51
|
Düsterhöft S, Künzel U, Freeman M. Rhomboid proteases in human disease: Mechanisms and future prospects. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2017; 1864:2200-2209. [PMID: 28460881 DOI: 10.1016/j.bbamcr.2017.04.016] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/23/2017] [Revised: 04/24/2017] [Accepted: 04/26/2017] [Indexed: 01/19/2023]
Abstract
Rhomboids are intramembrane serine proteases that cleave the transmembrane helices of substrate proteins, typically releasing luminal/extracellular domains from the membrane. They are conserved in all branches of life and there is a growing recognition of their association with a wide range of human diseases. Human rhomboids, for example, have been implicated in cancer, metabolic disease and neurodegeneration, while rhomboids in apicomplexan parasites appear to contribute to their invasion of host cells. Recent advances in our knowledge of the structure and the enzyme function of rhomboids, and increasing efforts to identify specific inhibitors, are beginning to provide important insight into the prospect of rhomboids becoming future therapeutic targets. This article is part of a Special Issue entitled: Proteolysis as a Regulatory Event in Pathophysiology edited by Stefan Rose-John.
Collapse
Affiliation(s)
- Stefan Düsterhöft
- Dunn School of Pathology, University of Oxford, OX1 3RE, United Kingdom
| | - Ulrike Künzel
- Dunn School of Pathology, University of Oxford, OX1 3RE, United Kingdom
| | - Matthew Freeman
- Dunn School of Pathology, University of Oxford, OX1 3RE, United Kingdom.
| |
Collapse
|
52
|
Koussis K, Goulielmaki E, Chalari A, Withers-Martinez C, Siden-Kiamos I, Matuschewski K, Loukeris TG. Targeted Deletion of a Plasmodium Site-2 Protease Impairs Life Cycle Progression in the Mammalian Host. PLoS One 2017; 12:e0170260. [PMID: 28107409 PMCID: PMC5249076 DOI: 10.1371/journal.pone.0170260] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2016] [Accepted: 01/01/2017] [Indexed: 12/26/2022] Open
Abstract
Site-2 proteases (S2P) belong to the M50 family of metalloproteases, which typically perform essential roles by mediating activation of membrane–bound transcription factors through regulated intramembrane proteolysis (RIP). Protease-dependent liberation of dormant transcription factors triggers diverse cellular responses, such as sterol regulation, Notch signalling and the unfolded protein response. Plasmodium parasites rely on regulated proteolysis for controlling essential pathways throughout the life cycle. In this study we examine the Plasmodium-encoded S2P in a murine malaria model and show that it is expressed in all stages of Plasmodium development. Localisation studies by endogenous gene tagging revealed that in all invasive stages the protein is in close proximity to the nucleus. Ablation of PbS2P by reverse genetics leads to reduced growth rates during liver and blood infection and, hence, virulence attenuation. Strikingly, absence of PbS2P was compatible with parasite life cycle progression in the mosquito and mammalian hosts under physiological conditions, suggesting redundant or dispensable roles in vivo.
Collapse
Affiliation(s)
- Konstantinos Koussis
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
- Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
- Malaria Biochemistry Laboratory, The Francis Crick Institute, London, United Kingdom
- * E-mail:
| | - Evi Goulielmaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | - Anna Chalari
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
- Department of Biology, University of Crete, Heraklion, Greece
| | | | - Inga Siden-Kiamos
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | - Kai Matuschewski
- Parasitology Unit, Max Planck Institute for Infection Biology, Berlin, Germany
- Institute of Biology, Humboldt University, Berlin, Germany
| | - Thanasis G. Loukeris
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| |
Collapse
|
53
|
Ashano E, Isewon I, Oyelade J, Adebiyi E. Cluster analysis of Plasmodium RNA-seq time-course data identifies stage-specific co-regulated biological processes and regulatory elements. F1000Res 2016; 5. [PMID: 27990252 PMCID: PMC5155496 DOI: 10.12688/f1000research.9093.1] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/29/2016] [Indexed: 12/26/2022] Open
Abstract
In this study, we interpreted RNA-seq time-course data of three developmental stages of Plasmodium species by clustering genes based on similarities in their expression profile without prior knowledge of the gene function. Functional enrichment of clusters of upregulated genes at specific time-points reveals potential targetable biological processes with information on their timings. We identified common consensus sequences that these clusters shared as potential points of coordinated transcriptional control. Five cluster groups showed upregulated profile patterns of biological interest. This included two clusters from the Intraerythrocytic Developmental Cycle (cluster 4 = 16 genes, and cluster 9 = 32 genes), one from the sexual development stage (cluster 2 = 851 genes), and two from the gamete-fertilization stage in the mosquito host (cluster 4 = 153 genes, and cluster 9 = 258 genes). The IDC expressed the least numbers of genes with only 1448 genes showing any significant activity of the 5020 genes (~29%) in the experiment. Gene ontology (GO) enrichment analysis of these clusters revealed a total of 671 uncharacterized genes implicated in 14 biological processes and components associated with these stages, some of which are currently being investigated as drug targets in on-going research. Five putative transcription regulatory binding motifs shared by members of each cluster were also identified, one of which was also identified in a previous study by separate researchers. Our study shows stage-specific genes and biological processes that may be important in antimalarial drug research efforts. In addition, timed-coordinated control of separate processes may explain the paucity of factors in parasites.
Collapse
Affiliation(s)
- Efejiro Ashano
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota, Ogun State, 110001, Nigeria
| | - Itunuoluwa Isewon
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota, Ogun State, 110001, Nigeria.,Department of Computer and Information Sciences, Covenant University, Ota, Ogun State, 110001, Nigeria
| | - Jelili Oyelade
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota, Ogun State, 110001, Nigeria.,Department of Computer and Information Sciences, Covenant University, Ota, Ogun State, 110001, Nigeria
| | - Ezekiel Adebiyi
- Covenant University Bioinformatics Research (CUBRe), Covenant University, Ota, Ogun State, 110001, Nigeria.,Department of Computer and Information Sciences, Covenant University, Ota, Ogun State, 110001, Nigeria.,Division of Applied Bioinformatics, German Cancer Research Center (DKFZ), Heidelberg,, 69120, Germany
| |
Collapse
|
54
|
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: 1.6] [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
|
55
|
The mechanism of force transmission at bacterial focal adhesion complexes. Nature 2016; 539:530-535. [PMID: 27749817 DOI: 10.1038/nature20121] [Citation(s) in RCA: 96] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2016] [Accepted: 09/28/2016] [Indexed: 12/14/2022]
Abstract
Various rod-shaped bacteria mysteriously glide on surfaces in the absence of appendages such as flagella or pili. In the deltaproteobacterium Myxococcus xanthus, a putative gliding motility machinery (the Agl-Glt complex) localizes to so-called focal adhesion sites (FASs) that form stationary contact points with the underlying surface. Here we show that the Agl-Glt machinery contains an inner-membrane motor complex that moves intracellularly along a right-handed helical path; when the machinery becomes stationary at FASs, the motor complex powers a left-handed rotation of the cell around its long axis. At FASs, force transmission requires cyclic interactions between the molecular motor and the adhesion proteins of the outer membrane via a periplasmic interaction platform, which presumably involves contractile activity of motor components and possible interactions with peptidoglycan. Our results provide a molecular model of bacterial gliding motility.
Collapse
|
56
|
Dogga SK, Soldati-Favre D. Biology of rhomboid proteases in infectious diseases. Semin Cell Dev Biol 2016; 60:38-45. [PMID: 27567708 DOI: 10.1016/j.semcdb.2016.08.020] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2016] [Revised: 08/04/2016] [Accepted: 08/23/2016] [Indexed: 12/26/2022]
Abstract
Rhomboids are a well-conserved class of intramembrane serine proteases found in all kingdoms of life, sharing a conserved core structure of at least six transmembrane (TM) domains that contain the catalytic serine-histidine dyad. The rhomboid proteases, which cleave membrane embedded substrates within their TM domains, are emerging as an important group of enzymes controlling a myriad of biological processes. These enzymes are found in a wide variety of pathogens manifesting important roles in their pathological processes. Accordingly, they have received considerable attention as potential targets for pharmacological intervention over the past few years. This review provides a general update on rhomboid proteases and their roles in pathogenesis of human infectious agents.
