1
|
Arora G, Černý J. Plasmodium proteases and their role in development of Malaria vaccines. ADVANCES IN PARASITOLOGY 2024; 126:253-273. [PMID: 39448193 DOI: 10.1016/bs.apar.2024.08.001] [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: 10/26/2024]
Abstract
Malaria remains a major health hazard for humans, despite the availability of efficacious antimalarial drugs and other interventions. Given that the disease is often deadly for children under 5 years and pregnant women living in malaria-endemic areas, an efficacious vaccine to prevent transmission and clinical disease would be ideal. Plasmodium, the causative agent of malaria, uses proteases and protease inhibitors to control and process to invade host, modulate host immunity, and for pathogenesis. Plasmodium parasites rely on these proteases for their development and survival, including feeding their metabolic needs and invasion of both mosquito and human tissues, and have thus been explored as potential targets for prophylaxis. In this chapter, we have discussed the potential of proteases like ROM4, SUB2, SERA4, SERA5, and others as vaccine candidates. We have also discussed the role of some protease inhibitors of plasmodium and mosquito origin. Inhibition of plasmodium proteases can interrupt the parasite development at many different stages therefore understanding their function is key to developing new drugs and malaria vaccines.
Collapse
Affiliation(s)
- Gunjan Arora
- Section of Infectious Diseases, Department of Internal Medicine, Yale School of Medicine, New Haven, CT, United States.
| | - Jiří Černý
- Centre for Infectious Animal Diseases, Faculty of Tropical AgriSciences, Czech University of Life Sciences Prague, Prague-Suchdol, Czechia
| |
Collapse
|
2
|
Withers-Martinez C, Lidumniece E, Hackett F, Collins CR, Taha Z, Blackman MJ, Jirgensons A. Peptidic Boronic Acid Plasmodium falciparum SUB1 Inhibitors with Improved Selectivity over Human Proteasome. J Med Chem 2024; 67:13033-13055. [PMID: 39051854 PMCID: PMC7616463 DOI: 10.1021/acs.jmedchem.4c01005] [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] [Indexed: 07/27/2024]
Abstract
Plasmodium falciparum subtilisin-like serine protease 1 (PfSUB1) is essential for egress of invasive merozoite forms of the parasite, rendering PfSUB1 an attractive antimalarial target. Here, we report studies aimed to improve drug-like properties of peptidic boronic acid PfSUB1 inhibitors including increased lipophilicity and selectivity over human proteasome (H20S). Structure-activity relationship investigations revealed that lipophilic P3 amino acid side chains as well as N-capping groups were well tolerated in retaining PfSUB1 inhibitory potency. At the P1 position, replacing the methyl group with a carboxyethyl substituent led to boralactone PfSUB1 inhibitors with remarkably improved selectivity over H20S. Combining lipophilic end-capping groups with the boralactone reduced the selectivity over H20S. However, compound 4c still showed >60-fold selectivity versus H20S and low nanomolar PfSUB1 inhibitory potency. Importantly, this compound inhibited the growth of a genetically modified P. falciparum line expressing reduced levels of PfSUB1 13-fold more efficiently compared to a wild-type parasite line.
Collapse
Affiliation(s)
| | | | - Fiona Hackett
- Malaria Biochemistry Laboratory, The Francis Crick Institute, LondonNW1 1AT, United Kingdom
| | - Christine R. Collins
- Malaria Biochemistry Laboratory, The Francis Crick Institute, LondonNW1 1AT, United Kingdom
| | - Zahie Taha
- Malaria Biochemistry Laboratory, The Francis Crick Institute, LondonNW1 1AT, United Kingdom
| | - Michael J. Blackman
- Malaria Biochemistry Laboratory, The Francis Crick Institute, LondonNW1 1AT, United Kingdom
- Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, LondonWC1E 7HT, United Kingdom
| | | |
Collapse
|
3
|
Schwarzer E, Skorokhod O. Post-Translational Modifications of Proteins of Malaria Parasites during the Life Cycle. Int J Mol Sci 2024; 25:6145. [PMID: 38892332 PMCID: PMC11173270 DOI: 10.3390/ijms25116145] [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: 05/01/2024] [Revised: 05/29/2024] [Accepted: 05/31/2024] [Indexed: 06/21/2024] Open
Abstract
Post-translational modifications (PTMs) are essential for regulating protein functions, influencing various fundamental processes in eukaryotes. These include, but are not limited to, cell signaling, protein trafficking, the epigenetic control of gene expression, and control of the cell cycle, as well as cell proliferation, differentiation, and interactions between cells. In this review, we discuss protein PTMs that play a key role in the malaria parasite biology and its pathogenesis. Phosphorylation, acetylation, methylation, lipidation and lipoxidation, glycosylation, ubiquitination and sumoylation, nitrosylation and glutathionylation, all of which occur in malarial parasites, are reviewed. We provide information regarding the biological significance of these modifications along all phases of the complex life cycle of Plasmodium spp. Importantly, not only the parasite, but also the host and vector protein PTMs are often crucial for parasite growth and development. In addition to metabolic regulations, protein PTMs can result in epitopes that are able to elicit both innate and adaptive immune responses of the host or vector. We discuss some existing and prospective results from antimalarial drug discovery trials that target various PTM-related processes in the parasite or host.
Collapse
Affiliation(s)
- Evelin Schwarzer
- Department of Oncology, University of Turin, Via Santena 5 bis, 10126 Turin, Italy;
| | - Oleksii Skorokhod
- Department of Life Sciences and Systems Biology, University of Turin, Via Accademia Albertina, 13, 10123 Turin, Italy
| |
Collapse
|
4
|
Mahanta PJ, Lhouvum K. Plasmodium falciparum proteases as new drug targets with special focus on metalloproteases. Mol Biochem Parasitol 2024; 258:111617. [PMID: 38554736 DOI: 10.1016/j.molbiopara.2024.111617] [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: 10/17/2023] [Revised: 02/15/2024] [Accepted: 03/10/2024] [Indexed: 04/02/2024]
Abstract
Malaria poses a significant global health threat particularly due to the prevalence of Plasmodium falciparum infection. With the emergence of parasite resistance to existing drugs including the recently discovered artemisinin, ongoing research seeks novel therapeutic avenues within the malaria parasite. Proteases are promising drug targets due to their essential roles in parasite biology, including hemoglobin digestion, merozoite invasion, and egress. While exploring the genomic landscape of Plasmodium falciparum, it has been revealed that there are 92 predicted proteases, with only approximately 14 of them having been characterized. These proteases are further distributed among 26 families grouped into five clans: aspartic proteases, cysteine proteases, metalloproteases, serine proteases, and threonine proteases. Focus on metalloprotease class shows further role in organelle processing for mitochondria and apicoplasts suggesting the potential of metalloproteases as viable drug targets. Holistic understanding of the parasite intricate life cycle and identification of potential drug targets are essential for developing effective therapeutic strategies against malaria and mitigating its devastating global impact.
Collapse
Affiliation(s)
| | - Kimjolly Lhouvum
- Department of Biotechnology, National Institute of Technology, Arunachal Pradesh, India.
| |
Collapse
|
5
|
Legru A, Batista FA, Puszko AK, Bouillon A, Maurel M, Martinez M, Ejjoummany A, Ortega Varga L, Adler P, Méchaly A, Hadjadj M, Sosnowski P, Hopfgartner G, Alzari PM, Blondel A, Haouz A, Barale JC, Hernandez JF. Insights from structure-activity relationships and the binding mode of peptidic α-ketoamide inhibitors of the malaria drug target subtilisin-like SUB1. Eur J Med Chem 2024; 269:116308. [PMID: 38503166 DOI: 10.1016/j.ejmech.2024.116308] [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: 12/11/2023] [Revised: 03/04/2024] [Accepted: 03/04/2024] [Indexed: 03/21/2024]
Abstract
Plasmodium multi-resistance, including against artemisinin, seriously threatens malaria treatment and control. Hence, new drugs are urgently needed, ideally targeting different parasitic stages, which are not yet targeted by current drugs. The SUB1 protease is involved in both hepatic and blood stages due to its essential role in the egress of parasites from host cells, and, as potential new target, it would meet the above criteria. We report here the synthesis as well as the biological and structural evaluation of substrate-based α-ketoamide SUB1 pseudopeptidic inhibitors encompassing positions P4-P2'. By individually substituting each position of the reference compound 1 (MAM-117, Ac-Ile-Thr-Ala-AlaCO-Asp-Glu (Oall)-NH2), we better characterized the structural determinants for SUB1 binding. We first identified compound 8 with IC50 values of 50 and 570 nM against Pv- and PfSUB1, respectively (about 3.5-fold higher potency compared to 1). Compound 8 inhibited P. falciparum merozoite egress in culture by 37% at 100 μM. By increasing the overall hydrophobicity of the compounds, we could improve the PfSUB1 inhibition level and antiparasitic activity, as shown with compound 40 (IC50 values of 12 and 10 nM against Pv- and PfSUB1, respectively, IC50 value of 23 μM on P. falciparum merozoite egress). We also found that 8 was highly selective towards SUB1 over three mammalian serine peptidases, supporting the promising value of this compound. Finally, several crystal 3D-structures of SUB1-inhibitor complexes, including with 8, were solved at high resolution to decipher the binding mode of these compounds.
