1
|
Moss WJ, Brusini L, Kuehnel R, Brochet M, Brown KM. Apicomplexan phosphodiesterases in cyclic nucleotide turnover: conservation, function, and therapeutic potential. mBio 2024; 15:e0305623. [PMID: 38132724 PMCID: PMC10865986 DOI: 10.1128/mbio.03056-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
Apicomplexa encompasses a large number of intracellular parasites infecting a wide range of animals. Cyclic nucleotide signaling is crucial for a variety of apicomplexan life stages and cellular processes. The cyclases and kinases that synthesize and respond to cyclic nucleotides (i.e., 3',5'-cyclic guanosine monophosphate and 3',5'-cyclic adenosine monophosphate) are highly conserved and essential throughout the parasite phylum. Growing evidence indicates that phosphodiesterases (PDEs) are also critical for regulating cyclic nucleotide signaling via cyclic nucleotide hydrolysis. Here, we discuss recent advances in apicomplexan PDE biology and opportunities for therapeutic interventions, with special emphasis on the major human apicomplexan parasite genera Plasmodium, Toxoplasma, Cryptosporidium, and Babesia. In particular, we show a highly flexible repertoire of apicomplexan PDEs associated with a wide range of cellular requirements across parasites and lifecycle stages. Despite this phylogenetic diversity, cellular requirements of apicomplexan PDEs for motility, host cell egress, or invasion are conserved. However, the molecular wiring of associated PDEs is extremely malleable suggesting that PDE diversity and redundancy are key for the optimization of cyclic nucleotide turnover to respond to the various environments encountered by each parasite and life stage. Understanding how apicomplexan PDEs are regulated and integrating multiple signaling systems into a unified response represent an untapped avenue for future exploration.
Collapse
Affiliation(s)
- William J. Moss
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| | - Lorenzo Brusini
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Ronja Kuehnel
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Mathieu Brochet
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland
| | - Kevin M. Brown
- Department of Microbiology and Immunology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
2
|
Rawat RS, Gupta A, Antil N, Bhatnagar S, Singh M, Rawat A, Prasad TSK, Sharma P. Protein kinase PfPK2 mediated signalling is critical for host erythrocyte invasion by malaria parasite. PLoS Pathog 2023; 19:e1011770. [PMID: 37988347 PMCID: PMC10662742 DOI: 10.1371/journal.ppat.1011770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 10/23/2023] [Indexed: 11/23/2023] Open
Abstract
Signalling pathways in malaria parasite remain poorly defined and major reason for this is the lack of understanding of the function of majority of parasite protein kinases and phosphatases in parasite signalling and its biology. In the present study, we have elucidated the function of Protein Kinase 2 (PfPK2), which is known to be indispensable for the survival of human malaria parasite Plasmodium falciparum. We demonstrate that it is involved in the invasion of host erythrocytes, which is critical for establishing infection. In addition, PfPK2 may also be involved in the maturation of the parasite post-invasion. PfPK2 regulates the release of microneme proteins like Apical Membrane Antigen 1 (AMA1), which facilitates the formation of Tight Junction between the merozoite and host erythrocyte- a key step in the process of invasion. Comparative phosphoproteomics studies revealed that PfPK2 may be involved in regulation of several key proteins involved in invasion and signalling. Furthermore, PfPK2 regulates the generation of cGMP and the release of calcium in the parasite, which are key second messengers for the process of invasion. These and other studies have shed light on a novel signalling pathway in which PfPK2 acts as an upstream regulator of important cGMP-calcium signalling, which plays an important role in parasite invasion.
Collapse
Affiliation(s)
- Rahul Singh Rawat
- Eukaryotic Gene Expression Laboratory, National Institute of Immunology, New Delhi, India
| | - Ankit Gupta
- Eukaryotic Gene Expression Laboratory, National Institute of Immunology, New Delhi, India
| | - Neelam Antil
- Institute of Bioinformatics, International Tech Park, Bangalore, India
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam, India
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Sonika Bhatnagar
- Eukaryotic Gene Expression Laboratory, National Institute of Immunology, New Delhi, India
| | - Monika Singh
- Eukaryotic Gene Expression Laboratory, National Institute of Immunology, New Delhi, India
| | - Akanksha Rawat
- Eukaryotic Gene Expression Laboratory, National Institute of Immunology, New Delhi, India
| | - T. S. Keshava Prasad
- Center for Systems Biology and Molecular Medicine, Yenepoya Research Centre, Yenepoya (Deemed to be University), Mangalore, India
| | - Pushkar Sharma
- Eukaryotic Gene Expression Laboratory, National Institute of Immunology, New Delhi, India
| |
Collapse
|
3
|
Santos BMD, Przyborski JM, Garcia CRS. Changes in K + Concentration as a Signaling Mechanism in the Apicomplexa Parasites Plasmodium and Toxoplasma. Int J Mol Sci 2023; 24:ijms24087276. [PMID: 37108438 PMCID: PMC10138558 DOI: 10.3390/ijms24087276] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 04/04/2023] [Accepted: 04/04/2023] [Indexed: 04/29/2023] Open
Abstract
During their life cycle, apicomplexan parasites pass through different microenvironments and encounter a range of ion concentrations. The discovery that the GPCR-like SR25 in Plasmodium falciparum is activated by a shift in potassium concentration indicates that the parasite can take advantage of its development by sensing different ionic concentrations in the external milieu. This pathway involves the activation of phospholipase C and an increase in cytosolic calcium. In the present report, we summarize the information available in the literature regarding the role of potassium ions during parasite development. A deeper understanding of the mechanisms that allow the parasite to cope with ionic potassium changes contributes to our knowledge about the cell cycle of Plasmodium spp.