Collapse
Affiliation(s)
- Sunil Kumar Dogga
- Department of Microbiology and Molecular Medicine, University of Geneva, CMU, 1 Rue Michel-Servet, CH-1211 Geneva 4, Switzerland.
| | - Dominique Soldati-Favre
- Department of Microbiology and Molecular Medicine, University of Geneva, CMU, 1 Rue Michel-Servet, CH-1211 Geneva 4, Switzerland
| |
Collapse
|
57
|
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: 54] [Impact Index Per Article: 6.0] [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
|
58
|
Kehrer J, Singer M, Lemgruber L, Silva PAGC, Frischknecht F, Mair GR. A Putative Small Solute Transporter Is Responsible for the Secretion of G377 and TRAP-Containing Secretory Vesicles during Plasmodium Gamete Egress and Sporozoite Motility. PLoS Pathog 2016; 12:e1005734. [PMID: 27427910 PMCID: PMC4948853 DOI: 10.1371/journal.ppat.1005734] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2016] [Accepted: 06/08/2016] [Indexed: 11/18/2022] Open
Abstract
Regulated protein secretion is required for malaria parasite life cycle progression and transmission between the mammalian host and mosquito vector. During transmission from the host to the vector, exocytosis of highly specialised secretory vesicles, such as osmiophilic bodies, is key to the dissolution of the red blood cell and parasitophorous vacuole membranes enabling gamete egress. The positioning of adhesins from the TRAP family, from micronemes to the sporozoite surface, is essential for gliding motility of the parasite and transmission from mosquito to mammalian host. Here we identify a conserved role for the putative pantothenate transporter PAT in Plasmodium berghei in vesicle fusion of two distinct classes of vesicles in gametocytes and sporozoites. PAT is a membrane component of osmiophilic bodies in gametocytes and micronemes in sporozoites. Despite normal formation and trafficking of osmiophilic bodies to the cell surface upon activation, PAT-deficient gametes fail to discharge their contents, remain intraerythrocytic and unavailable for fertilisation and further development in the mosquito. Sporozoites lacking PAT fail to secrete TRAP, are immotile and thus unable to infect the subsequent rodent host. Thus, P. berghei PAT appears to regulate exocytosis in two distinct populations of vesicles in two different life cycle forms rather than acting as pantothenic transporter during parasite transmission. Transmission of the malaria parasite between mosquito and host requires two different life cycle stages—the gametocyte and the sporozoite. In both parasite forms, transmission is dependent on exocytosis of stage-specific vesicles. In gametocytes these vesicles release proteins allowing egress from red blood cells and fertilization, and are hence needed to establish an infection in the mosquito. In contrast, proteins are secreted into the membrane of the sporozoite, where they play distinct roles during adhesion and motility, both crucial for transmission back into the mammalian host. Here we show that parasites lacking the putative small solute transporter PAT are still able to form vesicles in both parasite forms but are unable to fuse and secrete their contents. This results in impaired parasite transmission into and from the mosquito. Our work shows that a single protein can regulate the function of functionally distinct classes of vesicles in different life cycle forms of a parasite.
Collapse
Affiliation(s)
- Jessica Kehrer
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
| | - Mirko Singer
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
| | - Leandro Lemgruber
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
| | | | - Friedrich Frischknecht
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
- * E-mail: (FF); ; (GRM)
| | - Gunnar R. Mair
- Integrative Parasitology, Center for Infectious Diseases, University of Heidelberg Medical School, Heidelberg, Germany
- Instituto Medicina Molecular, Lisbon, Portugal
- * E-mail: (FF); ; (GRM)
| |
Collapse
|
59
|
Hopp CS, Balaban AE, Bushell ESC, Billker O, Rayner JC, Sinnis P. Palmitoyl transferases have critical roles in the development of mosquito and liver stages of Plasmodium. Cell Microbiol 2016; 18:1625-1641. [PMID: 27084458 DOI: 10.1111/cmi.12601] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2016] [Revised: 03/24/2016] [Accepted: 04/01/2016] [Indexed: 12/24/2022]
Abstract
As the Plasmodium parasite transitions between mammalian and mosquito host, it has to adjust quickly to new environments. Palmitoylation, a reversible and dynamic lipid post-translational modification, plays a central role in regulating this process and has been implicated with functions for parasite morphology, motility and host cell invasion. While proteins associated with the gliding motility machinery have been described to be palmitoylated, no palmitoyl transferase responsible for regulating gliding motility has previously been identified. Here, we characterize two palmityol transferases with gene tagging and gene deletion approaches. We identify DHHC3, a palmitoyl transferase, as a mediator of ookinete development, with a crucial role for gliding motility in ookinetes and sporozoites, and we co-localize the protein with a marker for the inner membrane complex in the ookinete stage. Ookinetes and sporozoites lacking DHHC3 are impaired in gliding motility and exhibit a strong phenotype in vivo; with ookinetes being significantly less infectious to their mosquito host and sporozoites being non-infectious to mice. Importantly, genetic complementation of the DHHC3-ko parasite completely restored virulence. We generated parasites lacking both DHHC3, as well as the palmitoyl transferase DHHC9, and found an enhanced phenotype for these double knockout parasites, allowing insights into the functional overlap and compensational nature of the large family of PbDHHCs. These findings contribute to our understanding of the organization and mechanism of the gliding motility machinery, which as is becoming increasingly clear, is mediated by palmitoylation.
Collapse
Affiliation(s)
- Christine S Hopp
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA. .,Wellcome Trust Sanger Institute, Hinxton, CB10 1SA, UK.
| | - Amanda E Balaban
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA
| | | | | | | | - Photini Sinnis
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, 21205, USA.
| |
Collapse
|
60
|
Swearingen KE, Lindner SE, Shi L, Shears MJ, Harupa A, Hopp CS, Vaughan AM, Springer TA, Moritz RL, Kappe SHI, Sinnis P. Interrogating the Plasmodium Sporozoite Surface: Identification of Surface-Exposed Proteins and Demonstration of Glycosylation on CSP and TRAP by Mass Spectrometry-Based Proteomics. PLoS Pathog 2016; 12:e1005606. [PMID: 27128092 PMCID: PMC4851412 DOI: 10.1371/journal.ppat.1005606] [Citation(s) in RCA: 141] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2015] [Accepted: 04/08/2016] [Indexed: 12/22/2022] Open
Abstract
Malaria parasite infection is initiated by the mosquito-transmitted sporozoite stage, a highly motile invasive cell that targets hepatocytes in the liver for infection. A promising approach to developing a malaria vaccine is the use of proteins located on the sporozoite surface as antigens to elicit humoral immune responses that prevent the establishment of infection. Very little of the P. falciparum genome has been considered as potential vaccine targets, and candidate vaccines have been almost exclusively based on single antigens, generating the need for novel target identification. The most advanced malaria vaccine to date, RTS,S, a subunit vaccine consisting of a portion of the major surface protein circumsporozoite protein (CSP), conferred limited protection in Phase III trials, falling short of community-established vaccine efficacy goals. In striking contrast to the limited protection seen in current vaccine trials, sterilizing immunity can be achieved by immunization with radiation-attenuated sporozoites, suggesting that more potent protection may be achievable with a multivalent protein vaccine. Here, we provide the most comprehensive analysis to date of proteins located on the surface of or secreted by Plasmodium falciparum salivary gland sporozoites. We used chemical labeling to isolate surface-exposed proteins on sporozoites and identified these proteins by mass spectrometry. We validated several of these targets and also provide evidence that components of the inner membrane complex are in fact surface-exposed and accessible to antibodies in live sporozoites. Finally, our mass spectrometry data provide the first direct evidence that the Plasmodium surface proteins CSP and TRAP are glycosylated in sporozoites, a finding that could impact the selection of vaccine antigens. Malaria remains one of the most important infectious diseases in the world, responsible for an estimated 500 million new cases and 600,000 deaths annually. The etiologic agents of the disease are protozoan parasites of the genus Plasmodium that have a complex cycle between mosquito and mammalian hosts. Though all clinical symptoms are attributable to the blood stages, it is only by attacking the transmission stages that we can make an impact on the economic and health burdens of malaria. Infection is initiated when mosquitoes inoculate sporozoites into the skin as they probe for blood. Sporozoites must locate blood vessels and enter the circulation to reach the liver where they invade and grow in hepatocytes. The inoculum is low and these early stages of infection are asymptomatic. Though the small amounts of material available for study has made large scale -omics studies difficult, killing the parasite at this stage would prevent infection and block downstream transmission to mosquitoes, thus preventing spread of disease. Here we use state-of-the-art biochemistry tools to identify the proteins on the sporozoite surface and find that two of the most studied proteins, CSP and TRAP, have post-translational modifications. These studies will aid investigations into the novel biology of sporozoites and importantly, significantly expand the pool of potential vaccine candidates.
