darren J Creek
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| |
Collapse
|
7
|
Creek D, Giannangelo C, Challis M, Siddiqui G, Edgar R, Malcolm T, Webb C, Drinkwater N, Vinh N, MacRaild C, Counihan N, Duffy S, Wittlin S, Devine S, Avery V, de Koning-Ward T, Scammells P, McGowan S. Chemoproteomics validates selective targeting of Plasmodium M1 alanyl aminopeptidase as an antimalarial strategy. RESEARCH SQUARE 2024:rs.3.rs-3251230. [PMID: 38746424 PMCID: PMC11092810 DOI: 10.21203/rs.3.rs-3251230/v3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
New antimalarial drug candidates that act via novel mechanisms are urgently needed to combat malaria drug resistance. Here, we describe the multi-omic chemical validation of Plasmodium M1 alanyl metalloaminopeptidase as an attractive drug target using the selective inhibitor, MIPS2673. MIPS2673 demonstrated potent inhibition of recombinant Plasmodium falciparum ( Pf A-M1) and Plasmodium vivax ( Pv A-M1) M1 metalloaminopeptidases, with selectivity over other Plasmodium and human aminopeptidases, and displayed excellent in vitro antimalarial activity with no significant host cytotoxicity. Orthogonal label-free chemoproteomic methods based on thermal stability and limited proteolysis of whole parasite lysates revealed that MIPS2673 solely targets Pf A-M1 in parasites, with limited proteolysis also enabling estimation of the binding site on Pf A-M1 to within ~5 Å of that determined by X-ray crystallography. Finally, functional investigation by untargeted metabolomics demonstrated that MIPS2673 inhibits the key role of Pf A-M1 in haemoglobin digestion. Combined, our unbiased multi-omic target deconvolution methods confirmed the on-target activity of MIPS2673, and validated selective inhibition of M1 alanyl metalloaminopeptidase as a promising antimalarial strategy.
Collapse
|
8
|
Wiser MF. The Digestive Vacuole of the Malaria Parasite: A Specialized Lysosome. Pathogens 2024; 13:182. [PMID: 38535526 PMCID: PMC10974218 DOI: 10.3390/pathogens13030182] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2024] [Revised: 02/14/2024] [Accepted: 02/17/2024] [Indexed: 02/11/2025] Open
Abstract
The malaria parasite resides within erythrocytes during one stage of its life cycle. During this intraerythrocytic period, the parasite ingests the erythrocyte cytoplasm and digests approximately two-thirds of the host cell hemoglobin. This digestion occurs within a lysosome-like organelle called the digestive vacuole. Several proteases are localized to the digestive vacuole and these proteases sequentially breakdown hemoglobin into small peptides, dipeptides, and amino acids. The peptides are exported into the host cytoplasm via the chloroquine-resistance transporter and an amino acid transporter has also been identified on the digestive vacuole membrane. The environment of the digestive vacuole also provides appropriate conditions for the biocrystallization of toxic heme into non-toxic hemozoin by a poorly understood process. Hemozoin formation is an attribute of Plasmodium and Haemoproteus and is not exhibited by other intraerythrocytic protozoan parasites. The efficient degradation of hemoglobin and detoxification of heme likely plays a major role in the high level of replication exhibited by malaria parasites within erythrocytes. Unique features of the digestive vacuole and the critical importance of nutrient acquisition provide therapeutic targets for the treatment of malaria.
Collapse
Affiliation(s)
- Mark F Wiser
- Department of Tropical Medicine and Infectious Disease, Tulane University School of Public Health and Tropical Medicine, New Orleans, LA 70112-2824, USA
| |
Collapse
|
9
|
Hagenah LM, Dhingra SK, Small-Saunders JL, Qahash T, Willems A, Schindler KA, Rangel GW, Gil-Iturbe E, Kim J, Akhundova E, Yeo T, Okombo J, Mancia F, Quick M, Roepe PD, Llinás M, Fidock DA. Additional PfCRT mutations driven by selective pressure for improved fitness can result in the loss of piperaquine resistance and altered Plasmodium falciparum physiology. mBio 2024; 15:e0183223. [PMID: 38059639 PMCID: PMC10790694 DOI: 10.1128/mbio.01832-23] [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: 07/12/2023] [Accepted: 10/31/2023] [Indexed: 12/08/2023] Open
Abstract
IMPORTANCE Our study leverages gene editing techniques in Plasmodium falciparum asexual blood stage parasites to profile novel mutations in mutant PfCRT, an important mediator of piperaquine resistance, which developed in Southeast Asian field isolates or in parasites cultured for long periods of time. We provide evidence that increased parasite fitness of these lines is the primary driver for the emergence of these PfCRT variants. These mutations differentially impact parasite susceptibility to piperaquine and chloroquine, highlighting the multifaceted effects of single point mutations in this transporter. Molecular features of drug resistance and parasite physiology were examined in depth using proteoliposome-based drug uptake studies and peptidomics, respectively. Energy minimization calculations, showing how these novel mutations might impact the PfCRT structure, suggested a small but significant effect on drug interactions. This study reveals the subtle interplay between antimalarial resistance, parasite fitness, PfCRT structure, and intracellular peptide availability in PfCRT-mediated parasite responses to changing drug selective pressures.
Collapse
Affiliation(s)
- Laura M. Hagenah
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, New York, USA
| | - Satish K. Dhingra
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, New York, USA
| | - Jennifer L. Small-Saunders
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, New York, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| | - Tarrick Qahash
- Department of Chemistry, Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Biochemistry and Molecular Biology and Huck Center for Malaria Research, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Andreas Willems
- Department of Chemistry, Georgetown University, Washington, DC, USA
- Department of Biochemistry and Cellular and Molecular Biology, Georgetown University, Washington, DC, USA
| | - Kyra A. Schindler
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, New York, USA
| | - Gabriel W. Rangel
- Department of Biochemistry and Molecular Biology and Huck Center for Malaria Research, Pennsylvania State University, University Park, Pennsylvania, USA
| | - Eva Gil-Iturbe
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York, USA
| | - Jonathan Kim
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, New York, USA
| | - Emiliya Akhundova
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, New York, USA
| | - Tomas Yeo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, New York, USA
| | - John Okombo
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, New York, USA
| | - Filippo Mancia
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, New York, USA
| | - Matthias Quick
- Department of Psychiatry, Columbia University Irving Medical Center, New York, New York, USA
- Department of Physiology and Cellular Biophysics, Columbia University Irving Medical Center, New York, New York, USA
- Area Neuroscience - Molecular Therapeutics, New York State Psychiatric Institute, New York, New York, USA
| | - Paul D. Roepe
- Department of Chemistry, Georgetown University, Washington, DC, USA
- Department of Biochemistry and Cellular and Molecular Biology, Georgetown University, Washington, DC, USA
| | - Manuel Llinás
- Department of Chemistry, Pennsylvania State University, University Park, Pennsylvania, USA
- Department of Biochemistry and Molecular Biology and Huck Center for Malaria Research, Pennsylvania State University, University Park, Pennsylvania, USA
| | - David A. Fidock
- Department of Microbiology and Immunology, Columbia University Irving Medical Center, New York, New York, USA
- Center for Malaria Therapeutics and Antimicrobial Resistance, Columbia University Irving Medical Center, New York, New York, USA
- Division of Infectious Diseases, Department of Medicine, Columbia University Irving Medical Center, New York, New York, USA
| |
Collapse
|
10
|
Schmidt S, Wichers-Misterek JS, Behrens HM, Birnbaum J, Henshall IG, Dröge J, Jonscher E, Flemming S, Castro-Peña C, Mesén-Ramírez P, Spielmann T. The Kelch13 compartment contains highly divergent vesicle trafficking proteins in malaria parasites. PLoS Pathog 2023; 19:e1011814. [PMID: 38039338 PMCID: PMC10718435 DOI: 10.1371/journal.ppat.1011814] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2023] [Revised: 12/13/2023] [Accepted: 11/09/2023] [Indexed: 12/03/2023] Open
Abstract
Single amino acid changes in the parasite protein Kelch13 (K13) result in reduced susceptibility of P. falciparum parasites to artemisinin and its derivatives (ART). Recent work indicated that K13 and other proteins co-localising with K13 (K13 compartment proteins) are involved in the endocytic uptake of host cell cytosol (HCCU) and that a reduction in HCCU results in reduced susceptibility to ART. HCCU is critical for parasite survival but is poorly understood, with the K13 compartment proteins among the few proteins so far functionally linked to this process. Here we further defined the composition of the K13 compartment by analysing more hits from a previous BioID, showing that MyoF and MCA2 as well as Kelch13 interaction candidate (KIC) 11 and 12 are found at this site. Functional analyses, tests for ART susceptibility as well as comparisons of structural similarities using AlphaFold2 predictions of these and previously identified proteins showed that vesicle trafficking and endocytosis domains were frequent in proteins involved in resistance or endocytosis (or both), comprising one group of K13 compartment proteins. While this strengthened the link of the K13 compartment to endocytosis, many proteins of this group showed unusual domain combinations and large parasite-specific regions, indicating a high level of taxon-specific adaptation of this process. Another group of K13 compartment proteins did not influence endocytosis or ART susceptibility and lacked detectable vesicle trafficking domains. We here identified the first protein of this group that is important for asexual blood stage development and showed that it likely is involved in invasion. Overall, this work identified novel proteins functioning in endocytosis and at the K13 compartment. Together with comparisons of structural predictions it provides a repertoire of functional domains at the K13 compartment that indicate a high level of adaption of endocytosis in malaria parasites.
Collapse
Affiliation(s)
- Sabine Schmidt
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | | | | | - Jakob Birnbaum
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | | | - Jana Dröge
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Ernst Jonscher
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Sven Flemming
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | | | | | - Tobias Spielmann
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| |
Collapse
|
11
|
Si W, Zhao Y, Qin X, Huang Y, Yu J, Liu X, Li Y, Yan X, Zhang Q, Sun J. What exactly does the PfK13 C580Y mutation in Plasmodium falciparum influence? Parasit Vectors 2023; 16:421. [PMID: 37974285 PMCID: PMC10652512 DOI: 10.1186/s13071-023-06024-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2023] [Accepted: 10/19/2023] [Indexed: 11/19/2023] Open
Abstract
BACKGROUND The emergence and spread of artemisinin resistance threaten global malaria control and elimination goals, and encourage research on the mechanisms of drug resistance in malaria parasites. Mutations in Plasmodium falciparum Kelch 13 (PfK13) protein are associated with artemisinin resistance, but the unique or common mechanism which results in this resistance is unclear. METHODS We analyzed the effects of the PfK13 mutation on the transcriptome and proteome of P. falciparum at different developmental stages. Additionally, the number of merozoites, hemozoin amount, and growth of P. falciparum 3D7C580Y and P. falciparum 3D7WT were compared. The impact of iron supplementation on the number of merozoites of P. falciparum 3D7C580Y was also examined. RESULTS We found that the PfK13 mutation did not significantly change glycolysis, TCA, pentose phosphate pathway, or oxidative phosphorylation, but did reduce the expression of reproduction- and DNA synthesis-related genes. The reduced number of merozoites, decreased level of hemozoin, and slowed growth of P. falciparum 3D7C580Y were consistent with these changes. Furthermore, adding iron supply could increase the number of the merozoites of P. falciparum 3D7C580Y. CONCLUSIONS These results revealed that the PfK13 mutation reduced hemoglobin ingestion, leading to artemisinin resistance, likely by decreasing the parasites' requirement for haem and iron. This study helps elucidate the mechanism of artemisinin resistance due to PfK13 mutations.