Collapse
Affiliation(s)
- Alice Legru
- Institut des Biomolécules Max Mousseron (IBMM), CNRS, Univ Montpellier, ENSCM, Montpellier, France
| | - Fernando A Batista
- Structural Microbiology, UMR3528, Institut Pasteur, CNRS, Université de Paris, Paris, France
| | - Anna K Puszko
- Institut des Biomolécules Max Mousseron (IBMM), CNRS, Univ Montpellier, ENSCM, Montpellier, France
| | - Anthony Bouillon
- Structural Microbiology, UMR3528, Institut Pasteur, CNRS, Université de Paris, Paris, France
| | - Manon Maurel
- Institut des Biomolécules Max Mousseron (IBMM), CNRS, Univ Montpellier, ENSCM, Montpellier, France
| | - Mariano Martinez
- Structural Microbiology, UMR3528, Institut Pasteur, CNRS, Université de Paris, Paris, France
| | - Abdelaziz Ejjoummany
- Institut des Biomolécules Max Mousseron (IBMM), CNRS, Univ Montpellier, ENSCM, Montpellier, France
| | - Laura Ortega Varga
- Structural Bioinformatic, UMR3528, Institut Pasteur, CNRS, Université de Paris, Paris, France
| | - Pauline Adler
- Institut des Biomolécules Max Mousseron (IBMM), CNRS, Univ Montpellier, ENSCM, Montpellier, France
| | - Ariel Méchaly
- Cristallography Platform-C2RT, UMR3528, Institut Pasteur, CNRS, Université de Paris, Paris, France
| | - Margot Hadjadj
- Institut des Biomolécules Max Mousseron (IBMM), CNRS, Univ Montpellier, ENSCM, Montpellier, France
| | - Piotr Sosnowski
- Department of Inorganic and Analytical Chemistry, University of Geneva, CH-1211, Geneva, Switzerland
| | - Gérard Hopfgartner
- Department of Inorganic and Analytical Chemistry, University of Geneva, CH-1211, Geneva, Switzerland
| | - Pedro M Alzari
- Structural Microbiology, UMR3528, Institut Pasteur, CNRS, Université de Paris, Paris, France
| | - Arnaud Blondel
- Structural Bioinformatic, UMR3528, Institut Pasteur, CNRS, Université de Paris, Paris, France
| | - Ahmed Haouz
- Cristallography Platform-C2RT, UMR3528, Institut Pasteur, CNRS, Université de Paris, Paris, France
| | - Jean-Christophe Barale
- Structural Microbiology, UMR3528, Institut Pasteur, CNRS, Université de Paris, Paris, France.
| | - Jean-François Hernandez
- Institut des Biomolécules Max Mousseron (IBMM), CNRS, Univ Montpellier, ENSCM, Montpellier, France.
| |
Collapse
|
6
|
Martinez M, Bouillon A, Brûlé S, Raynal B, Haouz A, Alzari PM, Barale JC. Prodomain-driven enzyme dimerization: a pH-dependent autoinhibition mechanism that controls Plasmodium Sub1 activity before merozoite egress. mBio 2024; 15:e0019824. [PMID: 38386597 PMCID: PMC10936178 DOI: 10.1128/mbio.00198-24] [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: 01/25/2024] [Accepted: 01/31/2024] [Indexed: 02/24/2024] Open
Abstract
Malaria symptoms are associated with the asexual multiplication of Plasmodium falciparum within human red blood cells (RBCs) and fever peaks coincide with the egress of daughter merozoites following the rupture of the parasitophorous vacuole (PV) and the RBC membranes. Over the last two decades, it has emerged that the release of competent merozoites is tightly regulated by a complex cascade of events, including the unusual multi-step activation mechanism of the pivotal subtilisin-like protease 1 (Sub1) that takes place in three different cellular compartments and remains poorly understood. Following an initial auto-maturation in the endoplasmic reticulum (ER) between its pro- and catalytic domains, the Sub1 prodomain (PD) undergoes further cleavages by the parasite aspartic protease plasmepsin X (PmX) within acidic secretory organelles that ultimately lead to full Sub1 activation upon discharge into the PV. Here, we report the crystal structure of full-length P. falciparum Sub1 (PfS1FL) and demonstrate, through structural, biochemical, and biophysical studies, that the atypical Plasmodium-specific Sub1 PD directly promotes the assembly of inactive enzyme homodimers at acidic pH, whereas Sub1 is primarily monomeric at neutral pH. Our results shed new light into the finely tuned Sub1 spatiotemporal activation during secretion, explaining how PmX processing and full activation of Sub1 can occur in different cellular compartments, and uncover a robust mechanism of pH-dependent subtilisin autoinhibition that plays a key role in P. falciparum merozoites egress from infected host cells.IMPORTANCEMalaria fever spikes are due to the rupture of infected erythrocytes, allowing the egress of Plasmodium sp. merozoites and further parasite propagation. This fleeting tightly regulated event involves a cascade of enzymes, culminating with the complex activation of the subtilisin-like protease 1, Sub1. Differently than other subtilisins, Sub1 activation strictly depends upon the processing by a parasite aspartic protease within acidic merozoite secretory organelles. However, Sub1 biological activity is required in the pH neutral parasitophorous vacuole, to prime effectors involved in the rupture of the vacuole and erythrocytic membranes. Here, we show that the unusual, parasite-specific Sub1 prodomain is directly responsible for its acidic-dependent dimerization and autoinhibition, required for protein secretion, before its full activation at neutral pH in a monomeric form. pH-dependent Sub1 dimerization defines a novel, essential regulatory element involved in the finely tuned spatiotemporal activation of the egress of competent Plasmodium merozoites.
Collapse
Affiliation(s)
- Mariano Martinez
- Unité de Microbiologie Structurale, Institut Pasteur, CNRS UMR 3528, Université Paris Cité, Paris, France
| | - Anthony Bouillon
- Unité de Microbiologie Structurale, Institut Pasteur, CNRS UMR 3528, Université Paris Cité, Paris, France
| | - Sébastien Brûlé
- Plate-forme de Biophysique Moleculaire-C2RT, Institut Pasteur, CNRS UMR 3528, Université Paris Cité, Paris, France
| | - Bertrand Raynal
- Plate-forme de Biophysique Moleculaire-C2RT, Institut Pasteur, CNRS UMR 3528, Université Paris Cité, Paris, France
| | - Ahmed Haouz
- Plate-forme de Cristallographie-C2RT, Institut Pasteur, CNRS UMR 3528, Université Paris Cité, Paris, France
| | - Pedro M. Alzari
- Unité de Microbiologie Structurale, Institut Pasteur, CNRS UMR 3528, Université Paris Cité, Paris, France
| | - Jean-Christophe Barale
- Unité de Microbiologie Structurale, Institut Pasteur, CNRS UMR 3528, Université Paris Cité, Paris, France
| |
Collapse
|
7
|
Scheiner M, Burda PC, Ingmundson A. Moving on: How malaria parasites exit the liver. Mol Microbiol 2024; 121:328-340. [PMID: 37602900 DOI: 10.1111/mmi.15141] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2023] [Revised: 08/03/2023] [Accepted: 08/04/2023] [Indexed: 08/22/2023]
Abstract
An essential step in the life cycle of malaria parasites is their egress from hepatocytes, which enables the transition from the asymptomatic liver stage to the pathogenic blood stage of infection. To exit the liver, Plasmodium parasites first disrupt the parasitophorous vacuole membrane that surrounds them during their intracellular replication. Subsequently, parasite-filled structures called merosomes emerge from the infected cell. Shrouded by host plasma membrane, like in a Trojan horse, parasites enter the vasculature undetected by the host immune system and travel to the lung where merosomes rupture, parasites are released, and the blood infection stage begins. This complex, multi-step process must be carefully orchestrated by the parasite and requires extensive manipulation of the infected host cell. This review aims to outline the known signaling pathways that trigger exit, highlight Plasmodium proteins that contribute to the release of liver-stage merozoites, and summarize the accompanying changes to the hepatic host cell.
Collapse
Affiliation(s)
- Mattea Scheiner
- Molecular Parasitology, Humboldt University Berlin, Berlin, Germany
| | - Paul-Christian Burda
- Centre for Structural Systems Biology, Hamburg, Germany
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- University of Hamburg, Hamburg, Germany
| | | |
Collapse
|
8
|
Abstract
Human malaria, caused by infection with Plasmodium parasites, remains one of the most important global public health problems, with the World Health Organization reporting more than 240 million cases and 600,000 deaths annually as of 2020 (World malaria report 2021). Our understanding of the biology of these parasites is critical for development of effective therapeutics and prophylactics, including both antimalarials and vaccines. Plasmodium is a protozoan organism that is intracellular for most of its life cycle. However, to complete its complex life cycle and to allow for both amplification and transmission, the parasite must egress out of the host cell in a highly regulated manner. This review discusses the major pathways and proteins involved in the egress events during the Plasmodium life cycle-merozoite and gametocyte egress out of red blood cells, sporozoite egress out of the oocyst, and merozoite egress out of the hepatocyte. The similarities, as well as the differences, between the various egress pathways of the parasite highlight both novel cell biology and potential therapeutic targets to arrest its life cycle.
Collapse
Affiliation(s)
- Jeffrey D Dvorin
- Division of Infectious Diseases, Boston Children's Hospital, Boston, Massachusetts, USA;
- Department of Pediatrics, Harvard Medical School, Boston, Massachusetts, USA
| | - Daniel E Goldberg
- Division of Infectious Diseases, Department of Medicine; and Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, Missouri, USA;
| |
Collapse
|
9
|
Peptidic boronic acids are potent cell-permeable inhibitors of the malaria parasite egress serine protease SUB1. Proc Natl Acad Sci U S A 2021; 118:2022696118. [PMID: 33975947 PMCID: PMC8157947 DOI: 10.1073/pnas.2022696118] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Malaria is a devastating infectious disease, which causes over 400,000 deaths per annum and impacts the lives of nearly half the world's population. The causative agent, a protozoan parasite, replicates within red blood cells (RBCs), eventually destroying the cells in a lytic process called egress to release a new generation of parasites. These invade fresh RBCs to repeat the cycle. Egress is regulated by an essential parasite subtilisin-like serine protease called SUB1. Here, we describe the development and optimization of substrate-based peptidic boronic acids that inhibit Plasmodium falciparum SUB1 with low nanomolar potency. Structural optimization generated membrane-permeable, slow off-rate inhibitors that prevent Pfalciparum egress through direct inhibition of SUB1 activity and block parasite replication in vitro at submicromolar concentrations. Our results validate SUB1 as a potential target for a new class of antimalarial drugs designed to prevent parasite replication and disease progression.
