1
|
Vigetti L, Touquet B, Debarre D, Rose T, Bureau L, Abdallah D, Dubacheva GV, Tardieux I. Submicrometre spatiotemporal characterization of the Toxoplasma adhesion strategy for gliding motility. Nat Microbiol 2024:10.1038/s41564-024-01818-3. [PMID: 39496912 DOI: 10.1038/s41564-024-01818-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Accepted: 08/21/2024] [Indexed: 11/06/2024]
Abstract
Toxoplasma gondii is a protozoan apicomplexan parasite that uses an adhesion-dependent mode of motility termed gliding to access host cells and disseminate into tissues. Previous studies on Apicomplexa motile morphotypes, including the T. gondii tachyzoite, have identified a cortical actin-myosin motor system that drives the rearward translocation of transmembrane adhesins, thus powering forward movement. However, this model is currently questioned. Here, combining micropatterning and tunable surface chemistry (to edit parasite surface ligands) with flow force and live or super-resolution imaging, we show that tachyzoites build only one apical anchoring contact with the substrate, over which it slides. Furthermore, we show that glycosaminoglycan-parasite interactions are sufficient to promote such force-productive contact and find that the apicobasal flow is set up independent of adhesin release and surface interactions. These findings should enable further characterization of the molecular functions at the T. gondii-substrate mechanosensitive interface and their comparison across apicomplexans.
Collapse
Affiliation(s)
- Luis Vigetti
- Institute for Advanced Biosciences, Biomechanics of Host-Parasite Cell Interactions Team, CNRS UMR 5309, INSERM U1209, Université Grenoble Alpes, Grenoble, France
| | - Bastien Touquet
- Institute for Advanced Biosciences, Biomechanics of Host-Parasite Cell Interactions Team, CNRS UMR 5309, INSERM U1209, Université Grenoble Alpes, Grenoble, France
| | - Delphine Debarre
- Laboratoire Interdisciplinaire de Physique, CNRS UMR 5588, Université Grenoble Alpes, Grenoble, France
| | - Thierry Rose
- Institut Pasteur, Université Paris Cité, Diagnostic Test Innovation and Development Core Facility, Paris, France
| | - Lionel Bureau
- Laboratoire Interdisciplinaire de Physique, CNRS UMR 5588, Université Grenoble Alpes, Grenoble, France
| | - Dima Abdallah
- Institute for Advanced Biosciences, Biomechanics of Host-Parasite Cell Interactions Team, CNRS UMR 5309, INSERM U1209, Université Grenoble Alpes, Grenoble, France
| | - Galina V Dubacheva
- Département de Chimie Moléculaire, CNRS UMR 5250, Université Grenoble Alpes, Grenoble, France.
| | - Isabelle Tardieux
- Institute for Advanced Biosciences, Biomechanics of Host-Parasite Cell Interactions Team, CNRS UMR 5309, INSERM U1209, Université Grenoble Alpes, Grenoble, France.
| |
Collapse
|
2
|
Sy Thau N, Nguyen TK, Truong NV, Chu TTH, Na SH, Moon RW, Lau YL, Nyunt MH, Park WS, Chun WJ, Lu F, Lee SK, Han JH, Han ET. Characterization of merozoite-specific thrombospondin-related anonymous protein (MTRAP) in Plasmodium vivax and P. knowlesi parasites. Front Cell Infect Microbiol 2024; 14:1354880. [PMID: 38465236 PMCID: PMC10920329 DOI: 10.3389/fcimb.2024.1354880] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 02/08/2024] [Indexed: 03/12/2024] Open
Abstract
Plasmodium vivax, the most widespread human malaria parasite, and P. knowlesi, an emerging Plasmodium that infects humans, are the phylogenetically closest malarial species that infect humans, which may induce cross-species reactivity across most co-endemic areas in Southeast Asia. The thrombospondin-related anonymous protein (TRAP) family is indispensable for motility and host cell invasion in the growth and development of Plasmodium parasites. The merozoite-specific TRAP (MTRAP), expressed in blood-stage merozoites, is supposed to be essential for human erythrocyte invasion. We aimed to characterize MTRAPs in blood-stage P. vivax and P. knowlesi parasites and ascertain their cross-species immunoreactivity. Recombinant P. vivax and P. knowlesi MTRAPs of full-length ectodomains were expressed in a mammalian expression system. The MTRAP-specific immunoglobulin G, obtained from immune animals, was used in an immunofluorescence assay for subcellular localization and invasion inhibitory activity in blood-stage parasites was determined. The cross-species humoral immune responses were analyzed in the sera of patients with P. vivax or P. knowlesi infections. The MTRAPs of P. vivax (PvMTRAP) and P. knowlesi (PkMTRAP) were localized on the rhoptry body of merozoites in blood-stage parasites. Both anti-PvMTRAP and anti-PkMTRAP antibodies inhibited erythrocyte invasion of blood-stage P. knowlesi parasites. The humoral immune response to PvMTRAP showed high immunogenicity, longevity, and cross-species immunoreactivity with P. knowlesi. MTRAPs are promising candidates for development of vaccines and therapeutics against vivax and knowlesi malaria.
Collapse
Affiliation(s)
- Nguyen Sy Thau
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon, Gangwon-do, Republic of Korea
| | - Tuyet-Kha Nguyen
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon, Gangwon-do, Republic of Korea
| | - Nguyen Van Truong
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon, Gangwon-do, Republic of Korea
| | - Thi-Thanh Hang Chu
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon, Gangwon-do, Republic of Korea
| | - Sung-Hun Na
- Department of Obstetrics and Gynecology, Kangwon National University School of Medicine, Chuncheon, Gangwon-d, Republic of Korea
| | - Robert W. Moon
- Department of Infection Biology, Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Yee Ling Lau
- Department of Parasitology, Faculty of Medicine, Universiti Malaya, Kuala Lumpur, Malaysia
| | | | - Won-Sun Park
- Department of Physiology, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Wan-Joo Chun
- Department of Pharmacology, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, Republic of Korea
| | - Feng Lu
- Department of Pathogen Biology and Immunology, School of Medicine, Yangzhou University, Yangzhou, Jiangsu, China
| | - Seong-Kyun Lee
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon, Gangwon-do, Republic of Korea
| | - Jin-Hee Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon, Gangwon-do, Republic of Korea
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon, Gangwon-do, Republic of Korea
| |
Collapse
|
3
|
Bhide AR, Surve DH, Jindal AB. Nanocarrier based active targeting strategies against erythrocytic stage of malaria. J Control Release 2023; 362:297-308. [PMID: 37625598 DOI: 10.1016/j.jconrel.2023.08.043] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2023] [Revised: 08/03/2023] [Accepted: 08/22/2023] [Indexed: 08/27/2023]
Abstract
The Global Technical Strategy for Malaria 2016-2030 aims to achieve a 90% reduction in malaria cases, and strategic planning and execution are crucial for accomplishing this target. This review aims to understand the complex interaction between erythrocytic receptors and parasites and to use this knowledge to actively target the erythrocytic stage of malaria. The review provides insight into the malaria life cycle, which involves various receptors such as glycophorin A, B, C, and D (GPA/B/C/D), complement receptor 1, basigin, semaphorin 7a, Band 3/ GPA, Kx, and heparan sulfate proteoglycan for parasite cellular binding and ingress in the erythrocytic and exo-erythrocytic stages. Synthetic peptides mimicking P. falciparum receptor binding ligands, human serum albumin, chondroitin sulfate, synthetic polymers, and lipids have been utilized as ligands and decorated onto nanocarriers for specific targeting to parasite-infected erythrocytes. The need of the hour for treatment and prophylaxis against malaria is a broadened horizon that includes multiple targeting strategies against the entry, proliferation, and transmission stages of the parasite. Platform technologies with established pre-clinical safety and efficacy should be translated into clinical evaluation and formulation scale-up. Future development should be directed towards nanovaccines as proactive tools against malaria infection.
Collapse
Affiliation(s)
- Atharva R Bhide
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Jhunjhunu, Rajasthan 333031, India
| | - Dhanashree H Surve
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA 01003, United States
| | - Anil B Jindal
- Department of Pharmacy, Birla Institute of Technology and Science Pilani, Pilani Campus, Jhunjhunu, Rajasthan 333031, India.
| |
Collapse
|
4
|
Baba M, Nozaki M, Tachibana M, Tsuboi T, Torii M, Ishino T. Rhoptry neck protein 4 plays important roles during Plasmodium sporozoite infection of the mammalian liver. mSphere 2023; 8:e0058722. [PMID: 37272704 PMCID: PMC10449513 DOI: 10.1128/msphere.00587-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 04/05/2023] [Indexed: 06/06/2023] Open
Abstract
During invasion, Plasmodium parasites secrete proteins from rhoptry and microneme apical end organelles, which have crucial roles in attaching to and invading target cells. A sporozoite stage-specific gene silencing system revealed that rhoptry neck protein 2 (RON2), RON4, and RON5 are important for sporozoite invasion of mosquito salivary glands. Here, we further investigated the roles of RON4 during sporozoite infection of the liver in vivo. Following intravenous inoculation of RON4-knockdown sporozoites into mice, we demonstrated that sporozoite RON4 has multiple functions during sporozoite traversal of sinusoidal cells and infection of hepatocytes. In vitro infection experiments using a hepatoma cell line revealed that secreted RON4 is involved in sporozoite adhesion to hepatocytes and has an important role in the early steps of hepatocyte infection. In addition, in vitro motility assays indicated that RON4 is required for sporozoite attachment to the substrate and the onset of migration. These findings indicate that RON4 is crucial for sporozoite migration toward and invasion of hepatocytes via attachment ability and motility.IMPORTANCEMalarial parasite transmission to mammals is established when sporozoites are inoculated by mosquitoes and migrate through the bloodstream to infect hepatocytes. Many aspects of the molecular mechanisms underpinning migration and cellular invasion remain largely unelucidated. By applying a sporozoite stage-specific gene silencing system in the rodent malarial parasite, Plasmodium berghei, we demonstrated that rhoptry neck protein 4 (RON4) is crucial for sporozoite infection of the liver in vivo. Combined with in vitro investigations, it was revealed that RON4 functions during a crossing of the sinusoidal cell layer and invading hepatocytes, at an early stage of liver infection, by mediating the sporozoite capacity for adhesion and the onset of motility. Since RON4 is also expressed in Plasmodium merozoites and Toxoplasma tachyzoites, our findings contribute to understanding the conserved invasion mechanisms of Apicomplexa parasites.
Collapse
Affiliation(s)
- Minami Baba
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime, Japan
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki University, Sakamoto, Nagasaki, Japan
| | - Mamoru Nozaki
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime, Japan
| | - Mayumi Tachibana
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime, Japan
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Ehime, Japan
| | - Motomi Torii
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime, Japan
| | - Tomoko Ishino
- Division of Molecular Parasitology, Proteo-Science Center, Ehime University, Toon, Ehime, Japan
- Department of Parasitology and Tropical Medicine, Graduate School of Medical and Dental Sciences, Tokyo Medical and Dental University (TMDU), Yushima, Bunkyo-ku, Tokyo, Japan
| |
Collapse
|
5
|
John A, M Bader S, Madiedo Soler N, Wiradiputri K, Tichkule S, Smyth ST, Ralph SA, Jex AR, Scott NE, Tonkin CJ, Goddard-Borger ED. Conservation, abundance, glycosylation profile, and localization of the TSP protein family in Cryptosporidium parvum. J Biol Chem 2023; 299:103006. [PMID: 36775128 PMCID: PMC10034466 DOI: 10.1016/j.jbc.2023.103006] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 02/05/2023] [Accepted: 02/06/2023] [Indexed: 02/12/2023] Open
Abstract
Cryptosporidium parvum is a zoonotic apicomplexan parasite and a common cause of diarrheal disease worldwide. The development of vaccines to prevent or limit infection remains an important goal for tackling cryptosporidiosis. At present, the only approved vaccine against any apicomplexan parasite targets a conserved adhesin possessing a thrombospondin repeat domain. C. parvum possesses 12 orthologous thrombospondin repeat domain-containing proteins known as CpTSP1-12, though little is known about these potentially important antigens. Here, we explore the architecture and conservation of the CpTSP protein family, as well as their abundance at the protein level within the sporozoite stage of the life cycle. We examine the glycosylation states of these proteins using a combination of glycopeptide enrichment techniques to demonstrate that these proteins are modified with C-, O-, and N-linked glycans. Using expansion microscopy, and an antibody against the C-linked mannose that is unique to the CpTSP protein family within C. parvum, we show that these proteins are found both on the cell surface and in structures that resemble the secretory pathway of C. parvum sporozoites. Finally, we generated a polyclonal antibody against CpTSP1 to show that it is found at the cell surface and within micronemes, in a pattern reminiscent of other apicomplexan motility-associated adhesins, and is present both in sporozoites and meronts. This work sheds new light on an understudied family of C. parvum proteins that are likely to be important to both parasite biology and the development of vaccines against cryptosporidiosis.