Collapse
Affiliation(s)
- Benedito M Dos Santos
- Department of Clinical and Toxicological Analysis, School of Pharmaceutical Sciences, University of Sao Paulo, Sao Paulo 05508-000, Brazil
| | - Jude M Przyborski
- Department of Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus-Liebig University, 35390 Gießen, Germany
| | - Célia R S Garcia
- Department of Biochemistry and Molecular Biology, Interdisciplinary Research Center, Justus-Liebig University, 35390 Gießen, Germany
| |
Collapse
|
4
|
de Oliveira LS, Alborghetti MR, Carneiro RG, Bastos IMD, Amino R, Grellier P, Charneau S. Calcium in the Backstage of Malaria Parasite Biology. Front Cell Infect Microbiol 2021; 11:708834. [PMID: 34395314 PMCID: PMC8355824 DOI: 10.3389/fcimb.2021.708834] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 07/14/2021] [Indexed: 12/26/2022] Open
Abstract
The calcium ion (Ca2+) is a ubiquitous second messenger involved in key biological processes in prokaryotes and eukaryotes. In Plasmodium species, Ca2+ signaling plays a central role in the parasite life cycle. It has been associated with parasite development, fertilization, locomotion, and host cell infection. Despite the lack of a canonical inositol-1,4,5-triphosphate receptor gene in the Plasmodium genome, pharmacological evidence indicates that inositol-1,4,5-triphosphate triggers Ca2+ mobilization from the endoplasmic reticulum. Other structures such as acidocalcisomes, food vacuole and mitochondria are proposed to act as supplementary intracellular Ca2+ reservoirs. Several Ca2+-binding proteins (CaBPs) trigger downstream signaling. Other proteins with no EF-hand motifs, but apparently involved with CaBPs, are depicted as playing an important role in the erythrocyte invasion and egress. It is also proposed that a cross-talk among kinases, which are not members of the family of Ca2+-dependent protein kinases, such as protein kinases G, A and B, play additional roles mediated indirectly by Ca2+ regulation. This statement may be extended for proteins directly related to invasion or egress, such as SUB1, ERC, IMC1I, IMC1g, GAP45 and EBA175. In this review, we update our understanding of aspects of Ca2+-mediated signaling correlated to the developmental stages of the malaria parasite life cycle.
Collapse
Affiliation(s)
- Lucas Silva de Oliveira
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
- UMR 7245 MCAM, Molécules de Communication et Adaptation des Micro-organismes, Muséum National d’Histoire Naturelle, CNRS, Équipe Parasites et Protistes Libres, Paris, France
| | - Marcos Rodrigo Alborghetti
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
- Brazilian Biosciences National Laboratory, Brazilian Center for Research in Energy and Materials, Campinas, Brazil
| | - Renata Garcia Carneiro
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | - Izabela Marques Dourado Bastos
- Laboratory of Host-Pathogen Interaction, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
| | - Rogerio Amino
- Unité Infection et Immunité Paludéennes, Institut Pasteur, Paris, France
| | - Philippe Grellier
- UMR 7245 MCAM, Molécules de Communication et Adaptation des Micro-organismes, Muséum National d’Histoire Naturelle, CNRS, Équipe Parasites et Protistes Libres, Paris, France
| | - Sébastien Charneau
- Laboratory of Biochemistry and Protein Chemistry, Department of Cell Biology, Institute of Biology, University of Brasilia, Brasilia, Brazil
| |
Collapse
|
5
|
Pereira PHS, Borges-Pereira L, Garcia CRS. Evidences of G Coupled-Protein Receptor (GPCR) Signaling in the human Malaria Parasite Plasmodium falciparum for Sensing its Microenvironment and the Role of Purinergic Signaling in Malaria Parasites. Curr Top Med Chem 2021; 21:171-180. [PMID: 32851963 DOI: 10.2174/1568026620666200826122716] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/15/2020] [Accepted: 07/20/2020] [Indexed: 11/22/2022]
Abstract
The nucleotides were discovered in the early 19th century and a few years later, the role of such molecules in energy metabolism and cell survival was postulated. In 1972, a pioneer work by Burnstock and colleagues suggested that ATP could also work as a neurotransmitter, which was known as the "purinergic hypothesis". The idea of ATP working as a signaling molecule faced initial resistance until the discovery of the receptors for ATP and other nucleotides, called purinergic receptors. Among the purinergic receptors, the P2Y family is of great importance because it comprises of G proteincoupled receptors (GPCRs). GPCRs are widespread among different organisms. These receptors work in the cells' ability to sense the external environment, which involves: to sense a dangerous situation or detect a pheromone through smell; the taste of food that should not be eaten; response to hormones that alter metabolism according to the body's need; or even transform light into an electrical stimulus to generate vision. Advances in understanding the mechanism of action of GPCRs shed light on increasingly promising treatments for diseases that have hitherto remained incurable, or the possibility of abolishing side effects from therapies widely used today.
Collapse
Affiliation(s)
- Pedro H S Pereira
- Department of Clinical and Toxicological Analyses, University of Sao Paulo, Sao Paulo, Brazil
| | - Lucas Borges-Pereira
- Department of Clinical and Toxicological Analyses, University of Sao Paulo, Sao Paulo, Brazil
| | - Célia R S Garcia
- Department of Clinical and Toxicological Analyses, University of Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
6
|
Guerra AJ, Zhang O, Bahr CME, Huynh MH, DelProposto J, Brown WC, Wawrzak Z, Koropatkin NM, Carruthers VB. Structural basis of Toxoplasma gondii perforin-like protein 1 membrane interaction and activity during egress. PLoS Pathog 2018; 14:e1007476. [PMID: 30513119 PMCID: PMC6294395 DOI: 10.1371/journal.ppat.1007476] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2018] [Revised: 12/14/2018] [Accepted: 11/19/2018] [Indexed: 11/19/2022] Open
Abstract
Intracellular pathogens must egress from the host cell to continue their infectious cycle. Apicomplexans are a phylum of intracellular protozoans that have evolved members of the membrane attack complex and perforin (MACPF) family of pore forming proteins to disrupt cellular membranes for traversing cells during tissue migration or egress from a replicative vacuole following intracellular reproduction. Previous work showed that the apicomplexan Toxoplasma gondii secretes a perforin-like protein (TgPLP1) that contains a C-terminal Domain (CTD) which is necessary for efficient parasite egress. However, the structural basis for CTD membrane binding and egress competency remained unknown. Here, we present evidence that TgPLP1 CTD prefers binding lipids that are abundant in the inner leaflet of the lipid bilayer. Additionally, solving the high-resolution crystal structure of the TgPLP1 APCβ domain within the CTD reveals an unusual double-layered β-prism fold that resembles only one other protein of known structure. Three direct repeat sequences comprise subdomains, with each constituting a wall of the β-prism fold. One subdomain features a protruding hydrophobic loop with an exposed tryptophan at its tip. Spectrophotometric measurements of intrinsic tryptophan fluorescence are consistent with insertion of the hydrophobic loop into a target membrane. Using CRISPR/Cas9 gene editing we show that parasite strains bearing mutations in the hydrophobic loop, including alanine substitution of the tip tryptophan, are equally deficient in egress as a strain lacking TgPLP1 altogether. Taken together our findings suggest a crucial role for the hydrophobic loop in anchoring TgPLP1 to the membrane to support its cytolytic activity and egress function. The intracellular parasite Toxoplasma gondii infects many hosts including humans. Infected people with a weak immune system can suffer severe disease when the parasite replicates uncontrolled via repeated cycles of cell invasion, intracellular growth, and exit, resulting in cell death. Previous studies showed that T. gondii encodes a pore-forming protein, TgPLP1, which contains an unusual domain that is crucial for efficient exit from both the parasite containing vacuole and the host cell. However, how TgPLP1 recognizes and binds to the appropriate membrane is unclear. Here we use a combination of biochemistry, structural biology, and parasitology to identify a preference of TgPLP1 for specific lipids and show that a loop within the structure of the membrane-binding domain inserts into the target membrane and is necessary for exit from the parasite containing vacuole. Our study sheds light into the determinants of membrane binding in TgPLP1 and may inform the overall mechanism of pore formation in similar systems.