Collapse
|
10
|
Abstract
All intracellular pathogens must escape (egress) from the confines of their host cell to disseminate and proliferate. The malaria parasite only replicates in an intracellular vacuole or in a cyst, and must undergo egress at four distinct phases during its complex life cycle, each time disrupting, in a highly regulated manner, the membranes or cyst wall that entrap the parasites. This Cell Science at a Glance article and accompanying poster summarises our current knowledge of the morphological features of egress across the Plasmodium life cycle, the molecular mechanisms that govern the process, and how researchers are working to exploit this knowledge to develop much-needed new approaches to malaria control. ![]()
Collapse
Affiliation(s)
- Michele S Y Tan
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London NW1 1AT, UK
| | - Michael J Blackman
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London NW1 1AT, UK .,Faculty of Infectious and Tropical Diseases, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| |
Collapse
|
11
|
Putrianti ED, Schmidt-Christensen A, Heussler V, Matuschewski K, Ingmundson A. A Plasmodium cysteine protease required for efficient transition from the liver infection stage. PLoS Pathog 2020; 16:e1008891. [PMID: 32956401 PMCID: PMC7529260 DOI: 10.1371/journal.ppat.1008891] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 10/01/2020] [Accepted: 08/15/2020] [Indexed: 01/23/2023] Open
Abstract
The transitions between developmental stages are critical points in the Plasmodium life cycle. The development of Plasmodium in the livers of their mammalian hosts bridges malaria transmission and the onset of clinical symptoms elicited by red blood cell infection. The egress of Plasmodium parasites from the liver must be a carefully orchestrated process to ensure a successful switch to the blood stage of infection. Cysteine protease activity is known to be required for liver-stage Plasmodium egress, but the crucial cysteine protease(s) remained unidentified. Here, we characterize a member of the papain-like cysteine protease family, Plasmodium berghei serine repeat antigen 4 (PbSERA4), that is required for efficient initiation of blood-stage infection. Through the generation PbSERA4-specific antisera and the creation of transgenic parasites expressing fluorescently tagged protein, we show that PbSERA4 is expressed and proteolytically processed in the liver and blood stages of infection. Targeted disruption of PbSERA4 results in viable and virulent blood-stage parasites. However, upon transmission from mosquitoes to mice, Pbsera4(-) parasites displayed a reduced capacity to initiate a new round of asexual blood-stage replication. Our results from cultured cells indicate that this defect results from an inability of the PbSERA4-deficient parasites to egress efficiently from infected cells at the culmination of liver-stage development. Protection against infection with wildtype P. berghei could be generated in animals in which Pbsera4(-) parasites failed to establish infection. Our findings confirm that liver-stage merozoite release is an active process and demonstrate that this parasite-encoded cysteine protease contributes to parasite escape from the liver. Plasmodium parasites cause over 200 million cases of malaria every year. When parasites are transmitted by mosquito bite, they initially colonize the liver before they move into the blood and cause disease. During successful transition from the liver into the blood, Plasmodium cloak themselves in host plasma membrane as they egress from the liver cells. Although some aspects of how Plasmodium exit their host hepatocytes appear unique, certain attributes are shared across diverse pathogens. For example, protease activity is required not only for multiple stages of Plasmodium exit, but is also involved in the egress of some bacteria and other protozoan. Here we characterize a protease in Plasmodium berghei that is expressed in the liver and conserved across Plasmodium species. Through gene targeting, we found PbSERA4 is required for efficient egress of Plasmodium from the liver. In the absence of this protease the transition between the liver and blood stages of growth is prolonged due to inefficient parasite release from liver cells. These findings provide new insights into the function of a conserved Plasmodium protease and into the process of Plasmodium escape from the liver.
Collapse
Affiliation(s)
- Elyzana Dewi Putrianti
- Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
- Metabolism of Microbial Pathogens, Robert Koch Institute, Berlin, Germany
| | - Anja Schmidt-Christensen
- Department of Experimental Medical Science, Lund University, Lund, Sweden
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Volker Heussler
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Kai Matuschewski
- Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
- Department of Molecular Parasitology, Institute of Biology, Humboldt University Berlin, Berlin, Germany
| | - Alyssa Ingmundson
- Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
- Department of Molecular Parasitology, Institute of Biology, Humboldt University Berlin, Berlin, Germany
- * E-mail:
| |
Collapse
|
12
|
A malaria parasite subtilisin propeptide-like protein is a potent inhibitor of the egress protease SUB1. Biochem J 2020; 477:525-540. [PMID: 31942933 PMCID: PMC6993865 DOI: 10.1042/bcj20190918] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 01/14/2020] [Accepted: 01/16/2020] [Indexed: 12/23/2022]
Abstract
Subtilisin-like serine peptidases (subtilases) play important roles in the life cycle of many organisms, including the protozoan parasites that are the causative agent of malaria, Plasmodium spp. As with other peptidases, subtilase proteolytic activity has to be tightly regulated in order to prevent potentially deleterious uncontrolled protein degradation. Maturation of most subtilases requires the presence of an N-terminal propeptide that facilitates folding of the catalytic domain. Following its proteolytic cleavage, the propeptide acts as a transient, tightly bound inhibitor until its eventual complete removal to generate active protease. Here we report the identification of a stand-alone malaria parasite propeptide-like protein, called SUB1-ProM, encoded by a conserved gene that lies in a highly syntenic locus adjacent to three of the four subtilisin-like genes in the Plasmodium genome. Template-based modelling and ab initio structure prediction showed that the SUB1-ProM core structure is most similar to the X-ray crystal structure of the propeptide of SUB1, an essential parasite subtilase that is discharged into the parasitophorous vacuole (PV) to trigger parasite release (egress) from infected host cells. Recombinant Plasmodium falciparum SUB1-ProM was found to be a fast-binding, potent inhibitor of P. falciparum SUB1, but not of the only other essential blood-stage parasite subtilase, SUB2, or of other proteases examined. Mass-spectrometry and immunofluorescence showed that SUB1-ProM is expressed in the PV of blood stage P. falciparum, where it may act as an endogenous inhibitor to regulate SUB1 activity in the parasite.
Collapse
|
13
|
Arisue N, Palacpac NMQ, Tougan T, Horii T. Characteristic features of the SERA multigene family in the malaria parasite. Parasit Vectors 2020; 13:170. [PMID: 32252804 PMCID: PMC7132891 DOI: 10.1186/s13071-020-04044-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2019] [Accepted: 03/27/2020] [Indexed: 02/28/2023] Open
Abstract
Serine repeat antigen (SERA) is conserved among species of the genus Plasmodium. Sera genes form a multigene family and are generally tandemly clustered on a single chromosome. Although all Plasmodium species encode multiple sera genes, the number varies between species. Among species, the members share similar sequences and gene organization. SERA possess a central papain-like cysteine protease domain, however, in some members, the active site cysteine residue is substituted with a serine. Recent studies implicate this gene family in a number of aspects in parasite biology and induction of protective immune response. This review summarizes the current understanding on this important gene family in several Plasmodium species. The Plasmodium falciparum (Pf)-sera family, for example, consists of nine gene members. Unlike other multigene families in Plasmodium species, Pf-sera genes do not exhibit antigenic variation. Pf-sera5 nucleotide diversity is also low. Moreover, although Pf-sera5 is highly transcribed during the blood stage of malaria infection, and a large amount is released into the host blood following schizont rupture, in malaria endemic countries the sero-positive rates for Pf-SERA5 are low, likely due to Pf-SERA5 binding of host proteins to avoid immune recognition. As an antigen, the N-terminal 47 kDa domain of Pf-SERA5 is a promising vaccine candidate currently undergoing clinical trials. Pf-SERA5 and Pf-SERA6, as well as P. berghei (Pb)-SERA3, and Pb-SERA5, have been investigated for their roles in parasite egress. Two P. yoelii SERA, which have a serine residue at the protease active center, are implicated in parasite virulence. Overall, these studies provide insight that during the evolution of the Plasmodium parasite, the sera gene family members have increased by gene duplication, and acquired various functions that enable the parasite to survive and successfully maintain infection in the host.![]()
Collapse
Affiliation(s)
- Nobuko Arisue
- Research Center for Infectious Disease Control, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan.
| | - Nirianne M Q Palacpac
- Department of Malaria Vaccine Development, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Takahiro Tougan
- Research Center for Infectious Disease Control, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
| | - Toshihiro Horii
- Department of Malaria Vaccine Development, Research Institute for Microbial Diseases, Osaka University, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
14
|
An Endoplasmic Reticulum CREC Family Protein Regulates the Egress Proteolytic Cascade in Malaria Parasites. mBio 2020; 11:mBio.03078-19. [PMID: 32098818 PMCID: PMC7042697 DOI: 10.1128/mbio.03078-19] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
The endoplasmic reticulum (ER) is thought to play an essential role during egress of malaria parasites because the ER is assumed to be required for biogenesis and secretion of egress-related organelles. However, no proteins localized to the parasite ER have been shown to play a role in egress of malaria parasites. In this study, we generated conditional mutants of the Plasmodium falciparum endoplasmic reticulum-resident calcium-binding protein (PfERC), a member of the CREC family. Knockdown of the PfERC gene showed that this gene is essential for asexual growth of P. falciparum Analysis of the intraerythrocytic life cycle revealed that PfERC is essential for parasite egress but is not required for protein trafficking or calcium storage. We found that PfERC knockdown prevents the rupture of the parasitophorous vacuole membrane. This is because PfERC knockdown inhibited the proteolytic maturation of the subtilisin-like serine protease SUB1. Using double mutant parasites, we showed that PfERC is required for the proteolytic maturation of the essential aspartic protease plasmepsin X, which is required for SUB1 cleavage. Further, we showed that processing of substrates downstream of the proteolytic cascade is inhibited by PfERC knockdown. Thus, these data establish that the ER-resident CREC family protein PfERC is a key early regulator of the egress proteolytic cascade of malaria parasites.IMPORTANCE The divergent eukaryotic parasites that cause malaria grow and divide within a vacuole inside a host cell, which they have to break open once they finish cell division. The egress of daughter parasites requires the activation of a proteolytic cascade, and a subtilisin-like protease initiates a proteolytic cascade to break down the membranes blocking egress. It is assumed that the parasite endoplasmic reticulum plays a role in this process, but the proteins in this organelle required for egress remain unknown. We have identified an early ER-resident regulator essential for the maturation of the recently discovered aspartic protease in the egress proteolytic cascade, plasmepsin X, which is required for maturation of the subtilisin-like protease. Conditional loss of PfERC results in the formation of immature and inactive egress proteases that are unable to breakdown the vacuolar membrane barring release of daughter parasites.