Collapse
Affiliation(s)
- Alan John
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Stefanie M Bader
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Niccolay Madiedo Soler
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Kharizta Wiradiputri
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Swapnil Tichkule
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Sean T Smyth
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Stuart A Ralph
- Department of Biochemistry and Pharmacology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Victoria, Australia
| | - Aaron R Jex
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Nichollas E Scott
- Department of Microbiology and Immunology, University of Melbourne at the Peter Doherty Institute for Infection and Immunity, Parkville, Victoria, Australia.
| | - Christopher J Tonkin
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia.
| | - Ethan D Goddard-Borger
- The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia; Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
6
|
Ibrahim A, Manko E, Dombrowski JG, Campos M, Benavente ED, Nolder D, Sutherland CJ, Nosten F, Fernandez D, Vélez-Tobón G, Castaño AT, Aguiar ACC, Pereira DB, da Silva Santos S, Suarez-Mutis M, Di Santi SM, Regina de Souza Baptista A, Dantas Machado RL, Marinho CR, Clark TG, Campino S. Population-based genomic study of Plasmodium vivax malaria in seven Brazilian states and across South America. LANCET REGIONAL HEALTH. AMERICAS 2023; 18:100420. [PMID: 36844008 PMCID: PMC9950661 DOI: 10.1016/j.lana.2022.100420] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 12/06/2022] [Accepted: 12/06/2022] [Indexed: 01/03/2023]
Abstract
Background Brazil is a unique and understudied setting for malaria, with complex foci of transmission associated with human and environmental conditions. An understanding of the population genomic diversity of P. vivax parasites across Brazil can support malaria control strategies. Methods Through whole genome sequencing of P. vivax isolates across 7 Brazilian states, we use population genomic approaches to compare genetic diversity within country (n = 123), continent (6 countries, n = 315) and globally (26 countries, n = 885). Findings We confirm that South American isolates are distinct, have more ancestral populations than the other global regions, with differentiating mutations in genes under selective pressure linked to antimalarial drugs (pvmdr1, pvdhfr-ts) and mosquito vectors (pvcrmp3, pvP45/48, pvP47). We demonstrate Brazil as a distinct parasite population, with signals of selection including ABC transporter (PvABCI3) and PHIST exported proteins. Interpretation Brazil has a complex population structure, with evidence of P. simium infections and Amazonian parasites separating into multiple clusters. Overall, our work provides the first Brazil-wide analysis of P. vivax population structure and identifies important mutations, which can inform future research and control measures. Funding AI is funded by an MRC LiD PhD studentship. TGC is funded by the Medical Research Council (Grant no. MR/M01360X/1, MR/N010469/1, MR/R025576/1, MR/R020973/1 and MR/X005895/1). SC is funded by Medical Research Council UK grants (MR/M01360X/1, MR/R025576/1, MR/R020973/1 and MR/X005895/1) and Bloomsbury SET (ref. CCF17-7779). FN is funded by The Shloklo Malaria Research Unit - part of the Mahidol Oxford Research Unit, supported by the Wellcome Trust (Grant no. 220211). ARSB is funded by São Paulo Research Foundation - FAPESP (Grant no. 2002/09546-1). RLDM is funded by Brazilian National Council for Scientific and Technological Development - CNPq (Grant no. 302353/2003-8 and 471605/2011-5); CRFM is funded by FAPESP (Grant no. 2020/06747-4) and CNPq (Grant no. 302917/2019-5 and 408636/2018-1); JGD is funded by FAPESP fellowships (2016/13465-0 and 2019/12068-5) and CNPq (Grant no. 409216/2018-6).
Collapse
Affiliation(s)
- Amy Ibrahim
- Faculty of Infectious & Tropical Diseases, London School of Hygiene
& Tropical Medicine, London, UK
| | - Emilia Manko
- Faculty of Infectious & Tropical Diseases, London School of Hygiene
& Tropical Medicine, London, UK
| | - Jamille G. Dombrowski
- Department of Parasitology, Institute of Biomedical Sciences, University
of São Paulo, São Paulo, Brazil
| | - Mónica Campos
- Faculty of Infectious & Tropical Diseases, London School of Hygiene
& Tropical Medicine, London, UK
| | - Ernest Diez Benavente
- Faculty of Infectious & Tropical Diseases, London School of Hygiene
& Tropical Medicine, London, UK
| | - Debbie Nolder
- Faculty of Infectious & Tropical Diseases, London School of Hygiene
& Tropical Medicine, London, UK
- Public Health England Malaria Reference Laboratory, London School of
Hygiene & Tropical Medicine, London, UK
| | - Colin J. Sutherland
- Faculty of Infectious & Tropical Diseases, London School of Hygiene
& Tropical Medicine, London, UK
- Public Health England Malaria Reference Laboratory, London School of
Hygiene & Tropical Medicine, London, UK
| | - Francois Nosten
- Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research
Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Tak,
Thailand
- Centre for Tropical Medicine and Global Health, Nuffield Department of
Clinical Medicine Research Building, University of Oxford Old Road Campus,
Oxford, UK
| | - Diana Fernandez
- Grupo Malaria, Facultad de Medicina, Universidad de Antioquia, Antioquia,
Colombia
| | - Gabriel Vélez-Tobón
- Grupo Malaria, Facultad de Medicina, Universidad de Antioquia, Antioquia,
Colombia
| | | | | | | | - Simone da Silva Santos
- Laboratório de Doenças Parasitárias, Institute Oswaldo Cruz - Fiocruz-
Rio de Janeiro, Brazil
| | - Martha Suarez-Mutis
- Laboratório de Doenças Parasitárias, Institute Oswaldo Cruz - Fiocruz-
Rio de Janeiro, Brazil
| | | | - Andrea Regina de Souza Baptista
- Centro de Investigação de Microrganismos – CIM, Departamento de
Microbiologia e Parasitologia, Universidade Federal Fluminense,
Brazil
| | - Ricardo Luiz Dantas Machado
- Centro de Investigação de Microrganismos – CIM, Departamento de
Microbiologia e Parasitologia, Universidade Federal Fluminense,
Brazil
| | - Claudio R.F. Marinho
- Department of Parasitology, Institute of Biomedical Sciences, University
of São Paulo, São Paulo, Brazil
| | - Taane G. Clark
- Faculty of Infectious & Tropical Diseases, London School of Hygiene
& Tropical Medicine, London, UK
- Faculty of Epidemiology & Population Health, London School of Hygiene
& Tropical Medicine, London, UK
| | - Susana Campino
- Faculty of Infectious & Tropical Diseases, London School of Hygiene
& Tropical Medicine, London, UK
| |
Collapse
|
7
|
Tryptophan C-mannosylation is critical for Plasmodium falciparum transmission. Nat Commun 2022; 13:4400. [PMID: 35906227 PMCID: PMC9338275 DOI: 10.1038/s41467-022-32076-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Accepted: 07/07/2022] [Indexed: 11/08/2022] Open
Abstract
Tryptophan C-mannosylation stabilizes proteins bearing a thrombospondin repeat (TSR) domain in metazoans. Here we show that Plasmodium falciparum expresses a DPY19 tryptophan C-mannosyltransferase in the endoplasmic reticulum and that DPY19-deficiency abolishes C-glycosylation, destabilizes members of the TRAP adhesin family and inhibits transmission to mosquitoes. Imaging P. falciparum gametogenesis in its entirety in four dimensions using lattice light-sheet microscopy reveals defects in ΔDPY19 gametocyte egress and exflagellation. While egress is diminished, ΔDPY19 microgametes still fertilize macrogametes, forming ookinetes, but these are abrogated for mosquito infection. The gametogenesis defects correspond with destabilization of MTRAP, which we show is C-mannosylated in P. falciparum, and the ookinete defect is concordant with defective CTRP secretion on the ΔDPY19 background. Genetic complementation of DPY19 restores ookinete infectivity, sporozoite production and C-mannosylation activity. Therefore, tryptophan C-mannosylation by DPY19 ensures TSR protein quality control at two lifecycle stages for successful transmission of the human malaria parasite. Here, Lopaticki et al. show that Plasmodium falciparum expresses a Dpy19 C-mannosyltransferase in the endoplasmic reticulum that glycosylates TSR domains. Functional characterization shows that PfDpy19 plays a critical role in transmission through mosquitoes as PfDpy19-deficiency abolishes C-glycosylation and destabilizes proteins relevant for gametogenesis and oocyst formation.
Collapse
|
8
|
ApiAP2 Gene-Network Regulates Gametocytogenesis in Plasmodium Parasites. Cell Microbiol 2022. [DOI: 10.1155/2022/5796578] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Malaria is a mosquito-borne infectious disease, caused by unicellular Apicomplexan protozoa of the genus Plasmodium. The sexual stage of Plasmodium is one of the most fascinating aspects of the Plasmodium life cycle, yet relatively less explored until now. The production of sexually fit gametocytes through gametocytogenesis is essential to the transmission of the Plasmodium parasite into an anopheline mosquito vector. Understanding how gametocytogenesis is regulated promotes the identification of novel drug targets and also the development of transmission-blocking vaccines that would help reduce the disease burden in endemic areas. Transcriptional regulation in Plasmodium parasites is primarily controlled by a family of twenty-seven Apicomplexan Apetela 2 (ApiAP2) genes which act in a cascade to enable the parasite to progress through its asexual replication as well as gametocytogenesis. Here, we review the latest progress made on members of the ApiAP2 family characterized as key players of the transcriptional machinery of gametocytes. Further, we will highlight the transcriptional regulation network of ApiAP2 genes at each stage of gametocytogenesis.
Collapse
|
9
|
Narwal SK, Nayak B, Mehra P, Mishra S. Protein kinase 9 is not required for completion of the Plasmodium berghei life cycle. Microbiol Res 2022; 260:127051. [DOI: 10.1016/j.micres.2022.127051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2021] [Revised: 04/11/2022] [Accepted: 04/20/2022] [Indexed: 10/18/2022]
|
10
|
Possenti A, Di Cristina M, Nicastro C, Lunghi M, Messina V, Piro F, Tramontana L, Cherchi S, Falchi M, Bertuccini L, Spano F. Functional Characterization of the Thrombospondin-Related Paralogous Proteins Rhoptry Discharge Factors 1 and 2 Unveils Phenotypic Plasticity in Toxoplasma gondii Rhoptry Exocytosis. Front Microbiol 2022; 13:899243. [PMID: 35756016 PMCID: PMC9218915 DOI: 10.3389/fmicb.2022.899243] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Accepted: 05/11/2022] [Indexed: 11/13/2022] Open
Abstract
To gain access to the intracellular cytoplasmic niche essential for their growth and replication, apicomplexan parasites such as Toxoplasma gondii rely on the timely secretion of two types of apical organelles named micronemes and rhoptries. Rhoptry proteins are key to host cell invasion and remodeling, however, the molecular mechanisms underlying the tight control of rhoptry discharge are poorly understood. Here, we report the identification and functional characterization of two novel T. gondii thrombospondin-related proteins implicated in rhoptry exocytosis. The two proteins, already annotated as MIC15 and MIC14, were renamed rhoptry discharge factor 1 (RDF1) and rhoptry discharge factor 2 (RDF2) and found to be exclusive of the Coccidia class of apicomplexan parasites. Furthermore, they were shown to have a paralogous relationship and share a C-terminal transmembrane domain followed by a short cytoplasmic tail. Immunofluorescence analysis of T. gondii tachyzoites revealed that RDF1 presents a diffuse punctate localization not reminiscent of any know subcellular compartment, whereas RDF2 was not detected. Using a conditional knockdown approach, we demonstrated that RDF1 loss caused a marked growth defect. The lack of the protein did not affect parasite gliding motility, host cell attachment, replication and egress, whereas invasion was dramatically reduced. Notably, while RDF1 depletion did not result in altered microneme exocytosis, rhoptry discharge was found to be heavily impaired. Interestingly, rhoptry secretion was reversed by spontaneous upregulation of the RDF2 gene in knockdown parasites grown under constant RDF1 repression. Collectively, our results identify RDF1 and RDF2 as additional key players in the pathway controlling rhoptry discharge. Furthermore, this study unveils a new example of compensatory mechanism contributing to phenotypic plasticity in T. gondii.
Collapse
Affiliation(s)
- Alessia Possenti
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Manlio Di Cristina
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Chiara Nicastro
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Matteo Lunghi
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Valeria Messina
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Federica Piro
- Department of Chemistry, Biology and Biotechnology, University of Perugia, Perugia, Italy
| | - Lorenzo Tramontana
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Simona Cherchi
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| | - Mario Falchi
- National AIDS Center, Istituto Superiore di Sanità, Rome, Italy
| | | | - Furio Spano
- Department of Infectious Diseases, Istituto Superiore di Sanità, Rome, Italy
| |
Collapse
|
11
|
Paoletta MS, Wilkowsky SE. Thrombospondin Related Anonymous Protein Superfamily in Vector-Borne Apicomplexans: The Parasite’s Toolkit for Cell Invasion. Front Cell Infect Microbiol 2022; 12:831592. [PMID: 35463644 PMCID: PMC9019593 DOI: 10.3389/fcimb.2022.831592] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 03/07/2022] [Indexed: 11/23/2022] Open
Abstract
Apicomplexan parasites transmitted by vectors, including Babesia spp. and Plasmodium spp., cause severe disease in both humans and animals. These parasites have a complex life cycle during which they migrate, invade, and replicate in contrasting hosts such as the mammal and the invertebrate vector. The interaction of parasites with the host cell is mediated by adhesive proteins which play a key role in the different cellular processes regarding successful progression of the life cycle. Thrombospondin related anonymous protein (TRAP) is a superfamily of adhesins that are involved in motility, invasion and egress of the parasite. These proteins are stored and released from apical organelles and have either one or two types of adhesive domains, namely thrombospondin type 1 repeat and von Willebrand factor type A, that upon secretion are located in the extracellular portion of the molecule. Proteins from the TRAP superfamily have been intensively studied in Plasmodium species and to a lesser extent in Babesia spp., where they have proven to be functionally relevant throughout the entire parasite’s journey both in the arthropod vector and in the mammalian host. In recent years new findings provided answers to the role of TRAP proteins and in some cases the function of these adhesins during the parasite’s life cycle was redefined. In this review we will discuss the current knowledge of the diverse roles of the TRAP superfamily in vector-borne parasites from Class Aconoidasida. We will focus on the varied approaches that allowed the understanding of protein function and the relevance of TRAP- superfamily throughout the entire parasite’s cell cycle.
Collapse
|
12
|
Beyer K, Kracht S, Kehrer J, Singer M, Klug D, Frischknecht F. Limited Plasmodium sporozoite gliding motility in the absence of TRAP family adhesins. Malar J 2021; 20:430. [PMID: 34717635 PMCID: PMC8557484 DOI: 10.1186/s12936-021-03960-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Accepted: 10/19/2021] [Indexed: 12/01/2022] Open
Abstract
Background Plasmodium sporozoites are the highly motile forms of malaria-causing parasites that are transmitted by the mosquito to the vertebrate host. Sporozoites need to enter and cross several cellular and tissue barriers for which they employ a set of surface proteins. Three of these proteins are members of the thrombospondin related anonymous protein (TRAP) family. Here, potential additive, synergistic or antagonistic roles of these adhesion proteins were investigated. Methods Four transgenic Plasmodium berghei parasite lines that lacked two or all three of the TRAP family adhesins TRAP, TLP and TREP were generated using positive–negative selection. The parasite lines were investigated for their capacity to attach to and move on glass, their ability to egress from oocysts and their capacity to enter mosquito salivary glands. One strain was in addition interrogated for its capacity to infect mice. Results The major phenotype of the TRAP single gene deletion dominates additional gene deletion phenotypes. All parasite lines including the one lacking all three proteins were able to conduct some form of active, if unproductive movement. Conclusions The individual TRAP-family adhesins appear to play functionally distinct roles during motility and infection. Other proteins must contribute to substrate adhesion and gliding motility. Graphical Abstract ![]()
Supplementary Information The online version contains supplementary material available at 10.1186/s12936-021-03960-3.