Collapse
Affiliation(s)
| | - Scott E. Lindner
- Center for Infectious Disease Research, formerly Seattle Biomedical Research Institute, Seattle, Washington, United States of America
- Center for Malaria Research, Pennsylvania State University, University Park, Pennsylvania, United States of America
| | - Lirong Shi
- Johns Hopkins Malaria Research Institute and Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Melanie J. Shears
- Johns Hopkins Malaria Research Institute and Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Anke Harupa
- Center for Infectious Disease Research, formerly Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | - Christine S. Hopp
- Johns Hopkins Malaria Research Institute and Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
| | - Ashley M. Vaughan
- Center for Infectious Disease Research, formerly Seattle Biomedical Research Institute, Seattle, Washington, United States of America
| | | | - Robert L. Moritz
- Institute for Systems Biology, Seattle, Washington, United States of America
- * E-mail: (RLM); (SHIK); (PS)
| | - Stefan H. I. Kappe
- Center for Infectious Disease Research, formerly Seattle Biomedical Research Institute, Seattle, Washington, United States of America
- * E-mail: (RLM); (SHIK); (PS)
| | - Photini Sinnis
- Johns Hopkins Malaria Research Institute and Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Johns Hopkins University, Baltimore, Maryland, United States of America
- * E-mail: (RLM); (SHIK); (PS)
| |
Collapse
|
61
|
Billman ZP, Kas A, Stone BC, Murphy SC. Defining rules of CD8(+) T cell expansion against pre-erythrocytic Plasmodium antigens in sporozoite-immunized mice. Malar J 2016; 15:238. [PMID: 27113469 PMCID: PMC4845300 DOI: 10.1186/s12936-016-1295-5] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2016] [Accepted: 04/14/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Whole Plasmodium sporozoites serve as both experimental tools and potentially as deployable vaccines in the fight against malaria infection. Live sporozoites infect hepatocytes and induce a diverse repertoire of CD8(+) T cell responses, some of which are capable of killing Plasmodium-infected hepatocytes. Previous studies in Plasmodium yoelii-immunized BALB/c mice showed that some CD8(+) T cell responses expanded with repeated parasite exposure, whereas other responses did not. RESULTS Here, similar outcomes were observed using known Plasmodium berghei epitopes in C57BL/6 mice. With the exception of the response to PbTRAP, IFNγ-producing T cell responses to most studied antigens, such as PbGAP50, failed to re-expand in mice immunized with two doses of irradiated P. berghei sporozoites. In an effort to boost secondary CD8(+) T cell responses, heterologous cross-species immunizations were performed. Alignment of P. yoelii 17XNL and P. berghei ANKA proteins revealed that >60 % of the amino acids in syntenic orthologous proteins are continuously homologous in fragments ≥8-amino acids long, suggesting that cross-species immunization could potentially trigger responses to a large number of common Class I epitopes. Heterologous immunization resulted in a larger liver burden than homologous immunization. Amongst seven tested antigen-specific responses, only CSP- and TRAP-specific CD8(+) T cell responses were expanded by secondary homologous sporozoite immunization and only those to the L3 ribosomal protein and S20 could be re-expanded by heterologous immunization. In general, heterologous late-arresting, genetically attenuated sporozoites were better at secondarily expanding L3-specific responses than were irradiated sporozoites. GAP50 and several other antigens shared between P. berghei and P. yoelii induced a large number of IFNγ-positive T cells during primary immunization, yet these responses could not be re-expanded by either homologous or heterologous secondary immunization. CONCLUSIONS These studies highlight how responses to different sporozoite antigens can markedly differ in recall following repeated sporozoite vaccinations. Cross-species immunization broadens the secondary response to sporozoites and may represent a novel strategy for candidate antigen discovery.
Collapse
Affiliation(s)
- Zachary P Billman
- Department of Laboratory Medicine, University of Washington, Seattle, WA, USA.,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Arnold Kas
- Department of Laboratory Medicine, University of Washington, Seattle, WA, USA.,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Brad C Stone
- Department of Laboratory Medicine, University of Washington, Seattle, WA, USA.,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA
| | - Sean C Murphy
- Department of Laboratory Medicine, University of Washington, Seattle, WA, USA. .,Department of Microbiology, University of Washington, Seattle, WA, USA. .,Center for Emerging and Re-emerging Infectious Diseases, University of Washington, Seattle, WA, USA.
| |
Collapse
|
62
|
Riestra AM, Gandhi S, Sweredoski MJ, Moradian A, Hess S, Urban S, Johnson PJ. A Trichomonas vaginalis Rhomboid Protease and Its Substrate Modulate Parasite Attachment and Cytolysis of Host Cells. PLoS Pathog 2015; 11:e1005294. [PMID: 26684303 PMCID: PMC4684317 DOI: 10.1371/journal.ppat.1005294] [Citation(s) in RCA: 38] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2015] [Accepted: 10/31/2015] [Indexed: 12/28/2022] Open
Abstract
Trichomonas vaginalis is an extracellular eukaryotic parasite that causes the most common, non-viral sexually transmitted infection worldwide. Although disease burden is high, molecular mechanisms underlying T. vaginalis pathogenesis are poorly understood. Here, we identify a family of putative T. vaginalis rhomboid proteases and demonstrate catalytic activity for two, TvROM1 and TvROM3, using a heterologous cell cleavage assay. The two T. vaginalis intramembrane serine proteases display different subcellular localization and substrate specificities. TvROM1 is a cell surface membrane protein and cleaves atypical model rhomboid protease substrates, whereas TvROM3 appears to localize to the Golgi apparatus and recognizes a typical model substrate. To identify TvROM substrates, we interrogated the T. vaginalis surface proteome using both quantitative proteomic and bioinformatic approaches. Of the nine candidates identified, TVAG_166850 and TVAG_280090 were shown to be cleaved by TvROM1. Comparison of amino acid residues surrounding the predicted cleavage sites of TvROM1 substrates revealed a preference for small amino acids in the predicted transmembrane domain. Over-expression of TvROM1 increased attachment to and cytolysis of host ectocervical cells. Similarly, mutations that block the cleavage of a TvROM1 substrate lead to its accumulation on the cell surface and increased parasite adherence to host cells. Together, these data indicate a role for TvROM1 and its substrate(s) in modulating attachment to and lysis of host cells, which are key processes in T. vaginalis pathogenesis. Trichomonas vaginalis, a common pathogen with a worldwide distribution, causes a sexually transmitted infection and exacerbates other diseases. Estimated to infect over a million people annually in the United States alone, the Center for Disease Control and Prevention categorized trichomoniasis as one of five neglected parasitic diseases in the US in 2014. Only one class of drug is available to treat T. vaginalis infection, making discovery of parasite factors contributing to host colonization critical for the development of new therapeutics. Here we report the first characterization of T. vaginalis intramembrane rhomboid proteases. One protease, TvROM1, is shown to increase the parasite’s association with and destruction of host cells. We further identified two TvROM1 substrates, one of which we demonstrate is involved in modulating host: parasite interactions. This study highlights the involvement of rhomboid proteases in T. vaginalis pathogenic processes, and provides further support for targeting parasite surface proteases for therapeutic intervention.