Collapse
Affiliation(s)
- Alfredo J. Guerra
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, United States of America
- * E-mail:
| | - Ou Zhang
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, United States of America
| | - Constance M. E. Bahr
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, United States of America
| | - My-Hang Huynh
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, United States of America
| | - James DelProposto
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States of America
| | - William C. Brown
- Life Sciences Institute, University of Michigan, Ann Arbor, MI, United States of America
| | - Zdzislaw Wawrzak
- Northwestern Synchrotron Research Center–LS-CAT, Northwestern University, Argonne, IL, United States of America
| | - Nicole M. Koropatkin
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, United States of America
| | - Vern B. Carruthers
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, MI, United States of America
| |
Collapse
|
7
|
Costa DM, Sá M, Teixeira AR, Loureiro I, Thouvenot C, Golba S, Amino R, Tavares J. TRSP is dispensable for the Plasmodium pre-erythrocytic phase. Sci Rep 2018; 8:15101. [PMID: 30305687 PMCID: PMC6180128 DOI: 10.1038/s41598-018-33398-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Accepted: 09/26/2018] [Indexed: 11/09/2022] Open
Abstract
Plasmodium sporozoites deposited in the skin following a mosquito bite must migrate and invade blood vessels to complete their development in the liver. Once in the bloodstream, sporozoites arrest in the liver sinusoids, but the molecular determinants that mediate this specific homing are not yet genetically defined. Here we investigate the involvement of the thrombospondin-related sporozoite protein (TRSP) in this process using knockout Plasmodium berghei parasites and in vivo bioluminescence imaging in mice. Resorting to a homing assay, trsp knockout sporozoites were found to arrest in the liver similar to control parasites. Moreover, we found no defects in the establishment of infection in mice following inoculation of trsp knockout sporozoites via intravenous and cutaneous injection or mosquito bite. Accordingly, mutant sporozoites were also able to successfully invade hepatocytes in vitro. Altogether, these results suggest TRSP may have a redundant role in the completion of the pre-erythrocytic phase of the malaria parasite. Nonetheless, identifying molecules with paramount roles in this phase could aid in the search for new antigens needed for the design of a protective vaccine against malaria.
Collapse
Affiliation(s)
- David Mendes Costa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, 4200-135, Portugal
| | - Mónica Sá
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, 4200-135, Portugal
| | - Ana Rafaela Teixeira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, 4200-135, Portugal
| | - Inês Loureiro
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal.,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, 4200-135, Portugal
| | - Catherine Thouvenot
- Center for Production and Infection of Anopheles, Institut Pasteur, Paris, 75015, France.,Ultrapole, Institut Pasteur, Paris, 75015, France
| | - Sylvain Golba
- Center for Production and Infection of Anopheles, Institut Pasteur, Paris, 75015, France
| | - Rogerio Amino
- Unit of Malaria Infection and Immunity, Institut Pasteur, Paris, 75015, France.
| | - Joana Tavares
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, 4200-135, Portugal. .,IBMC - Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, 4200-135, Portugal.
| |
Collapse
|
8
|
Darvill N, Dubois DJ, Rouse SL, Hammoudi PM, Blake T, Benjamin S, Liu B, Soldati-Favre D, Matthews S. Structural Basis of Phosphatidic Acid Sensing by APH in Apicomplexan Parasites. Structure 2018; 26:1059-1071.e6. [PMID: 29910186 PMCID: PMC6084407 DOI: 10.1016/j.str.2018.05.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2018] [Revised: 03/26/2018] [Accepted: 05/08/2018] [Indexed: 10/29/2022]
Abstract
Plasmodium falciparum and Toxoplasma gondii are obligate intracellular parasites that belong to the phylum of Apicomplexa and cause major human diseases. Their access to an intracellular lifestyle is reliant on the coordinated release of proteins from the specialized apical organelles called micronemes and rhoptries. A specific phosphatidic acid effector, the acylated pleckstrin homology domain-containing protein (APH) plays a central role in microneme exocytosis and thus is essential for motility, cell entry, and egress. TgAPH is acylated on the surface of the micronemes and recruited to phosphatidic acid (PA)-enriched membranes. Here, we dissect the atomic details of APH PA-sensing hub and its functional interaction with phospholipid membranes. We unravel the key determinant of PA recognition for the first time and show that APH inserts into and clusters multiple phosphate head-groups at the bilayer binding surface.
Collapse
Affiliation(s)
- Nick Darvill
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London SW7 2AZ, UK
| | - David J Dubois
- Department of Microbiology & Molecular Medicine, Faculty of Medicine, University of Geneva, 1 Rue Michel-Servet, 1211 Geneva, Switzerland
| | - Sarah L Rouse
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London SW7 2AZ, UK
| | - Pierre-Mehdi Hammoudi
- Department of Microbiology & Molecular Medicine, Faculty of Medicine, University of Geneva, 1 Rue Michel-Servet, 1211 Geneva, Switzerland
| | - Tom Blake
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London SW7 2AZ, UK
| | - Stefi Benjamin
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London SW7 2AZ, UK
| | - Bing Liu
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London SW7 2AZ, UK; BioBank, First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Dominique Soldati-Favre
- Department of Microbiology & Molecular Medicine, Faculty of Medicine, University of Geneva, 1 Rue Michel-Servet, 1211 Geneva, Switzerland.