Collapse
Affiliation(s)
- Konrad Beyer
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 344, 69120, Heidelberg, Germany
| | - Simon Kracht
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 344, 69120, Heidelberg, Germany
| | - Jessica Kehrer
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 344, 69120, Heidelberg, Germany.,German Center for Infection Research, Partner Site Heidelberg, 69120, Heidelberg, Germany
| | - Mirko Singer
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 344, 69120, Heidelberg, Germany.,Experimental Parasitology, Faculty of Veterinary Medicine, Ludwig-Maximilians-University Munich, Lena-Christ-Straße 48, Planegg, 82152, Munich, Germany
| | - Dennis Klug
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 344, 69120, Heidelberg, Germany.,Université de Strasbourg, CNRS UPR9022, INSERM U963, Institut de Biologie Moléculaire et Cellulaire, 67000, Strasbourg, France
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 344, 69120, Heidelberg, Germany. .,German Center for Infection Research, Partner Site Heidelberg, 69120, Heidelberg, Germany.
| |
Collapse
|
13
|
Real E, Howick VM, Dahalan FA, Witmer K, Cudini J, Andradi-Brown C, Blight J, Davidson MS, Dogga SK, Reid AJ, Baum J, Lawniczak MKN. A single-cell atlas of Plasmodium falciparum transmission through the mosquito. Nat Commun 2021; 12:3196. [PMID: 34045457 PMCID: PMC8159942 DOI: 10.1038/s41467-021-23434-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2021] [Accepted: 04/28/2021] [Indexed: 01/29/2023] Open
Abstract
Malaria parasites have a complex life cycle featuring diverse developmental strategies, each uniquely adapted to navigate specific host environments. Here we use single-cell transcriptomics to illuminate gene usage across the transmission cycle of the most virulent agent of human malaria - Plasmodium falciparum. We reveal developmental trajectories associated with the colonization of the mosquito midgut and salivary glands and elucidate the transcriptional signatures of each transmissible stage. Additionally, we identify both conserved and non-conserved gene usage between human and rodent parasites, which point to both essential mechanisms in malaria transmission and species-specific adaptations potentially linked to host tropism. Together, the data presented here, which are made freely available via an interactive website, provide a fine-grained atlas that enables intensive investigation of the P. falciparum transcriptional journey. As well as providing insights into gene function across the transmission cycle, the atlas opens the door for identification of drug and vaccine targets to stop malaria transmission and thereby prevent disease.
Collapse
Affiliation(s)
- Eliana Real
- Department of Life Sciences, Imperial College London, London, UK
| | - Virginia M Howick
- Parasites and Microbes Programme, Wellcome Sanger Institute, Hinxton, UK
- Institute of Biodiversity, Animal Health and Comparative Medicine, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
- Wellcome Centre for Integrative Parasitology, College of Medical Veterinary and Life Sciences, University of Glasgow, Glasgow, UK
| | - Farah A Dahalan
- Department of Life Sciences, Imperial College London, London, UK
| | - Kathrin Witmer
- Department of Life Sciences, Imperial College London, London, UK
- Parasites and Microbes Programme, Wellcome Sanger Institute, Hinxton, UK
| | - Juliana Cudini
- Parasites and Microbes Programme, Wellcome Sanger Institute, Hinxton, UK
| | - Clare Andradi-Brown
- Department of Life Sciences, Imperial College London, London, UK
- Department of Infectious Disease, Imperial College London, London, UK
| | - Joshua Blight
- Department of Life Sciences, Imperial College London, London, UK
| | - Mira S Davidson
- Department of Life Sciences, Imperial College London, London, UK
| | - Sunil Kumar Dogga
- Parasites and Microbes Programme, Wellcome Sanger Institute, Hinxton, UK
| | - Adam J Reid
- Parasites and Microbes Programme, Wellcome Sanger Institute, Hinxton, UK
| | - Jake Baum
- Department of Life Sciences, Imperial College London, London, UK.
| | - Mara K N Lawniczak
- Parasites and Microbes Programme, Wellcome Sanger Institute, Hinxton, UK.
| |
Collapse
|
14
|
Sekar V, Rivero A, Pigeault R, Gandon S, Drews A, Ahren D, Hellgren O. Gene regulation of the avian malaria parasite Plasmodium relictum, during the different stages within the mosquito vector. Genomics 2021; 113:2327-2337. [PMID: 34023365 DOI: 10.1016/j.ygeno.2021.05.021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 04/26/2021] [Accepted: 05/18/2021] [Indexed: 10/21/2022]
Abstract
The malaria parasite Plasmodium relictum is one of the most widespread species of avian malaria. As in the case of its human counterparts, bird Plasmodium undergoes a complex life cycle infecting two hosts: the arthropod vector and the vertebrate host. In this study, we examined transcriptomes of P. relictum (SGS1) during crucial timepoints within its vector, Culex pipiens quinquefasciatus. Differential gene-expression analyses identified genes linked to the parasites life-stages at: i) a few minutes after the blood meal is ingested, ii) during peak oocyst production phase, iii) during peak sporozoite phase and iv) during the late-stages of the infection. A large amount of genes coding for functions linked to host-immune invasion and multifunctional genes was active throughout the infection cycle. One gene associated with a conserved Plasmodium membrane protein with unknown function was upregulated throughout the parasite development in the vector, suggesting an important role in the successful completion of the sporogonic cycle. Gene expression analysis further identified genes, with unknown functions to be significantly differentially expressed during the infection in the vector as well as upregulation of reticulocyte-binding proteins, which raises the possibility of the multifunctionality of these RBPs. We establish the existence of highly stage-specific pathways being overexpressed during the infection. This first study of gene-expression of a non-human Plasmodium species in its vector provides a comprehensive insight into the molecular mechanisms of the common avian malaria parasite P. relictum and provides essential information on the evolutionary diversity in gene regulation of the Plasmodium's vector stages.
Collapse
Affiliation(s)
| | - Ana Rivero
- MIVEGEC (CNRS - Université de Montpellier - IRD), 34394 Montpellier, France; CREES (Centre de Recherche en Ecologie et Evolution de la Santé), 34394 Montpellier, France
| | - Romain Pigeault
- Department of Biology, Lund University, Sweden; Department of Ecology and Evolution, CH-1015 Lausanne, Switzerland
| | - Sylvain Gandon
- CEFE (CNRS - Université de Montpellier - Université Paul-Valéry - EPHE - IRD), Montpellier, France
| | - Anna Drews
- MEMEG, Department of Biology, Lund University, Sweden
| | - Dag Ahren
- National Bioinformatics Infrastructure Sweden (NBIS), SciLifeLab, Department of Biology, Lund, Sweden
| | - Olof Hellgren
- MEMEG, Department of Biology, Lund University, Sweden.
| |
Collapse
|
15
|
Knöckel J, Dundas K, Yang ASP, Galaway F, Metcalf T, Gemert GJV, Sauerwein RW, Rayner JC, Billker O, Wright GJ. Systematic Identification of Plasmodium Falciparum Sporozoite Membrane Protein Interactions Reveals an Essential Role for the p24 Complex in Host Infection. Mol Cell Proteomics 2021; 20:100038. [PMID: 33515807 PMCID: PMC7950211 DOI: 10.1074/mcp.ra120.002432] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 12/22/2020] [Indexed: 12/14/2022] Open
Abstract
Sporozoites are a motile form of malaria-causing Plasmodium falciparum parasites that migrate from the site of transmission in the dermis through the bloodstream to invade hepatocytes. Sporozoites interact with many cells within the host, but the molecular identity of these interactions and their role in the pathology of malaria is poorly understood. Parasite proteins that are secreted and embedded within membranes are known to be important for these interactions, but our understanding of how they interact with each other to form functional complexes is largely unknown. Here, we compile a library of recombinant proteins representing the repertoire of cell surface and secreted proteins from the P. falciparum sporozoite and use an assay designed to detect extracellular interactions to systematically identify complexes. We identify three protein complexes including an interaction between two components of the p24 complex that is involved in the trafficking of glycosylphosphatidylinositol-anchored proteins through the secretory pathway. Plasmodium parasites lacking either gene are strongly inhibited in the establishment of liver-stage infections. These findings reveal an important role for the p24 complex in malaria pathogenesis and show that the library of recombinant proteins represents a valuable resource to investigate P. falciparum sporozoite biology.
Collapse
Key Words
- avexis, avidity-based extracellular interaction screen
- csp, circumsporozoite protein
- gpi, glycosylphosphatidylinositol
- hbs, hepes-buffered saline
- hek, human embryonic kidney
- ivis, in vivo imaging system
- msp, merozoite surface protein
- piesp15, parasite-infected erythrocyte surface protein 15
- spr, surface plasmon resonance
- trap, thrombospondin-related anonymous protein
Collapse
Affiliation(s)
- Julia Knöckel
- Cell Surface Signalling Laboratory, Wellcome Sanger Institute, Cambridge, United Kingdom; Malaria Programme, Wellcome Sanger Institute, Cambridge, United Kingdom
| | - Kirsten Dundas
- Cell Surface Signalling Laboratory, Wellcome Sanger Institute, Cambridge, United Kingdom; Malaria Programme, Wellcome Sanger Institute, Cambridge, United Kingdom
| | - Annie S P Yang
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Francis Galaway
- Cell Surface Signalling Laboratory, Wellcome Sanger Institute, Cambridge, United Kingdom; Malaria Programme, Wellcome Sanger Institute, Cambridge, United Kingdom
| | - Tom Metcalf
- Malaria Programme, Wellcome Sanger Institute, Cambridge, United Kingdom
| | - Geert-Jan van Gemert
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Robert W Sauerwein
- Radboudumc Center for Infectious Diseases, Department of Medical Microbiology, Radboud University Medical Center, Nijmegen, The Netherlands
| | - Julian C Rayner
- Malaria Programme, Wellcome Sanger Institute, Cambridge, United Kingdom
| | - Oliver Billker
- Malaria Programme, Wellcome Sanger Institute, Cambridge, United Kingdom; The Laboratory for Molecular Infection Medicine Sweden (MIMS) and Department of Molecular Biology, Umeå University, Umeå, Sweden
| | - Gavin J Wright
- Cell Surface Signalling Laboratory, Wellcome Sanger Institute, Cambridge, United Kingdom; Malaria Programme, Wellcome Sanger Institute, Cambridge, United Kingdom; Department of Biology, Hull York Medical School, York Biomedical Research Institute, University of York, York, United Kingdom.
| |
Collapse
|
16
|
Montenegro VN, Paoletta MS, Jaramillo Ortiz JM, Suarez CE, Wilkowsky SE. Identification and characterization of a Babesia bigemina thrombospondin-related superfamily member, TRAP-1: a novel antigen containing neutralizing epitopes involved in merozoite invasion. Parasit Vectors 2020; 13:602. [PMID: 33261638 PMCID: PMC7705850 DOI: 10.1186/s13071-020-04469-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Accepted: 11/05/2020] [Indexed: 11/22/2022] Open
Abstract
Background Thrombospondin-related anonymous protein (TRAP) has been described as a potential vaccine candidate for several diseases caused by apicomplexan parasites. However, this protein and members of this family have not yet been characterized in Babesia bigemina, one of the most prevalent species causing bovine babesiosis. Methods The 3186-bp Babesia bigemina TRAP-1 (BbiTRAP-1) gene was identified by a bioinformatics search using the B. bovis TRAP-1 sequence. Members of the TRAP and TRAP-related protein families (TRP) were identified in Babesia and Theileria through a search of the TSP-1 adhesive domain, which is the hallmark motif in both proteins. Structural modeling and phylogenetic analysis were performed with the identified TRAP proteins. A truncated recombinant BbiTRAP-1 that migrates at approximately 107 kDa and specific antisera were produced and used in Western blot analysis and indirect fluorescent antibody tests (IFAT). B-cell epitopes with neutralizing activity in BbiTRAP-1 were defined by enzyme-linked immunosorbent assays (ELISA) and invasion assays. Results Three members of the TRAP family of proteins were identified in B. bigemina (BbiTRAP-1 to -3). All are type 1 transmembrane proteins containing the von Willebrand factor A (vWFA), thrombospondin type 1 (TSP-1), and cytoplasmic C-terminus domains, as well as transmembrane regions. The BbiTRAP-1 predicted structure also contains a metal ion-dependent adhesion site for interaction with the host cell. The TRP family in Babesia and Theileria species contains the canonical TSP-1 domain but lacks the vWFA domain and together with TRAP define a novel gene superfamily. A variable number of tandem repeat units are present in BbiTRAP-1 and could be used for strain genotyping. Western blot and IFAT analysis confirmed the expression of BbiTRAP-1 by blood-stage parasites. Partial recognition by a panel of sera from B. bigemina-infected cattle in ELISAs using truncated BbiTRAP-1 suggests that this protein is not an immunodominant antigen. Additionally, bovine anti-recombinant BbiTRAP-1 antibodies were found to be capable of neutralizing merozoite invasion in vitro. Conclusions We have identified the TRAP and TRP gene families in several Babesia and Theileria species and characterized BbiTRAP-1 as a novel antigen of B. bigemina. The functional relevance and presence of neutralization-sensitive B-cell epitopes suggest that BbiTRAP-1 could be included in tests for future vaccine candidates against B. bigemina.![]()
Collapse
Affiliation(s)
- Valeria Noely Montenegro
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO) (INTA-CONICET), De Los Reseros y Dr. Nicolás Repetto s/N, P.O. Box 25, B1712WAA, Castelar, Buenos Aires, Argentina
| | - Martina Soledad Paoletta
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO) (INTA-CONICET), De Los Reseros y Dr. Nicolás Repetto s/N, P.O. Box 25, B1712WAA, Castelar, Buenos Aires, Argentina
| | - José M Jaramillo Ortiz
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO) (INTA-CONICET), De Los Reseros y Dr. Nicolás Repetto s/N, P.O. Box 25, B1712WAA, Castelar, Buenos Aires, Argentina
| | - Carlos E Suarez
- Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, WA, 99164, USA.,Animal Disease Research Unit, U.S. Department of Agriculture-Agricultural Research Service (USDA-ARS), Washington State University, 3003 ADBF, P.O. Box 646630, Pullman, WA, 99164, USA
| | - Silvina Elizabeth Wilkowsky
- Instituto de Agrobiotecnología y Biología Molecular (IABIMO) (INTA-CONICET), De Los Reseros y Dr. Nicolás Repetto s/N, P.O. Box 25, B1712WAA, Castelar, Buenos Aires, Argentina.