Collapse
Affiliation(s)
- Angelica M. Riestra
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
| | - Shiv Gandhi
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Michael J. Sweredoski
- Proteome Exploration Laboratory, Division of Biology and Biological Engineering, Beckman Institute, California Institute of Technology, Pasadena, California, United States of America
| | - Annie Moradian
- Proteome Exploration Laboratory, Division of Biology and Biological Engineering, Beckman Institute, California Institute of Technology, Pasadena, California, United States of America
| | - Sonja Hess
- Proteome Exploration Laboratory, Division of Biology and Biological Engineering, Beckman Institute, California Institute of Technology, Pasadena, California, United States of America
| | - Sinisa Urban
- Department of Molecular Biology and Genetics, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
- Howard Hughes Medical Institute, Johns Hopkins University School of Medicine, Baltimore, Maryland, United States of America
| | - Patricia J. Johnson
- Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
- Molecular Biology Institute and Department of Microbiology, Immunology, and Molecular Genetics, University of California, Los Angeles, Los Angeles, California, United States of America
- * E-mail:
| |
Collapse
|
63
|
Riglar DT, Whitehead L, Cowman AF, Rogers KL, Baum J. Localisation-based imaging of malarial antigens during erythrocyte entry reaffirms a role for AMA1 but not MTRAP in invasion. J Cell Sci 2015; 129:228-42. [PMID: 26604223 PMCID: PMC4732298 DOI: 10.1242/jcs.177741] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2015] [Accepted: 11/16/2015] [Indexed: 01/17/2023] Open
Abstract
Microscopy-based localisation of proteins during malaria parasite (Plasmodium) invasion of the erythrocyte is widely used for tentative assignment of protein function. To date, however, imaging has been limited by the rarity of invasion events and the poor resolution available, given the micron size of the parasite, which leads to a lack of quantitative measures for definitive localisation. Here, using computational image analysis we have attempted to assign relative protein localisation during invasion using wide-field deconvolution microscopy. By incorporating three-dimensional information we present a detailed assessment of known parasite effectors predicted to function during entry but as yet untested or for which data are equivocal. Our method, termed longitudinal intensity profiling, resolves confusion surrounding the localisation of apical membrane antigen 1 (AMA1) at the merozoite–erythrocyte junction and predicts that the merozoite thrombospondin-related anonymous protein (MTRAP) is unlikely to play a direct role in the mechanics of entry, an observation supported with additional biochemical evidence. This approach sets a benchmark for imaging of complex micron-scale events and cautions against simplistic interpretations of small numbers of representative images for the assignment of protein function or prioritisation of candidates as therapeutic targets. Highlighted Article: Here we develop a high-definition imaging approach to dissect and assign function to proteins involved in the rapid process of malaria parasite invasion of the human erythrocyte.
Collapse
Affiliation(s)
- David T Riglar
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, 3052, Australia Department of Medical Biology, University of Melbourne, Victoria, 3050, Melbourne, Australia
| | - Lachlan Whitehead
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, 3052, Australia Department of Medical Biology, University of Melbourne, Victoria, 3050, Melbourne, Australia
| | - Alan F Cowman
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, 3052, Australia Department of Medical Biology, University of Melbourne, Victoria, 3050, Melbourne, Australia
| | - Kelly L Rogers
- The Walter and Eliza Hall Institute of Medical Research, Melbourne, Victoria, 3052, Australia Department of Medical Biology, University of Melbourne, Victoria, 3050, Melbourne, Australia
| | - Jake Baum
- Department of Life Sciences, Imperial College, London SW7 2AZ, UK
| |
Collapse
|
64
|
Abstract
In vivo imaging has revealed new details about how the malaria parasite enters the bloodstream.
Collapse
Affiliation(s)
- Pauline Formaglio
- Institute of Molecular and Clinical Immunology, Otto-von-Guericke University, Magdeburg, Germany
| | - Rogerio Amino
- Unit of Malaria Infection and Immunity, Department of Parasites and Insect Vectors, Institut Pasteur, Paris, France
| |
Collapse
|
65
|
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: 1.9] [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
|
66
|
Hopp CS, Chiou K, Ragheb DRT, Salman AM, Khan SM, Liu AJ, Sinnis P. Longitudinal analysis of Plasmodium sporozoite motility in the dermis reveals component of blood vessel recognition. eLife 2015; 4. [PMID: 26271010 PMCID: PMC4594146 DOI: 10.7554/elife.07789] [Citation(s) in RCA: 96] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Accepted: 08/12/2015] [Indexed: 11/26/2022] Open
Abstract
Malaria infection starts with injection of Plasmodium sporozoites by an Anopheles mosquito into the skin of the mammalian host. How sporozoites locate and enter a blood vessel is a critical, but poorly understood process. In this study, we examine sporozoite motility and their interaction with dermal blood vessels, using intravital microscopy in mice. Our data suggest that sporozoites exhibit two types of motility: in regions far from blood vessels, they exhibit ‘avascular motility’, defined by high speed and less confinement, while in the vicinity of blood vessels their motility is more constrained. We find that curvature of sporozoite tracks engaging with vasculature optimizes contact with dermal capillaries. Imaging of sporozoites with mutations in key adhesive proteins highlight the importance of the sporozoite's gliding speed and its ability to modulate adhesive properties for successful exit from the inoculation site. DOI:http://dx.doi.org/10.7554/eLife.07789.001 Malaria remains a devastating disease in many parts of the world. Malaria parasites enter the host via the skin, where they are deposited by infected mosquitoes as they look for blood. The parasites must exit the skin to reach the liver, where they multiply and ultimately infect red blood cells, where they cause the symptoms of the disease. In the skin, the parasites must move to find blood vessels that they enter to travel via the blood circulation to the liver. Only about 10–20% of parasites make it out of the skin, making this a bottleneck for the parasite. Scientists have been working to develop vaccines that would protect people against malaria. One way these could work would be to stop malaria parasites from leaving the skin and entering the blood vessels. But to do that, more needs to be learnt about how the parasites move in the skin and enter the blood vessels. Hopp et al., using a mouse model of malaria, created malaria parasites that produce a fluorescent protein that allows the parasites to be tracked after they have been injected into the skin of a mouse's ear. This revealed that the parasites have two ways of moving. After first being injected, the parasites move quickly and freely. The parasites slow down when they come close to a blood vessel and move on or around the vessel for some time before entering it. During this stage of movement, the parasites tend to move in paths that follow the curvature of the blood vessels, which may improve how well they make contact with the blood vessel surface and may enable them to find the areas of the vessels best suited for entry. Next, Hopp et al. investigated how two parasite mutants move through mouse skin. Both mutants had previously been found to be less likely than wild-type parasites to exit the inoculation site. Hopp et al. found that one of the mutants moves slowly after being injected and so explores a smaller tissue volume than normal and encounters fewer blood vessels. The second mutant parasite spends more time than normal moving on the surface of the blood vessels, but finds it difficult to enter them. Continuing this work will allow us to learn more about the interactions between the parasite and the blood vessels, which in turn could reveal key events that could be targeted by a vaccine. Furthermore, the significant amount of time that the parasites spend moving and looking for blood vessels in the skin could be a good time to target them with antibodies and prevent malaria infection. DOI:http://dx.doi.org/10.7554/eLife.07789.002
Collapse
Affiliation(s)
- Christine S Hopp
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, United States
| | - Kevin Chiou
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, United States
| | - Daniel R T Ragheb
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, United States
| | - Ahmed M Salman
- Department of Parasitology, Leiden Malaria Research Group, Leiden University Medical Center, Leiden, Netherlands
| | - Shahid M Khan
- Department of Parasitology, Leiden Malaria Research Group, Leiden University Medical Center, Leiden, Netherlands
| | - Andrea J Liu
- Department of Physics and Astronomy, University of Pennsylvania, Philadelphia, United States
| | - Photini Sinnis
- Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, United States
| |
Collapse
|
67
|
Reversible Conformational Change in the Plasmodium falciparum Circumsporozoite Protein Masks Its Adhesion Domains. Infect Immun 2015; 83:3771-80. [PMID: 26169272 DOI: 10.1128/iai.02676-14] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Accepted: 07/02/2015] [Indexed: 11/20/2022] Open
Abstract
The extended rod-like Plasmodium falciparum circumsporozoite protein (CSP) is comprised of three primary domains: a charged N terminus that binds heparan sulfate proteoglycans, a central NANP repeat domain, and a C terminus containing a thrombospondin-like type I repeat (TSR) domain. Only the last two domains are incorporated in RTS,S, the leading malaria vaccine in phase 3 trials that, to date, protects about 50% of vaccinated children against clinical disease. A seroepidemiological study indicated that the N-terminal domain might improve the efficacy of a new CSP vaccine. Using a panel of CSP-specific monoclonal antibodies, well-characterized recombinant CSPs, label-free quantitative proteomics, and in vitro inhibition of sporozoite invasion, we show that native CSP is N-terminally processed in the mosquito host and undergoes a reversible conformational change to mask some epitopes in the N- and C-terminal domains until the sporozoite interacts with the liver hepatocyte. Our findings show the importance of understanding processing and the biophysical change in conformation, possibly due to a mechanical or molecular signal, and may aid in the development of a new CSP vaccine.