| | - Steve Matthews
- Department of Life Sciences, Faculty of Natural Sciences, Imperial College London, London SW7 2AZ, UK; BioBank, First Affiliated Hospital, School of Medicine, Xi'an Jiaotong University, Xi'an, 710049, P. R. China.
| |
Collapse
|
9
|
Targeted Phenotypic Screening in Plasmodium falciparum and Toxoplasma gondii Reveals Novel Modes of Action of Medicines for Malaria Venture Malaria Box Molecules. mSphere 2018; 3:mSphere00534-17. [PMID: 29359192 PMCID: PMC5770543 DOI: 10.1128/msphere.00534-17] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Accepted: 12/20/2017] [Indexed: 01/23/2023] Open
Abstract
The Malaria Box collection includes 400 chemically diverse small molecules with documented potency against malaria parasite growth, but the underlying modes of action are largely unknown. Using complementary phenotypic screens against Plasmodium falciparum and Toxoplasma gondii, we report phenotype-specific hits based on inhibition of overall parasite growth, apicoplast segregation, and egress or host invasion, providing hitherto unavailable insights into the possible mechanisms affected. First, the Malaria Box library was screened against tachyzoite stage T. gondii and the half-maximal effective concentrations (EC50s) of molecules showing ≥80% growth inhibition at 10 µM were determined. Comparison of the EC50s for T. gondii and P. falciparum identified a subset of 24 molecules with nanomolar potency against both parasites. Thirty molecules that failed to induce acute growth inhibition in T. gondii tachyzoites in a 2-day assay caused delayed parasite death upon extended exposure, with at least three molecules interfering with apicoplast segregation during daughter cell formation. Using flow cytometry and microscopy-based examinations, we prioritized 26 molecules with the potential to inhibit host cell egress/invasion during asexual developmental stages of P. falciparum. None of the inhibitors affected digestive vacuole integrity, ruling out a mechanism mediated by broadly specific protease inhibitor activity. Interestingly, five of the plasmodial egress inhibitors inhibited ionophore-induced egress of T. gondii tachyzoites. These findings highlight the advantage of comparative and targeted phenotypic screens in related species as a means to identify lead molecules with a conserved mode of action. Further work on target identification and mechanism analysis will facilitate the development of antiparasitic compounds with cross-species efficacy. IMPORTANCE The phylum Apicomplexa includes many human and animal pathogens, such as Plasmodium falciparum (human malaria) and Toxoplasma gondii (human and animal toxoplasmosis). Widespread resistance to current antimalarials and the lack of a commercial vaccine necessitate novel pharmacological interventions with distinct modes of action against malaria. For toxoplasmosis, new drugs to effectively eliminate tissue-dwelling latent cysts of the parasite are needed. The Malaria Box antimalarial collection, managed and distributed by the Medicines for Malaria Venture, includes molecules of novel chemical classes with proven antimalarial efficacy. Using targeted phenotypic assays of P. falciparum and T. gondii, we have identified a subset of the Malaria Box molecules as potent inhibitors of plastid segregation and parasite invasion and egress, thereby providing early insights into their probable mode of action. Five molecules that inhibit the egress of both parasites have been identified for further mechanistic studies. Thus, the approach we have used to identify novel molecules with defined modes of action in multiple parasites can expedite the development of pan-active antiparasitic agents.
Collapse
|
10
|
Singh S, Chitnis CE. Molecular Signaling Involved in Entry and Exit of Malaria Parasites from Host Erythrocytes. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a026815. [PMID: 28507195 DOI: 10.1101/cshperspect.a026815] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
During the blood stage, Plasmodium spp. merozoites invade host red blood cells (RBCs), multiply, exit, and reinvade uninfected RBCs in a continuing cycle that is responsible for all the clinical symptoms associated with malaria. Entry into (invasion) and exit from (egress) RBCs are highly regulated processes that are mediated by an array of parasite proteins with specific functional roles. Many of these parasite proteins are stored in specialized apical secretory vesicles, and their timely release is critical for successful invasion and egress. For example, the discharge of parasite protein ligands to the apical surface of merozoites is required for interaction with host receptors to mediate invasion, and the timely discharge of proteases and pore-forming proteins helps in permeabilization and dismantling of limiting membranes during egress. This review focuses on our understanding of the signaling mechanisms that regulate apical organelle secretion during host cell invasion and egress by malaria parasites. The review also explores how understanding key signaling mechanisms in the parasite can open opportunities to develop novel strategies to target Plasmodium parasites and eliminate malaria.
Collapse
Affiliation(s)
- Shailja Singh
- Department of Parasites and Insect Vectors, Institut Pasteur, 75015 Paris, France.,Shiv Nadar University, Gautam Buddha Nagar, Uttar Pradesh 201314, India
| | - Chetan E Chitnis
- Department of Parasites and Insect Vectors, Institut Pasteur, 75015 Paris, France.,Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi 110067, India
| |
Collapse
|
11
|
Santos JM, Josling G, Ross P, Joshi P, Orchard L, Campbell T, Schieler A, Cristea IM, Llinás M. Red Blood Cell Invasion by the Malaria Parasite Is Coordinated by the PfAP2-I Transcription Factor. Cell Host Microbe 2017; 21:731-741.e10. [PMID: 28618269 PMCID: PMC5855115 DOI: 10.1016/j.chom.2017.05.006] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 02/16/2017] [Accepted: 05/23/2017] [Indexed: 10/19/2022]
Abstract
Obligate intracellular parasites must efficiently invade host cells in order to mature and be transmitted. For the malaria parasite Plasmodium falciparum, invasion of host red blood cells (RBCs) is essential. Here we describe a parasite-specific transcription factor PfAP2-I, belonging to the Apicomplexan AP2 (ApiAP2) family, that is responsible for regulating the expression of genes involved in RBC invasion. Our genome-wide analysis by ChIP-seq shows that PfAP2-I interacts with a specific DNA motif in the promoters of target genes. Although PfAP2-I contains three AP2 DNA-binding domains, only one is required for binding of the target genes during blood stage development. Furthermore, we find that PfAP2-I associates with several chromatin-associated proteins, including the Plasmodium bromodomain protein PfBDP1 and that complex formation is associated with transcriptional regulation. As a key regulator of red blood cell invasion, PfAP2-I represents a potential new antimalarial therapeutic target.