| |
Collapse
|
17
|
Ueti MW, Johnson WC, Kappmeyer LS, Herndon DR, Mousel MR, Reif KE, Taus NS, Ifeonu OO, Silva JC, Suarez CE, Brayton KA. Comparative analysis of gene expression between Babesia bovis blood stages and kinetes allowed by improved genome annotation. Int J Parasitol 2020; 51:123-136. [PMID: 33069745 DOI: 10.1016/j.ijpara.2020.08.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2020] [Revised: 07/29/2020] [Accepted: 08/04/2020] [Indexed: 12/23/2022]
Abstract
Throughout their life cycle, Babesia parasites alternate between a mammalian host, where they cause babesiosis, and the tick vector. Transition between hosts results in distinct environmental signals that influence patterns of gene expression, consistent with the morphological and functional changes operating in the parasites during their life stages. In addition, comparing differential patterns of gene expression among mammalian and tick parasite stages can provide clues for developing improved methods of control. Hereby, we upgraded the genome assembly of Babesia bovis, a bovine hemoparasite, closing a 139 kbp gap, and used RNA-Seq datasets derived from mammalian blood and tick kinete stages to update the genome annotation. Of the originally annotated genes, 1,254 required structural changes, and 326 new genes were identified, leading to a different predicted proteome compared to the original annotation. Next, the RNA-Seq data was used to identify B. bovis genes that were differentially expressed in the vertebrate and arthropod hosts. In blood stages, 28% of the genes were upregulated up to 300 fold, whereas 26% of the genes in kinetes, a tick stage, were upregulated up to >19,000 fold. We thus discovered differentially expressed genes that may play key biological roles and serve as suitable targets for the development of vaccines to control bovine babesiosis.
Collapse
Affiliation(s)
- Massaro W Ueti
- Animal Diseases Research Unit, USDA-ARS, Pullman, Washington, USA; Program in Vector-borne Diseases, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA; Paul G. Allen School for Global Animal Health, Washington State University, Pullman, Washington, USA.
| | | | | | - David R Herndon
- Animal Diseases Research Unit, USDA-ARS, Pullman, Washington, USA
| | - Michelle R Mousel
- Animal Diseases Research Unit, USDA-ARS, Pullman, Washington, USA; Paul G. Allen School for Global Animal Health, Washington State University, Pullman, Washington, USA
| | - Kathryn E Reif
- Animal Diseases Research Unit, USDA-ARS, Pullman, Washington, USA; Program in Vector-borne Diseases, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Naomi S Taus
- Animal Diseases Research Unit, USDA-ARS, Pullman, Washington, USA; Program in Vector-borne Diseases, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Olukemi O Ifeonu
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Joana C Silva
- Institute for Genome Sciences, University of Maryland School of Medicine, Baltimore, Maryland, USA; Department of Microbiology and Immunology, University of Maryland School of Medicine, Baltimore, Maryland, USA
| | - Carlos E Suarez
- Animal Diseases Research Unit, USDA-ARS, Pullman, Washington, USA; Program in Vector-borne Diseases, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA
| | - Kelly A Brayton
- Program in Vector-borne Diseases, Department of Veterinary Microbiology and Pathology, Washington State University, Pullman, Washington, USA; Paul G. Allen School for Global Animal Health, Washington State University, Pullman, Washington, USA.
| |
Collapse
|
18
|
Tartarelli I, Tinari A, Possenti A, Cherchi S, Falchi M, Dubey JP, Spano F. During host cell traversal and cell-to-cell passage, Toxoplasma gondii sporozoites inhabit the parasitophorous vacuole and posteriorly release dense granule protein-associated membranous trails. Int J Parasitol 2020; 50:1099-1115. [PMID: 32882286 DOI: 10.1016/j.ijpara.2020.06.012] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2020] [Revised: 05/21/2020] [Accepted: 06/23/2020] [Indexed: 10/23/2022]
Abstract
Toxoplasma gondii has a worldwide distribution and infects virtually all warm-blooded animals, including humans. Ingestion of the environmentally resistant oocyst stage, excreted only in the feces of cats, is central to transmission of this apicomplexan parasite. There is vast literature on the host and T. gondii tachyzoite (proliferative stage of the parasite) but little is known of the host-parasite interaction and conversion of the free-living stage (sporozoite inside the oocyst) to the parasitic stage. Here, we present events that follow invasion of host cells with T. gondii sporozoites by using immunofluorescence (IF) and transmission electron microscopy (TEM). Several human type cell cultures were infected with T. gondii sporozoites of the two genotypes (Type II, ME49 and Type III, VEG) most prevalent worldwide. For the first known time, using anti-rhoptry neck protein 4 (RON4) antibodies, the moving junction was visualized in sporozoites during the invasion process and shortly after its completion. Surprisingly, IF and TEM evaluation revealed that intracellular sporozoites release, at their posterior end, long membranous tails, herein named sporozoite-specific trails (SSTs). Differential permeabilization and IF experiments showed that the SSTs are associated with several dense granule proteins (GRAs) and that their membranous component is of parasite origin. Furthermore, TEM observations demonstrated that SST-associated sporozoites are delimited by a typical parasitophorous vacuole, which is retained during parasite exit from the host cell and during cell-to-cell passage. Our data strongly suggest that host cell traversal by T. gondii sporozoites relies on a novel force-producing mechanism, based on the massive extrusion at the parasite posterior pole of GRA-associated membranous material derived from the same pool of membranes forming the intravacuolar network.
Collapse
Affiliation(s)
- Irene Tartarelli
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Antonella Tinari
- Center for Gender-Specific Medicine, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Alessia Possenti
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Simona Cherchi
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Mario Falchi
- National AIDS Center, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy
| | - Jitender P Dubey
- U.S. Department of Agriculture, Agricultural Research Service, Beltsville Agricultural Research Center, Animal Parasitic Diseases Laboratory, Beltsville, Maryland 20705, United States
| | - Furio Spano
- Department of Infectious Diseases, Istituto Superiore di Sanità, Viale Regina Elena 299, 00161 Rome, Italy.
| |
Collapse
|
19
|
Li W, Wang M, Chen Y, Chen C, Liu X, Sun X, Jing C, Xu L, Yan R, Li X, Song X. EtMIC3 and its receptors BAG1 and ENDOUL are essential for site-specific invasion of Eimeria tenella in chickens. Vet Res 2020; 51:90. [PMID: 32678057 PMCID: PMC7367391 DOI: 10.1186/s13567-020-00809-6] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2020] [Accepted: 06/08/2020] [Indexed: 12/16/2022] Open
Abstract
Avian coccidian parasites exhibit a high degree of site specificity in different Eimeria species. Although the underlying mechanism is unclear, an increasing body of evidence suggests that site specificity is due to the interaction between microneme proteins (MICs) and their receptors on the surface of target host cells. In this study, the binding ability of E. tenella MICs (EtMICs) to different intestinal tissue was observed by immunofluorescence to identify the key surface molecule on the parasite responsible for the site specificity. Subsequently, we identified the corresponding host-cell receptors by yeast two-hybrid screening and glutathione-S-transferase pull-down experiments and the distribution of these receptors was observed by immunofluorescence in chicken intestinal tissues. Finally, we evaluated the efficacy of receptor antiserum against the infection of E. tenella in chickens. The results showed that EtMIC3 could only bind to the caecum while EtMIC1, EtMIC2, and EtAMA1 did not bind to any other intestinal tissues. Anti-serum to EtMIC3 was able to block the invasion of sporozoites with a blocking rate of 66.3%. The receptors for EtMIC3 were BCL2-associated athanogene 1 (BAG1) and Endonuclease polyU-specific-like (ENDOUL), which were mainly distributed in the caecum. BAG1 and ENDOUL receptor antiserum reduced weight loss and oocyst output following E. tenella infection, showing partial inhibition of E. tenella infection. These data elucidate the mechanism of site specificity for Eimeria infection and reveal a potential therapeutic avenue.
Collapse
Affiliation(s)
- Wenyu Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Mingyue Wang
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Yufeng Chen
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Chen Chen
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Xiaoqian Liu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Xiaoting Sun
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Chuanxu Jing
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Lixin Xu
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Ruofeng Yan
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Xiangrui Li
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China
| | - Xiaokai Song
- MOE Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, People's Republic of China.
| |
Collapse
|
20
|
Klug D, Goellner S, Kehrer J, Sattler J, Strauss L, Singer M, Lu C, Springer TA, Frischknecht F. Evolutionarily distant I domains can functionally replace the essential ligand-binding domain of Plasmodium TRAP. eLife 2020; 9:57572. [PMID: 32648541 PMCID: PMC7351488 DOI: 10.7554/elife.57572] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Accepted: 06/25/2020] [Indexed: 02/02/2023] Open
Abstract
Inserted (I) domains function as ligand-binding domains in adhesins that support cell adhesion and migration in many eukaryotic phyla. These adhesins include integrin αβ heterodimers in metazoans and single subunit transmembrane proteins in apicomplexans such as TRAP in Plasmodium and MIC2 in Toxoplasma. Here we show that the I domain of TRAP is essential for sporozoite gliding motility, mosquito salivary gland invasion and mouse infection. Its replacement with the I domain from Toxoplasma MIC2 fully restores tissue invasion and parasite transmission, while replacement with the aX I domain from human integrins still partially restores liver infection. Mutations around the ligand binding site allowed salivary gland invasion but led to inefficient transmission to the rodent host. These results suggest that apicomplexan parasites appropriated polyspecific I domains in part for their ability to engage with multiple ligands and to provide traction for emigration into diverse organs in distant phyla. Malaria is an infectious disease caused by single-celled parasites known as Plasmodium. Humans and other animals with backbones – such as birds, reptiles and rodents – can become hosts for these parasites if an infected female mosquito feeds on their blood. Likewise, healthy mosquitoes can in turn become infected with Plasmodium if they feed on the blood of an infected animal. To complete their life cycle, Plasmodium parasites within a mosquito must become spore-like cells called sporozoites. These sporozoites are highly mobile and can get into the mosquitoes’ salivary glands, meaning they can be passed on to a new host when the insect feeds. During a mosquito bite the sporozoites are spat into the skin of the potential host, where they then need to migrate rapidly to enter the bloodstream. Once in the blood, the sporozoites can then get into liver cells and begin a new infection. One protein called TRAP, which is found on the surface of the sporozoites, is important for their migration and the infection of the salivary glands or liver. Yet it was not known how this happens at the level of the individual proteins involved. Klug et al. have now tested how a part of the TRAP protein, called the I domain, contributes to the infection process. In the experiments, the I domain of TRAP was deleted which showed that the sporozoites need this domain to be able to move around and get into the host tissues. Without the I domain the sporozoites were stuck and could not successfully infect either the mosquitoes, the livers of mice, or human liver cells grown in the laboratory. Klug et al. then replaced the Plasmodium I domain of TRAP with the I domain from a distantly related parasite called Toxoplasma gondii, which causes a condition known as toxoplasmosis. The I domain from Toxoplasma allowed the Plasmodium parasites to infect the host tissues again. This observation was unexpected because Toxoplasma and Plasmodium parasites have evolved separately over the last 800 million years and Toxoplasma does not infect insects. These findings suggest that the I domain of TRAP evolved to bind several other proteins in different tissues and hosts. Future studies will investigate which other parasite proteins TRAP works with to guide sporozoites to the salivary glands or liver. Knowledge of how these proteins act together may lead to new approaches for treating or preventing malaria. For example, some treatments could stop sporozoites from entering liver cells.
Collapse
Affiliation(s)
- Dennis Klug
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany.,Université de Strasbourg, CNRS UPR9022, INSERM U963, Institut de Biologie Moléculaire et Cellulaire, Strasbourg, France
| | - Sarah Goellner
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany.,Department of Molecular Virology, Heidelberg University Medical School, Heidelberg, Germany
| | - Jessica Kehrer
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| | - Julia Sattler
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| | - Léanne Strauss
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| | - Mirko Singer
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany.,Experimental Parasitology, Faculty of Veterinary Medicine, Ludwig-Maximilians-Universität München, München, Germany
| | - Chafen Lu
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, United States
| | - Timothy A Springer
- Program in Cellular and Molecular Medicine, Children's Hospital Boston, and Departments of Biological Chemistry and Molecular Pharmacology and of Medicine, Harvard Medical School, Boston, United States
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Heidelberg, Germany
| |
Collapse
|
21
|
Ruiz JL, Gómez-Díaz E. The second life of Plasmodium in the mosquito host: gene regulation on the move. Brief Funct Genomics 2020; 18:313-357. [PMID: 31058281 DOI: 10.1093/bfgp/elz007] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2018] [Revised: 03/08/2019] [Accepted: 03/26/2019] [Indexed: 01/08/2023] Open
Abstract
Malaria parasites face dynamically changing environments and strong selective constraints within human and mosquito hosts. To survive such hostile and shifting conditions, Plasmodium switches transcriptional programs during development and has evolved mechanisms to adjust its phenotype through heterogeneous patterns of gene expression. In vitro studies on culture-adapted isolates have served to set the link between chromatin structure and functional gene expression. Yet, experimental evidence is limited to certain stages of the parasite in the vertebrate, i.e. blood, while the precise mechanisms underlying the dynamic regulatory landscapes during development and in the adaptation to within-host conditions remain poorly understood. In this review, we discuss available data on transcriptional and epigenetic regulation in Plasmodium mosquito stages in the context of sporogonic development and phenotypic variation, including both bet-hedging and environmentally triggered direct transcriptional responses. With this, we advocate the mosquito offers an in vivo biological model to investigate the regulatory networks, transcription factors and chromatin-modifying enzymes and their modes of interaction with regulatory sequences, which might be responsible for the plasticity of the Plasmodium genome that dictates stage- and cell type-specific blueprints of gene expression.