Collapse
|
68
|
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: 5.4] [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
|
69
|
Rastew E, Morf L, Singh U. Entamoeba histolytica rhomboid protease 1 has a role in migration and motility as validated by two independent genetic approaches. Exp Parasitol 2015; 154:33-42. [PMID: 25889553 DOI: 10.1016/j.exppara.2015.04.004] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2014] [Revised: 02/24/2015] [Accepted: 04/08/2015] [Indexed: 11/25/2022]
Abstract
Rhomboid proteins represent a recently discovered family of intramembrane proteases present in a broad range of organisms and with increasing links to human diseases. The enteric parasite Entamoeba histolytica has evolved multiple mechanisms to adapt to the human host environment and establish infection. Our recent studies identified EhROM1 as a functional E. histolytica rhomboid protease with roles in adhesion to and phagocytosis of host cells. Since those studies were performed in a non-virulent strain, roles in parasite virulence could not be assessed. We focused this study on the comparison and validation of two genetic manipulation techniques: overexpression of a dominant-negative catalytic mutant of EhROM1 and knock down of EhROM1 using a RNAi-based silencing approach followed by functional studies of phenotypic analyses in virulent parasites. Both the EhROM1 catalytic mutant and parasites with EhROM1 downregulation were reduced in cytotoxicity, hemolytic activity, and directional and non-directional transwell migration. Importantly, the role for EhROM1 in cell migration mimics similar roles for rhomboid proteases from mammalian and apicomplexan systems. However, the EhROM1 catalytic mutant and EhROM1 downregulation parasites had different phenotypes for erythrophagocytosis, while complement resistance was not affected in either strain. In summary, in this study we genetically manipulated E. histolytica rhomboid protease EhROM1 by two different approaches and identified similarly attenuated phenotypes by both approaches, suggesting a novel role for EhROM1 in amebic motility.
Collapse
Affiliation(s)
- Elena Rastew
- Department of Internal Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Laura Morf
- Department of Internal Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Upinder Singh
- Department of Internal Medicine, Stanford University School of Medicine, Stanford, CA, USA; Department of Microbiology and Immunology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
70
|
Abstract
Host cell invasion by Toxoplasma gondii and other apicomplexan parasites requires transmembrane adhesins that mediate binding to receptors on the substrate and host cell to facilitate motility and invasion. Rhomboid proteases (ROMs) are thought to cleave adhesins within their transmembrane segments, thus allowing the parasite to disengage from receptors and completely enter the host cell. To examine the specific roles of individual ROMs during invasion, we generated single, double, and triple knockouts for the three ROMs expressed in T. gondii tachyzoites. Analysis of these mutants demonstrated that ROM4 is the primary protease involved in adhesin processing and host cell invasion, whereas ROM1 or ROM5 plays negligible roles in these processes. Deletion of ROM4 blocked the shedding of adhesins such as MIC2 (microneme protein 2), causing them to accumulate on the surface of extracellular parasites. Increased surface adhesins led to nonproductive attachment, altered gliding motility, impaired moving junction formation, and reduced invasion efficiency. Despite the importance of ROM4 for efficient invasion, mutants lacking all three ROMs were viable and MIC2 was still efficiently removed from the surface of invaded mutant parasites, implying the existence of ROM-independent mechanisms for adhesin removal during invasion. Collectively, these results suggest that although ROM processing of adhesins is not absolutely essential, it is important for efficient host cell invasion by T. gondii. Apicomplexan parasites such as Toxoplasma gondii express surface proteins that bind host cell receptors to aid invasion. Many of these adhesins are subject to cleavage by rhomboid proteases (ROMs) within their transmembrane segments during invasion. Previous studies have demonstrated the importance of adhesin cleavage for parasite invasion and proposed that the ROMs responsible for processing would be essential for parasite survival. In T. gondii, ROM5 was thought to be the critical ROM for adhesin shedding due to its robust protease activity in vitro and posterior localization on the parasite surface. Here, we knocked out all three ROMs in T. gondii tachyzoites and found that ROM4, but not ROM5, was key for adhesin cleavage. However, none of the ROMs individually or in combination was essential for cell entry, further emphasizing that essential pathways such as invasion typically rely on redundant pathways to ensure survival.
Collapse
|
71
|
Lehmann C, Heitmann A, Mishra S, Burda PC, Singer M, Prado M, Niklaus L, Lacroix C, Ménard R, Frischknecht F, Stanway R, Sinnis P, Heussler V. A cysteine protease inhibitor of plasmodium berghei is essential for exo-erythrocytic development. PLoS Pathog 2014; 10:e1004336. [PMID: 25166051 PMCID: PMC4148452 DOI: 10.1371/journal.ppat.1004336] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2013] [Accepted: 07/08/2014] [Indexed: 11/18/2022] Open
Abstract
Plasmodium parasites express a potent inhibitor of cysteine proteases (ICP) throughout their life cycle. To analyze the role of ICP in different life cycle stages, we generated a stage-specific knockout of the Plasmodium berghei ICP (PbICP). Excision of the pbicb gene occurred in infective sporozoites and resulted in impaired sporozoite invasion of hepatocytes, despite residual PbICP protein being detectable in sporozoites. The vast majority of these parasites invading a cultured hepatocyte cell line did not develop to mature liver stages, but the few that successfully developed hepatic merozoites were able to initiate a blood stage infection in mice. These blood stage parasites, now completely lacking PbICP, exhibited an attenuated phenotype but were able to infect mosquitoes and develop to the oocyst stage. However, PbICP-negative sporozoites liberated from oocysts exhibited defective motility and invaded mosquito salivary glands in low numbers. They were also unable to invade hepatocytes, confirming that control of cysteine protease activity is of critical importance for sporozoites. Importantly, transfection of PbICP-knockout parasites with a pbicp-gfp construct fully reversed these defects. Taken together, in P. berghei this inhibitor of the ICP family is essential for sporozoite motility but also appears to play a role during parasite development in hepatocytes and erythrocytes.