Collapse
|
15
|
Caldelari R, Dogga S, Schmid MW, Franke-Fayard B, Janse CJ, Soldati-Favre D, Heussler V. Transcriptome analysis of Plasmodium berghei during exo-erythrocytic development. Malar J 2019; 18:330. [PMID: 31551073 PMCID: PMC6760107 DOI: 10.1186/s12936-019-2968-7] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Accepted: 09/17/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND The complex life cycle of malaria parasites requires well-orchestrated stage specific gene expression. In the vertebrate host the parasites grow and multiply by schizogony in two different environments: within erythrocytes and within hepatocytes. Whereas erythrocytic parasites are well-studied in this respect, relatively little is known about the exo-erythrocytic stages. METHODS In an attempt to fill this gap, genome wide RNA-seq analyses of various exo-erythrocytic stages of Plasmodium berghei including sporozoites, samples from a time-course of liver stage development and detached cells were performed. These latter contain infectious merozoites and represent the final step in exo-erythrocytic development. RESULTS The analysis represents the complete transcriptome of the entire life cycle of P. berghei parasites with temporal detailed analysis of the liver stage allowing comparison of gene expression across the progression of the life cycle. These RNA-seq data from different developmental stages were used to cluster genes with similar expression profiles, in order to infer their functions. A comparison with published data from other parasite stages confirmed stage-specific gene expression and revealed numerous genes that are expressed differentially in blood and exo-erythrocytic stages. One of the most exo-erythrocytic stage-specific genes was PBANKA_1003900, which has previously been annotated as a "gametocyte specific protein". The promoter of this gene drove high GFP expression in exo-erythrocytic stages, confirming its expression profile seen by RNA-seq. CONCLUSIONS The comparative analysis of the genome wide mRNA expression profiles of erythrocytic and different exo-erythrocytic stages could be used to improve the understanding of gene regulation in Plasmodium parasites and can be used to model exo-erythrocytic stage metabolic networks toward the identification of differences in metabolic processes during schizogony in erythrocytes and hepatocytes.
Collapse
Affiliation(s)
- Reto Caldelari
- Institute of Cell Biology, University of Bern, Bern, Switzerland.
| | - Sunil Dogga
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva CMU, Geneva, Switzerland
| | | | - Blandine Franke-Fayard
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Chris J Janse
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva CMU, Geneva, Switzerland
| | - Volker Heussler
- Institute of Cell Biology, University of Bern, Bern, Switzerland.
| |
Collapse
|
16
|
Pace T, Grasso F, Camarda G, Suarez C, Blackman MJ, Ponzi M, Olivieri A. The Plasmodium berghei serine protease PbSUB1 plays an important role in male gamete egress. Cell Microbiol 2019; 21:e13028. [PMID: 30941868 PMCID: PMC6766862 DOI: 10.1111/cmi.13028] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Revised: 03/07/2019] [Accepted: 03/28/2019] [Indexed: 01/10/2023]
Abstract
The Plasmodium subtilisin-like serine protease SUB1 is expressed in hepatic and both asexual and sexual blood parasite stages. SUB1 is required for egress of invasive forms of the parasite from both erythrocytes and hepatocytes, but its subcellular localisation, function, and potential substrates in the sexual stages are unknown. Here, we have characterised the expression profile and subcellular localisation of SUB1 in Plasmodium berghei sexual stages. We show that the protease is selectively expressed in mature male gametocytes and localises to secretory organelles known to be involved in gamete egress, called male osmiophilic bodies. We have investigated PbSUB1 function in the sexual stages by generating P. berghei transgenic lines deficient in PbSUB1 expression or enzyme activity in gametocytes. Our results demonstrate that PbSUB1 plays a role in male gamete egress. We also show for the first time that the PbSUB1 substrate PbSERA3 is expressed in gametocytes and processed by PbSUB1 upon gametocyte activation. Taken together, our results strongly suggest that PbSUB1 is not only a promising drug target for asexual stages but could also be an attractive malaria transmission-blocking target.
Collapse
Affiliation(s)
- Tomasino Pace
- Dipartimento di Malattie InfettiveIstituto Superiore di SanitàRomeItaly
| | - Felicia Grasso
- Dipartimento di Malattie InfettiveIstituto Superiore di SanitàRomeItaly
| | - Grazia Camarda
- Dipartimento di Malattie InfettiveIstituto Superiore di SanitàRomeItaly
| | - Catherine Suarez
- Malaria Biochemistry LaboratoryThe Francis Crick InstituteLondonUK
| | - Michael J. Blackman
- Malaria Biochemistry LaboratoryThe Francis Crick InstituteLondonUK
- Faculty of Infectious and Tropical DiseasesLondon School of Hygiene and Tropical MedicineLondonUK
| | - Marta Ponzi
- Dipartimento di Malattie InfettiveIstituto Superiore di SanitàRomeItaly
| | - Anna Olivieri
- Dipartimento di Malattie InfettiveIstituto Superiore di SanitàRomeItaly
| |
Collapse
|
17
|
Choudhary HH, Gupta R, Mishra S. PKAc is not required for the preerythrocytic stages of Plasmodium berghei. Life Sci Alliance 2019; 2:2/3/e201900352. [PMID: 31142638 PMCID: PMC6545604 DOI: 10.26508/lsa.201900352] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Revised: 05/22/2019] [Accepted: 05/22/2019] [Indexed: 12/12/2022] Open
Abstract
The mutant salivary gland sporozoites lacking PKAc are able to glide, invade hepatocytes, and mature into hepatic merozoites, which release successfully from the merosome, however, fail to initiate blood stage infection when inoculated into mice. Plasmodium sporozoites invade hepatocytes to initiate infection in the mammalian host. In the infected hepatocytes, sporozoites undergo rapid expansion and differentiation, resulting in the formation and release of thousands of invasive merozoites into the bloodstream. Both sporozoites and merozoites invade their host cells by activation of a signaling cascade followed by discharge of micronemal content. cAMP-dependent protein kinase catalytic subunit (PKAc)–mediated signaling plays an important role in merozoite invasion of erythrocytes, but its role during other stages of the parasite remains unknown. Becaused of the essentiality of PKAc in blood stages, we generated conditional mutants of PKAc by disrupting the gene in Plasmodium berghei sporozoites. The mutant salivary gland sporozoites were able to glide, invaded hepatocytes, and matured into hepatic merozoites which were released successfully from merosome, however failed to initiate blood stage infection when inoculated into mice. Our results demonstrate that malaria parasite complete preerythrocytic stages development without PKAc, raising the possibility that the PKAc independent signaling operates in preerythrocytic stages of P. berghei.
Collapse
Affiliation(s)
| | - Roshni Gupta
- Division of Parasitology, CSIR-Central Drug Research Institute, Lucknow, India
| | - Satish Mishra
- Division of Parasitology, CSIR-Central Drug Research Institute, Lucknow, India .,Academy of Scientific and Innovative Research, Ghaziabad, India
| |
Collapse
|
18
|
Dubois DJ, Soldati-Favre D. Biogenesis and secretion of micronemes in Toxoplasma gondii. Cell Microbiol 2019; 21:e13018. [PMID: 30791192 DOI: 10.1111/cmi.13018] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2018] [Revised: 02/11/2019] [Accepted: 02/17/2019] [Indexed: 12/20/2022]
Abstract
One of the hallmarks of the parasitic phylum of Apicomplexa is the presence of highly specialised, apical secretory organelles, called the micronemes and rhoptries that play critical roles in ensuring survival and dissemination. Upon exocytosis, the micronemes release adhesin complexes, perforins, and proteases that are crucially implicated in egress from infected cells, gliding motility, migration across biological barriers, and host cell invasion. Recent studies on Toxoplasma gondii and Plasmodium species have shed more light on the signalling events and the machinery that trigger microneme secretion. Intracellular cyclic nucleotides, calcium level, and phosphatidic acid act as key mediators of microneme exocytosis, and several downstream effectors have been identified. Here, we review the key steps of microneme biogenesis and exocytosis, summarising the still fractal knowledge at the molecular level regarding the fusion event with the parasite plasma membrane.
Collapse
Affiliation(s)
- David J Dubois
- Department of Microbiology and Molecular Medicine, University of Geneva CMU, Geneva, Switzerland
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, University of Geneva CMU, Geneva, Switzerland
| |
Collapse
|
19
|
Obrova K, Cyrklaff M, Frank R, Mair GR, Mueller AK. Transmission of the malaria parasite requires ferlin for gamete egress from the red blood cell. Cell Microbiol 2019; 21:e12999. [PMID: 30597708 DOI: 10.1111/cmi.12999] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2018] [Revised: 11/15/2018] [Accepted: 12/09/2018] [Indexed: 02/02/2023]
Abstract
Ferlins mediate calcium-dependent vesicular fusion. Although conserved throughout eukaryotic evolution, their function in unicellular organisms including apicomplexan parasites is largely unknown. Here, we define a crucial role for a ferlin-like protein (FLP) in host-to-vector transmission of the rodent malaria parasite Plasmodium berghei. Infection of the mosquito vectors requires the formation of free gametes and their fertilisation in the mosquito midgut. Mature gametes will only emerge upon secretion of factors that stimulate the disruption of the red blood cell membrane and the parasitophorous vacuole membrane. Genetic depletion of FLP in sexual stages leads to a complete life cycle arrest in the mosquito. Although mature gametes form normally, mutants lacking FLP remain trapped in the red blood cell. The egress defect is rescued by detergent-mediated membrane lysis. In agreement with ferlin vesicular localisation, HA-tagged FLP labels intracellular speckles, which relocalise to the cell periphery during gamete maturation. Our data define FLP as a novel critical factor for Plasmodium fertilisation and transmission and suggest an evolutionarily conserved example of ferlin-mediated exocytosis.