Collapse
Affiliation(s)
- Joana Mendonca Santos
- Department of Biochemistry and Molecular Biology and Huck Center for Malaria Research, Pennsylvania State University, State College, PA 16802, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Gabrielle Josling
- Department of Biochemistry and Molecular Biology and Huck Center for Malaria Research, Pennsylvania State University, State College, PA 16802, USA
| | - Philipp Ross
- Department of Biochemistry and Molecular Biology and Huck Center for Malaria Research, Pennsylvania State University, State College, PA 16802, USA
| | - Preeti Joshi
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Lindsey Orchard
- Department of Biochemistry and Molecular Biology and Huck Center for Malaria Research, Pennsylvania State University, State College, PA 16802, USA
| | - Tracey Campbell
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Ariel Schieler
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA
| | - Ileana M Cristea
- Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA
| | - Manuel Llinás
- Department of Biochemistry and Molecular Biology and Huck Center for Malaria Research, Pennsylvania State University, State College, PA 16802, USA; Department of Molecular Biology, Princeton University, Princeton, NJ 08544, USA; Lewis-Sigler Institute for Integrative Genomics, Princeton University, Princeton, NJ 08544, USA; Department of Chemistry and Huck Center for Infectious Disease Dynamics, Pennsylvania State University, State College, PA 16802, USA.
| |
Collapse
|
12
|
Gregarine infection accelerates larval development of the cat fleaCtenocephalides felis(Bouché). Parasitology 2017; 144:419-425. [DOI: 10.1017/s0031182016002122] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
SUMMARYA high degree of specialization between host and parasite is a well-known outcome of a long history of coevolution, and it is strikingly illustrated in a coordination of their life cycles. In some cases, the arms race ensued at the establishment of a symbiotic relationship results in the adoption of manipulative strategies by the parasite. We have already learned thatSteinina ctenocephali, a gregarine living in the alimentary canal of cat flea,Ctenocephalides felisfollows its phenology and metamorphosis. Despite these findings the outcome of their symbiotic partnership (mutualist, parasitic or commensal) remains unclear. To address this important question, we measured life history parameters of the flea in the presence of varying infection intensities of gregarine oocysts in laboratory conditions. We found that neither the emergence nor survival rate of fleas was affected by harbouring the gregarines. More surprisingly, our results show that flea larvae infected with gregarines developed faster and emerged earlier than the control group. This gregarine therefore joins the selected group of protists that can modify physiological host traits and provides not only new model taxa to be explored in an evolutionary scenario, but also potential development of control strategies of cat flea.
Collapse
|
13
|
Phosphatidic Acid-Mediated Signaling Regulates Microneme Secretion in Toxoplasma. Cell Host Microbe 2016; 19:349-60. [PMID: 26962945 DOI: 10.1016/j.chom.2016.02.006] [Citation(s) in RCA: 108] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2015] [Revised: 09/28/2015] [Accepted: 02/17/2016] [Indexed: 11/24/2022]
Abstract
The obligate intracellular lifestyle of apicomplexan parasites necessitates an invasive phase underpinned by timely and spatially controlled secretion of apical organelles termed micronemes. In Toxoplasma gondii, extracellular potassium levels and other stimuli trigger a signaling cascade culminating in phosphoinositide-phospholipase C (PLC) activation, which generates the second messengers diacylglycerol (DAG) and IP3 and ultimately results in microneme secretion. Here we show that a delicate balance between DAG and its downstream product, phosphatidic acid (PA), is essential for controlling microneme release. Governing this balance is the apicomplexan-specific DAG-kinase-1, which interconverts PA and DAG, and whose depletion impairs egress and causes parasite death. Additionally, we identify an acylated pleckstrin-homology (PH) domain-containing protein (APH) on the microneme surface that senses PA during microneme secretion and is necessary for microneme exocytosis. As APH is conserved in Apicomplexa, these findings highlight a potentially widely used mechanism in which key lipid mediators regulate microneme exocytosis.
Collapse
|
14
|
Aniweh Y, Gao X, Gunalan K, Preiser PR. PfRH2b specific monoclonal antibodies inhibit merozoite invasion. Mol Microbiol 2016; 102:386-404. [DOI: 10.1111/mmi.13468] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/18/2016] [Indexed: 11/28/2022]
Affiliation(s)
- Yaw Aniweh
- Division of Molecular Genetics and Cell biology, School of Biological Sciences; Nanyang Technological University; 637551 Singapore
| | - Xiaohong Gao
- Division of Molecular Genetics and Cell biology, School of Biological Sciences; Nanyang Technological University; 637551 Singapore
| | - Karthigayan Gunalan
- Division of Molecular Genetics and Cell biology, School of Biological Sciences; Nanyang Technological University; 637551 Singapore
| | - Peter R. Preiser
- Division of Molecular Genetics and Cell biology, School of Biological Sciences; Nanyang Technological University; 637551 Singapore
| |
Collapse
|
15
|
Ruiz A, Pérez D, Muñoz MC, Molina JM, Taubert A, Jacobs-Lorena M, Vega-Rodríguez J, López AM, Hermosilla C. Targeting essential Eimeria ninakohlyakimovae sporozoite ligands for caprine host endothelial cell invasion with a phage display peptide library. Parasitol Res 2015; 114:4327-31. [PMID: 26341796 DOI: 10.1007/s00436-015-4666-x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2015] [Accepted: 08/06/2015] [Indexed: 01/14/2023]
Abstract
Eimeria ninakohlyakimovae is an important coccidian parasite of goats which causes severe diarrhoea in young animals. Specific molecules that mediate E. ninakohlyakimovae host interactions and molecular mechanisms involved in the pathogenesis are still unknown. Although strong circumstantial evidence indicates that E. ninakohlyakimovae sporozoite interactions with caprine endothelial host cells (ECs) are specific, hardly any information is available about the interacting molecules that confer host cell specificity. In this study, we describe a novel method to identify surface proteins of caprine umbilical vein endothelial cells (CUVEC) using a phage display library. After several panning rounds, we identified a number of peptides that specifically bind to the surface of CUVEC. Importantly, caprine endothelial cell peptide 2 (PCEC2) and PCEC5 selectively reduced the infection rate by E. ninakohlyakimovae sporozoites. These preliminary data give new insight for the molecular identification of ligands involved in the interaction between E. ninakohlyakimovae sporozoites and host ECs. Further studies using this phage approach might be useful to identify new potential target molecules for the development of anti-coccidial drugs or even new vaccine strategies.