Collapse
Affiliation(s)
| | - Anna Heitmann
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Satish Mishra
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | | | - Mirko Singer
- University of Heidelberg Medical School, Heidelberg, Germany
| | - Monica Prado
- Bernhard Nocht Institute for Tropical Medicine, Hamburg, Germany
| | - Livia Niklaus
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Céline Lacroix
- Institute Pasteur, Unité de Biologie et Génétique du Paludisme, Paris, France
| | - Robert Ménard
- Institute Pasteur, Unité de Biologie et Génétique du Paludisme, Paris, France
| | | | - Rebecca Stanway
- Institute of Cell Biology, University of Bern, Bern, Switzerland
| | - Photini Sinnis
- Johns Hopkins Bloomberg School of Public Health, Baltimore, Maryland, United States of America
| | - Volker Heussler
- Institute of Cell Biology, University of Bern, Bern, Switzerland
- * E-mail:
| |
Collapse
|
72
|
SSP3 is a novel Plasmodium yoelii sporozoite surface protein with a role in gliding motility. Infect Immun 2014; 82:4643-53. [PMID: 25156733 DOI: 10.1128/iai.01800-14] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Plasmodium sporozoites develop within oocysts in the mosquito midgut wall and then migrate to the salivary glands. After transmission, they embark on a complex journey to the mammalian liver, where they infect hepatocytes. Proteins on the sporozoite surface likely mediate multiple steps of this journey, yet only a few sporozoite surface proteins have been described. Here, we characterize a novel, conserved sporozoite surface protein (SSP3) in the rodent malaria parasite Plasmodium yoelii. SSP3 is a putative type I transmembrane protein unique to Plasmodium. By using epitope tagging and SSP3-specific antibodies in conjunction with immunofluorescence microscopy, we showed that SSP3 is expressed in mosquito midgut oocyst sporozoites, exhibiting an intracellular localization. In sporozoites derived from the mosquito salivary glands, however, SSP3 localized predominantly to the sporozoite surface as determined by immunoelectron microscopy. However, the ectodomain of SSP3 appeared to be inaccessible to antibodies in nonpermeabilized salivary gland sporozoites. Antibody-induced shedding of the major surface protein circumsporozoite protein (CSP) exposed the SSP3 ectodomain to antibodies in some sporozoites. Targeted deletion of SSP3 adversely affected in vitro sporozoite gliding motility, which, surprisingly, impacted neither their cell traversal capacity, host cell invasion in vitro, nor infectivity in vivo. Together, these data reveal a previously unappreciated complexity of the Plasmodium sporozoite surface proteome and the roles of surface proteins in distinct biological activities of sporozoites.
Collapse
|
73
|
Ocholla H, Preston MD, Mipando M, Jensen ATR, Campino S, MacInnis B, Alcock D, Terlouw A, Zongo I, Oudraogo JB, Djimde AA, Assefa S, Doumbo OK, Borrmann S, Nzila A, Marsh K, Fairhurst RM, Nosten F, Anderson TJC, Kwiatkowski DP, Craig A, Clark TG, Montgomery J. Whole-genome scans provide evidence of adaptive evolution in Malawian Plasmodium falciparum isolates. J Infect Dis 2014; 210:1991-2000. [PMID: 24948693 PMCID: PMC4241944 DOI: 10.1093/infdis/jiu349] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Background Selection by host immunity and antimalarial drugs has driven extensive adaptive evolution in Plasmodium falciparum and continues to produce ever-changing landscapes of genetic variation. Methods We performed whole-genome sequencing of 69 P. falciparum isolates from Malawi and used population genetics approaches to investigate genetic diversity and population structure and identify loci under selection. Results High genetic diversity (π = 2.4 × 10−4), moderately high multiplicity of infection (2.7), and low linkage disequilibrium (500-bp) were observed in Chikhwawa District, Malawi, an area of high malaria transmission. Allele frequency–based tests provided evidence of recent population growth in Malawi and detected potential targets of host immunity and candidate vaccine antigens. Comparison of the sequence variation between isolates from Malawi and those from 5 geographically dispersed countries (Kenya, Burkina Faso, Mali, Cambodia, and Thailand) detected population genetic differences between Africa and Asia, within Southeast Asia, and within Africa. Haplotype-based tests of selection to sequence data from all 6 populations identified signals of directional selection at known drug-resistance loci, including pfcrt, pfdhps, pfmdr1, and pfgch1. Conclusions The sequence variations observed at drug-resistance loci reflect differences in each country's historical use of antimalarial drugs and may be useful in formulating local malaria treatment guidelines.
Collapse
Affiliation(s)
- Harold Ocholla
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme Liverpool School of Tropical Medicine, Pembroke Place, Liverpool
| | - Mark D Preston
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine
| | - Mwapatsa Mipando
- Department of Physiology, College of Medicine, University of Malawi, Blantyre
| | - Anja T R Jensen
- Centre for Medical Parasitology, Department of International Health, Immunology and Microbiology, University of Copenhagen Department of Infectious Diseases, Copenhagen University Hospital, Denmark
| | | | | | | | - Anja Terlouw
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme Liverpool School of Tropical Medicine, Pembroke Place, Liverpool
| | - Issaka Zongo
- Institut de Recherche en Sciences de la Sant, Bobo-Dioulasso, Burkina Faso
| | | | - Abdoulaye A Djimde
- Wellcome Trust Sanger Institute, Hinxton Malaria Research and Training Centre, Faculty of Medicine, Pharmacy and Dentistry, University of Bamako, Mali
| | - Samuel Assefa
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine
| | - Ogobara K Doumbo
- Malaria Research and Training Centre, Faculty of Medicine, Pharmacy and Dentistry, University of Bamako, Mali
| | | | - Alexis Nzila
- Department of Biology, King Fahd University of Petroleum and Minerals, Dhahran, Saudi Arabia
| | - Kevin Marsh
- KEMRI-Wellcome Trust Research Programme, Kilifi, Kenya
| | - Rick M Fairhurst
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, Maryland
| | - Francois Nosten
- Centre for Tropical Medicine, Nuffield Department of Medicine, University of Oxford, United Kingdom Shoklo Malaria Research Unit, Mahidol-Oxford Tropical Medicine Research Unit, Faculty of Tropical Medicine, Mahidol University, Mae Sot, Thailand
| | | | - Dominic P Kwiatkowski
- Wellcome Trust Sanger Institute, Hinxton Wellcome Trust Centre for Human Genetics, University of Oxford, United Kingdom
| | - Alister Craig
- Liverpool School of Tropical Medicine, Pembroke Place, Liverpool
| | - Taane G Clark
- Faculty of Infectious and Tropical Diseases, London School of Hygiene and Tropical Medicine
| | - Jacqui Montgomery
- Malawi-Liverpool-Wellcome Trust Clinical Research Programme Liverpool School of Tropical Medicine, Pembroke Place, Liverpool
| |
Collapse
|
74
|
Zheng J, Gong P, Jia H, Li M, Zhang G, Zhang X, Li J. Eimeria tenella rhomboid 3 has a potential role in microneme protein cleavage. Vet Parasitol 2014; 201:146-9. [PMID: 24529829 DOI: 10.1016/j.vetpar.2014.01.010] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2013] [Revised: 01/02/2014] [Accepted: 01/19/2014] [Indexed: 01/04/2023]
Abstract
Invasion in several apicomplexan parasites, including Eimeria tenella, is accompanied by shedding of surface adhesins by intramembrane proteolysis mediated by rhomboid protease. We have previously identified E. tenella rhomboid 3 (EtROM3), but its precise role has not been elucidated. In this study, the interactions between EtROM3 and microneme (MIC) proteins were analyzed using the yeast two hybrid technique. The results showed that c-Myc-ROM3 fusion protein interacted with EtMIC4 protein in co-transformed AH109 yeasts, which was further confirmed by immunoprecipitation assay. Smaller EtMIC4 band from co-transformed cells suggested that EtROM3 was an active protease and involved in the cleavage of EtMIC4.