Collapse
Affiliation(s)
- Wenwen Si
- School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Yuemeng Zhao
- School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Xixi Qin
- School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Yixuan Huang
- School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Jing Yu
- School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Xiao Liu
- School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Yanna Li
- School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Xiaoli Yan
- School of Medicine, Tongji University, Shanghai, People's Republic of China
| | - Qingfeng Zhang
- School of Medicine, Tongji University, Shanghai, People's Republic of China.
| | - Jun Sun
- School of Medicine, Tongji University, Shanghai, People's Republic of China.
| |
Collapse
|
12
|
Henshall IG, Spielmann T. Critical interdependencies between Plasmodium nutrient flux and drugs. Trends Parasitol 2023; 39:936-944. [PMID: 37716852 PMCID: PMC10580322 DOI: 10.1016/j.pt.2023.08.008] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/18/2023]
Abstract
Nutrient import and waste efflux are critical dependencies for intracellular Plasmodium falciparum parasites. Nutrient transport proteins are often lineage specific and can provide unique targets for antimalarial drug development. P. falciparum nutrient transport pathways can be a double-edged sword for the parasite, not only mediating the import of nutrients and excretion of waste products but also providing an access route for drugs. Here we briefly summarise the nutrient acquisition pathways of intracellular P. falciparum blood-stage parasites and then highlight how these pathways influence many aspects relevant to antimalarial drugs, resulting in complex and often underappreciated interdependencies.
Collapse
Affiliation(s)
| | - Tobias Spielmann
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany.
| |
Collapse
|
13
|
Berger F, Gomez GM, Sanchez CP, Posch B, Planelles G, Sohraby F, Nunes-Alves A, Lanzer M. pH-dependence of the Plasmodium falciparum chloroquine resistance transporter is linked to the transport cycle. Nat Commun 2023; 14:4234. [PMID: 37454114 PMCID: PMC10349806 DOI: 10.1038/s41467-023-39969-2] [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: 11/16/2022] [Accepted: 07/05/2023] [Indexed: 07/18/2023] Open
Abstract
The chloroquine resistance transporter, PfCRT, of the human malaria parasite Plasmodium falciparum is sensitive to acidic pH. Consequently, PfCRT operates at 60% of its maximal drug transport activity at the pH of 5.2 of the digestive vacuole, a proteolytic organelle from which PfCRT expels drugs interfering with heme detoxification. Here we show by alanine-scanning mutagenesis that E207 is critical for pH sensing. The E207A mutation abrogates pH-sensitivity, while preserving drug substrate specificity. Substituting E207 with Asp or His, but not other amino acids, restores pH-sensitivity. Molecular dynamics simulations and kinetics analyses suggest an allosteric binding model in which PfCRT can accept both protons and chloroquine in a partial noncompetitive manner, with increased proton concentrations decreasing drug transport. Further simulations reveal that E207 relocates from a peripheral to an engaged location during the transport cycle, forming a salt bridge with residue K80. We propose that the ionized carboxyl group of E207 acts as a hydrogen acceptor, facilitating transport cycle progression, with pH sensing as a by-product.
Collapse
Affiliation(s)
- Fiona Berger
- Center of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | - Guillermo M Gomez
- Center of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | - Cecilia P Sanchez
- Center of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | - Britta Posch
- Center of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany
| | - Gabrielle Planelles
- INSERM, Centre de Recherche des Cordeliers, Unité 1138, CNRS ERL8228, Université Pierre et Marie Curie and Université Paris-Descartes, Paris, 75006, France
| | - Farzin Sohraby
- Institute of Chemistry, Technische Universität Berlin, Straße des 17. Juni 135, 10623, Berlin, Germany
| | - Ariane Nunes-Alves
- Institute of Chemistry, Technische Universität Berlin, Straße des 17. Juni 135, 10623, Berlin, Germany.
| | - Michael Lanzer
- Center of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Im Neuenheimer Feld 324, 69120, Heidelberg, Germany.
| |
Collapse
|
14
|
Ghosh S, Kundu R, Chandana M, Das R, Anand A, Beura S, Bobde RC, Jain V, Prabhu SR, Behera PK, Mohanty AK, Chakrapani M, Satyamoorthy K, Suryawanshi AR, Dixit A, Padmanaban G, Nagaraj VA. Distinct evolution of type I glutamine synthetase in Plasmodium and its species-specific requirement. Nat Commun 2023; 14:4216. [PMID: 37452051 PMCID: PMC10349072 DOI: 10.1038/s41467-023-39670-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 06/22/2023] [Indexed: 07/18/2023] Open
Abstract
Malaria parasite lacks canonical pathways for amino acid biosynthesis and depends primarily on hemoglobin degradation and extracellular resources for amino acids. Interestingly, a putative gene for glutamine synthetase (GS) is retained despite glutamine being an abundant amino acid in human and mosquito hosts. Here we show Plasmodium GS has evolved as a unique type I enzyme with distinct structural and regulatory properties to adapt to the asexual niche. Methionine sulfoximine (MSO) and phosphinothricin (PPT) inhibit parasite GS activity. GS is localized to the parasite cytosol and abundantly expressed in all the life cycle stages. Parasite GS displays species-specific requirement in Plasmodium falciparum (Pf) having asparagine-rich proteome. Targeting PfGS affects asparagine levels and inhibits protein synthesis through eIF2α phosphorylation leading to parasite death. Exposure of artemisinin-resistant Pf parasites to MSO and PPT inhibits the emergence of viable parasites upon artemisinin treatment.
Collapse
Affiliation(s)
- Sourav Ghosh
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
- Regional Centre for Biotechnology, Faridabad, 121001, Haryana, India
| | - Rajib Kundu
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
- Regional Centre for Biotechnology, Faridabad, 121001, Haryana, India
| | - Manjunatha Chandana
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
- School of Biotechnology, Kalinga Institute of Industrial Technology, Bhubaneswar, 751024, Odisha, India
| | - Rahul Das
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
- Regional Centre for Biotechnology, Faridabad, 121001, Haryana, India
| | - Aditya Anand
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
- Regional Centre for Biotechnology, Faridabad, 121001, Haryana, India
| | - Subhashree Beura
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
| | - Ruchir Chandrakant Bobde
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
- Regional Centre for Biotechnology, Faridabad, 121001, Haryana, India
| | - Vishal Jain
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
| | - Sowmya Ramakant Prabhu
- Department of Biotechnology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | | | - Akshaya Kumar Mohanty
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
- Ispat General Hospital, Sector 19, Rourkela, 769005, Odisha, India
| | - Mahabala Chakrapani
- Department of Medicine, Kasturba Medical College, Mangalore, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | - Kapaettu Satyamoorthy
- Department of Cell and Molecular Biology, Manipal School of Life Sciences, Manipal Academy of Higher Education, Manipal, 576104, Karnataka, India
| | | | - Anshuman Dixit
- Infectious Disease Biology, Institute of Life Sciences, Bhubaneswar, 751023, Odisha, India
| | - Govindarajan Padmanaban
- Department of Biochemistry, Indian Institute of Science, Bangalore, 560012, Karnataka, India
| | | |
Collapse
|
15
|
Jonsdottir TK, Elsworth B, Cobbold S, Gabriela M, Ploeger E, Parkyn Schneider M, Charnaud SC, Dans MG, McConville M, Bullen HE, Crabb BS, Gilson PR. PTEX helps efficiently traffic haemoglobinases to the food vacuole in Plasmodium falciparum. PLoS Pathog 2023; 19:e1011006. [PMID: 37523385 PMCID: PMC10414648 DOI: 10.1371/journal.ppat.1011006] [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: 11/14/2022] [Revised: 08/10/2023] [Accepted: 07/16/2023] [Indexed: 08/02/2023] Open
Abstract
A key element of Plasmodium biology and pathogenesis is the trafficking of ~10% of the parasite proteome into the host red blood cell (RBC) it infects. To cross the parasite-encasing parasitophorous vacuole membrane, exported proteins utilise a channel-forming protein complex termed the Plasmodium translocon of exported proteins (PTEX). PTEX is obligatory for parasite survival, both in vitro and in vivo, suggesting that at least some exported proteins have essential metabolic functions. However, to date only one essential PTEX-dependent process, the new permeability pathways, has been described. To identify other essential PTEX-dependant proteins/processes, we conditionally knocked down the expression of one of its core components, PTEX150, and examined which pathways were affected. Surprisingly, the food vacuole mediated process of haemoglobin (Hb) digestion was substantially perturbed by PTEX150 knockdown. Using a range of transgenic parasite lines and approaches, we show that two major Hb proteases; falcipain 2a and plasmepsin II, interact with PTEX core components, implicating the translocon in the trafficking of Hb proteases. We propose a model where these proteases are translocated into the PV via PTEX in order to reach the cytostome, located at the parasite periphery, prior to food vacuole entry. This work offers a second mechanistic explanation for why PTEX function is essential for growth of the parasite within its host RBC.
Collapse
Affiliation(s)
- Thorey K. Jonsdottir
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
- Department of Immunology and Microbiology, University of Melbourne, Melbourne, Australia
| | - Brendan Elsworth
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
| | - Simon Cobbold
- Department of Biochemistry and Molecular Biology, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Melbourne, Australia
| | - Mikha Gabriela
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
- School of Medicine, Deakin University, Geelong, Australia
| | - Ellen Ploeger
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
| | | | - Sarah C. Charnaud
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
| | - Madeline G. Dans
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
| | - Malcolm McConville
- Department of Biochemistry and Molecular Biology, Bio21 Institute of Molecular Science and Biotechnology, University of Melbourne, Melbourne, Australia
| | - Hayley E. Bullen
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
- Department of Immunology and Microbiology, University of Melbourne, Melbourne, Australia
| | - Brendan S. Crabb
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
- Department of Immunology and Microbiology, University of Melbourne, Melbourne, Australia
- Department of Immunology and Pathology, Monash University, Melbourne, Australia
| | - Paul R. Gilson
- Malaria Virulence and Drug Discovery Group, Burnet Institute, Melbourne, Australia
- Department of Immunology and Microbiology, University of Melbourne, Melbourne, Australia
| |
Collapse
|
16
|
Aguado ME, Izquierdo M, González-Matos M, Varela AC, Méndez Y, Del Rivero MA, Rivera DG, González-Bacerio J. Parasite Metalo-aminopeptidases as Targets in Human Infectious Diseases. Curr Drug Targets 2023; 24:416-461. [PMID: 36825701 DOI: 10.2174/1389450124666230224140724] [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: 08/25/2022] [Revised: 12/25/2022] [Accepted: 01/02/2023] [Indexed: 02/25/2023]
Abstract
BACKGROUND Parasitic human infectious diseases are a worldwide health problem due to the increased resistance to conventional drugs. For this reason, the identification of novel molecular targets and the discovery of new chemotherapeutic agents are urgently required. Metalo- aminopeptidases are promising targets in parasitic infections. They participate in crucial processes for parasite growth and pathogenesis. OBJECTIVE In this review, we describe the structural, functional and kinetic properties, and inhibitors, of several parasite metalo-aminopeptidases, for their use as targets in parasitic diseases. CONCLUSION Plasmodium falciparum M1 and M17 aminopeptidases are essential enzymes for parasite development, and M18 aminopeptidase could be involved in hemoglobin digestion and erythrocyte invasion and egression. Trypanosoma cruzi, T. brucei and Leishmania major acidic M17 aminopeptidases can play a nutritional role. T. brucei basic M17 aminopeptidase down-regulation delays the cytokinesis. The inhibition of Leishmania basic M17 aminopeptidase could affect parasite viability. L. donovani methionyl aminopeptidase inhibition prevents apoptosis but not the parasite death. Decrease in Acanthamoeba castellanii M17 aminopeptidase activity produces cell wall structural modifications and encystation inhibition. Inhibition of Babesia bovis growth is probably related to the inhibition of the parasite M17 aminopeptidase, probably involved in host hemoglobin degradation. Schistosoma mansoni M17 aminopeptidases inhibition may affect parasite development, since they could participate in hemoglobin degradation, surface membrane remodeling and eggs hatching. Toxoplasma gondii M17 aminopeptidase inhibition could attenuate parasite virulence, since it is apparently involved in the hydrolysis of cathepsin Cs- or proteasome-produced dipeptides and/or cell attachment/invasion processes. These data are relevant to validate these enzymes as targets.