Collapse
Affiliation(s)
- A Ruiz
- Parasitology Unit, Department of Animal Pathology, Faculty of Veterinary Medicine, University of Las Palmas de Gran Canaria, 35416, Arucas, Las Palmas, Spain.
| | - D Pérez
- Parasitology Unit, Department of Animal Pathology, Faculty of Veterinary Medicine, University of Las Palmas de Gran Canaria, 35416, Arucas, Las Palmas, Spain
| | - M C Muñoz
- Parasitology Unit, Department of Animal Pathology, Faculty of Veterinary Medicine, University of Las Palmas de Gran Canaria, 35416, Arucas, Las Palmas, Spain
| | - J M Molina
- Parasitology Unit, Department of Animal Pathology, Faculty of Veterinary Medicine, University of Las Palmas de Gran Canaria, 35416, Arucas, Las Palmas, Spain
| | - A Taubert
- Institute of Parasitology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Giessen, Germany
| | - M Jacobs-Lorena
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - J Vega-Rodríguez
- The W. Harry Feinstone Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, MD, USA
| | - A M López
- Parasitology Unit, Department of Animal Pathology, Faculty of Veterinary Medicine, University of Las Palmas de Gran Canaria, 35416, Arucas, Las Palmas, Spain
| | - C Hermosilla
- Institute of Parasitology, Faculty of Veterinary Medicine, Justus Liebig University Giessen, Giessen, Germany
| |
Collapse
|
16
|
Garg S, Agarwal S, Dabral S, Kumar N, Sehrawat S, Singh S. Visualization and quantification of Plasmodium falciparum intraerythrocytic merozoites. SYSTEMS AND SYNTHETIC BIOLOGY 2015; 9:23-6. [PMID: 26702305 DOI: 10.1007/s11693-015-9167-9] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2014] [Accepted: 03/10/2015] [Indexed: 11/25/2022]
Abstract
Malaria, a leading parasitic killer, is caused by Plasmodium spp. The pathology of the disease starts when Plasmodium merozoites infect erythrocytes to form rings, that matures through a large trophozoite form and develop into schizonts containing multiple merozoites. The number of intra-erythrocytic merozoites is a key-determining factor for multiplication rate of the parasite. Counting of intraerythrocytic merozoites by classical 2-D microscopy method is error prone due to insufficient representation of merozoite in one optical plane of a schizont. Here, we report an alternative 3-D microscopy based automated method for counting of intraerythrocytic merozoites in entire volume of schizont. This method offers a considerable amount of advantages in terms of both, ease and accuracy.
Collapse
Affiliation(s)
- Swati Garg
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, 110067 India
| | - Shalini Agarwal
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, 110067 India
| | - Surbhi Dabral
- Malaria Group, International Centre for Genetic Engineering and Biotechnology (ICGEB), New Delhi, 110067 India
| | - Naveen Kumar
- Department of Life Sciences, Shiv Nadar University, Gautam Budha Nagar, 203207 UP India
| | - Seema Sehrawat
- Department of Life Sciences, Shiv Nadar University, Gautam Budha Nagar, 203207 UP India
| | - Shailja Singh
- Department of Life Sciences, Shiv Nadar University, Gautam Budha Nagar, 203207 UP India
| |
Collapse
|
17
|
Lourido S, Moreno SNJ. The calcium signaling toolkit of the Apicomplexan parasites Toxoplasma gondii and Plasmodium spp. Cell Calcium 2014; 57:186-93. [PMID: 25605521 DOI: 10.1016/j.ceca.2014.12.010] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2014] [Accepted: 12/15/2014] [Indexed: 12/21/2022]
Abstract
Apicomplexan parasites have complex life cycles, frequently split between different hosts and reliant on rapid responses as the parasites react to changing environmental conditions. Calcium ion (Ca(2+)) signaling is consequently essential for the cellular and developmental changes that support Apicomplexan parasitism. Apicomplexan genomes reveal a rich repertoire of genes involved in calcium signaling, although many of the genes responsible for observed physiological changes remain unknown. There is evidence, for example, for the presence of a nifedipine-sensitive calcium entry mechanism in Toxoplasma, but the molecular components involved in Ca(2+) entry in both Toxoplasma and Plasmodium, have not been identified. The major calcium stores are the endoplasmic reticulum (ER), the acidocalcisomes, and the plant-like vacuole in Toxoplasma, or the food vacuole in Plasmodium spp. Pharmacological evidence suggests that Ca(2+) release from intracellular stores may be mediated by inositol 1,4,5-trisphosphate (IP3) or cyclic ADP ribose (cADPR) although there is no molecular evidence for the presence of receptors for these second messengers in the parasites. Several Ca(2+)-ATPases are present in Apicomplexans and a putative mitochondrial Ca(2+)/H(+) exchanger has been identified. Apicomplexan genomes contain numerous genes encoding Ca(2+)-binding proteins, with the notable expansion of calcium-dependent protein kinases (CDPKs), whose study has revealed roles in gliding motility, microneme secretion, host cell invasion and egress, and parasite differentiation. Microneme secretion has also been shown to depend on the C2 domain containing protein DOC2 in both Plasmodium spp. and Toxoplasma, providing further evidence for the complex transduction of Ca(2+) signals in these organisms. The characterization of these pathways could lead to the discovery of novel drug targets and to a better understanding of the role of Ca(2+) in these parasites.
Collapse
Affiliation(s)
- Sebastian Lourido
- Whitehead Institute for Biomedical Research, Cambridge, MA 02142, USA
| | - Silvia N J Moreno
- Center for Tropical and Emerging Global Diseases, Department of Cellular Biology, University of Georgia, Athens, GA 30602, USA.
| |
Collapse
|
18
|
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
|
19
|
Tavares J, Amino R, Ménard R. The role of MACPF proteins in the biology of malaria and other apicomplexan parasites. Subcell Biochem 2014; 80:241-253. [PMID: 24798015 DOI: 10.1007/978-94-017-8881-6_12] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/03/2023]
Abstract
Apicomplexans are eukaryotic parasites of major medical and veterinary importance. They have complex life cycles through frequently more than one host, interact with many cell types in their hosts, and can breach host cell membranes during parasite traversal of, or egress from, host cells. Some of these parasites make a strikingly heavy use of the pore-forming MACPF domain, and encode up to 10 different MACPF domain-containing proteins. In this chapter, we focus on the two most studied and medically important apicomplexans, Plasmodium and Toxoplasma, and describe the known functions of their MACPF polypeptide arsenal. Apicomplexan MACPF proteins appear to be involved in a variety of membrane-damaging events, making them an attractive model to dissect the structure-function relationships of the MACPF domain.