Collapse
Affiliation(s)
- Klara Obrova
- Center for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Marek Cyrklaff
- Center for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Roland Frank
- Center for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany
| | - Gunnar R Mair
- Department of Biomedical Sciences, Iowa State University, Ames, Iowa, USA
| | - Ann-Kristin Mueller
- Center for Infectious Diseases, Parasitology Unit, Heidelberg University Hospital, Heidelberg, Germany.,German Center for Infectious Diseases (DZIF), Universitätsklinikum Heidelberg, Heidelberg, Germany
| |
Collapse
|
20
|
Armistead JS, Jennison C, O'Neill MT, Lopaticki S, Liehl P, Hanson KK, Annoura T, Rajasekaran P, Erickson SM, Tonkin CJ, Khan SM, Mota MM, Boddey JA. Plasmodium falciparum
subtilisin-like ookinete protein SOPT plays an important and conserved role during ookinete infection of the Anopheles stephensi
midgut. Mol Microbiol 2018; 109:458-473. [DOI: 10.1111/mmi.13993] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/22/2018] [Indexed: 11/27/2022]
Affiliation(s)
- Jennifer S. Armistead
- The Walter and Eliza Hall Institute of Medical Research; Parkville 3052 Australia
- Department of Medical Biology; The University of Melbourne; Parkville 3052 Australia
| | - Charlie Jennison
- The Walter and Eliza Hall Institute of Medical Research; Parkville 3052 Australia
- Department of Medical Biology; The University of Melbourne; Parkville 3052 Australia
| | - Matthew T. O'Neill
- The Walter and Eliza Hall Institute of Medical Research; Parkville 3052 Australia
| | - Sash Lopaticki
- The Walter and Eliza Hall Institute of Medical Research; Parkville 3052 Australia
| | - Peter Liehl
- Instituto de Medicina Molecular, Faculdade de Medicina; Universidade de Lisboa; 1649-028 Lisbon Portugal
| | - Kirsten K. Hanson
- Instituto de Medicina Molecular, Faculdade de Medicina; Universidade de Lisboa; 1649-028 Lisbon Portugal
| | - Takeshi Annoura
- Leiden Malaria Research Group, Parasitology; Leiden University Medical Centre; 2333ZA Leiden the Netherlands
| | - Pravin Rajasekaran
- The Walter and Eliza Hall Institute of Medical Research; Parkville 3052 Australia
- Department of Medical Biology; The University of Melbourne; Parkville 3052 Australia
| | - Sara M. Erickson
- The Walter and Eliza Hall Institute of Medical Research; Parkville 3052 Australia
- Department of Medical Biology; The University of Melbourne; Parkville 3052 Australia
| | - Christopher J. Tonkin
- The Walter and Eliza Hall Institute of Medical Research; Parkville 3052 Australia
- Department of Medical Biology; The University of Melbourne; Parkville 3052 Australia
| | - Shahid M. Khan
- Leiden Malaria Research Group, Parasitology; Leiden University Medical Centre; 2333ZA Leiden the Netherlands
| | - Maria M. Mota
- Instituto de Medicina Molecular, Faculdade de Medicina; Universidade de Lisboa; 1649-028 Lisbon Portugal
| | - Justin A. Boddey
- The Walter and Eliza Hall Institute of Medical Research; Parkville 3052 Australia
- Department of Medical Biology; The University of Melbourne; Parkville 3052 Australia
| |
Collapse
|
21
|
Pino P, Caldelari R, Mukherjee B, Vahokoski J, Klages N, Maco B, Collins CR, Blackman MJ, Kursula I, Heussler V, Brochet M, Soldati-Favre D. A multistage antimalarial targets the plasmepsins IX and X essential for invasion and egress. Science 2018; 358:522-528. [PMID: 29074775 DOI: 10.1126/science.aaf8675] [Citation(s) in RCA: 103] [Impact Index Per Article: 17.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2017] [Accepted: 09/18/2017] [Indexed: 12/20/2022]
Abstract
Regulated exocytosis by secretory organelles is important for malaria parasite invasion and egress. Many parasite effector proteins, including perforins, adhesins, and proteases, are extensively proteolytically processed both pre- and postexocytosis. Here we report the multistage antiplasmodial activity of the aspartic protease inhibitor hydroxyl-ethyl-amine-based scaffold compound 49c. This scaffold inhibits the preexocytosis processing of several secreted rhoptry and microneme proteins by targeting the corresponding maturases plasmepsins IX (PMIX) and X (PMX), respectively. Conditional excision of PMIX revealed its crucial role in invasion, and recombinantly active PMIX and PMX cleave egress and invasion factors in a 49c-sensitive manner.
Collapse
Affiliation(s)
- Paco Pino
- Department of Microbiology and Molecular Medicine, Faculty of Medicine-University of Geneva, Centre Médical Universitaire (CMU), 1211 Geneva, Switzerland.
| | - Reto Caldelari
- Institute of Cell Biology, University of Bern, 3012 Bern, Switzerland
| | - Budhaditya Mukherjee
- Department of Microbiology and Molecular Medicine, Faculty of Medicine-University of Geneva, Centre Médical Universitaire (CMU), 1211 Geneva, Switzerland
| | - Juha Vahokoski
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway
| | - Natacha Klages
- Department of Microbiology and Molecular Medicine, Faculty of Medicine-University of Geneva, Centre Médical Universitaire (CMU), 1211 Geneva, Switzerland
| | - Bohumil Maco
- Department of Microbiology and Molecular Medicine, Faculty of Medicine-University of Geneva, Centre Médical Universitaire (CMU), 1211 Geneva, Switzerland
| | - Christine R Collins
- Malaria Biochemistry Laboratory, The Francis Crick Institute, Mill Hill, London NW1 1AT, UK
| | - Michael J Blackman
- Malaria Biochemistry Laboratory, The Francis Crick Institute, Mill Hill, London NW1 1AT, UK.,Department of Pathogen Molecular Biology, London School of Hygiene & Tropical Medicine, London WC1E 7HT, UK
| | - Inari Kursula
- Department of Biomedicine, University of Bergen, Jonas Lies vei 91, 5009 Bergen, Norway.,Biocenter Oulu and Faculty of Biochemistry and Molecular Medicine, University of Oulu, Aapistie 7, 90220 Oulu, Finland
| | - Volker Heussler
- Institute of Cell Biology, University of Bern, 3012 Bern, Switzerland
| | - Mathieu Brochet
- Department of Microbiology and Molecular Medicine, Faculty of Medicine-University of Geneva, Centre Médical Universitaire (CMU), 1211 Geneva, Switzerland
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, Faculty of Medicine-University of Geneva, Centre Médical Universitaire (CMU), 1211 Geneva, Switzerland.
| |
Collapse
|
22
|
Gural N, Mancio-Silva L, He J, Bhatia SN. Engineered Livers for Infectious Diseases. Cell Mol Gastroenterol Hepatol 2017; 5:131-144. [PMID: 29322086 PMCID: PMC5756057 DOI: 10.1016/j.jcmgh.2017.11.005] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/18/2017] [Accepted: 11/02/2017] [Indexed: 01/18/2023]
Abstract
Engineered liver systems come in a variety of platform models, from 2-dimensional cocultures of primary human hepatocytes and stem cell-derived progeny, to 3-dimensional organoids and humanized mice. Because of the species-specificity of many human hepatropic pathogens, these engineered systems have been essential tools for biologic discovery and therapeutic agent development in the context of liver-dependent infectious diseases. Although improvement of existing models is always beneficial, and the addition of a robust immune component is a particular need, at present, considerable progress has been made using this combination of research platforms. We highlight advances in the study of hepatitis B and C viruses and malaria-causing Plasmodium falciparum and Plasmodium vivax parasites, and underscore the importance of pairing the most appropriate model system and readout modality with the particular experimental question at hand, without always requiring a platform that recapitulates human physiology in its entirety.
Collapse
Key Words
- 2D, 2-dimensional
- 3D
- 3D, 3-dimensional
- EBOV, Ebola virus
- Falciparum
- HBC, hepatitis C virus
- HBV
- HBV, hepatitis B virus
- HCV
- HLC, hepatocyte-like cells
- Hepatotropic
- LASV, Lassa virus
- Liver
- Liver Models
- MPCC, micropatterned coculture system
- Malaria
- PCR, polymerase chain reaction
- Pathogen
- SACC, self-assembling coculture
- Vivax
- iHLC, induced pluripotent stem cell–derived hepatocyte-like cells
- in vitro
- in vivo
Collapse
Affiliation(s)
- Nil Gural
- Harvard-MIT Department of Health Sciences and Technology, Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Boston, Massachusetts,Koch Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Liliana Mancio-Silva
- Koch Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Jiang He
- Koch Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts
| | - Sangeeta N. Bhatia
- Koch Institute, Massachusetts Institute of Technology, Cambridge, Massachusetts,Institute for Medical Engineering and Science, Massachusetts Institute of Technology, Cambridge, Massachusetts,Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, Massachusetts,Department of Medicine, Brigham and Women’s Hospital, Boston, Massachusetts,Broad Institute, Cambridge, Massachusetts,Howard Hughes Medical Institute, Chevy Chase, Maryland,Correspondence Address correspondence to: Sangeeta N. Bhatia, MD, PhD, Koch Institute for Integrative Cancer, Research at MIT, Building 76, Room 473, 500 Main Street, Cambridge, Massachusetts 02142.
| |
Collapse
|
23
|
Zuck M, Austin LS, Danziger SA, Aitchison JD, Kaushansky A. The Promise of Systems Biology Approaches for Revealing Host Pathogen Interactions in Malaria. Front Microbiol 2017; 8:2183. [PMID: 29201016 PMCID: PMC5696578 DOI: 10.3389/fmicb.2017.02183] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2017] [Accepted: 10/24/2017] [Indexed: 12/18/2022] Open
Abstract
Despite global eradication efforts over the past century, malaria remains a devastating public health burden, causing almost half a million deaths annually (WHO, 2016). A detailed understanding of the mechanisms that control malaria infection has been hindered by technical challenges of studying a complex parasite life cycle in multiple hosts. While many interventions targeting the parasite have been implemented, the complex biology of Plasmodium poses a major challenge, and must be addressed to enable eradication. New approaches for elucidating key host-parasite interactions, and predicting how the parasite will respond in a variety of biological settings, could dramatically enhance the efficacy and longevity of intervention strategies. The field of systems biology has developed methodologies and principles that are well poised to meet these challenges. In this review, we focus our attention on the Liver Stage of the Plasmodium lifecycle and issue a “call to arms” for using systems biology approaches to forge a new era in malaria research. These approaches will reveal insights into the complex interplay between host and pathogen, and could ultimately lead to novel intervention strategies that contribute to malaria eradication.