Collapse
Affiliation(s)
- Jun Zheng
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China; National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Pengtao Gong
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Honglin Jia
- National Key Laboratory of Veterinary Biotechnology, Harbin Veterinary Research Institute, Chinese Academy of Agricultural Sciences, Harbin, China
| | - Mingying Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Guocai Zhang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Xichen Zhang
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China
| | - Jianhua Li
- Key Laboratory of Zoonosis, Ministry of Education, College of Veterinary Medicine, Jilin University, Changchun, China.
| |
Collapse
|
75
|
Terkawi MA, Ratthanophart J, Salama A, AbouLaila M, Asada M, Ueno A, Alhasan H, Guswanto A, Masatani T, Yokoyama N, Nishikawa Y, Xuan X, Igarashi I. Molecular characterization of a new Babesia bovis thrombospondin-related anonymous protein (BbTRAP2). PLoS One 2013; 8:e83305. [PMID: 24349483 PMCID: PMC3862764 DOI: 10.1371/journal.pone.0083305] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2013] [Accepted: 11/02/2013] [Indexed: 11/18/2022] Open
Abstract
A gene encoding a Babesia bovis protein that shares significant degree of similarity to other apicomplexan thrombospondin-related anonymous proteins (TRAPs) was found in the genomic database and designated as BbTRAP2. Recombinant protein containing a conserved region of BbTRAP2 was produced in E. coli. A high antigenicity of recombinant BbTRAP2 (rBbTRAP2) was observed with field B. bovis-infected bovine sera collected from geographically different regions of the world. Moreover, antiserum against rBbTRAP2 specifically reacted with the authentic protein by Western blot analysis and an indirect fluorescent antibody test. Three bands corresponding to 104-, 76-, and 44-kDa proteins were identified in the parasite lysates and two bands of 76- and 44-kDa proteins were detected in the supernatant of cultivated parasites, indicating that BbTRAP2 was proteolytically processed and shed into the culture. Apical and surface localizations of BbTRAP2 were observed in the intracellular and extracellular parasites, respectively, by confocal laser microscopic examination. Moreover, native BbTRAP2 was precipitated by bovine erythrocytes, suggesting its role in the attachment to erythrocytes. Furthermore, the specific antibody to rBbTRAP2 inhibited the growth of B. bovis in a concentration-dependent manner. Consistently, pre-incubation of the free merozoites with the antibody to rBbTRAP2 resulted in an inhibition of the parasite invasion into host erythrocytes. Interestingly, the antibody to rBbTRAP2 was the most inhibitive for the parasite’s growth as compared to those of a set of antisera produced against different recombinant proteins, including merozoite surface antigen 2c (BbMSA-2c), rhoptry-associated protein 1 C-terminal (BbRAP-1CT), and spherical body protein 1 (BbSBP-1). These results suggest that BbTRAP2 might be a potential candidate for development of a subunit vaccine against B. bovis infection.
Collapse
Affiliation(s)
- Mohamad Alaa Terkawi
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Jadsada Ratthanophart
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Akram Salama
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
- Department of Animal Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Sadat City University, Minoufiya, Egypt
| | - Mahmoud AbouLaila
- Department of Parasitology, Faculty of Veterinary Medicine, Sadat City University, Minoufiya, Egypt
| | - Masahito Asada
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Akio Ueno
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Hend Alhasan
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Azirwan Guswanto
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Tatsunori Masatani
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Naoaki Yokoyama
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Yoshifumi Nishikawa
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Xuenan Xuan
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Ikuo Igarashi
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
- * E-mail:
| |
Collapse
|
76
|
Sibley LD. The roles of intramembrane proteases in protozoan parasites. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2013; 1828:2908-15. [PMID: 24099008 DOI: 10.1016/j.bbamem.2013.04.017] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2013] [Revised: 04/02/2013] [Accepted: 04/11/2013] [Indexed: 11/30/2022]
Abstract
Intramembrane proteolysis is widely conserved throughout different forms of life, with three major types of proteases being known for their ability to cleave peptide bonds directly within the transmembrane domains of their substrates. Although intramembrane proteases have been extensively studied in humans and model organisms, they have only more recently been investigated in protozoan parasites, where they turn out to play important and sometimes unexpected roles. Signal peptide peptidases are involved in endoplasmic reticulum (ER) quality control and signal peptide degradation from exported proteins. Recent studies suggest that repurposing inhibitors developed for blocking presenilins may be useful for inhibiting the growth of Plasmodium, and possibly other protozoan parasites, by blocking signal peptide peptidases. Rhomboid proteases, originally described in the fly, are also widespread in parasites, and are especially expanded in apicomplexans. Their study in parasites has revealed novel roles that expand our understanding of how these proteases function. Within this diverse group of parasites, rhomboid proteases contribute to processing of adhesins involved in attachment, invasion, intracellular replication, phagocytosis, and immune evasion, placing them at the vertex of host-parasite interactions. This article is part of a Special Issue entitled: Intramembrane Proteases.
Collapse
Affiliation(s)
- L David Sibley
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
77
|
Abstract
Malaria is transmitted when motile sporozoites are injected into the dermis by an infected female Anopheles mosquito. Inside the mosquito vector, sporozoites egress from midgut-associated oocysts and eventually penetrate the acinar cells of salivary glands. Parasite-encoded factors with exclusive vital roles in the insect vector can be studied by classical reverse genetics. Here, we characterized the in vivo roles of Plasmodium berghei falstatin/ICP (inhibitor of cysteine proteases). This protein was previously suggested to act as a protease inhibitor during erythrocyte invasion. We show by targeted gene disruption that loss of ICP function does not affect growth inside the mammalian host but causes a complete defect in sporozoite transmission. Sporogony occurred normally in icp(−) parasites, but hemocoel sporozoites showed a defect in continuous gliding motility and infectivity for salivary glands, which are prerequisites for sporozoite transmission to the mammalian host. Absence of ICP correlates with enhanced cleavage of circumsporozoite protein, in agreement with a role as a protease regulator. We conclude that ICP is essential for only the final stages of sporozoite maturation inside the mosquito vector. This study is the first genetic evidence that an ICP is necessary for the productive motility of a eukaryotic parasitic cell. Cysteine proteases and their inhibitors are considered ideal drug targets for the treatment of a wide range of diseases, including cancer and parasitic infections. In protozoan parasites, including Leishmania, Trypanosoma, and Plasmodium, cysteine proteases play important roles in life cycle progression. A mouse malaria model provides an unprecedented opportunity to study the roles of a parasite-encoded inhibitor of cysteine proteases (ICP) over the entire parasite life cycle. By precise gene deletion, we found no evidence that ICP influences disease progression or parasite virulence. Instead, we discovered that this factor is necessary for parasite movement and malaria transmission from mosquitoes to mammals. This finding in a fast-moving unicellular protozoan has important implications for malaria intervention strategies and the roles of ICPs in the regulation of eukaryotic cell migration.
Collapse
|
78
|
Walker DM, Oghumu S, Gupta G, McGwire BS, Drew ME, Satoskar AR. Mechanisms of cellular invasion by intracellular parasites. Cell Mol Life Sci 2013; 71:1245-63. [PMID: 24221133 DOI: 10.1007/s00018-013-1491-1] [Citation(s) in RCA: 108] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2013] [Revised: 10/04/2013] [Accepted: 10/07/2013] [Indexed: 12/22/2022]
Abstract
Numerous disease-causing parasites must invade host cells in order to prosper. Collectively, such pathogens are responsible for a staggering amount of human sickness and death throughout the world. Leishmaniasis, Chagas disease, toxoplasmosis, and malaria are neglected diseases and therefore are linked to socio-economical and geographical factors, affecting well-over half the world's population. Such obligate intracellular parasites have co-evolved with humans to establish a complexity of specific molecular parasite-host cell interactions, forming the basis of the parasite's cellular tropism. They make use of such interactions to invade host cells as a means to migrate through various tissues, to evade the host immune system, and to undergo intracellular replication. These cellular migration and invasion events are absolutely essential for the completion of the lifecycles of these parasites and lead to their for disease pathogenesis. This review is an overview of the molecular mechanisms of protozoan parasite invasion of host cells and discussion of therapeutic strategies, which could be developed by targeting these invasion pathways. Specifically, we focus on four species of protozoan parasites Leishmania, Trypanosoma cruzi, Plasmodium, and Toxoplasma, which are responsible for significant morbidity and mortality.