Collapse
Affiliation(s)
- José L Ruiz
- Instituto de Parasitología y Biomedicina López-Neyra (IPBLN), Consejo Superior de Investigaciones Científicas, Granada, Spain
| | - Elena Gómez-Díaz
- Instituto de Parasitología y Biomedicina López-Neyra (IPBLN), Consejo Superior de Investigaciones Científicas, Granada, Spain
| |
Collapse
|
22
|
Arévalo-Pinzón G, Garzón-Ospina D, Pulido FA, Bermúdez M, Forero-Rodríguez J, Rodríguez-Mesa XM, Reyes-Guarín LP, Suárez CF, Patarroyo MA. Plasmodium vivax Cell Traversal Protein for Ookinetes and Sporozoites (CelTOS) Functionally Restricted Regions Are Involved in Specific Host-Pathogen Interactions. Front Cell Infect Microbiol 2020; 10:119. [PMID: 32266169 PMCID: PMC7105572 DOI: 10.3389/fcimb.2020.00119] [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: 12/27/2019] [Accepted: 03/04/2020] [Indexed: 11/13/2022] Open
Abstract
Following the injection of Plasmodium sporozoites by a female Anopheles mosquito into the dermis, they become engaged on a long journey to hepatic tissue where they must migrate through different types of cell to become established in parasitophorous vacuoles in hepatocytes. Studies have shown that proteins such as cell traversal protein for Plasmodium ookinetes and sporozoites (CelTOS) play a crucial role in cell-traversal ability. Although CelTOS has been extensively studied in various species and included in pre-clinical assays it remains unknown which P. vivax CelTOS (PvCelTOS) regions are key in its interaction with traversed or target cells (Kupffer or hepatocytes) and what type of pressure, association and polymorphism these important regions could have to improve their candidacy as important vaccine antigens. This work has described producing a recombinant PvCelTOS which was recognized by ~30% P. vivax-infected individuals, thereby confirming its ability for inducing a natural immune response. PvCelTOS' genetic diversity in Colombia and its ability to interact with HeLa (traversal cell) and/or HepG2 cell (target cell) external membrane have been assessed. One region in the PvCelTOS amino-terminal region and another in its C-terminus were seen to be participating in host-pathogen interactions. These regions had important functional constraint signals (ω < 0.3 and several sites under negative selection) and were able to inhibit specific rPvCelTOS binding to HeLa cells. This led to suggesting that sequences between aa 41-60 (40833) and 141-160 (40838) represent promising candidates for an anti-P. vivax subunit-based vaccine.
Collapse
Affiliation(s)
- Gabriela Arévalo-Pinzón
- Receptor-Ligand Department, Fundacion Instituto de Inmunologia de Colombia (FIDIC), Bogota, Colombia.,School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia
| | - Diego Garzón-Ospina
- PhD Programme in Biomedical and Biological Sciences, Universidad del Rosario, Bogota, Colombia
| | - Fredy A Pulido
- Receptor-Ligand Department, Fundacion Instituto de Inmunologia de Colombia (FIDIC), Bogota, Colombia
| | - Maritza Bermúdez
- Receptor-Ligand Department, Fundacion Instituto de Inmunologia de Colombia (FIDIC), Bogota, Colombia
| | - Johanna Forero-Rodríguez
- Molecular Biology and Immunology Department, Fundacion Instituto de Immunologia de Colombia (FIDIC), Bogota, Colombia
| | - Xandy M Rodríguez-Mesa
- Molecular Biology and Immunology Department, Fundacion Instituto de Immunologia de Colombia (FIDIC), Bogota, Colombia
| | - Leidy P Reyes-Guarín
- Molecular Biology and Immunology Department, Fundacion Instituto de Immunologia de Colombia (FIDIC), Bogota, Colombia
| | - Carlos F Suárez
- School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia.,Biomathematics Department, Fundacion Instituto de Immunologia de Colombia (FIDIC), Bogota, Colombia
| | - Manuel A Patarroyo
- School of Medicine and Health Sciences, Universidad del Rosario, Bogota, Colombia.,Molecular Biology and Immunology Department, Fundacion Instituto de Immunologia de Colombia (FIDIC), Bogota, Colombia
| |
Collapse
|
23
|
Horta MF, Andrade LO, Martins-Duarte ÉS, Castro-Gomes T. Cell invasion by intracellular parasites - the many roads to infection. J Cell Sci 2020; 133:133/4/jcs232488. [PMID: 32079731 DOI: 10.1242/jcs.232488] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Intracellular parasites from the genera Toxoplasma, Plasmodium, Trypanosoma, Leishmania and from the phylum Microsporidia are, respectively, the causative agents of toxoplasmosis, malaria, Chagas disease, leishmaniasis and microsporidiosis, illnesses that kill millions of people around the globe. Crossing the host cell plasma membrane (PM) is an obstacle these parasites must overcome to establish themselves intracellularly and so cause diseases. The mechanisms of cell invasion are quite diverse and include (1) formation of moving junctions that drive parasites into host cells, as for the protozoans Toxoplasma gondii and Plasmodium spp., (2) subversion of endocytic pathways used by the host cell to repair PM, as for Trypanosoma cruzi and Leishmania, (3) induction of phagocytosis as for Leishmania or (4) endocytosis of parasites induced by specialized structures, such as the polar tubes present in microsporidian species. Understanding the early steps of cell entry is essential for the development of vaccines and drugs for the prevention or treatment of these diseases, and thus enormous research efforts have been made to unveil their underlying biological mechanisms. This Review will focus on these mechanisms and the factors involved, with an emphasis on the recent insights into the cell biology of invasion by these pathogens.
Collapse
Affiliation(s)
- Maria Fátima Horta
- Departamento de Bioquímica e Imunologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| | - Luciana Oliveira Andrade
- Departamento de Morfologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| | - Érica Santos Martins-Duarte
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| | - Thiago Castro-Gomes
- Departamento de Parasitologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Belo Horizonte, CEP 31270-901, Brazil
| |
Collapse
|
24
|
Transcriptomics and proteomics reveal two waves of translational repression during the maturation of malaria parasite sporozoites. Nat Commun 2019; 10:4964. [PMID: 31673027 PMCID: PMC6823429 DOI: 10.1038/s41467-019-12936-6] [Citation(s) in RCA: 75] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Accepted: 10/09/2019] [Indexed: 02/08/2023] Open
Abstract
Plasmodium sporozoites are transmitted from infected mosquitoes to mammals, and must navigate the host skin and vasculature to infect the liver. This journey requires distinct proteomes. Here, we report the dynamic transcriptomes and proteomes of both oocyst sporozoites and salivary gland sporozoites in both rodent-infectious Plasmodium yoelii parasites and human-infectious Plasmodium falciparum parasites. The data robustly define mRNAs and proteins that are upregulated in oocyst sporozoites (UOS) or upregulated in infectious sporozoites (UIS) within the salivary glands, including many that are essential for sporozoite functions in the vector and host. Moreover, we find that malaria parasites use two overlapping, extensive, and independent programs of translational repression across sporozoite maturation to temporally regulate protein expression. Together with gene-specific validation experiments, these data indicate that two waves of translational repression are implemented and relieved at different times during sporozoite maturation, migration and infection, thus promoting their successful development and vector-to-host transition. Here, the authors report transcriptomes and proteomes of oocyst sporozoite and salivary gland sporozoite stages in rodent-infectious Plasmodium yoelii parasites and human infectious Plasmodium falciparum parasites and define two waves of translational repression during sporozoite maturation.
Collapse
|
25
|
A Plasmodium falciparum C-mannosyltransferase is dispensable for parasite asexual blood stage development. Parasitology 2019; 146:1767-1772. [PMID: 31559936 PMCID: PMC6939167 DOI: 10.1017/s0031182019001380] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
C-mannosylation was recently identified in the thrombospondin-related anonymous protein (TRAP) from Plasmodium falciparum salivary gland sporozoites. A candidate P. falciparum C-mannosyltransferase (PfDPY-19) was demonstrated to modify thrombospondin type 1 repeat (TSR) domains in vitro, exhibiting a different acceptor specificity than their mammalian counterparts. According to the described minimal acceptor of PfDPY19, several TSR domain-containing proteins of P. falciparum could be C-mannosylated in vivo. However, the relevance of this protein modification for the parasite viability remains unknown. In the present study, we used CRISPR/Cas9 technology to generate a PfDPY19 null mutant, demonstrating that this glycosyltransferase is not essential for the asexual blood development of the parasite. PfDPY19 gene disruption was not associated with a growth phenotype, not even under endoplasmic reticulum-stressing conditions that could impair protein folding. The data presented in this work strongly suggest that PfDPY19 is unlikely to play a critical role in the asexual blood stages of the parasite, at least under in vitro conditions.
Collapse
|
26
|
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
|
27
|
Abstract
Plasmodium sporozoites are injected into the skin as mosquitoes probe for blood. From here, they migrate through the dermis to find blood vessels which they enter in order to be rapidly carried to the liver, where they invade hepatocytes and develop into the next life cycle stage, the exoerythrocytic stage. Once sporozoites enter the blood circulation, they are found in hepatocytes within minutes. In contrast, sporozoite exit from the inoculation site resembles a slow trickle and occurs over several hours. Thus, sporozoites spend the majority of their extracellular time at the inoculation site, raising the hypothesis that this is when the malarial parasite is most vulnerable to antibody-mediated destruction. Here, we investigate this hypothesis and demonstrate that the neutralizing capacity of circulating antibodies is greater at the inoculation site than in the blood circulation. Furthermore, these antibodies are working, at least in part, by impacting sporozoite motility at the inoculation site. Using actively and passively immunized mice, we found that most parasites are either immobilized at the site of injection or display reduced motility, particularly in their net displacement. We also found that antibodies severely impair the entry of sporozoites into the bloodstream. Overall, our data suggest that antibodies targeting the migratory sporozoite exert a large proportion of their protective effect at the inoculation site.IMPORTANCE Studies in experimental animal models and humans have shown that antibodies against Plasmodium sporozoites abolish parasite infectivity and provide sterile immunity. While it is well documented that these antibodies can be induced after immunization with attenuated parasites or subunit vaccines, the mechanisms by and location in which they neutralize parasites have not been fully elucidated. Here, we report studies indicating that these antibodies display a significant portion of their protective effect in the skin after injection of sporozoites and that one mechanism by which they work is by impairing sporozoite motility, thus diminishing their ability to reach blood vessels. These results suggest that immune protection against malaria begins at the earliest stages of parasite infection and emphasize the need of performing parasite challenge in the skin for the evaluation of protective immunity.
Collapse
|
28
|
Plasmodium genomics: an approach for learning about and ending human malaria. Parasitol Res 2018; 118:1-27. [PMID: 30402656 DOI: 10.1007/s00436-018-6127-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2018] [Accepted: 10/19/2018] [Indexed: 12/31/2022]
Abstract
Malaria causes high levels of morbidity and mortality in human beings worldwide. According to the World Health Organization (WHO), about half a million people die of this disease each year. Malaria is caused by six species of parasites belonging to the Plasmodium genus: P. falciparum, P. knowlesi, P. vivax, P. malariae, P. ovale curtisi, and P. ovale wallikeri. Currently, malaria is being kept under control with varying levels of elimination success in different countries. The development of new molecular tools as well as the use of next-generation sequencing (NGS) technologies and novel bioinformatic approaches has improved our knowledge of malarial epidemiology, diagnosis, treatment, vaccine development, and surveillance strategies. In this work, the genetics and genomics of human malarias have been analyzed. Since the first P. falciparum genome was sequenced in 2002, various population-level genetic and genomic surveys, together with transcriptomic and proteomic studies, have shown the importance of molecular approaches in supporting malaria elimination.
Collapse
|
29
|
Schleicher TR, Yang J, Freudzon M, Rembisz A, Craft S, Hamilton M, Graham M, Mlambo G, Tripathi AK, Li Y, Cresswell P, Sinnis P, Dimopoulos G, Fikrig E. A mosquito salivary gland protein partially inhibits Plasmodium sporozoite cell traversal and transmission. Nat Commun 2018; 9:2908. [PMID: 30046053 PMCID: PMC6060088 DOI: 10.1038/s41467-018-05374-3] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 06/28/2018] [Indexed: 12/24/2022] Open
Abstract
The key step during the initiation of malaria is for motile Plasmodium parasites to exit the host dermis and infect the liver. During transmission, the parasites in the form of sporozoites, are injected together with mosquito saliva into the skin. However, the contribution of vector saliva to sporozoite activity during the establishment of the initial infection of the liver is poorly understood. Here we identify a vector protein by mass spectrometry, with similarity to the human gamma interferon inducible thiol reductase (GILT), that is associated with saliva sporozoites of infected Anopheles mosquitoes and has a negative impact on the speed and cell traversal activity of Plasmodium. This protein, referred to as mosquito GILT (mosGILT) represents an example of a protein found in mosquito saliva that may negatively influence sporozoite movement in the host and could lead to new approaches to prevent malaria.
Collapse
Affiliation(s)
- Tyler R Schleicher
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, 06520, USA
| | - Jing Yang
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, 06520, USA
| | - Marianna Freudzon
- Department of Dermatology, Yale University School of Medicine, New Haven, Connecticut, 06520, USA
| | - Alison Rembisz
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, 06520, USA
| | - Samuel Craft
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, 06520, USA
| | - Madeleine Hamilton
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, 06520, USA
| | - Morven Graham
- Yale Center for Cellular and Molecular Imaging, Yale University School of Medicine, New Haven, Connecticut, 06510, USA
| | - Godfree Mlambo
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, 21205, USA
| | - Abhai K Tripathi
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, 21205, USA
| | - Yue Li
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, 06520, USA
| | - Peter Cresswell
- Department of Immunobiology, Yale University School of Medicine, New Haven, Connecticut, 06520, USA
| | - Photini Sinnis
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, 21205, USA
| | - George Dimopoulos
- Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, 21205, USA
| | - Erol Fikrig
- Section of Infectious Diseases, Department of Internal Medicine, Yale University School of Medicine, New Haven, Connecticut, 06520, USA.