Collapse
Affiliation(s)
- Joana Tavares
- Institut Pasteur, Unité de Biologie et Génétique du Paludisme, Département de Parasitologie et Mycologie, 28 rue du Docteur Roux, 75015, Paris, France
| | | | | |
Collapse
|
20
|
Pollo-Oliveira L, Post H, Acencio ML, Lemke N, van den Toorn H, Tragante V, Heck AJR, Altelaar AFM, Yatsuda AP. Unravelling the Neospora caninum secretome through the secreted fraction (ESA) and quantification of the discharged tachyzoite using high-resolution mass spectrometry-based proteomics. Parasit Vectors 2013; 6:335. [PMID: 24267406 PMCID: PMC4182915 DOI: 10.1186/1756-3305-6-335] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2013] [Accepted: 11/15/2013] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The apicomplexan parasite Neospora caninum causes neosporosis, a disease that leads to abortion or stillbirth in cattle, generating an economic impact on the dairy and beef cattle trade. As an obligatory intracellular parasite, N. caninum needs to invade the host cell in an active manner to survive. The increase in parasite cytosolic Ca2+ upon contact with the host cell mediates critical events, including the exocytosis of phylum-specific secretory organelles and the activation of the parasite invasion motor. Because invasion is considered a requirement for pathogen survival and replication within the host, the identification of secreted proteins (secretome) involved in invasion may be useful to reveal interesting targets for therapeutic intervention. METHODS To chart the currently missing N. caninum secretome, we employed mass spectrometry-based proteomics to identify proteins present in the N. caninum tachyzoite using two different approaches. The first approach was identifying the proteins present in the tachyzoite-secreted fraction (ESA). The second approach was determining the relative quantification through peptide stable isotope labelling of the tachyzoites submitted to an ethanol secretion stimulus (discharged tachyzoite), expecting to identify the secreted proteins among the down-regulated group. RESULTS As a result, 615 proteins were identified at ESA and 2,011 proteins quantified at the discharged tachyzoite. We have analysed the connection between the secreted and the down-regulated proteins and searched for putative regulators of the secretion process among the up-regulated proteins. An interaction network was built by computational prediction involving the up- and down-regulated proteins. The mass spectrometry proteomics data have been deposited to the ProteomeXchange with identifier PXD000424. CONCLUSIONS The comparison between the protein abundances in ESA and their measure in the discharged tachyzoite allowed for a more precise identification of the most likely secreted proteins. Information from the network interaction and up-regulated proteins was important to recognise key proteins potentially involved in the metabolic regulation of secretion. Our results may be helpful to guide the selection of targets to be investigated against Neospora caninum and other Apicomplexan organisms.
Collapse
Affiliation(s)
- Letícia Pollo-Oliveira
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto e Núcleo de Apoio à Pesquisa em Produtos Naturais e Sintéticos (NPPNS), Universidade de São Paulo, Av do Café , s/n, Ribeirão Preto, SP 14040-903, Brazil
| | - Harm Post
- Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Centre for Biomolecular Research, Utrecht University, Padualaan 8, Utrecht 3884 CH, The Netherlands
- Netherlands Proteomics Centre, Padualaan 8, Utrecht 3884 CH, The Netherlands
| | - Marcio Luis Acencio
- Botucatu Institute of Biosciences, UNESP - Univ Estadual Paulista, Distrito de Rubião Jr, s/n, Botucatu, São Paulo 18918-970, Brazil
| | - Ney Lemke
- Botucatu Institute of Biosciences, UNESP - Univ Estadual Paulista, Distrito de Rubião Jr, s/n, Botucatu, São Paulo 18918-970, Brazil
| | - Henk van den Toorn
- Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Centre for Biomolecular Research, Utrecht University, Padualaan 8, Utrecht 3884 CH, The Netherlands
- Netherlands Proteomics Centre, Padualaan 8, Utrecht 3884 CH, The Netherlands
| | - Vinicius Tragante
- Division of Heart and Lungs, Department of Cardiology, University Medical Center Utrecht, Utrecht, The Netherlands
- Division of Biomedical Genetics, Department of Medical Genetics, University Medical Center Utrecht, Utrecht, The Netherlands
| | - Albert JR Heck
- Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Centre for Biomolecular Research, Utrecht University, Padualaan 8, Utrecht 3884 CH, The Netherlands
- Netherlands Proteomics Centre, Padualaan 8, Utrecht 3884 CH, The Netherlands
| | - AF Maarten Altelaar
- Biomolecular Mass Spectrometry and Proteomics, Utrecht Institute for Pharmaceutical Sciences and Bijvoet Centre for Biomolecular Research, Utrecht University, Padualaan 8, Utrecht 3884 CH, The Netherlands
- Netherlands Proteomics Centre, Padualaan 8, Utrecht 3884 CH, The Netherlands
| | - Ana Patrícia Yatsuda
- Faculdade de Ciências Farmacêuticas de Ribeirão Preto e Núcleo de Apoio à Pesquisa em Produtos Naturais e Sintéticos (NPPNS), Universidade de São Paulo, Av do Café , s/n, Ribeirão Preto, SP 14040-903, Brazil
| |
Collapse
|
21
|
Roiko MS, Carruthers VB. Functional dissection of Toxoplasma gondii perforin-like protein 1 reveals a dual domain mode of membrane binding for cytolysis and parasite egress. J Biol Chem 2013; 288:8712-8725. [PMID: 23376275 DOI: 10.1074/jbc.m113.450932] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The recently discovered role of a perforin-like protein (PLP1) for rapid host cell egress by the protozoan parasite Toxoplasma gondii expanded the functional diversity of pore-forming proteins. Whereas PLP1 was found to be necessary for rapid egress and pathogenesis, the sufficiency for and mechanism of membrane attack were yet unknown. Here we further dissected the PLP1 knock-out phenotype, the mechanism of PLP1 pore formation, and the role of each domain by genetic complementation. We found that PLP1 is sufficient for membrane disruption and has a conserved mechanism of pore formation through target membrane binding and oligomerization to form large, multimeric membrane-embedded complexes. The highly conserved, central MACPF domain and the β-sheet-rich C-terminal domain were required for activity. Loss of the unique N-terminal extension reduced lytic activity and led to a delay in rapid egress, but did not significantly decrease virulence, suggesting that small amounts of lytic activity are sufficient for pathogenesis. We found that both N- and C-terminal domains have membrane binding activity, with the C-terminal domain being critical for function. This dual mode of membrane association may promote PLP1 activity and parasite egress in the diverse cell types in which this parasite replicates.