Collapse
Affiliation(s)
- Mirtha E Aguado
- Center for Protein Studies, Faculty of Biology, University of Havana, Calle 25 #455 Entre I y J, 10400, Vedado, La Habana, Cuba
| | - Maikel Izquierdo
- Center for Protein Studies, Faculty of Biology, University of Havana, Calle 25 #455 Entre I y J, 10400, Vedado, La Habana, Cuba
| | - Maikel González-Matos
- Center for Protein Studies, Faculty of Biology, University of Havana, Calle 25 #455 Entre I y J, 10400, Vedado, La Habana, Cuba
| | - Ana C Varela
- Center for Protein Studies, Faculty of Biology, University of Havana, Calle 25 #455 Entre I y J, 10400, Vedado, La Habana, Cuba
| | - Yanira Méndez
- Center for Natural Products Research, Faculty of Chemistry, University of Havana, Zapata y G, 10400, La Habana, Cuba
| | - Maday A Del Rivero
- Center for Protein Studies, Faculty of Biology, University of Havana, Calle 25 #455 Entre I y J, 10400, Vedado, La Habana, Cuba
| | - Daniel G Rivera
- Center for Natural Products Research, Faculty of Chemistry, University of Havana, Zapata y G, 10400, La Habana, Cuba
| | - Jorge González-Bacerio
- Center for Protein Studies, Faculty of Biology, University of Havana, Calle 25 #455 Entre I y J, 10400, Vedado, La Habana, Cuba
- Department of Biochemistry, Faculty of Biology, University of Havana, calle 25 #455 entre I y J, 10400, Vedado, La Habana, Cuba
| |
Collapse
|
17
|
Edgar RCS, Siddiqui G, Hjerrild K, Malcolm TR, Vinh NB, Webb CT, Holmes C, MacRaild CA, Chernih HC, Suen WW, Counihan NA, Creek DJ, Scammells PJ, McGowan S, de Koning-Ward TF. Genetic and chemical validation of Plasmodium falciparum aminopeptidase PfA-M17 as a drug target in the hemoglobin digestion pathway. eLife 2022; 11:e80813. [PMID: 36097817 PMCID: PMC9470162 DOI: 10.7554/elife.80813] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2022] [Accepted: 08/02/2022] [Indexed: 11/18/2022] Open
Abstract
Plasmodium falciparum, the causative agent of malaria, remains a global health threat as parasites continue to develop resistance to antimalarial drugs used throughout the world. Accordingly, drugs with novel modes of action are desperately required to combat malaria. P. falciparum parasites infect human red blood cells where they digest the host's main protein constituent, hemoglobin. Leucine aminopeptidase PfA-M17 is one of several aminopeptidases that have been implicated in the last step of this digestive pathway. Here, we use both reverse genetics and a compound specifically designed to inhibit the activity of PfA-M17 to show that PfA-M17 is essential for P. falciparum survival as it provides parasites with free amino acids for growth, many of which are highly likely to originate from hemoglobin. We further show that loss of PfA-M17 results in parasites exhibiting multiple digestive vacuoles at the trophozoite stage. In contrast to other hemoglobin-degrading proteases that have overlapping redundant functions, we validate PfA-M17 as a potential novel drug target.
Collapse
Affiliation(s)
- Rebecca CS Edgar
- School of Medicine, Deakin UniversityGeelongAustralia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin UniversityGeelongAustralia
| | - Ghizal Siddiqui
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | | | - Tess R Malcolm
- Biomedicine Discovery Institute and Department of Microbiology, Monash UniversityClaytonAustralia
- Centre to Impact AMR, Monash UniversityMelbourneAustralia
| | - Natalie B Vinh
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Chaille T Webb
- Biomedicine Discovery Institute and Department of Microbiology, Monash UniversityClaytonAustralia
- Centre to Impact AMR, Monash UniversityMelbourneAustralia
| | - Clare Holmes
- CSIRO Australian Centre for Disease PreparednessGeelongAustralia
| | - Christopher A MacRaild
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Hope C Chernih
- School of Medicine, Deakin UniversityGeelongAustralia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin UniversityGeelongAustralia
| | - Willy W Suen
- CSIRO Australian Centre for Disease PreparednessGeelongAustralia
| | - Natalie A Counihan
- School of Medicine, Deakin UniversityGeelongAustralia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin UniversityGeelongAustralia
| | - Darren J Creek
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Peter J Scammells
- Medicinal Chemistry, Monash Institute of Pharmaceutical Sciences, Monash UniversityParkvilleAustralia
| | - Sheena McGowan
- Biomedicine Discovery Institute and Department of Microbiology, Monash UniversityClaytonAustralia
- Centre to Impact AMR, Monash UniversityMelbourneAustralia
| | - Tania F de Koning-Ward
- School of Medicine, Deakin UniversityGeelongAustralia
- The Institute for Mental and Physical Health and Clinical Translation, Deakin UniversityGeelongAustralia
| |
Collapse
|
18
|
Brenda CT, Norma RF, Marcela RL, Nelly LV, Teresa I F. Action mechanisms of metallic compounds on Plasmodium spp. J Trace Elem Med Biol 2022; 73:127028. [PMID: 35797926 DOI: 10.1016/j.jtemb.2022.127028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 06/10/2022] [Accepted: 06/20/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND Malaria is a parasitic disease with the highest morbidity and mortality worldwide. Unfortunately, during the last decades, the causal agent, Plasmodium spp., has developed resistance to chloroquine and artemisinin. For this reason, metallic compounds have been proposed as an optional treatment since they have shown a potential antimalarial effect with diverse action mechanisms in the parasite and the host. OBJECTIVE To show the possible targets of metallic compounds in Plasmodium spp. CONCLUSION The metallic compounds are an option attractive to treatment for the malaria, for its low cost and its great activity to reduce parasitemia; however is necessary more studies principally in vivo in order to know the interactions that it can have in an experimental model.
Collapse
Affiliation(s)
- Casarrubias-Tabarez Brenda
- Departamento de Biología Celular y TIsular, Facultad de Medicina, UNAM, Av. Ciudad Universitaria 3000, Coyoacan, C.P. 04510 Mexico City, Mexico.
| | - Rivera-Fernández Norma
- Departamento de Microbiología y Parasitología, UNAM, Av. Ciudad Universitaria 3000, Coyoacan, C.P. 04510 Mexico City, Mexico.
| | - Rojas-Lemus Marcela
- Departamento de Biología Celular y TIsular, Facultad de Medicina, UNAM, Av. Ciudad Universitaria 3000, Coyoacan, C.P. 04510 Mexico City, Mexico.
| | - López-Valdez Nelly
- Departamento de Biología Celular y TIsular, Facultad de Medicina, UNAM, Av. Ciudad Universitaria 3000, Coyoacan, C.P. 04510 Mexico City, Mexico.
| | - Fortoul Teresa I
- Departamento de Biología Celular y TIsular, Facultad de Medicina, UNAM, Av. Ciudad Universitaria 3000, Coyoacan, C.P. 04510 Mexico City, Mexico.
| |
Collapse
|
19
|
Sanchez CP, Manson EDT, Moliner Cubel S, Mandel L, Weidt SK, Barrett MP, Lanzer M. The Knock-Down of the Chloroquine Resistance Transporter PfCRT Is Linked to Oligopeptide Handling in Plasmodium falciparum. Microbiol Spectr 2022; 10:e0110122. [PMID: 35867395 PMCID: PMC9431119 DOI: 10.1128/spectrum.01101-22] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Accepted: 06/27/2022] [Indexed: 11/20/2022] Open
Abstract
The chloroquine resistance transporter, PfCRT, is an essential factor during intraerythrocytic development of the human malaria parasite Plasmodium falciparum. PfCRT resides at the digestive vacuole of the parasite, where hemoglobin taken up by the parasite from its host cell is degraded. PfCRT can acquire several mutations that render PfCRT a drug transporting system expelling compounds targeting hemoglobin degradation from the digestive vacuole. The non-drug related function of PfCRT is less clear, although a recent study has suggested a role in oligopeptide transport based on studies conducted in a heterologous expression system. The uncertainty about the natural function of PfCRT is partly due to a lack of a null mutant and a dearth of functional assays in the parasite. Here, we report on the generation of a conditional PfCRT knock-down mutant in P. falciparum. The mutant accumulated oligopeptides 2 to at least 8 residues in length under knock-down conditions, as shown by comparative global metabolomics. The accumulated oligopeptides were structurally diverse, had an isoelectric point between 4.0 and 5.4 and were electrically neutral or carried a single charge at the digestive vacuolar pH of 5.2. Fluorescently labeled dipeptides and live cell imaging identified the digestive vacuole as the compartment where oligopeptides accumulated. Our findings suggest a function of PfCRT in oligopeptide transport across the digestive vacuolar membrane in P. falciparum and associated with it a role in nutrient acquisition and the maintenance of the colloid osmotic balance. IMPORTANCE The chloroquine resistance transporter, PfCRT, is important for the survival of the human malaria parasite Plasmodium falciparum. It increases the tolerance to many antimalarial drugs, and it is essential for the development of the parasite within red blood cells. While we understand the role of PfCRT in drug resistance in ever increasing detail, the non-drug resistance functions are still debated. Identifying the natural substrate of PfCRT has been hampered by a paucity of functional assays to test putative substrates in the parasite system and the absence of a parasite mutant deficient for the PfCRT encoding gene. By generating a conditional PfCRT knock-down mutant, together with comparative metabolomics and uptake studies using fluorescently labeled oligopeptides, we could show that PfCRT is an oligopeptide transporter. The oligopeptides were structurally diverse and were electrically neutral or carried a single charge. Our data support a function of PfCRT in oligopeptide transport.
Collapse
Affiliation(s)
- Cecilia P. Sanchez
- Center of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | | | - Sonia Moliner Cubel
- Center of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Heidelberg, Germany
| | | | - Stefan K. Weidt
- Glasgow Polyomics, University of Glasgow, Wolfson Wohl Cancer Research Centre, Glasgow, United Kingdom
| | - Michael P. Barrett
- Glasgow Polyomics, University of Glasgow, Wolfson Wohl Cancer Research Centre, Glasgow, United Kingdom
- The Wellcome Centre for Integrative Parasitology, Institute for Infection, Immunity and Inflammation, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow, United Kingdom
| | - Michael Lanzer
- Center of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, Heidelberg, Germany
| |
Collapse
|
20
|
Babesia, Theileria, Plasmodium and Hemoglobin. Microorganisms 2022; 10:microorganisms10081651. [PMID: 36014069 PMCID: PMC9414693 DOI: 10.3390/microorganisms10081651] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2022] [Revised: 08/08/2022] [Accepted: 08/11/2022] [Indexed: 12/03/2022] Open
Abstract
The Propagation of Plasmodium spp. and Babesia/Theileria spp. vertebrate blood stages relies on the mediated acquisition of nutrients available within the host’s red blood cell (RBC). The cellular processes of uptake, trafficking and metabolic processing of host RBC proteins are thus crucial for the intraerythrocytic development of these parasites. In contrast to malarial Plasmodia, the molecular mechanisms of uptake and processing of the major RBC cytoplasmic protein hemoglobin remain widely unexplored in intraerythrocytic Babesia/Theileria species. In the paper, we thus provide an updated comparison of the intraerythrocytic stage feeding mechanisms of these two distantly related groups of parasitic Apicomplexa. As the associated metabolic pathways including proteolytic degradation and networks facilitating heme homeostasis represent attractive targets for diverse antimalarials, and alterations in these pathways underpin several mechanisms of malaria drug resistance, our ambition is to highlight some fundamental differences resulting in different implications for parasite management with the potential for novel interventions against Babesia/Theileria infections.