- Howard Hughes Medical Institute, Chevy Chase, Maryland, 20815, USA.
| |
Collapse
|
30
|
Boucher LE, Hopp CS, Muthinja JM, Frischknecht F, Bosch J. Discovery of Plasmodium (M)TRAP-Aldolase Interaction Stabilizers Interfering with Sporozoite Motility and Invasion. ACS Infect Dis 2018; 4:620-634. [PMID: 29411968 DOI: 10.1021/acsinfecdis.7b00225] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
As obligate, intracellular parasites, Plasmodium spp. rely on invasion of host cells in order to replicate and continue their life cycle. The parasite needs to traverse the dermis and endothelium of blood vessels, invade hepatocytes and red blood cells, traverse the mosquito midgut, and enter the salivary glands to continue the cycle of infection and transmission. To traverse and invade cells, the parasite employs an actomyosin motor at the core of a larger invasion machinery complex known as the glideosome. The complex is comprised of multiple protein-protein interactions linking the motor to the internal cytoskeletal network of the parasite and to the extracellular adhesins, which directly contact the host tissue or cell surface. One key interaction is between the cytoplasmic tails of the thrombospondin related anonymous protein (TRAP) and aldolase, a bridging protein to the motor. Here, we present results from screening the Medicines for Malaria Venture (MMV) library of 400 compounds against this key protein-protein interaction. Using a surface plasmon resonance screen, we have identified several compounds that modulate the dynamics of the interaction between TRAP and aldolase. These compounds have been validated in vitro by studying their effects on sporozoite gliding motility and hepatocyte invasion. One of the MMV compounds identified reduced invasion levels by 89% at the lowest concentration tested (16 μM) and severely inhibited gliding at even lower concentrations (5 μM). By targeting protein-protein interactions, we investigated an under-explored area of parasite biology and general drug development, to identify potential antimalarial lead compounds.
Collapse
Affiliation(s)
- Lauren E. Boucher
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, Maryland 21205, United States
- Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, Maryland 21205, United States
| | - Christine S. Hopp
- Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, Maryland 21205, United States
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, Maryland 21205, United States
- Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, 5625 Fishers Lane, Rockville, Maryland 20852, United States
| | - Julianne Mendi Muthinja
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, Heidelberg University Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Jürgen Bosch
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, Maryland 21205, United States
- Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, 615 N. Wolfe Street, Baltimore, Maryland 21205, United States
- InterRayBio, LLC, 1812 Ashland Avenue, Baltimore, Maryland 21205, United States
| |
Collapse
|
31
|
Abstract
Malaria parasites require certain host nutrients for growth and survival. In this project, we examined the role of the human vitamin biotin in all stages of the malaria life cycle. We cultured blood- and liver-stage malaria parasites in the absence of biotin and found that, whereas blood-stage replication was unaffected, liver-stage parasites deprived of biotin were no longer capable of establishing a blood-stage infection. Interestingly, biotin depletion resulted in more severe developmental defects than the genetic disruption of parasite biotin metabolism. This finding suggests that host biotin metabolism also contributes to parasite development. Because neither the parasite nor the human host can synthesize biotin, parasite infectivity may be affected by the nutritional status of the host. Acetyl-CoA carboxylase (ACC) is a biotin-dependent enzyme that is the target of several classes of herbicides. Malaria parasites contain a plant-like ACC, and this is the only protein predicted to be biotinylated in the parasite. We found that ACC is expressed in the apicoplast organelle in liver- and blood-stage malaria parasites; however, it is activated through biotinylation only in the liver stages. Consistent with this observation, deletion of the biotin ligase responsible for ACC biotinylation does not impede blood-stage growth, but results in late liver-stage developmental defects. Biotin depletion increases the severity of the developmental defects, demonstrating that parasite and host biotin metabolism are required for normal liver-stage progression. This finding may link the development of liver-stage malaria parasites to the nutritional status of the host, as neither the parasite nor the human host can synthesize biotin.
Collapse
|
32
|
Garzón-Ospina D, Buitrago SP, Ramos AE, Patarroyo MA. Identifying Potential Plasmodium vivax Sporozoite Stage Vaccine Candidates: An Analysis of Genetic Diversity and Natural Selection. Front Genet 2018; 9:10. [PMID: 29422913 PMCID: PMC5788960 DOI: 10.3389/fgene.2018.00010] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2017] [Accepted: 01/09/2018] [Indexed: 11/30/2022] Open
Abstract
Parasite antigen genetic diversity represents a great obstacle when designing a vaccine against malaria caused by Plasmodium vivax. Selecting vaccine candidate antigens has been focused on those fulfilling a role in invasion and which are conserved, thus avoiding specific-allele immune responses. Most antigens described to date belong to the blood stage, thereby blocking parasite development within red blood cells, whilst studying antigens from other stages has been quite restricted. Antigens from different parasite stages are required for developing a completely effective vaccine; thus, pre-erythrocyte stage antigens able to block the first line of infection becoming established should also be taken into account. However, few antigens from this stage have been studied to date. Several P. falciparum sporozoite antigens are involved in invasion. Since 77% of genes are orthologous amongst Plasmodium parasites, P. vivax sporozoite antigen orthologs to those of P. falciparum might be present in its genome. Although these genes might have high genetic diversity, conserved functionally-relevant regions (ideal for vaccine development) could be predicted by comparing genetic diversity patterns and evolutionary rates. This study was thus aimed at searching for putative P. vivax sporozoite genes so as to analyse their genetic diversity for determining their potential as vaccine candidates. Several DNA sequence polymorphism estimators were computed at each locus. The evolutionary force (drift, selection and recombination) drawing the genetic diversity pattern observed was also determined by using tests based on polymorphism frequency spectrum as well as the type of intra- and inter-species substitutions. Likewise, recombination was assessed both indirectly and directly. The results showed that sporozoite genes were more conserved than merozoite genes evaluated to date. Putative domains implied in cell traversal, gliding motility and hepatocyte interaction had a negative selection signal, being conserved amongst different species in the genus. PvP52, PvP36, PvSPATR, PvPLP1, PvMCP1, PvTLP, PvCelTOS, and PvMB2 antigens or functionally restricted regions within them would thus seem promising vaccine candidates and could be used when designing a pre-erythrocyte and/or multi-stage vaccine against P. vivax to avoid allele-specific immune responses that could reduce vaccine efficacy.
Collapse
Affiliation(s)
- Diego Garzón-Ospina
- Molecular Biology and Immunology Laboratory, Fundación Instituto de Inmunología de Colombia, Bogotá, Colombia.,PhD Programme in Biomedical and Biological Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| | - Sindy P Buitrago
- Molecular Biology and Immunology Laboratory, Fundación Instituto de Inmunología de Colombia, Bogotá, Colombia
| | - Andrea E Ramos
- Molecular Biology and Immunology Laboratory, Fundación Instituto de Inmunología de Colombia, Bogotá, Colombia
| | - Manuel A Patarroyo
- Molecular Biology and Immunology Laboratory, Fundación Instituto de Inmunología de Colombia, Bogotá, Colombia.,Basic Sciences Department, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá, Colombia
| |
Collapse
|
33
|
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
|
34
|
Chakraborty S, Roy S, Mistry HU, Murthy S, George N, Bhandari V, Sharma P. Potential Sabotage of Host Cell Physiology by Apicomplexan Parasites for Their Survival Benefits. Front Immunol 2017; 8:1261. [PMID: 29081773 PMCID: PMC5645534 DOI: 10.3389/fimmu.2017.01261] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 09/21/2017] [Indexed: 12/26/2022] Open
Abstract
Plasmodium, Toxoplasma, Cryptosporidium, Babesia, and Theileria are the major apicomplexan parasites affecting humans or animals worldwide. These pathogens represent an excellent example of host manipulators who can overturn host signaling pathways for their survival. They infect different types of host cells and take charge of the host machinery to gain nutrients and prevent itself from host attack. The mechanisms by which these pathogens modulate the host signaling pathways are well studied for Plasmodium, Toxoplasma, Cryptosporidium, and Theileria, except for limited studies on Babesia. Theileria is a unique pathogen taking into account the way it modulates host cell transformation, resulting in its clonal expansion. These parasites majorly modulate similar host signaling pathways, however, the disease outcome and effect is different among them. In this review, we discuss the approaches of these apicomplexan to manipulate the host–parasite clearance pathways during infection, invasion, survival, and egress.
Collapse
Affiliation(s)
| | - Sonti Roy
- National Institute of Animal Biotechnology (NIAB-DBT), Hyderabad, India
| | - Hiral Uday Mistry
- National Institute of Animal Biotechnology (NIAB-DBT), Hyderabad, India
| | - Shweta Murthy
- National Institute of Animal Biotechnology (NIAB-DBT), Hyderabad, India
| | - Neena George
- National Institute of Animal Biotechnology (NIAB-DBT), Hyderabad, India
| | | | - Paresh Sharma
- National Institute of Animal Biotechnology (NIAB-DBT), Hyderabad, India
| |
Collapse
|
35
|
Protein O-fucosylation in Plasmodium falciparum ensures efficient infection of mosquito and vertebrate hosts. Nat Commun 2017; 8:561. [PMID: 28916755 PMCID: PMC5601480 DOI: 10.1038/s41467-017-00571-y] [Citation(s) in RCA: 45] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 07/11/2017] [Indexed: 01/14/2023] Open
Abstract
O-glycosylation of the Plasmodium sporozoite surface proteins CSP and TRAP was recently identified, but the role of this modification in the parasite life cycle and its relevance to vaccine design remain unclear. Here, we identify the Plasmodium protein O-fucosyltransferase (POFUT2) responsible for O-glycosylating CSP and TRAP. Genetic disruption of POFUT2 in Plasmodium falciparum results in ookinetes that are attenuated for colonizing the mosquito midgut, an essential step in malaria transmission. Some POFUT2-deficient parasites mature into salivary gland sporozoites although they are impaired for gliding motility, cell traversal, hepatocyte invasion, and production of exoerythrocytic forms in humanized chimeric liver mice. These defects can be attributed to destabilization and incorrect trafficking of proteins bearing thrombospondin repeats (TSRs). Therefore, POFUT2 plays a similar role in malaria parasites to that in metazoans: it ensures the trafficking of Plasmodium TSR proteins as part of a non-canonical glycosylation-dependent endoplasmic reticulum protein quality control mechanism. The role of O-glycosylation in the malaria life cycle is largely unknown. Here, the authors identify a Plasmodium protein O-fucosyltransferase and show that it is important for normal trafficking of a subset of surface proteins, particularly CSP and TRAP, and efficient infection of mosquito and vertebrate hosts.
Collapse
|
36
|
Gras S, Jackson A, Woods S, Pall G, Whitelaw J, Leung JM, Ward GE, Roberts CW, Meissner M. Parasites lacking the micronemal protein MIC2 are deficient in surface attachment and host cell egress, but remain virulent in vivo. Wellcome Open Res 2017. [PMID: 28630943 PMCID: PMC5473411 DOI: 10.12688/wellcomeopenres.11594.2] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background: Micronemal proteins of the thrombospondin-related anonymous protein (TRAP) family are believed to play essential roles during gliding motility and host cell invasion by apicomplexan parasites, and currently represent major vaccine candidates against
Plasmodium falciparum, the causative agent of malaria. However, recent evidence suggests that they play multiple and different roles than previously assumed. Here, we analyse a null mutant for MIC2, the TRAP homolog in
Toxoplasma gondii.
Methods: We performed a careful analysis of parasite motility in a 3D-environment, attachment under shear stress conditions, host cell invasion and
in vivo virulence.
Results: We verified the role of MIC2 in efficient surface attachment, but were unable to identify any direct function of MIC2 in sustaining gliding motility or host cell invasion once initiated. Furthermore, we find that deletion of
mic2 causes a slightly delayed infection
in vivo, leading only to mild attenuation of virulence; like with wildtype parasites, inoculation with even low numbers of
mic2 KO parasites causes lethal disease in mice. However, deletion of
mic2 causes delayed host cell egress
in vitro, possibly via disrupted signal transduction pathways.
Conclusions: We confirm a critical role of MIC2 in parasite attachment to the surface, leading to reduced parasite motility and host cell invasion. However, MIC2 appears to not be critical for gliding motility or host cell invasion, since parasite speed during these processes is unaffected. Furthermore, deletion of MIC2 leads only to slight attenuation of the parasite.