Collapse
Affiliation(s)
- Meghan Zuck
- Center for Infectious Disease Research, formerly Seattle Biomedical Research Institute, Seattle, WA, United States
| | - Laura S Austin
- Center for Infectious Disease Research, formerly Seattle Biomedical Research Institute, Seattle, WA, United States
| | - Samuel A Danziger
- Center for Infectious Disease Research, formerly Seattle Biomedical Research Institute, Seattle, WA, United States.,Institute for Systems Biology, Seattle, WA, United States
| | - John D Aitchison
- Center for Infectious Disease Research, formerly Seattle Biomedical Research Institute, Seattle, WA, United States.,Institute for Systems Biology, Seattle, WA, United States
| | - Alexis Kaushansky
- Center for Infectious Disease Research, formerly Seattle Biomedical Research Institute, Seattle, WA, United States.,Department of Global Health, University of Washington, Seattle, WA, United States
| |
Collapse
|
24
|
|
25
|
Deu E. Proteases as antimalarial targets: strategies for genetic, chemical, and therapeutic validation. FEBS J 2017; 284:2604-2628. [PMID: 28599096 PMCID: PMC5575534 DOI: 10.1111/febs.14130] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2017] [Revised: 04/29/2017] [Accepted: 06/06/2017] [Indexed: 01/17/2023]
Abstract
Malaria is a devastating parasitic disease affecting half of the world's population. The rapid emergence of resistance against new antimalarial drugs, including artemisinin-based therapies, has made the development of drugs with novel mechanisms of action extremely urgent. Proteases are enzymes proven to be well suited for target-based drug development due to our knowledge of their enzymatic mechanisms and active site structures. More importantly, Plasmodium proteases have been shown to be involved in a variety of pathways that are essential for parasite survival. However, pharmacological rather than target-based approaches have dominated the field of antimalarial drug development, in part due to the challenge of robustly validating Plasmodium targets at the genetic level. Fortunately, over the last few years there has been significant progress in the development of efficient genetic methods to modify the parasite, including several conditional approaches. This progress is finally allowing us not only to validate essential genes genetically, but also to study their molecular functions. In this review, I present our current understanding of the biological role proteases play in the malaria parasite life cycle. I also discuss how the recent advances in Plasmodium genetics, the improvement of protease-oriented chemical biology approaches, and the development of malaria-focused pharmacological assays, can be combined to achieve a robust biological, chemical and therapeutic validation of Plasmodium proteases as viable drug targets.
Collapse
Affiliation(s)
- Edgar Deu
- Chemical Biology Approaches to Malaria LaboratoryThe Francis Crick InstituteLondonUK
| |
Collapse
|
26
|
Plasmodium berghei PIMMS2 Promotes Ookinete Invasion of the Anopheles gambiae Mosquito Midgut. Infect Immun 2017; 85:IAI.00139-17. [PMID: 28559405 PMCID: PMC5520436 DOI: 10.1128/iai.00139-17] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Accepted: 05/11/2017] [Indexed: 12/21/2022] Open
Abstract
Mosquito midgut stages of the malaria parasite present an attractive biological system to study host-parasite interactions and develop interventions to block disease transmission. Mosquito infection ensues upon oocyst development that follows ookinete invasion and traversal of the mosquito midgut epithelium. Here, we report the characterization of PIMMS2 (Plasmodium invasion of mosquito midgut screen candidate 2), a Plasmodium berghei protein with structural similarities to subtilisin-like proteins. PIMMS2 orthologs are present in the genomes of all plasmodia and are mapped between the subtilisin-encoding genes SUB1 and SUB3. P. berghei PIMMS2 is specifically expressed in zygotes and ookinetes and is localized on the ookinete surface. Loss of PIMMS2 function through gene disruption by homologous recombination leads to normal development of motile ookinetes that exhibit a severely impaired capacity to traverse the mosquito midgut and transform to oocysts. Genetic complementation of the disrupted locus with a mutated PIMMS2 allele reveals that amino acid residues corresponding to the putative subtilisin-like catalytic triad are important but not essential for protein function. Our data demonstrate that PIMMS2 is a novel ookinete-specific protein that promotes parasite traversal of the mosquito midgut epithelium and establishment of mosquito infection.
Collapse
|
27
|
Mishra M, Mishra VK, Kashaw V, Iyer AK, Kashaw SK. Comprehensive review on various strategies for antimalarial drug discovery. Eur J Med Chem 2016; 125:1300-1320. [PMID: 27886547 DOI: 10.1016/j.ejmech.2016.11.025] [Citation(s) in RCA: 65] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2016] [Revised: 11/07/2016] [Accepted: 11/11/2016] [Indexed: 01/14/2023]
Abstract
The resistance of malaria parasites to existing drugs carries on growing and progressively limiting our ability to manage this severe disease and finally lead to a massive global health burden. Till now, malaria control has relied upon the traditional quinoline, antifolate and artemisinin compounds. Very few new antimalarials were developed in the past 50 years. Among recent approaches, identification of novel chemotherapeutic targets, exploration of natural products with medicinal significance, covalent bitherapy having a dual mode of action into a single hybrid molecule and malaria vaccine development are explored heavily. The proper execution of these approaches and proper investment from international agencies will accelerate the discovery of drugs that provide new hope for the control or eventual eradication of this global infectious disease. This review explores various strategies for assessment and development of new antimalarial drugs. Current status and scientific value of previous approaches are systematically reviewed and new approaches provide a pragmatic forecast for future developments are introduced as well.
Collapse
Affiliation(s)
- Mitali Mishra
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar, MP, India
| | - Vikash K Mishra
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar, MP, India
| | - Varsha Kashaw
- SVN Institute of Pharmaceutical Sciences, SVN University, Sagar, MP, India
| | - Arun K Iyer
- Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, USA
| | - Sushil Kumar Kashaw
- Department of Pharmaceutical Sciences, Dr. Harisingh Gour University (A Central University), Sagar, MP, India; Use-inspired Biomaterials & Integrated Nano Delivery (U-BiND) Systems Laboratory, Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, USA.
| |
Collapse
|
28
|
Yan X, Tao G, Liu X, Ji Y, Suo X. Calcium-dependent microneme protein discharge and in vitro egress of Eimeria tenella sporozoites. Exp Parasitol 2016; 170:193-197. [PMID: 27680997 DOI: 10.1016/j.exppara.2016.09.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2015] [Revised: 09/02/2016] [Accepted: 09/15/2016] [Indexed: 11/17/2022]
Abstract
Egress is a vital step in the endogenous development of apicomplexan parasites, as it assures the parasites exit from consumed host cells and entry into fresh ones. However, little information has previously been reported on this step of Eimeria spp. In this study, we investigated in vitro egress of Eimeria tenella sporozoites triggered by acetaldehyde. We found that addition of exogenous acetaldehyde induces egress of sporozoites from primary chicken kidney cells (PCKs) and stimulate secretion of E. tenella microneme 2 protein (EtMic 2). Moreover, by using cellular calcium inhibitors, we further proved that these processes were dependent on the intracellular calcium of the parasites. Our findings provide clues to the study of interaction between eimerian parasites and their hosts.
Collapse
Affiliation(s)
- Xinlei Yan
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, 100193, China; National Animal Protozoa Laboratory & College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Geru Tao
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, 100193, China; National Animal Protozoa Laboratory & College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Xianyong Liu
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, 100193, China; National Animal Protozoa Laboratory & College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China; Key Laboratory of Zoonosis of Ministry of Agriculture & College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Yongsheng Ji
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, 100193, China; National Animal Protozoa Laboratory & College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China
| | - Xun Suo
- State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, 100193, China; National Animal Protozoa Laboratory & College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China; Key Laboratory of Zoonosis of Ministry of Agriculture & College of Veterinary Medicine, China Agricultural University, Beijing, 100193, China.
| |
Collapse
|
29
|
Kehrer J, Frischknecht F, Mair GR. Proteomic Analysis of the Plasmodium berghei Gametocyte Egressome and Vesicular bioID of Osmiophilic Body Proteins Identifies Merozoite TRAP-like Protein (MTRAP) as an Essential Factor for Parasite Transmission. Mol Cell Proteomics 2016; 15:2852-62. [PMID: 27371728 PMCID: PMC5013303 DOI: 10.1074/mcp.m116.058263] [Citation(s) in RCA: 67] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 06/17/2016] [Indexed: 01/08/2023] Open
Abstract
Malaria transmission from an infected host to the mosquito vector requires the uptake of intraerythrocytic sexual precursor cells into the mosquito midgut. For the release of mature extracellular gametes two membrane barriers-the parasite parasitophorous vacuole membrane and the host red blood cell membrane-need to be dissolved. Membrane lysis occurs after the release of proteins from specialized secretory vesicles including osmiophilic bodies. In this study we conducted proteomic analyses of the P. berghei gametocyte egressome and developed a vesicular bioID approach to identify hitherto unknown proteins with a potential function in gametocyte egress. This first Plasmodium gametocyte egressome includes the proteins released by the parasite during the lysis of the parasitophorous vacuole membrane and red blood cell membrane. BioID of the osmiophilic body protein MDV1/PEG3 revealed a vesicular proteome of these gametocyte-specific secretory vesicles. Fluorescent protein tagging and gene deletion approaches were employed to validate and identify a set of novel factors essential for this lysis and egress process. Our study provides the first in vivo bioID for a rodent malaria parasite and together with the first Plasmodium gametocyte egressome identifies MTRAP as a novel factor essential for mosquito transmission. Our data provide an important resource for proteins potentially involved in a key step of gametogenesis.
Collapse
Affiliation(s)
- Jessica Kehrer
- From the ‡Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Friedrich Frischknecht
- From the ‡Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Gunnar R Mair
- From the ‡Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| |
Collapse
|
30
|
Brochet M, Billker O. Calcium signalling in malaria parasites. Mol Microbiol 2016; 100:397-408. [PMID: 26748879 DOI: 10.1111/mmi.13324] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/05/2016] [Indexed: 12/24/2022]
Abstract
Ca(2+) is a ubiquitous intracellular messenger in malaria parasites with important functions in asexual blood stages responsible for malaria symptoms, the preceding liver-stage infection and transmission through the mosquito. Intracellular messengers amplify signals by binding to effector molecules that trigger physiological changes. The characterisation of some Ca(2+) effector proteins has begun to provide insights into the vast range of biological processes controlled by Ca(2+) signalling in malaria parasites, including host cell egress and invasion, protein secretion, motility and cell cycle regulation. Despite the importance of Ca(2+) signalling during the life cycle of malaria parasites, little is known about Ca(2+) homeostasis. Recent findings highlighted that upstream of stage-specific Ca(2+) effectors is a conserved interplay between second messengers to control critical intracellular Ca(2+) signals throughout the life cycle. The identification of the molecular mechanisms integrating stage-transcending mechanisms of Ca(2+) homeostasis in a network of stage-specific regulator and effector pathways now represents a major challenge for a meaningful understanding of Ca(2+) signalling in malaria parasites.