Collapse
|
21
|
Matz JM. Plasmodium’s bottomless pit: properties and functions of the malaria parasite's digestive vacuole. Trends Parasitol 2022; 38:525-543. [DOI: 10.1016/j.pt.2022.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2021] [Revised: 02/21/2022] [Accepted: 02/21/2022] [Indexed: 11/30/2022]
|
22
|
Adderley J, O'Donoghue F, Doerig C, Davis S. MAPPINGS, a tool for network analysis of large phospho-signalling datasets: application to host erythrocyte response to Plasmodium infection. CURRENT RESEARCH IN MICROBIAL SCIENCES 2022; 3:100149. [PMID: 35909628 PMCID: PMC9325900 DOI: 10.1016/j.crmicr.2022.100149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 05/12/2022] [Accepted: 06/20/2022] [Indexed: 11/17/2022] Open
Abstract
The complexity of signal transduction networks in eukaryotic cells, superimposed to very large datasets generated by “omics” approaches (notably phosphor-proteomics), calls for tools to identify pathways that are mobilised under specific conditions, including infection by intracellular pathogens. This has become a bottleneck in various biology fields, from cancer through developmental biology to infectious diseases. We developed MAPPINGS, a computational tool to extract meaning from large phosphosignalling datasets, and used it to analyse host erythrocyte response to infection with malaria parasites, leading to the identification of host cell pathways that are activated by Plasmodium. MAPPINGS uses random walks to identify chains of phosphorylation events occurring much more or much less frequently than expected, and highlights pathways of phosphorylation that work synergistically, providing a rapid interpretation of the most critical pathways in any phosphosiganlling dataset.
Large datasets of phosphorylation interactions are constantly being generated, but deciphering the complex network structure hidden in these datasets remains challenging. Many phosphorylation interactions occurring in human cells have been identified and constitute the basis for the known phosphorylation interaction network. We overlayed onto this network phosphorylation datasets obtained from an antibody microarray approach aimed at determining changes in phospho-signalling of host erythrocytes, during infection with the malaria parasite Plasmodium falciparum. We designed a pathway analysis tool denoted MAPPINGS that uses random walks to identify chains of phosphorylation events occurring much more or much less frequently than expected. MAPPINGS highlights pathways of phosphorylation that work synergistically, providing a rapid interpretation of the most critical pathways in each dataset. MAPPINGS confirmed several signalling interactions previously shown to be modulated by infection, and revealed additional interactions which could form the basis of numerous future studies. The MAPPINGS analysis strategy described here is widely applicable to comparative phosphorylation datasets in any context, such as response of cells to infection, treatment, or comparison between differentiation stages of any cellular population.
Collapse
Affiliation(s)
- Jack Adderley
- School of Health and Biomedical Sciences, RMIT University, Bundoora VIC 3083, Australia
- Corresponding authors.
| | - Finn O'Donoghue
- School of Science, RMIT University, Melbourne, VIC 3053, Australia
| | - Christian Doerig
- School of Health and Biomedical Sciences, RMIT University, Bundoora VIC 3083, Australia
- Corresponding authors.
| | - Stephen Davis
- School of Science, RMIT University, Melbourne, VIC 3053, Australia
| |
Collapse
|
23
|
Baptista V, Costa MS, Calçada C, Silva M, Gil JP, Veiga MI, Catarino SO. The Future in Sensing Technologies for Malaria Surveillance: A Review of Hemozoin-Based Diagnosis. ACS Sens 2021; 6:3898-3911. [PMID: 34735120 DOI: 10.1021/acssensors.1c01750] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Early and effective malaria diagnosis is vital to control the disease spread and to prevent the emergence of severe cases and death. Currently, malaria diagnosis relies on optical microscopy and immuno-rapid tests; however, these require a drop of blood, are time-consuming, or are not specific and sensitive enough for reliable detection of low-level parasitaemia. Thus, there is an urge for simpler, prompt, and accurate alternative diagnostic methods. Particularly, hemozoin has been increasingly recognized as an attractive biomarker for malaria detection. As the disease proliferates, parasites digest host hemoglobin, in the process releasing toxic haem that is detoxified into an insoluble crystal, the hemozoin, which accumulates along with infection progression. Given its magnetic, optical, and acoustic unique features, hemozoin has been explored for new label-free diagnostic methods. Thereby, herein, we review the hemozoin-based malaria detection methods and critically discuss their challenges and potential for the development of an ideal diagnostic device.
Collapse
Affiliation(s)
- Vitória Baptista
- Microelectromechanical Systems Research Unit (CMEMS-UMinho), School of Engineering, University of Minho, Campus de Azurém, 4800-058 Guimarães, Portugal
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s − PT Government Associate Laboratory, 4805-017 Braga/Guimarães, Portugal
| | - Mariana S. Costa
- Microelectromechanical Systems Research Unit (CMEMS-UMinho), School of Engineering, University of Minho, Campus de Azurém, 4800-058 Guimarães, Portugal
| | - Carla Calçada
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s − PT Government Associate Laboratory, 4805-017 Braga/Guimarães, Portugal
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, 3004-504 Coimbra, Portugal
| | - Miguel Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s − PT Government Associate Laboratory, 4805-017 Braga/Guimarães, Portugal
| | - José Pedro Gil
- Stockholm Malaria Center, Department of Microbiology and Tumour Cell Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Maria Isabel Veiga
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Campus de Gualtar, 4710-057 Braga, Portugal
- ICVS/3B’s − PT Government Associate Laboratory, 4805-017 Braga/Guimarães, Portugal
| | - Susana O. Catarino
- Microelectromechanical Systems Research Unit (CMEMS-UMinho), School of Engineering, University of Minho, Campus de Azurém, 4800-058 Guimarães, Portugal
| |
Collapse
|
24
|
González-Bacerio J, Izquierdo M, Aguado ME, Varela AC, González-Matos M, Del Rivero MA. Using microbial metalo-aminopeptidases as targets in human infectious diseases. MICROBIAL CELL 2021; 8:239-246. [PMID: 34692819 PMCID: PMC8485470 DOI: 10.15698/mic2021.10.761] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 03/23/2021] [Revised: 07/22/2021] [Accepted: 07/28/2021] [Indexed: 11/13/2022]
Abstract
Several microbial metalo-aminopeptidases are emerging as novel targets for the treatment of human infectious diseases. Some of them are well validated as targets and some are not; some are essential enzymes and others are important for virulence and pathogenesis. For another group, it is not clear if their enzymatic activity is involved in the critical functions that they mediate. But one aspect has been established: they display relevant roles in bacteria and protozoa that could be targeted for therapeutic purposes. This work aims to describe these biological functions for several microbial metalo-aminopeptidases.
Collapse
Affiliation(s)
- Jorge González-Bacerio
- Center for Protein Studies, Faculty of Biology, University of Havana, calle 25 #455 entre I y J, 10400, Vedado, La Habana, Cuba.,Department of Biochemistry, Faculty of Biology, University of Havana, calle 25 #455 entre I y J, 10400, Vedado, La Habana, Cuba
| | - Maikel Izquierdo
- Center for Protein Studies, Faculty of Biology, University of Havana, calle 25 #455 entre I y J, 10400, Vedado, La Habana, Cuba
| | - Mirtha Elisa Aguado
- Center for Protein Studies, Faculty of Biology, University of Havana, calle 25 #455 entre I y J, 10400, Vedado, La Habana, Cuba
| | - Ana C Varela
- Center for Protein Studies, Faculty of Biology, University of Havana, calle 25 #455 entre I y J, 10400, Vedado, La Habana, Cuba
| | - Maikel González-Matos
- Center for Protein Studies, Faculty of Biology, University of Havana, calle 25 #455 entre I y J, 10400, Vedado, La Habana, Cuba
| | - Maday Alonso Del Rivero
- Center for Protein Studies, Faculty of Biology, University of Havana, calle 25 #455 entre I y J, 10400, Vedado, La Habana, Cuba
| |
Collapse
|
25
|
Garten M, Beck JR. Structured to conquer: transport across the Plasmodium parasitophorous vacuole. Curr Opin Microbiol 2021; 63:181-188. [PMID: 34375857 PMCID: PMC8463430 DOI: 10.1016/j.mib.2021.07.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2021] [Revised: 07/13/2021] [Accepted: 07/19/2021] [Indexed: 11/20/2022]
Abstract
During the vertebrate stage of the Plasmodium life cycle, obligate intracellular malaria parasites establish a vacuolar niche for replication, first within host hepatocytes at the pre-patent liver-stage and subsequently in erythrocytes during the pathogenic blood-stage. Survival in this protective microenvironment requires diverse transport mechanisms that enable the parasite to transcend the vacuolar barrier. Effector proteins exported out of the vacuole modify the erythrocyte membrane, increasing access to serum nutrients which then cross the vacuole membrane through a nutrient-permeable channel, supporting rapid parasite growth. This review highlights the most recent insights into the organization of the parasite vacuole to facilitate the solute, lipid and effector protein trafficking that establishes a nutrition pipeline in the terminally differentiated, organelle-free red blood cell.
Collapse
Affiliation(s)
- Matthias Garten
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, 20892, USA.
| | - Josh R Beck
- Department of Biomedical Sciences, Iowa State University, Ames, IA, 50011, USA.
| |
Collapse
|
26
|
Sinha S, Gautam CS, Sehgal R. L-cysteine whether a nutritional booster or a radical scavenger for Plasmodium. Trop Parasitol 2021; 11:19-24. [PMID: 34195056 PMCID: PMC8213117 DOI: 10.4103/tp.tp_20_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 10/13/2020] [Accepted: 10/24/2020] [Indexed: 11/13/2022] Open
Abstract
Introduction: Plasmodium falciparum is the most noxious species among other Plasmodium species that cause malaria. Attention is required to understand more about the pathophysiology and parasite biology to obscure this disease. The fact is, very little is known about the nutritional requirement in sense of carbohydrate, lipid, nucleic acid, and amino acid metabolism that regulate the growth of parasite and out of this, studies related to the metabolism of amino acid are exceptionally limited. Out of several amino acids, L-cysteine is essential for the continuous erythrocytic growth of Plasmodium. However, the exact role of L-cysteine in regulating the growth of Plasmodium is unknown. Here, we tried to investigate how does L-cysteine affects the growth of Plasmodium in in vitro culture, and also the study was aimed to find whether there is a synergism with chloroquine on the Plasmodium growth in vitro. Materials and Methods: Parasite inhibition assay based on schizont maturation inhibition following WHO protocol on P. falciparum chloroquine-sensitive strain (MRC-2) was employed to determine IC50 value and drug interaction pattern was shown through fractional inhibitory concentration index. Results: Inhibitory effect of L-cysteine hydrochloride on Plasmodium growth was depicted with IC50 1.152 ± 0.287 μg/mL and the most synergistic pattern of interaction was shown with chloroquine. Conclusions: The present study anticipates two important findings, firstly inconsistent results from previous findings and secondly, synergistic effect with chloroquine suggests its potency that may be used as an add-on therapy along with chloroquine. However, further study is needed to validate the above findings in vivo models.