Collapse
Affiliation(s)
- Simon Gras
- Wellcome Trust Centre For Molecular Parasitology, Institute of Infection, Immunity & Inflammation, Glasgow Biomedical Research Centre, University of Glasgow, Glasgow, G12 8TA, UK
| | - Allison Jackson
- Wellcome Trust Centre For Molecular Parasitology, Institute of Infection, Immunity & Inflammation, Glasgow Biomedical Research Centre, University of Glasgow, Glasgow, G12 8TA, UK
| | - Stuart Woods
- Strathclyde Institute of Pharmacy and Biomedical Sciences, Glasgow, G4 0RE, UK
| | - Gurman Pall
- Wellcome Trust Centre For Molecular Parasitology, Institute of Infection, Immunity & Inflammation, Glasgow Biomedical Research Centre, University of Glasgow, Glasgow, G12 8TA, UK
| | - Jamie Whitelaw
- Wellcome Trust Centre For Molecular Parasitology, Institute of Infection, Immunity & Inflammation, Glasgow Biomedical Research Centre, University of Glasgow, Glasgow, G12 8TA, UK
| | - Jacqueline M Leung
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA.,Department of Microbiology and Molecular Genetics, College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Gary E Ward
- Department of Microbiology and Molecular Genetics, College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Craig W Roberts
- Strathclyde Institute of Pharmacy and Biomedical Sciences, Glasgow, G4 0RE, UK
| | - Markus Meissner
- Wellcome Trust Centre For Molecular Parasitology, Institute of Infection, Immunity & Inflammation, Glasgow Biomedical Research Centre, University of Glasgow, Glasgow, G12 8TA, UK
| |
Collapse
|
37
|
Gras S, Jackson A, Woods S, Pall G, Whitelaw J, Leung JM, Ward GE, Roberts CW, Meissner M. Parasites lacking the micronemal protein MIC2 are deficient in surface attachment and host cell egress, but remain virulent in vivo. Wellcome Open Res 2017. [PMID: 28630943 DOI: 10.12688/wellcomeopenres.11594.1] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Background: Micronemal proteins of the thrombospondin-related anonymous protein (TRAP) family are believed to play essential roles during gliding motility and host cell invasion by apicomplexan parasites, and currently represent major vaccine candidates against Plasmodium falciparum, the causative agent of malaria. However, recent evidence suggests that they play multiple and different roles than previously assumed. Here, we analyse a null mutant for MIC2, the TRAP homolog in Toxoplasma gondii. Methods: We performed a careful analysis of parasite motility in a 3D-environment, attachment under shear stress conditions, host cell invasion and in vivo virulence. Results: We verified the role of MIC2 in efficient surface attachment, but were unable to identify any direct function of MIC2 in sustaining gliding motility or host cell invasion once initiated. Furthermore, we find that deletion of mic2 causes a slightly delayed infection in vivo, leading only to mild attenuation of virulence; like with wildtype parasites, inoculation with even low numbers of mic2 KO parasites causes lethal disease in mice. However, deletion of mic2 causes delayed host cell egress in vitro, possibly via disrupted signal transduction pathways. Conclusions: We confirm a critical role of MIC2 in parasite attachment to the surface, leading to reduced parasite motility and host cell invasion. However, MIC2 appears to not be critical for gliding motility or host cell invasion, since parasite speed during these processes is unaffected. Furthermore, deletion of MIC2 leads only to slight attenuation of the parasite.
Collapse
Affiliation(s)
- Simon Gras
- Wellcome Trust Centre For Molecular Parasitology, Institute of Infection, Immunity & Inflammation, Glasgow Biomedical Research Centre, University of Glasgow, Glasgow, G12 8TA, UK
| | - Allison Jackson
- Wellcome Trust Centre For Molecular Parasitology, Institute of Infection, Immunity & Inflammation, Glasgow Biomedical Research Centre, University of Glasgow, Glasgow, G12 8TA, UK
| | - Stuart Woods
- Strathclyde Institute of Pharmacy and Biomedical Sciences, Glasgow, G4 0RE, UK
| | - Gurman Pall
- Wellcome Trust Centre For Molecular Parasitology, Institute of Infection, Immunity & Inflammation, Glasgow Biomedical Research Centre, University of Glasgow, Glasgow, G12 8TA, UK
| | - Jamie Whitelaw
- Wellcome Trust Centre For Molecular Parasitology, Institute of Infection, Immunity & Inflammation, Glasgow Biomedical Research Centre, University of Glasgow, Glasgow, G12 8TA, UK
| | - Jacqueline M Leung
- Department of Biology, Indiana University, Bloomington, IN, 47405, USA.,Department of Microbiology and Molecular Genetics, College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Gary E Ward
- Department of Microbiology and Molecular Genetics, College of Medicine, University of Vermont, Burlington, VT, 05405, USA
| | - Craig W Roberts
- Strathclyde Institute of Pharmacy and Biomedical Sciences, Glasgow, G4 0RE, UK
| | - Markus Meissner
- Wellcome Trust Centre For Molecular Parasitology, Institute of Infection, Immunity & Inflammation, Glasgow Biomedical Research Centre, University of Glasgow, Glasgow, G12 8TA, UK
| |
Collapse
|
38
|
Santos JM, Egarter S, Zuzarte-Luís V, Kumar H, Moreau CA, Kehrer J, Pinto A, da Costa M, Franke-Fayard B, Janse CJ, Frischknecht F, Mair GR. Malaria parasite LIMP protein regulates sporozoite gliding motility and infectivity in mosquito and mammalian hosts. eLife 2017; 6:e24109. [PMID: 28525314 PMCID: PMC5438254 DOI: 10.7554/elife.24109] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2016] [Accepted: 04/29/2017] [Indexed: 12/11/2022] Open
Abstract
Gliding motility allows malaria parasites to migrate and invade tissues and cells in different hosts. It requires parasite surface proteins to provide attachment to host cells and extracellular matrices. Here, we identify the Plasmodium protein LIMP (the name refers to a gliding phenotype in the sporozoite arising from epitope tagging of the endogenous protein) as a key regulator for adhesion during gliding motility in the rodent malaria model P. berghei. Transcribed in gametocytes, LIMP is translated in the ookinete from maternal mRNA, and later in the sporozoite. The absence of LIMP reduces initial mosquito infection by 50%, impedes salivary gland invasion 10-fold, and causes a complete absence of liver invasion as mutants fail to attach to host cells. GFP tagging of LIMP caused a limping defect during movement with reduced speed and transient curvature changes of the parasite. LIMP is an essential motility and invasion factor necessary for malaria transmission.
Collapse
Affiliation(s)
- Jorge M Santos
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Edifício Egas Moniz, Av. Prof. Egas Moniz, Lisbon, Portugal
| | - Saskia Egarter
- Parasitology, Department of Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
| | - Vanessa Zuzarte-Luís
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Edifício Egas Moniz, Av. Prof. Egas Moniz, Lisbon, Portugal
| | - Hirdesh Kumar
- Parasitology, Department of Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
- Kusuma School of Biological Sciences, Indian Institute of Technology Delhi, New Delhi, India
| | - Catherine A Moreau
- Parasitology, Department of Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
| | - Jessica Kehrer
- Parasitology, Department of Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
| | - Andreia Pinto
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Edifício Egas Moniz, Av. Prof. Egas Moniz, Lisbon, Portugal
| | - Mário da Costa
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Edifício Egas Moniz, Av. Prof. Egas Moniz, Lisbon, Portugal
| | - 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
| | - Friedrich Frischknecht
- Parasitology, Department of Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
| | - Gunnar R Mair
- Instituto de Medicina Molecular, Faculdade de Medicina da Universidade de Lisboa, Edifício Egas Moniz, Av. Prof. Egas Moniz, Lisbon, Portugal
- Parasitology, Department of Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
| |
Collapse
|
39
|
Frischknecht F, Matuschewski K. Plasmodium Sporozoite Biology. Cold Spring Harb Perspect Med 2017; 7:cshperspect.a025478. [PMID: 28108531 DOI: 10.1101/cshperspect.a025478] [Citation(s) in RCA: 54] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Plasmodium sporozoite transmission is a critical population bottleneck in parasite life-cycle progression and, hence, a target for prophylactic drugs and vaccines. The recent progress of a candidate antisporozoite subunit vaccine formulation to licensure highlights the importance of sporozoite transmission intervention in the malaria control portfolio. Sporozoites colonize mosquito salivary glands, migrate through the skin, penetrate blood vessels, breach the liver sinusoid, and invade hepatocytes. Understanding the molecular and cellular mechanisms that mediate the remarkable sporozoite journey in the invertebrate vector and the vertebrate host can inform evidence-based next-generation drug development programs and immune intervention strategies.
Collapse
Affiliation(s)
- Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, 69120 Heidelberg, Germany
| | - Kai Matuschewski
- Department of Molecular Parasitology, Institute of Biology, Humboldt University Berlin, 10115 Berlin, Germany
| |
Collapse
|
40
|
Jimah JR, Salinas ND, Sala-Rabanal M, Jones NG, Sibley LD, Nichols CG, Schlesinger PH, Tolia NH. Malaria parasite CelTOS targets the inner leaflet of cell membranes for pore-dependent disruption. eLife 2016; 5. [PMID: 27906127 PMCID: PMC5132341 DOI: 10.7554/elife.20621] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2016] [Accepted: 11/14/2016] [Indexed: 01/02/2023] Open
Abstract
Apicomplexan parasites contain a conserved protein CelTOS that, in malaria parasites, is essential for traversal of cells within the mammalian host and arthropod vector. However, the molecular role of CelTOS is unknown because it lacks sequence similarity to proteins of known function. Here, we determined the crystal structure of CelTOS and discovered CelTOS resembles proteins that bind to and disrupt membranes. In contrast to known membrane disruptors, CelTOS has a distinct architecture, specifically binds phosphatidic acid commonly present within the inner leaflet of plasma membranes, and potently disrupts liposomes composed of phosphatidic acid by forming pores. Microinjection of CelTOS into cells resulted in observable membrane damage. Therefore, CelTOS is unique as it achieves nearly universal inner leaflet cellular activity to enable the exit of parasites from cells during traversal. By providing novel molecular insight into cell traversal by apicomplexan parasites, our work facilitates the design of therapeutics against global pathogens.
Collapse
Affiliation(s)
- John R Jimah
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, United States
| | - Nichole D Salinas
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, United States
| | - Monica Sala-Rabanal
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, United States.,Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, United States
| | - Nathaniel G Jones
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, United States
| | - L David Sibley
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, United States
| | - Colin G Nichols
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, United States.,Center for the Investigation of Membrane Excitability Diseases, Washington University School of Medicine, Saint Louis, United States
| | - Paul H Schlesinger
- Department of Cell Biology and Physiology, Washington University School of Medicine, Saint Louis, United States
| | - Niraj H Tolia
- Department of Molecular Microbiology, Washington University School of Medicine, Saint Louis, United States.,Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, Saint Louis, United States
| |
Collapse
|
41
|
Yang ASP, Boddey JA. Molecular mechanisms of host cell traversal by malaria sporozoites. Int J Parasitol 2016; 47:129-136. [PMID: 27825827 DOI: 10.1016/j.ijpara.2016.09.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Revised: 07/22/2016] [Accepted: 09/05/2016] [Indexed: 11/19/2022]
Abstract
Malaria is a pernicious infectious disease caused by apicomplexan parasites of the genus Plasmodium. Each year, malaria afflicts over 200million people, causing considerable morbidity, loss to gross domestic product of endemic countries, and more than 420,000 deaths. A central feature of the virulence of malaria parasites is the ability of sporozoite forms injected by a mosquito to navigate from the inoculation site in the skin through host tissues to infect the liver. The ability for sporozoites to traverse through different host cell types is very important for the successful development of parasites within the mammalian host. Over the past decade, our understanding of the role of host cell traversal has become clearer through important studies with rodent models of malaria. However, we still do not understand the stepwise process of host cell entry and exit or know the molecular mechanisms governing each step. We know even less about cell traversal by malaria parasite species that infect humans. Here, we review current knowledge regarding the role and molecular mechanisms of sporozoite cell traversal and highlight recent advances that prompt new ways of thinking about this important process.
Collapse
Affiliation(s)
- Annie S P Yang
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia
| | - Justin A Boddey
- The Walter and Eliza Hall Institute of Medical Research, Parkville, VIC 3052, Australia; Department of Medical Biology, University of Melbourne, Parkville, VIC 3052, Australia.
| |
Collapse
|
42
|
Govindasamy K, Jebiwott S, Jaijyan DK, Davidow A, Ojo KK, Van Voorhis WC, Brochet M, Billker O, Bhanot P. Invasion of hepatocytes by Plasmodium sporozoites requires cGMP-dependent protein kinase and calcium dependent protein kinase 4. Mol Microbiol 2016; 102:349-363. [PMID: 27425827 DOI: 10.1111/mmi.13466] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/17/2016] [Indexed: 01/24/2023]
Abstract
Invasion of hepatocytes by sporozoites is essential for Plasmodium to initiate infection of the mammalian host. The parasite's subsequent intracellular differentiation in the liver is the first developmental step of its mammalian cycle. Despite their biological significance, surprisingly little is known of the signalling pathways required for sporozoite invasion. We report that sporozoite invasion of hepatocytes requires signalling through two second-messengers - cGMP mediated by the parasite's cGMP-dependent protein kinase (PKG), and Ca2+ , mediated by the parasite's calcium-dependent protein kinase 4 (CDPK4). Sporozoites expressing a mutated form of Plasmodium berghei PKG or carrying a deletion of the CDPK4 gene are defective in invasion of hepatocytes. Using specific and potent inhibitors of Plasmodium PKG and CDPK4, we demonstrate that PKG and CDPK4 are required for sporozoite motility, and that PKG regulates the secretion of TRAP, an adhesin that is essential for motility. Chemical inhibition of PKG decreases parasite egress from hepatocytes by inhibiting either the formation or release of merosomes. In contrast, genetic inhibition of CDPK4 does not significantly decrease the number of merosomes. By revealing the requirement for PKG and CDPK4 in Plasmodium sporozoite invasion, our work enables a better understanding of kinase pathways that act in different Plasmodium stages.
Collapse
Affiliation(s)
- K Govindasamy
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers - New Jersey Medical School, Newark, NJ, USA
| | - S Jebiwott
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers - New Jersey Medical School, Newark, NJ, USA
| | - D K Jaijyan
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers - New Jersey Medical School, Newark, NJ, USA
| | - A Davidow
- Department of Biostatistics, Rutgers - School of Public Health, Newark, NJ, USA
| | - K K Ojo
- Division of Allergy and Infectious Diseases, Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - W C Van Voorhis
- Division of Allergy and Infectious Diseases, Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - M Brochet
- CNRS UMR5235 -Université Montpellier 2, 34095, Montpellier, France.,University of Geneva, Faculty of Medicine, Department of Microbiology and Molecular Medicine, Switzerland
| | - O Billker
- Wellcome Trust Sanger Institute, Malaria Programme, CB10 1SA, Hinxton, UK
| | - P Bhanot
- Department of Microbiology, Biochemistry and Molecular Genetics, Rutgers - New Jersey Medical School, Newark, NJ, USA.
| |
Collapse
|
43
|
Moreira CK, Naissant B, Coppi A, Bennett BL, Aime E, Franke-Fayard B, Janse CJ, Coppens I, Sinnis P, Templeton TJ. The Plasmodium PHIST and RESA-Like Protein Families of Human and Rodent Malaria Parasites. PLoS One 2016; 11:e0152510. [PMID: 27022937 PMCID: PMC4811531 DOI: 10.1371/journal.pone.0152510] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2014] [Accepted: 03/15/2016] [Indexed: 11/19/2022] Open
Abstract
The phist gene family has members identified across the Plasmodium genus, defined by the presence of a domain of roughly 150 amino acids having conserved aromatic residues and an all alpha-helical structure. The family is highly amplified in P. falciparum, with 65 predicted genes in the genome of the 3D7 isolate. In contrast, in the rodent malaria parasite P. berghei 3 genes are identified, one of which is an apparent pseudogene. Transcripts of the P. berghei phist genes are predominant in schizonts, whereas in P. falciparum transcript profiles span different asexual blood stages and gametocytes. We pursued targeted disruption of P. berghei phist genes in order to characterize a simplistic model for the expanded phist gene repertoire in P. falciparum. Unsuccessful attempts to disrupt P. berghei PBANKA_114540 suggest that this phist gene is essential, while knockout of phist PBANKA_122900 shows an apparent normal progression and non-essential function throughout the life cycle. Epitope-tagging of P. falciparum and P. berghei phist genes confirmed protein export to the erythrocyte cytoplasm and localization with a punctate pattern. Three P. berghei PEXEL/HT-positive exported proteins exhibit at least partial co-localization, in support of a common vesicular compartment in the cytoplasm of erythrocytes infected with rodent malaria parasites.