Collapse
Affiliation(s)
- Marijo S Roiko
- Cell and Molecular Biology Program, University of Michigan, Ann Arbor, Michigan 48109-5630; Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan 48109-5630
| | - Vern B Carruthers
- Department of Microbiology and Immunology, University of Michigan, Ann Arbor, Michigan 48109-5630.
| |
Collapse
|
22
|
Lindner SE, Swearingen KE, Harupa A, Vaughan AM, Sinnis P, Moritz RL, Kappe SHI. Total and putative surface proteomics of malaria parasite salivary gland sporozoites. Mol Cell Proteomics 2013; 12:1127-43. [PMID: 23325771 DOI: 10.1074/mcp.m112.024505] [Citation(s) in RCA: 141] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Malaria infections of mammals are initiated by the transmission of Plasmodium salivary gland sporozoites during an Anopheles mosquito vector bite. Sporozoites make their way through the skin and eventually to the liver, where they infect hepatocytes. Blocking this initial stage of infection is a promising malaria vaccine strategy. Therefore, comprehensively elucidating the protein composition of sporozoites will be invaluable in identifying novel targets for blocking infection. Previous efforts to identify the proteins expressed in Plasmodium mosquito stages were hampered by the technical difficulty of separating the parasite from its vector; without effective purifications, the large majority of proteins identified were of vector origin. Here we describe the proteomic profiling of highly purified salivary gland sporozoites from two Plasmodium species: human-infective Plasmodium falciparum and rodent-infective Plasmodium yoelii. The combination of improved sample purification and high mass accuracy mass spectrometry has facilitated the most complete proteome coverage to date for a pre-erythrocytic stage of the parasite. A total of 1991 P. falciparum sporozoite proteins and 1876 P. yoelii sporozoite proteins were identified, with >86% identified with high sequence coverage. The proteomic data were used to confirm the presence of components of three features critical for sporozoite infection of the mammalian host: the sporozoite motility and invasion apparatus (glideosome), sporozoite signaling pathways, and the contents of the apical secretory organelles. Furthermore, chemical labeling and identification of proteins on live sporozoites revealed previously uncharacterized complexity of the putative sporozoite surface-exposed proteome. Taken together, the data constitute the most comprehensive analysis to date of the protein expression of salivary gland sporozoites and reveal novel potential surface-exposed proteins that might be valuable targets for antibody blockage of infection.
Collapse
Affiliation(s)
- Scott E Lindner
- Malaria Program, Seattle Biomedical Research Institute, 307 Westlake Avenue North, Suite 500, Seattle, Washington 98109, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Hopp CS, Bowyer PW, Baker DA. The role of cGMP signalling in regulating life cycle progression of Plasmodium. Microbes Infect 2012; 14:831-7. [PMID: 22613210 PMCID: PMC3484397 DOI: 10.1016/j.micinf.2012.04.011] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2012] [Revised: 04/13/2012] [Accepted: 04/17/2012] [Indexed: 11/25/2022]
Abstract
The 3′-5′-cyclic guanosine monophosphate (cGMP)-dependent protein kinase (PKG) is the main mediator of cGMP signalling in the malaria parasite. This article reviews the role of PKG in Plasmodium falciparum during gametogenesis and blood stage schizont rupture, as well as the role of the Plasmodium berghei orthologue in ookinete differentiation and motility, and liver stage schizont development. The current views on potential effector proteins downstream of PKG and the mechanisms that may regulate cyclic nucleotide levels are presented.
Collapse
Affiliation(s)
- Christine S Hopp
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, Keppel Street, London WC1E 7HT, UK
| | | | | |
Collapse
|
24
|
Purinoceptor signaling in malaria-infected erythrocytes. Microbes Infect 2012; 14:779-86. [PMID: 22580091 DOI: 10.1016/j.micinf.2012.04.009] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2012] [Revised: 04/13/2012] [Accepted: 04/13/2012] [Indexed: 01/25/2023]
Abstract
Human erythrocytes are endowed with ATP release pathways and metabotropic and ionotropic purinoceptors. This review summarizes the pivotal function of purinergic signaling in erythrocyte control of vascular tone, in hemolytic septicemia, and in malaria. In malaria, the intraerythrocytic parasite exploits the purinergic signaling of its host to adapt the erythrocyte to its requirements.
Collapse
|
25
|
Generation of second messengers in Plasmodium. Microbes Infect 2012; 14:787-95. [PMID: 22584103 DOI: 10.1016/j.micinf.2012.04.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2011] [Revised: 04/17/2012] [Accepted: 04/18/2012] [Indexed: 02/05/2023]
Abstract
Signalling in malaria parasites is a field of growing interest as its components may prove to be valuable drug targets, especially when one considers the burden of a disease that is responsible for up to 500 million infections annually. The scope of this review is to discuss external stimuli in the parasite life cycle and the upstream machinery responsible for translating them into intracellular responses, focussing particularly on the calcium signalling pathway.
Collapse
|
26
|
Holder AA, Mohd Ridzuan MA, Green JL. Calcium dependent protein kinase 1 and calcium fluxes in the malaria parasite. Microbes Infect 2012; 14:825-30. [PMID: 22584104 DOI: 10.1016/j.micinf.2012.04.006] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2012] [Revised: 04/11/2012] [Accepted: 04/11/2012] [Indexed: 01/20/2023]
Abstract
Calcium dependent protein kinases (CDPKs) are found only in plants and alveolates and are distinguished from other kinases by an activation domain that binds calcium directly. Plants contain families of these kinases and their functions are modulated by post translational modifications as well as calcium activation. Apicomplexan parasites also contain CDPK families and this review is focused on CDPK1 in Plasmodium spp. This enzyme has been implicated in parasite motility and host cell invasion and at least two substrates associated with the actomyosin motor complex have been identified. By analogy with the plant CDPKs we propose that its activity is modulated both by post translational modifications and by its subcellular location in a compartment within the parasite's pellicle, which may regulate the calcium concentration required for activation.
Collapse
Affiliation(s)
- Anthony A Holder
- Division of Parasitology, MRC National Institute for Medical Research, The Ridgeway, Mill Hill, London NW7 1AA, UK.
| | | | | |
Collapse
|