Collapse
Affiliation(s)
- Shweta Sinha
- Department of Medical Parasitology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| | - C S Gautam
- Department of Pharmacology, Government Medical College and Hospital, Chandigarh, India
| | - Rakesh Sehgal
- Department of Medical Parasitology, Postgraduate Institute of Medical Education and Research, Chandigarh, India
| |
Collapse
|
27
|
Abstract
Obligate intracellular malaria parasites reside within a vacuolar compartment generated during invasion which is the principal interface between pathogen and host. To subvert their host cell and support their metabolism, these parasites coordinate a range of transport activities at this membrane interface that are critically important to parasite survival and virulence, including nutrient import, waste efflux, effector protein export, and uptake of host cell cytosol. Here, we review our current understanding of the transport mechanisms acting at the malaria parasite vacuole during the blood and liver-stages of development with a particular focus on recent advances in our understanding of effector protein translocation into the host cell by the Plasmodium Translocon of EXported proteins (PTEX) and small molecule transport by the PTEX membrane-spanning pore EXP2. Comparison to Toxoplasma gondii and other related apicomplexans is provided to highlight how similar and divergent mechanisms are employed to fulfill analogous transport activities.
Collapse
Affiliation(s)
- Josh R. Beck
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, United States of America
| | - Chi-Min Ho
- Department of Microbiology and Immunology, Vagelos College of Physicians and Surgeons, Columbia University, New York, New York, United States of America
| |
Collapse
|
28
|
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: 0.8] [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
|
29
|
Bouyer G, Barbieri D, Dupuy F, Marteau A, Sissoko A, N'Dri ME, Neveu G, Bedault L, Khodabux N, Roman D, Houzé S, Siciliano G, Alano P, Martins RM, Lopez-Rubio JJ, Clain J, Duval R, Egée S, Lavazec C. Plasmodium falciparum sexual parasites regulate infected erythrocyte permeability. Commun Biol 2020; 3:726. [PMID: 33262483 PMCID: PMC7708629 DOI: 10.1038/s42003-020-01454-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Accepted: 10/30/2020] [Indexed: 11/28/2022] Open
Abstract
To ensure the transport of nutrients necessary for their survival, Plasmodium falciparum parasites increase erythrocyte permeability to diverse solutes. These new permeation pathways (NPPs) have been extensively characterized in the pathogenic asexual parasite stages, however the existence of NPPs has never been investigated in gametocytes, the sexual stages responsible for transmission to mosquitoes. Here, we show that NPPs are still active in erythrocytes infected with immature gametocytes and that this activity declines along gametocyte maturation. Our results indicate that NPPs are regulated by cyclic AMP (cAMP) signaling cascade, and that the decrease in cAMP levels in mature stages results in a slowdown of NPP activity. We also show that NPPs facilitate the uptake of artemisinin derivatives and that phosphodiesterase (PDE) inhibitors can reactivate NPPs and increase drug uptake in mature gametocytes. These processes are predicted to play a key role in P. falciparum gametocyte biology and susceptibility to antimalarials. Bouyer et al. report that the new permeation pathways (NPP), responsible of modulating erythrocyte permeability to diverse solutes and considered only to be in pathogenic asexual stages of P. falciparum, are also active in erythrocytes infected with immature gametocytes and this activity declines with gametocyte maturation. NPPs are regulated by the cAMP signalling cascade, and the decrease in cAMP levels in mature stages slows NPP activity.
Collapse
Affiliation(s)
- Guillaume Bouyer
- Sorbonne Université, CNRS UMR 8227, Station Biologique de Roscoff, Roscoff, France.,Laboratoire d'excellence GR-Ex, Paris, France
| | - Daniela Barbieri
- Laboratoire d'excellence GR-Ex, Paris, France.,Université de Paris, Inserm U1016, CNRS UMR 8104, Institut Cochin, Paris, France
| | - Florian Dupuy
- Laboratoire d'excellence GR-Ex, Paris, France.,Université de Paris, Inserm U1016, CNRS UMR 8104, Institut Cochin, Paris, France
| | - Anthony Marteau
- Laboratoire d'excellence GR-Ex, Paris, France.,Université de Paris, Inserm U1016, CNRS UMR 8104, Institut Cochin, Paris, France
| | - Abdoulaye Sissoko
- Laboratoire d'excellence GR-Ex, Paris, France.,Université de Paris, IRD 261, MERIT, Paris, France
| | - Marie-Esther N'Dri
- Laboratoire d'excellence GR-Ex, Paris, France.,Université de Paris, Inserm U1016, CNRS UMR 8104, Institut Cochin, Paris, France
| | - Gaelle Neveu
- Laboratoire d'excellence GR-Ex, Paris, France.,Université de Paris, Inserm U1016, CNRS UMR 8104, Institut Cochin, Paris, France
| | - Laurianne Bedault
- Laboratoire d'excellence GR-Ex, Paris, France.,Université de Paris, Inserm U1016, CNRS UMR 8104, Institut Cochin, Paris, France
| | - Nabiha Khodabux
- Laboratoire d'excellence GR-Ex, Paris, France.,Université de Paris, Inserm U1016, CNRS UMR 8104, Institut Cochin, Paris, France
| | - Diana Roman
- Laboratoire d'excellence GR-Ex, Paris, France.,Université de Paris, IRD 261, MERIT, Paris, France
| | - Sandrine Houzé
- Laboratoire d'excellence GR-Ex, Paris, France.,Université de Paris, IRD 261, MERIT, Paris, France
| | | | | | - Rafael M Martins
- Université de Montpellier 1 & 2, CNRS 5290, IRD 224, MIVEGEC, Montpellier, France
| | | | - Jérome Clain
- Laboratoire d'excellence GR-Ex, Paris, France.,Université de Paris, IRD 261, MERIT, Paris, France
| | - Romain Duval
- Laboratoire d'excellence GR-Ex, Paris, France.,Université de Paris, IRD 261, MERIT, Paris, France
| | - Stéphane Egée
- Sorbonne Université, CNRS UMR 8227, Station Biologique de Roscoff, Roscoff, France.,Laboratoire d'excellence GR-Ex, Paris, France
| | - Catherine Lavazec
- Laboratoire d'excellence GR-Ex, Paris, France. .,Université de Paris, Inserm U1016, CNRS UMR 8104, Institut Cochin, Paris, France.
| |
Collapse
|
30
|
Gómez F, Silva LS, Teixeira DE, Agero U, Pinheiro AAS, Viana NB, Pontes B. Plasmodium falciparum maturation across the intra-erythrocytic cycle shifts the soft glassy viscoelastic properties of red blood cells from a liquid-like towards a solid-like behavior. Exp Cell Res 2020; 397:112370. [PMID: 33186602 DOI: 10.1016/j.yexcr.2020.112370] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 11/06/2020] [Accepted: 11/09/2020] [Indexed: 11/26/2022]
Abstract
The mechanical properties of erythrocytes have been investigated by different techniques. However, there are few reports on how the viscoelasticity of these cells varies during malaria disease. Here, we quantitatively map the viscoelastic properties of Plasmodium falciparum-parasitized human erythrocytes. We apply new methodologies based on optical tweezers to measure the viscoelastic properties and defocusing microscopy to measure the erythrocyte height profile, the overall cell volume, and its form factor, a crucial parameter to convert the complex elastic constant into complex shear modulus. The storage and loss shear moduli are obtained for each stage of parasite maturation inside red blood cells, while the former increase, the latter decrease. Employing a soft glassy rheology model, we obtain the power-law exponent for the storage and loss shear moduli, characterizing the soft glassy features of red blood cells in each parasite maturation stage. Ring forms present a liquid-like behavior, with a slightly lower power-law exponent than healthy erythrocytes, whereas trophozoite and schizont stages exhibit increasingly solid-like behaviors. Finally, the surface elastic shear moduli, low-frequency surface viscosities, and shape recovery relaxation times all increase not only in a stage-dependent manner but also when compared to healthy red blood cells. Overall, the results call attention to the soft glassy characteristics of Plasmodium falciparum-parasitized erythrocyte membrane and may provide a basis for future studies to better understand malaria disease from a mechanobiological perspective.
Collapse
Affiliation(s)
- Fran Gómez
- Instituto de Física, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-972, Brazil; Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Leandro S Silva
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Douglas E Teixeira
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Ubirajara Agero
- Instituto de Ciências Exatas, Departamento de Física, Universidade Federal de Minas Gerais, Belo Horizonte, MG, 31270-901, Brazil
| | - Ana Acácia S Pinheiro
- Instituto de Biofísica Carlos Chagas Filho, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil
| | - Nathan B Viana
- Instituto de Física, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-972, Brazil; Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil.
| | - Bruno Pontes
- Centro Nacional de Biologia Estrutural e Bioimagem (CENABIO), Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil; Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, RJ, 21941-902, Brazil.
| |
Collapse
|
31
|
Pádua TA, Souza MC. Heme on Pulmonary Malaria: Friend or Foe? Front Immunol 2020; 11:1835. [PMID: 32983096 PMCID: PMC7477073 DOI: 10.3389/fimmu.2020.01835] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2020] [Accepted: 07/08/2020] [Indexed: 12/19/2022] Open
Abstract
Malaria is a hemolytic disease that, in severe cases, can compromise multiple organs. Pulmonary distress is a common symptom observed in severe malaria caused by Plasmodium vivax or Plasmodium falciparum. However, biological components involved in the development of lung malaria are poorly studied. In experimental models of pulmonary malaria, it was observed that parasitized red blood cell-congested pulmonary capillaries are related to intra-alveolar hemorrhages and inflammatory cell infiltration. Thus, it is very likely that hemolysis participates in malaria-induced acute lung injury. During malaria, heme assumes different biochemical structures such as hemin and hemozoin (biocrystallized structure of heme inside Plasmodium sp.). Each heme-derived structure triggers a different biological effect: on the one hand, hemozoin found in lung tissue is responsible for the infiltration of inflammatory cells and consequent tissue injury; on the other hand, heme stimulates heme oxygenase-1 (HO-1) expression and CO production, which protect mice from severe malaria. In this review, we discuss the biological mechanism involved in the dual role of heme response in experimental malaria-induced acute lung injury.
Collapse
Affiliation(s)
- Tatiana Almeida Pádua
- Laboratory of Applied Pharmacology, Institute of Drug Technology (Farmanguinhos), Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| | - Mariana Conceição Souza
- Laboratory of Applied Pharmacology, Institute of Drug Technology (Farmanguinhos), Oswaldo Cruz Foundation, Rio de Janeiro, Brazil
| |
Collapse
|
32
|
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: 9.4] [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
|
33
|
Giannangelo C, Siddiqui G, De Paoli A, Anderson BM, Edgington-Mitchell LE, Charman SA, Creek DJ. System-wide biochemical analysis reveals ozonide antimalarials initially act by disrupting Plasmodium falciparum haemoglobin digestion. PLoS Pathog 2020; 16:e1008485. [PMID: 32589689 PMCID: PMC7347234 DOI: 10.1371/journal.ppat.1008485] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 07/09/2020] [Accepted: 05/13/2020] [Indexed: 01/23/2023] Open
Abstract
Ozonide antimalarials, OZ277 (arterolane) and OZ439 (artefenomel), are synthetic peroxide-based antimalarials with potent activity against the deadliest malaria parasite, Plasmodium falciparum. Here we used a "multi-omics" workflow, in combination with activity-based protein profiling (ABPP), to demonstrate that peroxide antimalarials initially target the haemoglobin (Hb) digestion pathway to kill malaria parasites. Time-dependent metabolomic profiling of ozonide-treated P. falciparum infected red blood cells revealed a rapid depletion of short Hb-derived peptides followed by subsequent alterations in lipid and nucleotide metabolism, while untargeted peptidomics showed accumulation of longer Hb-derived peptides. Quantitative proteomics and ABPP assays demonstrated that Hb-digesting proteases were increased in abundance and activity following treatment, respectively. Ozonide-induced depletion of short Hb-derived peptides was less extensive in a drug-treated K13-mutant artemisinin resistant parasite line (Cam3.IIR539T) than in the drug-treated isogenic sensitive strain (Cam3.IIrev), further confirming the association between ozonide activity and Hb catabolism. To demonstrate that compromised Hb catabolism may be a primary mechanism involved in ozonide antimalarial activity, we showed that parasites forced to rely solely on Hb digestion for amino acids became hypersensitive to short ozonide exposures. Quantitative proteomics analysis also revealed parasite proteins involved in translation and the ubiquitin-proteasome system were enriched following drug treatment, suggestive of the parasite engaging a stress response to mitigate ozonide-induced damage. Taken together, these data point to a mechanism of action involving initial impairment of Hb catabolism, and indicate that the parasite regulates protein turnover to manage ozonide-induced damage.