Collapse
Affiliation(s)
- Cristina K. Moreira
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10021, United States of America
| | - Bernina Naissant
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10021, United States of America
| | - Alida Coppi
- Department of Medical Parasitology, NYU School of Medicine, New York, NY, 10010, United States of America
| | - Brandy L. Bennett
- Department of Medical Parasitology, NYU School of Medicine, New York, NY, 10010, United States of America
| | - Elena Aime
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Blandine Franke-Fayard
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Chris J. Janse
- Leiden Malaria Research Group, Department of Parasitology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Isabelle Coppens
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, United States of America
| | - Photini Sinnis
- Department of Medical Parasitology, NYU School of Medicine, New York, NY, 10010, United States of America
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, United States of America
| | - Thomas J. Templeton
- Department of Microbiology and Immunology, Weill Cornell Medical College, New York, NY 10021, United States of America
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN), Nagasaki 852-8523, Japan
- * E-mail:
| |
Collapse
|
44
|
Quadt KA, Streichfuss M, Moreau CA, Spatz JP, Frischknecht F. Coupling of Retrograde Flow to Force Production During Malaria Parasite Migration. ACS NANO 2016; 10:2091-2102. [PMID: 26792112 DOI: 10.1021/acsnano.5b06417] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
Migration of malaria parasites is powered by a myosin motor that moves actin filaments, which in turn link to adhesive proteins spanning the plasma membrane. The retrograde flow of these adhesins appears to be coupled to forward locomotion. However, the contact dynamics between the parasite and the substrate as well as the generation of forces are complex and their relation to retrograde flow is unclear. Using optical tweezers we found retrograde flow rates up to 15 μm/s contrasting with parasite average speeds of 1-2 μm/s. We found that a surface protein, TLP, functions in reducing retrograde flow for the buildup of adhesive force and that actin dynamics appear optimized for the generation of force but not for maximizing the speed of retrograde flow. These data uncover that TLP acts by modulating actin dynamics or actin filament organization and couples retrograde flow to force production in malaria parasites.
Collapse
Affiliation(s)
- Katharina A Quadt
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School , Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Martin Streichfuss
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School , Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
- University of Heidelberg , Department of Biophysical Chemistry and Max Planck Institute for Intelligent Systems, Department of New Materials and Biosystems, Heisenbergstr. 3, 70569 Stuttgart, Germany
| | - Catherine A Moreau
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School , Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Joachim P Spatz
- University of Heidelberg , Department of Biophysical Chemistry and Max Planck Institute for Intelligent Systems, Department of New Materials and Biosystems, Heisenbergstr. 3, 70569 Stuttgart, Germany
| | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School , Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| |
Collapse
|
45
|
Risco-Castillo V, Topçu S, Marinach C, Manzoni G, Bigorgne A, Briquet S, Baudin X, Lebrun M, Dubremetz JF, Silvie O. Malaria Sporozoites Traverse Host Cells within Transient Vacuoles. Cell Host Microbe 2015; 18:593-603. [DOI: 10.1016/j.chom.2015.10.006] [Citation(s) in RCA: 89] [Impact Index Per Article: 9.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Revised: 08/31/2015] [Accepted: 10/02/2015] [Indexed: 12/28/2022]
|
46
|
Gliding motility in apicomplexan parasites. Semin Cell Dev Biol 2015; 46:135-42. [DOI: 10.1016/j.semcdb.2015.09.020] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2015] [Accepted: 09/25/2015] [Indexed: 11/22/2022]
|
47
|
Nemetski SM, Cardozo TJ, Bosch G, Weltzer R, O'Malley K, Ejigiri I, Kumar KA, Buscaglia CA, Nussenzweig V, Sinnis P, Levitskaya J, Bosch J. Inhibition by stabilization: targeting the Plasmodium falciparum aldolase-TRAP complex. Malar J 2015; 14:324. [PMID: 26289816 PMCID: PMC4545932 DOI: 10.1186/s12936-015-0834-9] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2015] [Accepted: 08/02/2015] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Emerging resistance of the malaria parasite Plasmodium to current therapies underscores the critical importance of exploring novel strategies for disease eradication. Plasmodium species are obligate intracellular protozoan parasites. They rely on an unusual form of substrate-dependent motility for their migration on and across host-cell membranes and for host cell invasion. This peculiar motility mechanism is driven by the 'glideosome', an actin-myosin associated, macromolecular complex anchored to the inner membrane complex of the parasite. Myosin A, actin, aldolase, and thrombospondin-related anonymous protein (TRAP) constitute the molecular core of the glideosome in the sporozoite, the mosquito stage that brings the infection into mammals. METHODS Virtual library screening of a large compound library against the PfAldolase-TRAP complex was used to identify candidate compounds that stabilize and prevent the disassembly of the glideosome. The mechanism of these compounds was confirmed by biochemical, biophysical and parasitological methods. RESULTS A novel inhibitory effect on the parasite was achieved by stabilizing a protein-protein interaction within the glideosome components. Compound 24 disrupts the gliding and invasive capabilities of Plasmodium parasites in in vitro parasite assays. A high-resolution, ternary X-ray crystal structure of PfAldolase-TRAP in complex with compound 24 confirms the mode of interaction and serves as a platform for future ligand optimization. CONCLUSION This proof-of-concept study presents a novel approach to anti-malarial drug discovery and design. By strengthening a protein-protein interaction within the parasite, an avenue towards inhibiting a previously "undruggable" target is revealed and the motility motor responsible for successful invasion of host cells is rendered inactive. This study provides new insights into the malaria parasite cell invasion machinery and convincingly demonstrates that liver cell invasion is dramatically reduced by 95 % in the presence of the small molecule stabilizer compound 24.
Collapse
Affiliation(s)
- Sondra Maureen Nemetski
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, USA. .,Department of Pediatrics, Phyllis and David Komansky Center for Children's Health, New York-Presbyterian Hospital-Weill Cornell Medical College, New York, USA.
| | - Timothy J Cardozo
- Department of Biochemistry and Molecular Pharmacology, New York University School of Medicine, New York, USA. .,Institute for Systems Genetics, New York University School of Medicine, New York, USA.
| | - Gundula Bosch
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, USA. .,Johns Hopkins Malaria Research Institute (JHMRI), Baltimore, USA.
| | - Ryan Weltzer
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, USA. .,Johns Hopkins Malaria Research Institute (JHMRI), Baltimore, USA.
| | - Kevin O'Malley
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, USA. .,Johns Hopkins Malaria Research Institute (JHMRI), Baltimore, USA.
| | - Ijeoma Ejigiri
- Department of Medical Parasitology, New York University School of Medicine, New York, USA.
| | - Kota Arun Kumar
- Michael Heidelberg Division of Pathology of Infectious Diseases, Department of Pathology, New York University School of Medicine, New York, USA. .,Department of Animal Sciences, School of Life Sciences, University of Hyderabad, Hyderabad, 500046, India.
| | - Carlos A Buscaglia
- Instituto de Investigaciones Biotecnológicas-Instituto Tecnológico de Chascomús (IIB-INTECH), Universidad Nacional de General San Martín-CONICET, 1650, San Martín, Buenos Aires, Argentina.
| | - Victor Nussenzweig
- Michael Heidelberg Division of Pathology of Infectious Diseases, Department of Pathology, New York University School of Medicine, New York, USA.
| | - Photini Sinnis
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, USA. .,Department of Medical Parasitology, New York University School of Medicine, New York, USA. .,Johns Hopkins Malaria Research Institute (JHMRI), Baltimore, USA.
| | - Jelena Levitskaya
- Department of Molecular Microbiology and Immunology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, USA. .,Johns Hopkins Malaria Research Institute (JHMRI), Baltimore, USA.
| | - Jürgen Bosch
- Department of Biochemistry and Molecular Biology, Johns Hopkins University Bloomberg School of Public Health, Baltimore, USA. .,Johns Hopkins Malaria Research Institute (JHMRI), Baltimore, USA.
| |
Collapse
|
48
|
Douglas RG, Amino R, Sinnis P, Frischknecht F. Active migration and passive transport of malaria parasites. Trends Parasitol 2015; 31:357-62. [PMID: 26001482 DOI: 10.1016/j.pt.2015.04.010] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2015] [Revised: 04/22/2015] [Accepted: 04/23/2015] [Indexed: 11/16/2022]
Abstract
Malaria parasites undergo a complex life cycle between their hosts and vectors. During this cycle the parasites invade different types of cells, migrate across barriers, and transfer from one host to another. Recent literature hints at a misunderstanding of the difference between active, parasite-driven migration and passive, circulation-driven movement of the parasite or parasite-infected cells in the various bodily fluids of mosquito and mammalian hosts. Because both active migration and passive transport could be targeted in different ways to interfere with the parasite, a distinction between the two ways the parasite uses to get from one location to another is essential. We discuss the two types of motion needed for parasite dissemination and elaborate on how they could be targeted by future vaccines or drugs.
Collapse
Affiliation(s)
- Ross G Douglas
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany
| | - Rogerio Amino
- Unité de Biologie et Génétique du Paludisme, Département Parasites et Insectes Vecteurs, Institut Pasteur, 25-28 Rue du Dr Roux, 75015 Paris, France
| | - Photini Sinnis
- Johns Hopkins Malaria Research Institute, Department of Molecular Microbiology and Immunology, Bloomberg School of Public Health, 615 North Wolfe Street, Baltimore, MD 21205, USA
| | - Freddy Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Im Neuenheimer Feld 324, 69120 Heidelberg, Germany.
| |
Collapse
|
49
|
Boucher LE, Bosch J. The apicomplexan glideosome and adhesins - Structures and function. J Struct Biol 2015; 190:93-114. [PMID: 25764948 PMCID: PMC4417069 DOI: 10.1016/j.jsb.2015.02.008] [Citation(s) in RCA: 71] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2014] [Revised: 02/20/2015] [Accepted: 02/26/2015] [Indexed: 01/10/2023]
Abstract
The apicomplexan family of pathogens, which includes Plasmodium spp. and Toxoplasma gondii, are primarily obligate intracellular parasites and invade multiple cell types. These parasites express extracellular membrane protein receptors, adhesins, to form specific pathogen-host cell interaction complexes. Various adhesins are used to invade a variety of cell types. The receptors are linked to an actomyosin motor, which is part of a complex comprised of many proteins known as the invasion machinery or glideosome. To date, reviews on invasion have focused primarily on the molecular pathways and signals of invasion, with little or no structural information presented. Over 75 structures of parasite receptors and glideosome proteins have been deposited with the Protein Data Bank. These structures include adhesins, motor proteins, bridging proteins, inner membrane complex and cytoskeletal proteins, as well as co-crystal structures with peptides and antibodies. These structures provide information regarding key interactions necessary for target receptor engagement, machinery complex formation, how force is transmitted, and the basis of inhibitory antibodies. Additionally, these structures can provide starting points for the development of antibodies and inhibitory molecules targeting protein-protein interactions, with the aim to inhibit invasion. This review provides an overview of the parasite adhesin protein families, the glideosome components, glideosome architecture, and discuss recent work regarding alternative models.
Collapse
Affiliation(s)
- Lauren E Boucher
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD 21205, USA; Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD 21205, USA.
| | - Jürgen Bosch
- Department of Biochemistry and Molecular Biology, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD 21205, USA; Johns Hopkins Malaria Research Institute, Johns Hopkins Bloomberg School of Public Health, 615 N Wolfe St, Baltimore, MD 21205, USA.
| |
Collapse
|
50
|
Karkowska-Kuleta J, Kozik A. Moonlighting proteins as virulence factors of pathogenic fungi, parasitic protozoa and multicellular parasites. Mol Oral Microbiol 2014; 29:270-83. [PMID: 25131723 DOI: 10.1111/omi.12078] [Citation(s) in RCA: 50] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/31/2014] [Indexed: 01/03/2023]
Abstract
The delicate balance between eukaryotic pathogens and their human hosts during the initiation and development of infection is a complex process involving many diverse interactions. Different infectious agents, including pathogenic fungi, parasitic protozoa and multicellular parasites, directly interact through their cell surface with epithelial or endothelial cells of the human host as well as various proteinaceous host ligands such as extracellular matrix or plasma proteins. Eukaryotic pathogens possess a number of virulence factors but a relatively recently recognized and particularly interesting group of factors capable of enhancing virulence is the set of so-called 'moonlighting proteins'. This term was coined for a relatively large collection of housekeeping enzymes lacking special targeting motifs that would determine their extracellular localization, but that are often present at the cell surface of pathogen. Several such enzymes with key metabolic functions in glycolysis, the pentose phosphate cycle or other fundamental intracellular processes perform entirely new, non-catalytic roles often associated with adhesion to host ligands. Our current study summarizes some of the current knowledge of interesting moonlighting proteins which play putative or confirmed roles as virulence factors in pathogenic fungi, parasitic protozoa and multicellular parasites.
Collapse
Affiliation(s)
- J Karkowska-Kuleta
- Department of Analytical Biochemistry, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | | |
Collapse
|