Collapse
Affiliation(s)
- Mathieu Brochet
- Faculty of Medicine, Department of Microbiology and Molecular Medicine, University of Geneva, 1 Rue Michel-Servet, CH-1211 Geneva 4, Switzerland.,UMR5235 CNRS-Université Montpellier 2, 34095, Montpellier, France
| | - Oliver Billker
- Wellcome Trust Sanger Institute, Malaria Programme, CB10 1SA, Hinxton, UK
| |
Collapse
|
31
|
Brogi S, Giovani S, Brindisi M, Gemma S, Novellino E, Campiani G, Blackman MJ, Butini S. In silico study of subtilisin-like protease 1 (SUB1) from different Plasmodium species in complex with peptidyl-difluorostatones and characterization of potent pan-SUB1 inhibitors. J Mol Graph Model 2016; 64:121-130. [PMID: 26826801 PMCID: PMC5276822 DOI: 10.1016/j.jmgm.2016.01.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2015] [Revised: 12/04/2015] [Accepted: 01/16/2016] [Indexed: 11/23/2022]
Abstract
Homology models of four SUB1 orthologues from P. falciparum species were produced. We analyzed the binding mode of our previous difluorostatone inhibitors to six SUB1. In vitro activity of our difluorostatone-based inhibitors was correctly predicted. We derived a structure-based pan-SUB1 pharmacophore, and validated it in silico. We confirmed that development of pan-SUB1 inhibitors is a feasible task.
Plasmodium falciparum subtilisin-like protease 1 (SUB1) is a novel target for the development of innovative antimalarials. We recently described the first potent difluorostatone-based inhibitors of the enzyme ((4S)-(N-((N-acetyl-l-lysyl)-l-isoleucyl-l-threonyl-l-alanyl)-2,2-difluoro-3-oxo-4-aminopentanoyl)glycine (1) and (4S)-(N-((N-acetyl-l-isoleucyl)-l-threonyl-l-alanylamino)-2,2-difluoro-3-oxo-4-aminopentanoyl)glycine (2)). As a continuation of our efforts towards the definition of the molecular determinants of enzyme-inhibitor interaction, we herein propose the first comprehensive computational investigation of the SUB1 catalytic core from six different Plasmodium species, using homology modeling and molecular docking approaches. Investigation of the differences in the binding sites as well as the interactions of our inhibitors 1,2 with all SUB1 orthologues, allowed us to highlight the structurally relevant regions of the enzyme that could be targeted for developing pan-SUB1 inhibitors. According to our in silico predictions, compounds 1,2 have been demonstrated to be potent inhibitors of SUB1 from all three major clinically relevant Plasmodium species (P. falciparum, P. vivax, and P. knowlesi). We next derived multiple structure-based pharmacophore models that were combined in an inclusive pan-SUB1 pharmacophore (SUB1-PHA). This latter was validated by applying in silico methods, showing that it may be useful for the future development of potent antimalarial agents.
Collapse
Affiliation(s)
- Simone Brogi
- European Research Centre for Drug Discovery and Development (NatSynDrugs), University of Siena, via Aldo Moro 2, 53100, Siena, Italy; Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena, via Aldo Moro 2, 53100, Siena, Italy; Centro Interuniversitario di Ricerche sulla Malaria (CIRM), University of Perugia, Perugia, Italy
| | - Simone Giovani
- European Research Centre for Drug Discovery and Development (NatSynDrugs), University of Siena, via Aldo Moro 2, 53100, Siena, Italy; Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena, via Aldo Moro 2, 53100, Siena, Italy; Centro Interuniversitario di Ricerche sulla Malaria (CIRM), University of Perugia, Perugia, Italy
| | - Margherita Brindisi
- European Research Centre for Drug Discovery and Development (NatSynDrugs), University of Siena, via Aldo Moro 2, 53100, Siena, Italy; Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena, via Aldo Moro 2, 53100, Siena, Italy; Centro Interuniversitario di Ricerche sulla Malaria (CIRM), University of Perugia, Perugia, Italy
| | - Sandra Gemma
- European Research Centre for Drug Discovery and Development (NatSynDrugs), University of Siena, via Aldo Moro 2, 53100, Siena, Italy; Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena, via Aldo Moro 2, 53100, Siena, Italy; Centro Interuniversitario di Ricerche sulla Malaria (CIRM), University of Perugia, Perugia, Italy.
| | - Ettore Novellino
- European Research Centre for Drug Discovery and Development (NatSynDrugs), University of Siena, via Aldo Moro 2, 53100, Siena, Italy; Dipartimento di Farmacia, University of Naples Federico II, Via D. Montesano 49, 80131, Naples, Italy
| | - Giuseppe Campiani
- European Research Centre for Drug Discovery and Development (NatSynDrugs), University of Siena, via Aldo Moro 2, 53100, Siena, Italy; Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena, via Aldo Moro 2, 53100, Siena, Italy; Centro Interuniversitario di Ricerche sulla Malaria (CIRM), University of Perugia, Perugia, Italy.
| | - Michael J Blackman
- Division of Parasitology, MRC National Institute for Medical Research, Mill Hill, London NW7 1AA, UK
| | - Stefania Butini
- European Research Centre for Drug Discovery and Development (NatSynDrugs), University of Siena, via Aldo Moro 2, 53100, Siena, Italy; Dipartimento di Biotecnologie, Chimica e Farmacia, University of Siena, via Aldo Moro 2, 53100, Siena, Italy; Centro Interuniversitario di Ricerche sulla Malaria (CIRM), University of Perugia, Perugia, Italy
| |
Collapse
|
32
|
Kalanon M, Bargieri D, Sturm A, Matthews K, Ghosh S, Goodman CD, Thiberge S, Mollard V, McFadden GI, Ménard R, Koning‐Ward TF. The
Plasmodium
translocon of exported proteins component EXP2 is critical for establishing a patent malaria infection in mice. Cell Microbiol 2015; 18:399-412. [DOI: 10.1111/cmi.12520] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2015] [Revised: 08/26/2015] [Accepted: 08/31/2015] [Indexed: 12/12/2022]
Affiliation(s)
- Ming Kalanon
- Molecular and Medical Research Unit, School of MedicineDeakin University Waurn Ponds Geelong Victoria 3216 Australia
| | - Daniel Bargieri
- Unité de Biologie et Génétique du PaludismeInstitut Pasteur Paris France
- Department of Parasitology, Institute of Biomedical SciencesUniversity of São Paulo São Paulo SP Brazil
| | - Angelika Sturm
- School of BioSciencesThe University of Melbourne Parkville Victoria 3010 Australia
| | - Kathryn Matthews
- Molecular and Medical Research Unit, School of MedicineDeakin University Waurn Ponds Geelong Victoria 3216 Australia
| | - Sreejoyee Ghosh
- Molecular and Medical Research Unit, School of MedicineDeakin University Waurn Ponds Geelong Victoria 3216 Australia
| | | | - Sabine Thiberge
- Unité de Biologie et Génétique du PaludismeInstitut Pasteur Paris France
| | - Vanessa Mollard
- School of BioSciencesThe University of Melbourne Parkville Victoria 3010 Australia
| | - Geoffrey I. McFadden
- School of BioSciencesThe University of Melbourne Parkville Victoria 3010 Australia
| | - Robert Ménard
- Unité de Biologie et Génétique du PaludismeInstitut Pasteur Paris France
| | - Tania F. Koning‐Ward
- Molecular and Medical Research Unit, School of MedicineDeakin University Waurn Ponds Geelong Victoria 3216 Australia
| |
Collapse
|
33
|
Pieperhoff MS, Pall GS, Jiménez-Ruiz E, Das S, Melatti C, Gow M, Wong EH, Heng J, Müller S, Blackman MJ, Meissner M. Conditional U1 Gene Silencing in Toxoplasma gondii. PLoS One 2015; 10:e0130356. [PMID: 26090798 PMCID: PMC4474610 DOI: 10.1371/journal.pone.0130356] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2014] [Accepted: 05/18/2015] [Indexed: 11/19/2022] Open
Abstract
The functional characterisation of essential genes in apicomplexan parasites, such as Toxoplasma gondii or Plasmodium falciparum, relies on conditional mutagenesis systems. Here we present a novel strategy based on U1 snRNP-mediated gene silencing. U1 snRNP is critical in pre-mRNA splicing by defining the exon-intron boundaries. When a U1 recognition site is placed into the 3'-terminal exon or adjacent to the termination codon, pre-mRNA is cleaved at the 3'-end and degraded, leading to an efficient knockdown of the gene of interest (GOI). Here we describe a simple method that combines endogenous tagging with DiCre-mediated positioning of U1 recognition sites adjacent to the termination codon of the GOI which leads to a conditional knockdown of the GOI upon rapamycin-induction. Specific knockdown mutants of the reporter gene GFP and several endogenous genes of T. gondii including the clathrin heavy chain gene 1 (chc1), the vacuolar protein sorting gene 26 (vps26), and the dynamin-related protein C gene (drpC) were silenced using this approach and demonstrate the potential of this technology. We also discuss advantages and disadvantages of this method in comparison to other technologies in more detail.