Collapse
Affiliation(s)
- Carlo Giannangelo
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Ghizal Siddiqui
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Amanda De Paoli
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Bethany M. Anderson
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
| | - Laura E. Edgington-Mitchell
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Parkville, Victoria, Australia
- Department of Maxillofacial Surgery, College of Dentistry, New York University, New York, New York, United States of America
| | - Susan A. Charman
- Centre for Drug Candidate Optimisation, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| | - Darren J. Creek
- Drug Delivery, Disposition and Dynamics, Monash Institute of Pharmaceutical Sciences, Monash University, Parkville, Victoria, Australia
| |
Collapse
|
34
|
Spielmann T, Gras S, Sabitzki R, Meissner M. Endocytosis in Plasmodium and Toxoplasma Parasites. Trends Parasitol 2020; 36:520-532. [DOI: 10.1016/j.pt.2020.03.010] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2020] [Revised: 03/25/2020] [Accepted: 03/26/2020] [Indexed: 02/08/2023]
|
35
|
Aminobenzosuberone derivatives as PfA-M1 inhibitors: Molecular recognition and antiplasmodial evaluation. Bioorg Chem 2020; 98:103750. [DOI: 10.1016/j.bioorg.2020.103750] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Revised: 01/27/2020] [Accepted: 03/09/2020] [Indexed: 12/16/2022]
|
36
|
Fröhlich B, Yang Y, Thoma J, Czajor J, Lansche C, Sanchez C, Lanzer M, Cloetens P, Tanaka M. Nanofocused Scanning X-ray Fluorescence Microscopy Revealing an Effect of Heterozygous Hemoglobin S and C on Biochemical Activities in Plasmodium falciparum-Infected Erythrocytes. Anal Chem 2020; 92:5765-5771. [PMID: 32202408 DOI: 10.1021/acs.analchem.9b05111] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
While there is ample evidence suggesting that carriers of heterozygous hemoglobin S and C are protected from life-threatening malaria, little is known about the underlying biochemical mechanisms at the single cell level. Using nanofocused scanning X-ray fluorescence microscopy, we quantify the spatial distribution of individual elements in subcellular compartments, including Fe, S, P, Zn, and Cu, in Plasmodium falciparum-infected (P. falciparum-infected) erythrocytes carrying the wild type or variant hemoglobins. Our data indicate that heterozygous hemoglobin S and C significantly modulate biochemical reactions in parasitized erythrocytes, such as aberrant hemozoin mineralization and a delay in hemoglobin degradation. The label-free scanning X-ray fluorescence imaging has great potential to quantify the spatial distribution of elements in subcellular compartments of P. falciparum-infected erythrocytes and unravel the biochemical mechanisms underpinning disease and protective traits.
Collapse
Affiliation(s)
- Benjamin Fröhlich
- Physical Chemistry of Biosystems, Institute of Physical Chemistry, Heidelberg University, D-69120 Heidelberg, Germany
| | - Yang Yang
- The European Synchrotron Radiation Facility (ESRF), 38043 Grenoble, France.,National Synchrotron Light Source II, Brookhaven National Laboratory, Upton, New York 11973, United States
| | - Judith Thoma
- Physical Chemistry of Biosystems, Institute of Physical Chemistry, Heidelberg University, D-69120 Heidelberg, Germany
| | - Julian Czajor
- Physical Chemistry of Biosystems, Institute of Physical Chemistry, Heidelberg University, D-69120 Heidelberg, Germany
| | - Christine Lansche
- Department of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, D-69120 Heidelberg, Germany
| | - Cecilia Sanchez
- Department of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, D-69120 Heidelberg, Germany
| | - Michael Lanzer
- Department of Infectious Diseases, Parasitology, Universitätsklinikum Heidelberg, D-69120 Heidelberg, Germany
| | - Peter Cloetens
- The European Synchrotron Radiation Facility (ESRF), 38043 Grenoble, France
| | - Motomu Tanaka
- Physical Chemistry of Biosystems, Institute of Physical Chemistry, Heidelberg University, D-69120 Heidelberg, Germany.,Center for Integrative Medicine and Physics, Institute for Advanced Study, Kyoto University, 606-8501 Kyoto, Japan
| |
Collapse
|
37
|
Brown AC, Moore CC, Guler JL. Cholesterol-dependent enrichment of understudied erythrocytic stages of human Plasmodium parasites. Sci Rep 2020; 10:4591. [PMID: 32165667 PMCID: PMC7067793 DOI: 10.1038/s41598-020-61392-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2019] [Accepted: 02/26/2020] [Indexed: 01/01/2023] Open
Abstract
For intracellular pathogens, the host cell provides needed protection and nutrients. A major challenge of intracellular parasite research is collection of high parasite numbers separated from host contamination. This situation is exemplified by the malaria parasite, which spends a substantial part of its life cycle inside erythrocytes as rings, trophozoites, and schizonts, before egress and reinvasion. Erythrocytic Plasmodium parasite forms refractory to enrichment remain understudied due to high host contamination relative to low parasite numbers. Here, we present a method for separating all stages of Plasmodium-infected erythrocytes through lysis and removal of uninfected erythrocytes. The Streptolysin O-Percoll (SLOPE) method is effective on previously inaccessible forms, including circulating rings from malaria-infected patients and artemisinin-induced quiescent parasites. SLOPE can be used on multiple parasite species, under multiple media formulations, and lacks measurable impacts on parasite viability. We demonstrate erythrocyte membrane cholesterol levels modulate the preferential lysis of uninfected host cells by SLO, and therefore modulate the effectiveness of SLOPE. Targeted metabolomics of SLOPE-enriched ring stage samples confirms parasite-derived metabolites are increased and contaminating host material is reduced compared to non-enriched samples. Due to consumption of cholesterol by other intracellular bacteria and protozoa, SLOPE holds potential for improving research on organisms beyond Plasmodium.
Collapse
Affiliation(s)
- Audrey C Brown
- Department of Biology, University of Virginia, Charlottesville, VA, USA
| | - Christopher C Moore
- Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA
| | - Jennifer L Guler
- Department of Biology, University of Virginia, Charlottesville, VA, USA. .,Division of Infectious Diseases and International Health, University of Virginia, Charlottesville, VA, USA.
| |
Collapse
|
38
|
Oyegue-Liabagui SL, Imboumy-Limoukou RK, Kouna CL, Bangueboussa F, Schmitt M, Florent I, Lekana-Douki JB. IgG antibody response against Plasmodium falciparum aminopeptidase 1 antigen in Gabonese children living in Makokou and Franceville. Clin Exp Immunol 2020; 200:287-298. [PMID: 32027020 DOI: 10.1111/cei.13425] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/15/2020] [Indexed: 11/28/2022] Open
Abstract
The search for novel chemical classes of anti-malarial compounds to cope with the current state of chemoresistance of malaria parasites has led to the identification of Plasmodium falciparum aminopeptidase 1 (PfA-M1) as a new therapeutic target. PfA-M1, known to be involved in the hemoglobin digestion cascade which helps to provide most of the amino acids necessary to the parasite's metabolism, is currently considered as a promising target for anti-malarial chemotherapy. However, its immunogenic properties have not yet been tested in the Gabonese population. In Gabon, the prevalence of malaria remains three times higher in semi-urban areas (60·12%) than in urban areas (17·06%). We show that malaria-specific PfA-M1 antibodies are present in children and increase with the level of infection. Children living in semi-urban areas have higher anti-PfA-M1 antibody titers (0·14 ± 0·02 AU) than those living in urban areas (0·08 ± 0·02 AU, P = 0·03), and their antibody titers increase with age (P < 0·0001). Moreover, anti-PfA-M1 antibody titers decrease in children with hyperparasitemia (0·027 ± 0·055 AU) but they remain high in children with low parasite density (0·21 ± 0·034 AU, P = 0·034). In conclusion, our results suggest that malaria-specific PfA-M1 antibodies may play an important role in the immune response of the host against P. falciparum in Gabonese children. Further studies on the role of PfA-M1 during anemia are needed.
Collapse
Affiliation(s)
- S L Oyegue-Liabagui
- Laboratoire de Recherches en Immunologie, Parasitologie et Microbiologie, Ecole Doctorale Régionale d'Afrique Centrale en Infectiologie Tropicale (ECODRAC), Université des Sciences et Techniques de Masuku, Franceville, Gabon
| | - R-K Imboumy-Limoukou
- Unité d'Evolution Epidémiologie et Résistances Parasitaires (UNEEREP), Centre International de Recherches Médicales de Franceville (CIRMF), Franceville, Gabon
| | - C L Kouna
- Unité d'Evolution Epidémiologie et Résistances Parasitaires (UNEEREP), Centre International de Recherches Médicales de Franceville (CIRMF), Franceville, Gabon
| | - F Bangueboussa
- Laboratoire de Recherches en Immunologie, Parasitologie et Microbiologie, Ecole Doctorale Régionale d'Afrique Centrale en Infectiologie Tropicale (ECODRAC), Université des Sciences et Techniques de Masuku, Franceville, Gabon.,Unité d'Evolution Epidémiologie et Résistances Parasitaires (UNEEREP), Centre International de Recherches Médicales de Franceville (CIRMF), Franceville, Gabon
| | - M Schmitt
- Université de Haute-Alsace, Université de Strasbourg, Mulhouse, France
| | - I Florent
- Molécules de Communication et Adaptation des Microorganismes (MCAM, UMR 7245), Muséum National d'Histoire Naturelle, Paris, France
| | - J B Lekana-Douki
- Unité d'Evolution Epidémiologie et Résistances Parasitaires (UNEEREP), Centre International de Recherches Médicales de Franceville (CIRMF), Franceville, Gabon.,Département de Parasitologie-Mycologie, Université des Sciences de la Santé, Libreville, Gabon
| |
Collapse
|
39
|
Goldberg DE, Zimmerberg J. Hardly Vacuous: The Parasitophorous Vacuolar Membrane of Malaria Parasites. Trends Parasitol 2020; 36:138-146. [PMID: 31866184 PMCID: PMC6937376 DOI: 10.1016/j.pt.2019.11.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2019] [Revised: 11/27/2019] [Accepted: 11/28/2019] [Indexed: 12/30/2022]
Abstract
When a malaria parasite invades a host erythrocyte it pushes itself in and invaginates a portion of the host membrane, thereby sealing itself inside and establishing itself in the resulting vacuole. The parasitophorous vacuolar membrane (PVM) that surrounds the parasite is modified by the parasite, using its secretory organelles. To survive within this enveloping membrane, the organism must take in nutrients, secrete wastes, export proteins into the host cell, and eventually egress. Here, we review current understanding of the unique solutions Plasmodium has evolved to these challenges and discuss the remaining questions.
Collapse
Affiliation(s)
- Daniel E Goldberg
- Departments of Medicine and Molecular Microbiology, Washington University School of Medicine, St Louis, MO, USA.
| | - Joshua Zimmerberg
- Section on Integrative Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
40
|
Martin RE. The transportome of the malaria parasite. Biol Rev Camb Philos Soc 2019; 95:305-332. [PMID: 31701663 DOI: 10.1111/brv.12565] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2019] [Revised: 10/02/2019] [Accepted: 10/04/2019] [Indexed: 12/15/2022]
Abstract
Membrane transport proteins, also known as transporters, control the movement of ions, nutrients, metabolites, and waste products across the membranes of a cell and are central to its biology. Proteins of this type also serve as drug targets and are key players in the phenomenon of drug resistance. The malaria parasite has a relatively reduced transportome, with only approximately 2.5% of its genes encoding transporters. Even so, assigning functions and physiological roles to these proteins, and ascertaining their contributions to drug action and drug resistance, has been very challenging. This review presents a detailed critique and synthesis of the disruption phenotypes, protein subcellular localisations, protein functions (observed or predicted), and links to antimalarial drug resistance for each of the parasite's transporter genes. The breadth and depth of the gene disruption data are particularly impressive, with at least one phenotype determined in the parasite's asexual blood stage for each transporter gene, and multiple phenotypes available for 76% of the genes. Analysis of the curated data set revealed there to be relatively little redundancy in the Plasmodium transportome; almost two-thirds of the parasite's transporter genes are essential or required for normal growth in the asexual blood stage of the parasite, and this proportion increased to 78% when the disruption phenotypes available for the other parasite life stages were included in the analysis. These observations, together with the finding that 22% of the transportome is implicated in the parasite's resistance to existing antimalarials and/or drugs within the development pipeline, indicate that transporters are likely to serve, or are already serving, as drug targets. Integration of the different biological and bioinformatic data sets also enabled the selection of candidates for transport processes known to be essential for parasite survival, but for which the underlying proteins have thus far remained undiscovered. These include potential transporters of pantothenate, isoleucine, or isopentenyl diphosphate, as well as putative anion-selective channels that may serve as the pore component of the parasite's 'new permeation pathways'. Other novel insights into the parasite's biology included the identification of transporters for the potential development of antimalarial treatments, transmission-blocking drugs, prophylactics, and genetically attenuated vaccines. The syntheses presented herein set a foundation for elucidating the functions and physiological roles of key members of the Plasmodium transportome and, ultimately, to explore and realise their potential as therapeutic targets.
Collapse
Affiliation(s)
- Rowena E Martin
- Research School of Biology, Australian National University, Canberra, Australian Capital Territory, Australia
| |
Collapse
|
41
|
Shaikh IU, Patel RK, Mevada VA, Gupta VK, Jadeja RN. Binary and Ternary Zinc(II) Complexes of Acyl Pyrazolones: Synthesis, Spectroscopic Analysis, Crystal Structure and Antimalarial Activity. ChemistrySelect 2019. [DOI: 10.1002/slct.201901058] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Affiliation(s)
- Irfan U. Shaikh
- Department of ChemistryFaculty of ScienceThe M.S. University of Baroda, Vadodara 390 002 India
| | - Rajesh K. Patel
- Department of BioscienceVeer Narmad South Gujarat University Surat- 395007 India
| | - Vishal A. Mevada
- Super Computing FacilityVeer Narmad South Gujarat University Surat- 395007 India
| | - Vivek K. Gupta
- Post-Graduate Department of Physics & ElectronicsUniversity of Jammu Jammu Tawi 180 006 India
| | - Rajendrasinh N. Jadeja
- Department of ChemistryFaculty of ScienceThe M.S. University of Baroda, Vadodara 390 002 India
| |
Collapse
|
42
|
Kennedy K, Cobbold SA, Hanssen E, Birnbaum J, Spillman NJ, McHugh E, Brown H, Tilley L, Spielmann T, McConville MJ, Ralph SA. Delayed death in the malaria parasite Plasmodium falciparum is caused by disruption of prenylation-dependent intracellular trafficking. PLoS Biol 2019; 17:e3000376. [PMID: 31318858 PMCID: PMC6667170 DOI: 10.1371/journal.pbio.3000376] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2019] [Revised: 07/30/2019] [Accepted: 07/01/2019] [Indexed: 02/07/2023] Open
Abstract
Apicomplexan parasites possess a plastid organelle called the apicoplast. Inhibitors that selectively target apicoplast housekeeping functions, including DNA replication and protein translation, are lethal for the parasite, and several (doxycycline, clindamycin, and azithromycin) are in clinical use as antimalarials. A major limitation of such drugs is that treated parasites only arrest one intraerythrocytic development cycle (approximately 48 hours) after treatment commences, a phenotype known as the ‘delayed death’ effect. The molecular basis of delayed death is a long-standing mystery in parasitology, and establishing the mechanism would aid rational clinical implementation of apicoplast-targeted drugs. Parasites undergoing delayed death transmit defective apicoplasts to their daughter cells and cannot produce the sole, blood-stage essential metabolic product of the apicoplast: the isoprenoid precursor isopentenyl-pyrophosphate. How the isoprenoid precursor depletion kills the parasite remains unknown. We investigated the requirements for the range of isoprenoids in the human malaria parasite Plasmodium falciparum and characterised the molecular and morphological phenotype of parasites experiencing delayed death. Metabolomic profiling reveals disruption of digestive vacuole function in the absence of apicoplast derived isoprenoids. Three-dimensional electron microscopy reveals digestive vacuole fragmentation and the accumulation of cytostomal invaginations, characteristics common in digestive vacuole disruption. We show that digestive vacuole disruption results from a defect in the trafficking of vesicles to the digestive vacuole. The loss of prenylation of vesicular trafficking proteins abrogates their membrane attachment and function and prevents the parasite from feeding. Our data show that the proximate cause of delayed death is an interruption of protein prenylation and consequent cellular trafficking defects. After treatment with drugs that target apicoplast functions, malaria parasites are initially superficially healthy and go on to infect new erythrocytes. This cell biology study shows that the parasites subsequently die in their second cycle due to trafficking defects caused by depletion of prenyl groups.
Collapse
Affiliation(s)
- Kit Kennedy
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, Australia
| | - Simon A. Cobbold
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, Australia
| | - Eric Hanssen
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, Australia
- Advanced Microscopy Facility, Bio21 Molecular Science and Biotechnology Institute, Victoria, Australia
| | - Jakob Birnbaum
- Molecular Biology and Immunology Section, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Natalie J. Spillman
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, Australia
| | - Emma McHugh
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, Australia
| | - Hannah Brown
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, Australia
| | - Leann Tilley
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, Australia
| | - Tobias Spielmann
- Molecular Biology and Immunology Section, Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Malcolm J. McConville
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, Australia
| | - Stuart A. Ralph
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, The University of Melbourne, Victoria, Australia
- * E-mail:
| |
Collapse
|
43
|
Inhibitory Mechanisms of DHA/CQ on pH and Iron Homeostasis of Erythrocytic Stage Growth of Plasmodium Falciparum. Molecules 2019; 24:molecules24101941. [PMID: 31137574 PMCID: PMC6571875 DOI: 10.3390/molecules24101941] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2019] [Revised: 05/11/2019] [Accepted: 05/13/2019] [Indexed: 12/19/2022] Open
Abstract
Malaria is an infectious disease caused by Plasmodium group. The mechanisms of antimalarial drugs DHA/CQ are still unclear today. The inhibitory effects (IC50) of single treatments with DHA/CQ or V-ATPase inhibitor Baf-A1 or combination treatments by DHA/CQ combined with Baf-A1 on the growth of Plasmodium falciparum strain 3D7 was investigated. Intracellular cytoplasmic pH and labile iron pool (LIP) were labeled by pH probe BCECF, AM and iron probe calcein, AM, the fluorescence of the probes was measured by FCM. The effects of low doses of DHA (0.2 nM, 0.4 nM, 0.8 nM) on gene expression of V-ATPases (vapE, vapA, vapG) located in the membrane of DV were tested by RT-qPCR. DHA combined with Baf-A1 showed a synergism effect (CI = 0.524) on the parasite growth in the concentration of IC50. Intracellular pH and irons were effected significantly by different doses of DHA/Baf-A1. Intracellular pH was decreased by CQ combined with Baf-A1 in the concentration of IC50. Intracellular LIP was increased by DHA combined with Baf-A1 in the concentration of 20 IC50. The expression of gene vapA was down-regulated by all low doses of DHA (0.2/0.4/0.8 nM) significantly (p < 0.001) and the expression of vapG/vapE were up-regulated by 0.8 nM DHA significantly (p < 0.001). Interacting with ferrous irons, affecting the DV membrane proton pumping and acidic pH or cytoplasmic irons homeostasis may be the antimalarial mechanism of DHA while CQ showed an effect on cytoplasmic pH of parasite in vitro. Lastly, this article provides us preliminary results and a new idea for antimalarial drugs combination and new potential antimalarial combination therapies.
Collapse
|
44
|
Musyoka TM, Njuguna JN, Tastan Bishop Ö. Comparing sequence and structure of falcipains and human homologs at prodomain and catalytic active site for malarial peptide based inhibitor design. Malar J 2019; 18:159. [PMID: 31053072 PMCID: PMC6500056 DOI: 10.1186/s12936-019-2790-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2018] [Accepted: 04/23/2019] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Falcipains are major cysteine proteases of Plasmodium falciparum involved in haemoglobin degradation and remain attractive anti-malarial drug targets. Several inhibitors against these proteases have been identified, yet none of them has been approved for malaria treatment. Other Plasmodium species also possess highly homologous proteins to falcipains. For selective therapeutic targeting, identification of sequence and structure differences with homologous human cathepsins is necessary. The substrate processing activity of these proteins is tightly controlled via a prodomain segment occluding the active site which is chopped under low pH conditions exposing the catalytic site. Current work characterizes these proteases to identify residues mediating the prodomain regulatory function for the design of peptide based anti-malarial inhibitors. METHODS Sequence and structure variations between prodomain regions of plasmodial proteins and human cathepsins were determined using in silico approaches. Additionally, evolutionary clustering of these proteins was evaluated using phylogenetic analysis. High quality partial zymogen protein structures were modelled using homology modelling and residue interaction analysis performed between the prodomain segment and mature domain to identify key interacting residues between these two domains. The resulting information was used to determine short peptide sequences which could mimic the inherent regulatory function of the prodomain regions. Through flexible docking, the binding affinity of proposed peptides on the proteins studied was evaluated. RESULTS Sequence, evolutionary and motif analyses showed important differences between plasmodial and human proteins. Residue interaction analysis identified important residues crucial for maintaining prodomain integrity across the different proteins as well as the pro-segment responsible for inhibitory mechanism. Binding affinity of suggested peptides was highly dependent on their residue composition and length. CONCLUSIONS Despite the conserved structural and catalytic mechanism between human cathepsins and plasmodial proteases, current work revealed significant differences between the two protein groups which may provide valuable information for selective anti-malarial inhibitor development. Part of this study aimed to design peptide inhibitors based on endogenous inhibitory portions of protease prodomains as a novel aspect. Even though peptide inhibitors may not be practical solutions to malaria at this stage, the approach followed and results offer a promising means to find new malarial inhibitors.