Collapse
Affiliation(s)
- Manuela S. Pieperhoff
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, Glasgow, Lanarkshire, United Kingdom
| | - Gurman S. Pall
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, Glasgow, Lanarkshire, United Kingdom
| | - Elena Jiménez-Ruiz
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, Glasgow, Lanarkshire, United Kingdom
| | - Sujaan Das
- Division of Parasitology, MRC National Institute for Medical Research, Mill Hill, London, United Kingdom
| | - Carmen Melatti
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, Glasgow, Lanarkshire, United Kingdom
| | - Matthew Gow
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, Glasgow, Lanarkshire, United Kingdom
| | - Eleanor H. Wong
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, Glasgow, Lanarkshire, United Kingdom
| | - Joanne Heng
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, Glasgow, Lanarkshire, United Kingdom
| | - Sylke Müller
- Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, Glasgow, Lanarkshire, United Kingdom
| | - Michael J. Blackman
- Division of Parasitology, MRC National Institute for Medical Research, Mill Hill, London, United Kingdom
- * E-mail: (MM); (MJB)
| | - Markus Meissner
- Wellcome Trust Centre for Molecular Parasitology, Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary and Life Sciences, Glasgow, Lanarkshire, United Kingdom
- * E-mail: (MM); (MJB)
| |
Collapse
|
34
|
de Koning-Ward TF, Gilson PR, Crabb BS. Advances in molecular genetic systems in malaria. Nat Rev Microbiol 2015; 13:373-87. [DOI: 10.1038/nrmicro3450] [Citation(s) in RCA: 91] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
35
|
Burda PC, Roelli MA, Schaffner M, Khan SM, Janse CJ, Heussler VT. A Plasmodium phospholipase is involved in disruption of the liver stage parasitophorous vacuole membrane. PLoS Pathog 2015; 11:e1004760. [PMID: 25786000 PMCID: PMC4364735 DOI: 10.1371/journal.ppat.1004760] [Citation(s) in RCA: 65] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2014] [Accepted: 02/22/2015] [Indexed: 11/18/2022] Open
Abstract
The coordinated exit of intracellular pathogens from host cells is a process critical to the success and spread of an infection. While phospholipases have been shown to play important roles in bacteria host cell egress and virulence, their role in the release of intracellular eukaryotic parasites is largely unknown. We examined a malaria parasite protein with phospholipase activity and found it to be involved in hepatocyte egress. In hepatocytes, Plasmodium parasites are surrounded by a parasitophorous vacuole membrane (PVM), which must be disrupted before parasites are released into the blood. However, on a molecular basis, little is known about how the PVM is ruptured. We show that Plasmodium berghei phospholipase, PbPL, localizes to the PVM in infected hepatocytes. We provide evidence that parasites lacking PbPL undergo completely normal liver stage development until merozoites are produced but have a defect in egress from host hepatocytes. To investigate this further, we established a live-cell imaging-based assay, which enabled us to study the temporal dynamics of PVM rupture on a quantitative basis. Using this assay we could show that PbPL-deficient parasites exhibit impaired PVM rupture, resulting in delayed parasite egress. A wild-type phenotype could be re-established by gene complementation, demonstrating the specificity of the PbPL deletion phenotype. In conclusion, we have identified for the first time a Plasmodium phospholipase that is important for PVM rupture and in turn for parasite exit from the infected hepatocyte and therefore established a key role of a parasite phospholipase in egress. Leaving their host cell is a crucial process for intracellular pathogens, allowing successful infection of other cells and thereby spreading of infection. Plasmodium parasites infect hepatocytes and red blood cells, and inside these cells they are contained within a vacuole like many other intracellular pathogens. Before parasites can infect other cells, the surrounding parasitophorous vacuole membrane (PVM) needs to be ruptured. However, little is known about this process on a molecular level and Plasmodium proteins mediating lysis of the PVM during parasite egress have not so far been identified. In this study, we characterize a Plasmodium phospholipase and show that it localizes to the PVM of parasites within hepatocytes. We demonstrate that parasites lacking this protein have a defect in rupture of the PVM and thereby in host cell egress. In conclusion, our study shows for the first time that a phospholipase plays a role in PVM disruption of an intracellular eukaryotic parasite.
Collapse
Affiliation(s)
- Paul-Christian Burda
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- Graduate School of Cellular Biology, University of Bern, Bern, Switzerland
- * E-mail:
| | | | - Marco Schaffner
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Shahid M. Khan
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | - Chris J. Janse
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Center, Leiden, The Netherlands
| | | |
Collapse
|
36
|
Bastianelli G, Bouillon A, Nguyen C, Le-Nguyen D, Nilges M, Barale JC. Computational design of protein-based inhibitors of Plasmodium vivax subtilisin-like 1 protease. PLoS One 2014; 9:e109269. [PMID: 25343504 PMCID: PMC4208747 DOI: 10.1371/journal.pone.0109269] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Accepted: 08/16/2014] [Indexed: 01/24/2023] Open
Abstract
BACKGROUND Malaria remains a major global health concern. The development of novel therapeutic strategies is critical to overcome the selection of multiresistant parasites. The subtilisin-like protease (SUB1) involved in the egress of daughter Plasmodium parasites from infected erythrocytes and in their subsequent invasion into fresh erythrocytes has emerged as an interesting new drug target. FINDINGS Using a computational approach based on homology modeling, protein-protein docking and mutation scoring, we designed protein-based inhibitors of Plasmodium vivax SUB1 (PvSUB1) and experimentally evaluated their inhibitory activity. The small peptidic trypsin inhibitor EETI-II was used as scaffold. We mutated residues at specific positions (P4 and P1) and calculated the change in free-energy of binding with PvSUB1. In agreement with our predictions, we identified a mutant of EETI-II (EETI-II-P4LP1W) with a Ki in the medium micromolar range. CONCLUSIONS Despite the challenges related to the lack of an experimental structure of PvSUB1, the computational protocol we developed in this study led to the design of protein-based inhibitors of PvSUB1. The approach we describe in this paper, together with other examples, demonstrates the capabilities of computational procedures to accelerate and guide the design of novel proteins with interesting therapeutic applications.
Collapse
Affiliation(s)
- Giacomo Bastianelli
- Institut Pasteur, Unité de Bioinformatique Structurale, Département de Biologie Structurale et Chimie, Paris, France
- CNRS UMR 3528, Paris, France
| | - Anthony Bouillon
- Institut Pasteur, Unité d’Immunologie Moléculaires des Parasites, Département de Parasitologie et de Mycologie & CNRS URA 2581, Paris, France
- CNRS, URA2581, Paris, France
| | | | | | - Michael Nilges
- Institut Pasteur, Unité de Bioinformatique Structurale, Département de Biologie Structurale et Chimie, Paris, France
- CNRS UMR 3528, Paris, France
| | - Jean-Christophe Barale
- Institut Pasteur, Unité d’Immunologie Moléculaires des Parasites, Département de Parasitologie et de Mycologie & CNRS URA 2581, Paris, France
- CNRS, URA2581, Paris, France
| |
Collapse
|
37
|
Giganti D, Bouillon A, Tawk L, Robert F, Martinez M, Crublet E, Weber P, Girard-Blanc C, Petres S, Haouz A, Hernandez JF, Mercereau-Puijalon O, Alzari PM, Barale JC. A novel Plasmodium-specific prodomain fold regulates the malaria drug target SUB1 subtilase. Nat Commun 2014; 5:4833. [PMID: 25204226 DOI: 10.1038/ncomms5833] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2014] [Accepted: 07/29/2014] [Indexed: 11/09/2022] Open
Abstract
The Plasmodium subtilase SUB1 plays a pivotal role during the egress of malaria parasites from host hepatocytes and erythrocytes. Here we report the crystal structure of full-length SUB1 from the human-infecting parasite Plasmodium vivax, revealing a bacterial-like catalytic domain in complex with a Plasmodium-specific prodomain. The latter displays a novel architecture with an amino-terminal insertion that functions as a 'belt', embracing the catalytic domain to further stabilize the quaternary structure of the pre-protease, and undergoes calcium-dependent autoprocessing during subsequent activation. Although dispensable for recombinant enzymatic activity, the SUB1 'belt' could not be deleted in Plasmodium berghei, suggesting an essential role of this domain for parasite development in vivo. The SUB1 structure not only provides a valuable platform to develop new anti-malarial candidates against this promising drug target, but also defines the Plasmodium-specific 'belt' domain as a key calcium-dependent regulator of SUB1 during parasite egress from host cells.
Collapse
Affiliation(s)
- David Giganti
- 1] Institut Pasteur, Unité de Microbiologie Structurale, Département de Biologie Structurale et Chimie, F-75015 Paris, France [2] CNRS UMR 3528, F-75015 Paris, France
| | - Anthony Bouillon
- 1] Institut Pasteur, Unité d'Immunologie Moléculaires des Parasites, Département de Parasitologie et de Mycologie, F-75015 Paris, France [2] CNRS URA 2581, F-75015 Paris, France
| | - Lina Tawk
- 1] Institut Pasteur, Unité d'Immunologie Moléculaires des Parasites, Département de Parasitologie et de Mycologie, F-75015 Paris, France [2] CNRS URA 2581, F-75015 Paris, France
| | - Fabienne Robert
- 1] Institut Pasteur, Unité d'Immunologie Moléculaires des Parasites, Département de Parasitologie et de Mycologie, F-75015 Paris, France [2] CNRS URA 2581, F-75015 Paris, France
| | - Mariano Martinez
- 1] Institut Pasteur, Unité de Microbiologie Structurale, Département de Biologie Structurale et Chimie, F-75015 Paris, France [2] CNRS UMR 3528, F-75015 Paris, France
| | - Elodie Crublet
- Institut Pasteur, Proteopole &CNRS UMR 3528, F-75015 Paris, France
| | - Patrick Weber
- Institut Pasteur, Proteopole &CNRS UMR 3528, F-75015 Paris, France
| | | | - Stéphane Petres
- Institut Pasteur, Proteopole &CNRS UMR 3528, F-75015 Paris, France
| | - Ahmed Haouz
- Institut Pasteur, Proteopole &CNRS UMR 3528, F-75015 Paris, France
| | - Jean-François Hernandez
- Faculté de Pharmacie, Institut des Biomolécules Max Mousseron, UMR5247, CNRS, Universités Montpellier 1 &2, 15 avenue Charles Flahault, 34093 Montpellier cedex 5, France
| | - Odile Mercereau-Puijalon
- 1] Institut Pasteur, Unité d'Immunologie Moléculaires des Parasites, Département de Parasitologie et de Mycologie, F-75015 Paris, France [2] CNRS URA 2581, F-75015 Paris, France
| | - Pedro M Alzari
- 1] Institut Pasteur, Unité de Microbiologie Structurale, Département de Biologie Structurale et Chimie, F-75015 Paris, France [2] CNRS UMR 3528, F-75015 Paris, France [3] Institut Pasteur, Proteopole &CNRS UMR 3528, F-75015 Paris, France
| | - Jean-Christophe Barale
- 1] Institut Pasteur, Unité d'Immunologie Moléculaires des Parasites, Département de Parasitologie et de Mycologie, F-75015 Paris, France [2] CNRS URA 2581, F-75015 Paris, France
| |
Collapse
|
38
|
Serine Proteases of Malaria Parasite Plasmodium falciparum: Potential as Antimalarial Drug Targets. Interdiscip Perspect Infect Dis 2014; 2014:453186. [PMID: 24799897 PMCID: PMC3988940 DOI: 10.1155/2014/453186] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2013] [Revised: 01/02/2014] [Accepted: 01/07/2014] [Indexed: 01/08/2023] Open
Abstract
Malaria is a major global parasitic disease and a cause of enormous mortality and morbidity. Widespread drug resistance against currently available antimalarials warrants the identification of novel drug targets and development of new drugs. Malarial proteases are a group of molecules that serve as potential drug targets because of their essentiality for parasite life cycle stages and feasibility of designing specific inhibitors against them. Proteases belonging to various mechanistic classes are found in P. falciparum, of which serine proteases are of particular interest due to their involvement in parasite-specific processes of egress and invasion. In P. falciparum, a number of serine proteases belonging to chymotrypsin, subtilisin, and rhomboid clans are found. This review focuses on the potential of P. falciparum serine proteases as antimalarial drug targets.
Collapse
|