Collapse
Affiliation(s)
- Thommas Mutemi Musyoka
- Research Unit in Bioinformatics (RUBi), Department of Biochemistry and Microbiology, Rhodes University, P.O. Box 94, Grahamstown, 6140, South Africa
| | - Joyce Njoki Njuguna
- Research Unit in Bioinformatics (RUBi), Department of Biochemistry and Microbiology, Rhodes University, P.O. Box 94, Grahamstown, 6140, South Africa
| | - Özlem Tastan Bishop
- Research Unit in Bioinformatics (RUBi), Department of Biochemistry and Microbiology, Rhodes University, P.O. Box 94, Grahamstown, 6140, South Africa.
| |
Collapse
|
45
|
Istvan ES, Das S, Bhatnagar S, Beck JR, Owen E, Llinas M, Ganesan SM, Niles JC, Winzeler E, Vaidya AB, Goldberg DE. Plasmodium Niemann-Pick type C1-related protein is a druggable target required for parasite membrane homeostasis. eLife 2019; 8:40529. [PMID: 30888318 PMCID: PMC6424564 DOI: 10.7554/elife.40529] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Accepted: 02/05/2019] [Indexed: 01/05/2023] Open
Abstract
Plasmodium parasites possess a protein with homology to Niemann-Pick Type C1 proteins (Niemann-Pick Type C1-Related protein, NCR1). We isolated parasites with resistance-conferring mutations in Plasmodium falciparum NCR1 (PfNCR1) during selections with three diverse small-molecule antimalarial compounds and show that the mutations are causative for compound resistance. PfNCR1 protein knockdown results in severely attenuated growth and confers hypersensitivity to the compounds. Compound treatment or protein knockdown leads to increased sensitivity of the parasite plasma membrane (PPM) to the amphipathic glycoside saponin and engenders digestive vacuoles (DVs) that are small and malformed. Immuno-electron microscopy and split-GFP experiments localize PfNCR1 to the PPM. Our experiments show that PfNCR1 activity is critically important for the composition of the PPM and is required for DV biogenesis, suggesting PfNCR1 as a novel antimalarial drug target. Editorial note: This article has been through an editorial process in which the authors decide how to respond to the issues raised during peer review. The Reviewing Editor's assessment is that all the issues have been addressed (see decision letter).
Collapse
Affiliation(s)
- Eva S Istvan
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, United States.,Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, United States
| | - Sudipta Das
- Department of Microbiology and Immunology, Center for Molecular Parasitology, Drexel University College of Medicine, Philadelphia, United States
| | - Suyash Bhatnagar
- Department of Microbiology and Immunology, Center for Molecular Parasitology, Drexel University College of Medicine, Philadelphia, United States
| | - Josh R Beck
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, United States.,Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, United States
| | - Edward Owen
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, United States.,Huck Center for Malaria Research, Pennsylvania State University, University Park, United States.,Department of Chemistry, Pennsylvania State University, University Park, United States
| | - Manuel Llinas
- Department of Biochemistry and Molecular Biology, Pennsylvania State University, University Park, United States.,Huck Center for Malaria Research, Pennsylvania State University, University Park, United States.,Department of Chemistry, Pennsylvania State University, University Park, United States
| | - Suresh M Ganesan
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, United States
| | - Jacquin C Niles
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, United States
| | - Elizabeth Winzeler
- Department of Pediatrics, University of California San Diego School of Medicine, La Jolla, United States
| | - Akhil B Vaidya
- Department of Microbiology and Immunology, Center for Molecular Parasitology, Drexel University College of Medicine, Philadelphia, United States
| | - Daniel E Goldberg
- Department of Medicine, Division of Infectious Diseases, Washington University School of Medicine, Saint Louis, United States.,Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, United States
| |
Collapse
|
46
|
Evidence for Regulation of Hemoglobin Metabolism and Intracellular Ionic Flux by the Plasmodium falciparum Chloroquine Resistance Transporter. Sci Rep 2018; 8:13578. [PMID: 30206341 PMCID: PMC6134138 DOI: 10.1038/s41598-018-31715-9] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Accepted: 08/22/2018] [Indexed: 11/30/2022] Open
Abstract
Plasmodium falciparum multidrug resistance constitutes a major obstacle to the global malaria elimination campaign. Specific mutations in the Plasmodium falciparum chloroquine resistance transporter (PfCRT) mediate resistance to the 4-aminoquinoline drug chloroquine and impact parasite susceptibility to several partner agents used in current artemisinin-based combination therapies, including amodiaquine. By examining gene-edited parasites, we report that the ability of the wide-spread Dd2 PfCRT isoform to mediate chloroquine and amodiaquine resistance is substantially reduced by the addition of the PfCRT L272F mutation, which arose under blasticidin selection. We also provide evidence that L272F confers a significant fitness cost to asexual blood stage parasites. Studies with amino acid-restricted media identify this mutant as a methionine auxotroph. Metabolomic analysis also reveals an accumulation of short, hemoglobin-derived peptides in the Dd2 + L272F and Dd2 isoforms, compared with parasites expressing wild-type PfCRT. Physiologic studies with the ionophores monensin and nigericin support an impact of PfCRT isoforms on Ca2+ release, with substantially reduced Ca2+ levels observed in Dd2 + L272F parasites. Our data reveal a central role for PfCRT in regulating hemoglobin catabolism, amino acid availability, and ionic balance in P. falciparum, in addition to its role in determining parasite susceptibility to heme-binding 4-aminoquinoline drugs.
Collapse
|
47
|
Kumar S, Bhardwaj TR, Prasad DN, Singh RK. Drug targets for resistant malaria: Historic to future perspectives. Biomed Pharmacother 2018; 104:8-27. [PMID: 29758416 DOI: 10.1016/j.biopha.2018.05.009] [Citation(s) in RCA: 58] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 04/22/2018] [Accepted: 05/07/2018] [Indexed: 01/05/2023] Open
Abstract
New antimalarial targets are the prime need for the discovery of potent drug candidates. In order to fulfill this objective, antimalarial drug researches are focusing on promising targets in order to develop new drug candidates. Basic metabolism and biochemical process in the malaria parasite, i.e. Plasmodium falciparum can play an indispensable role in the identification of these targets. But, the emergence of resistance to antimalarial drugs is an escalating comprehensive problem with the progress of antimalarial drug development. The development of resistance has highlighted the need for the search of novel antimalarial molecules. The pharmaceutical industries are committed to new drug development due to the global recognition of this life threatening resistance to the currently available antimalarial therapy. The recent developments in the understanding of parasite biology are exhilarating this resistance issue which is further being ignited by malaria genome project. With this background of information, this review was aimed to highlights and provides useful information on various present and promising treatment approaches for resistant malaria, new progresses, pursued by some innovative targets that have been explored till date. This review also discusses modern and futuristic multiple approaches to antimalarial drug discovery and development with pictorial presentations highlighting the various targets, that could be exploited for generating promising new drugs in the future for drug resistant malaria.
Collapse
Affiliation(s)
- Sahil Kumar
- School of Pharmacy and Emerging Sciences, Baddi University of Emerging Sciences & Technology, Baddi, Dist. Solan, 173205, Himachal Pradesh, India
| | - T R Bhardwaj
- School of Pharmacy and Emerging Sciences, Baddi University of Emerging Sciences & Technology, Baddi, Dist. Solan, 173205, Himachal Pradesh, India
| | - D N Prasad
- Department of Pharmaceutical Chemistry, Shivalik College of Pharmacy, Nangal, Dist. Rupnagar, 140126, Punjab, India
| | - Rajesh K Singh
- Department of Pharmaceutical Chemistry, Shivalik College of Pharmacy, Nangal, Dist. Rupnagar, 140126, Punjab, India.
| |
Collapse
|
48
|
McGovern OL, Rivera-Cuevas Y, Kannan G, Narwold AJ, Carruthers VB. Intersection of endocytic and exocytic systems in Toxoplasma gondii. Traffic 2018; 19:336-353. [PMID: 29437275 DOI: 10.1111/tra.12556] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2017] [Revised: 02/09/2018] [Accepted: 02/10/2018] [Indexed: 12/12/2022]
Abstract
Host cytosolic proteins are endocytosed by Toxoplasma gondii and degraded in its lysosome-like compartment, the vacuolar compartment (VAC), but the dynamics and route of endocytic trafficking remain undefined. Conserved endocytic components and plant-like features suggest T. gondii endocytic trafficking involves transit through early and late endosome-like compartments (ELCs) and potentially the trans-Golgi network (TGN) as in plants. However, exocytic trafficking to regulated secretory organelles, micronemes and rhoptries, also proceeds through ELCs and requires classical endocytic components, including a dynamin-related protein, DrpB. Here, we show that host cytosolic proteins are endocytosed within 7 minutes post-invasion, trafficked through ELCs en route to the VAC, and degraded within 30 minutes. We could not definitively interpret if ingested protein is trafficked through the TGN. We also found that parasites ingest material from the host cytosol throughout the parasite cell cycle. Ingested host proteins colocalize with immature microneme proteins, proM2AP and proMIC5, in transit to the micronemes, but not with the immature rhoptry protein proRON4, indicating that endocytic trafficking of ingested protein intersects with exocytic trafficking of microneme proteins. Finally, we show that conditional expression of a DrpB dominant negative mutant increases T. gondii ingestion of host-derived proteins, suggesting that DrpB is not required for parasite endocytosis.
Collapse
Affiliation(s)
- Olivia L McGovern
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, Michigan
| | - Yolanda Rivera-Cuevas
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, Michigan
| | - Geetha Kannan
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, Michigan
| | - Andrew J Narwold
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, Michigan
| | - Vern B Carruthers
- Department of Microbiology and Immunology, University of Michigan School of Medicine, Ann Arbor, Michigan
| |
Collapse
|
49
|
Himangini, Pathak DP, Sharma V, Kumar S. Designing novel inhibitors against falcipain-2 of Plasmodium falciparum. Bioorg Med Chem Lett 2018; 28:1566-1569. [PMID: 29602682 DOI: 10.1016/j.bmcl.2018.03.058] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 03/15/2018] [Accepted: 03/21/2018] [Indexed: 12/29/2022]
Abstract
Coumarin containing pyrazoline derivatives have been synthesized and tested as inhibitors of in vitro development of a chloroquine-sensitive (MRC-02) and chloroquine-resistant (RKL-2) strain of Plasmodium falciparum and in vivo Plasmodium berghei malaria. Docking study was also done on cysteine protease falcipain-2 which showed that the binding pose of C-14 molecule and epoxysuccinate, inhibitor of falcipain-2, binds in the similar pattern. The most active antimalarial compound was 3-(1-benzoyl-5-(4-flurophenyl)-4,5-dihydro-1H-pyrazol-3yl)-7-(diethyamino)-2H-chromen-2-one C-14, with an IC50 of 4.21 µg/ml provided complete protection to the infected mice at 24 mg/kg X 4 days respectively.
Collapse
Affiliation(s)
- Himangini
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research, Sector-3, Pushp Vihar, Delhi 17, India.
| | - Dharam Pal Pathak
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research, Sector-3, Pushp Vihar, Delhi 17, India
| | - Vidushi Sharma
- Department of Pharmaceutical Chemistry, Delhi Institute of Pharmaceutical Sciences and Research, Sector-3, Pushp Vihar, Delhi 17, India
| | - Sachin Kumar
- Department of Pharmacology, Delhi Institute of Pharmaceutical Sciences and Research, Sector-3, Pushp Vihar, Delhi 17, India
| |
Collapse
|
50
|
Gazanion E, Vergnes B. Protozoan Parasite Auxotrophies and Metabolic Dependencies. EXPERIENTIA SUPPLEMENTUM (2012) 2018; 109:351-375. [PMID: 30535605 DOI: 10.1007/978-3-319-74932-7_9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Diseases caused by protozoan parasites have a major impact on world health. These early branching eukaryotes cause significant morbidity and mortality in humans and livestock. During evolution, protozoan parasites have evolved toward complex life cycles in multiple host organisms with different nutritional resources. The conservation of functional metabolic pathways required for these successive environments is therefore a prerequisite for parasitic lifestyle. Nevertheless, parasitism drives genome evolution toward gene loss and metabolic dependencies (including strict auxotrophy), especially for obligatory intracellular parasites. In this chapter, we will compare and contrast how protozoan parasites have perfected this metabolic adaptation by focusing on specific auxotrophic pathways and scavenging strategies used by clinically relevant apicomplexan and trypanosomatid parasites to access host's nutritional resources. We will further see how these metabolic dependencies have in turn been exploited for therapeutic purposes against these human pathogens.
Collapse
Affiliation(s)
- Elodie Gazanion
- MIVEGEC, IRD, CNRS, University of Montpellier, Montpellier, France
| | - Baptiste Vergnes
- MIVEGEC, IRD, CNRS, University of Montpellier, Montpellier, France.
| |
Collapse
|