Collapse
Affiliation(s)
- Dawn M Walker
- Department of Microbial Infection and Immunity, Wexner Medical Center, The Ohio State University, Columbus, OH, 43210, USA
| | | | | | | | | | | |
Collapse
|
79
|
TREM2 governs Kupffer cell activation and explains belr1 genetic resistance to malaria liver stage infection. Proc Natl Acad Sci U S A 2013; 110:19531-6. [PMID: 24218563 DOI: 10.1073/pnas.1306873110] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Plasmodium liver stage infection is a target of interest for the treatment of and vaccination against malaria. Here we used forward genetics to search for mechanisms underlying natural host resistance to infection and identified triggering receptor expressed on myeloid cells 2 (TREM2) and MHC class II molecules as determinants of Plasmodium berghei liver stage infection in mice. Locus belr1 confers resistance to malaria liver stage infection. The use of newly derived subcongenic mouse lines allowed to map belr1 to a 4-Mb interval on mouse chromosome 17 that contains the Trem2 gene. We show that Trem2 expression in the nonparenchymal liver cells closely correlates with resistance to liver stage infection, implicating TREM2 as a mediator of the belr1 genetic effect. Trem2-deficient mice are more susceptible to liver stage infection than their WT counterparts. We found that Kupffer cells are the principle cells expressing TREM2 in the liver, and that Trem2(-/-) Kupffer cells display altered functional activation on exposure to P. berghei sporozoites. TREM2 expression in Kupffer cells contributes to the limitation of parasite expansion in isolated hepatocytes in vitro, potentially explaining the increased susceptibility of Trem2(-/-) mice to liver stage infection. The MHC locus was also found to control liver parasite burden, possibly owing to the expression of MHC class II molecules in hepatocytes. Our findings implicate unexpected Kupffer-hepatocyte cross-talk in the control Plasmodium liver stage infection and demonstrate that TREM2 is involved in host responses against the malaria parasite.
Collapse
|
80
|
Lawton JC, Benson RA, Garside P, Brewer JM. Using lymph node transplantation as an approach to image cellular interactions between the skin and draining lymph nodes during parasitic infections. Parasitol Int 2013; 63:165-70. [PMID: 23892176 PMCID: PMC3863950 DOI: 10.1016/j.parint.2013.07.010] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Accepted: 07/16/2013] [Indexed: 12/24/2022]
Abstract
The growing use of protozoan parasites expressing fluorescent reporter genes, together with advances in microscopy, is enabling visualisation of their behaviour and functions within the host from the very earliest stages of infection with previously unparalleled spatiotemporal resolution. These developments have begun to provide novel insights, which are informing our understanding of where host immune responses may be initiated, which cells are involved and the types of response that are elicited. Here we will review some of these recent observations that highlight the importance of cellular communication between the site of infection and the draining lymph node (dLN) in establishing infection and immunity. We also highlight a number of remaining challenges and unknowns that arise through our inability to follow and fate map the journey of a single cell between spatially separated tissue sites. In response to these challenges, we review a recently described experimental strategy that extends the spatial and temporal limits of previous imaging approaches, most significantly allowing longitudinal analysis of cellular migration between the skin and draining lymph nodes in vivo, without the requirement for invasive surgery. Role of skin and LN communication in establishing infection and immunity A technique to extend the spatial and temporal limits of tissue imaging This approach does not require invasive surgery for imaging. We can follow the journey of a single cell between spatially separated tissue sites. We outline future applications for this approach.
Collapse
Affiliation(s)
- Jennifer C Lawton
- Wellcome Trust Centre For Molecular Parasitology, Institute of Infection, Immunity & Inflammation, College of Medical, Veterinary & Life Sciences, University of Glasgow, Glasgow, G12 8TA, United Kingdom
| | | | | | | |
Collapse
|
81
|
Frénal K, Soldati-Favre D. [The glideosome, a unique machinery that assists the Apicomplexa in gliding into host cells]. Med Sci (Paris) 2013; 29:515-22. [PMID: 23732101 DOI: 10.1051/medsci/2013295015] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
Protozoan parasites belonging to the phylum Apicomplexa are of considerable medical and veterinary significance. These obligate intracellular parasites use a unique form of locomotion to traverse biological barriers and actively invade in and egress from host cells. An actin-myosin-based complex named "glideosome" drives this unusual substrate-dependent motility, which is essential for the establishment of the infection. The mechanisms involved in motility, invasion and egress are conserved throughout the phylum. This article describes the current knowledge on the invasion process of two experimentally tractable apicomplexan parasites: Toxoplasma gondii and Plasmodium falciparum.
Collapse
Affiliation(s)
- Karine Frénal
- Département de microbiologie et médecine moléculaire, faculté de médecine, université de Genève, centre médical universitaire, 1 rue Michel Servet, 1211 Genève, Suisse.
| | | |
Collapse
|
82
|
Lin JW, Meireles P, Prudêncio M, Engelmann S, Annoura T, Sajid M, Chevalley-Maurel S, Ramesar J, Nahar C, Avramut CMC, Koster AJ, Matuschewski K, Waters AP, Janse CJ, Mair GR, Khan SM. Loss-of-function analyses defines vital and redundant functions of the Plasmodium rhomboid protease family. Mol Microbiol 2013; 88:318-38. [PMID: 23490234 DOI: 10.1111/mmi.12187] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/13/2013] [Indexed: 11/26/2022]
Abstract
Rhomboid-like proteases cleave membrane-anchored proteins within their transmembrane domains. In apicomplexan parasites substrates include molecules that function in parasite motility and host cell invasion. While two Plasmodium rhomboids, ROM1 and ROM4, have been examined, the roles of the remaining six rhomboids during the malaria parasite's life cycle are unknown. We present systematic gene deletion analyses of all eight Plasmodium rhomboid-like proteins as a means to discover stage-specific phenotypes and potential functions in the rodent malaria model, P. berghei. Four rhomboids (ROM4, 6, 7 and 8) are refractory to gene deletion, suggesting an essential role during asexual blood stage development. In contrast ROM1, 3, 9 and 10 were dispensable for blood stage development and exhibited no, subtle or severe defects in mosquito or liver development. Parasites lacking ROM9 and ROM10 showed no major phenotypic defects. Parasites lacking ROM1 presented a delay in blood stage patency following liver infection, but in contrast to a previous study blood stage parasites had similar growth and virulence characteristics as wild type parasites. Parasites lacking ROM3 in mosquitoes readily established oocysts but failed to produce sporozoites. ROM3 is the first apicomplexan rhomboid identified to play a vital role in sporogony.
Collapse
Affiliation(s)
- Jing-Wen Lin
- Leiden Malaria Research Group (Parasitology), Leiden University Medical Centre, Leiden, The Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
83
|
Shape change in the receptor for gliding motility in Plasmodium sporozoites. Proc Natl Acad Sci U S A 2012; 109:21420-5. [PMID: 23236185 DOI: 10.1073/pnas.1218581109] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Sporozoite gliding motility and invasion of mosquito and vertebrate host cells in malaria is mediated by thrombospondin repeat anonymous protein (TRAP). Tandem von Willebrand factor A (VWA) and thrombospondin type I repeat (TSR) domains in TRAP connect through proline-rich stalk, transmembrane, and cytoplasmic domains to the parasite actin-dependent motility apparatus. We crystallized fragments containing the VWA and TSR domains from Plasmodium vivax and Plasmodium falciparum in different crystal lattices. TRAP VWA domains adopt closed and open conformations, and bind a Mg(2+) ion at a metal ion-dependent adhesion site implicated in ligand binding. Metal ion coordination in the open state is identical to that seen in the open high-affinity state of integrin I domains. The closed VWA conformation associates with a disordered TSR domain. In contrast, the open VWA conformation crystallizes with an extensible β ribbon and ordered TSR domain. The extensible β ribbon is composed of disulfide-bonded segments N- and C-terminal to the VWA domain that are largely drawn out of the closed VWA domain in a 15 Å movement to the open conformation. The extensible β ribbon and TSR domain overlap at a conserved interface. The VWA, extensible β ribbon, and TSR domains adopt a highly elongated overall orientation that would be stabilized by tensile force exerted across a ligand-receptor complex by the actin motility apparatus of the sporozoite. Our results provide insights into regulation of "stick-and-slip" parasite motility and for development of sporozoite subunit vaccines.
Collapse
|
84
|
The skin: where malaria infection and the host immune response begin. Semin Immunopathol 2012; 34:787-92. [PMID: 23053392 DOI: 10.1007/s00281-012-0345-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 09/13/2012] [Indexed: 10/27/2022]
Abstract
Infection by malaria parasites begins with the inoculation of sporozoites into the skin of the host. The early events following sporozoite deposition in the dermis are critical for both the establishment of malaria infection and for the induction of protective immune responses. The initial sporozoite inoculum is generally low, and only a small percentage of these sporozoites successfully reach the liver and grow to the next life cycle stage, making this a significant bottleneck for the parasite. Recent studies highlight the importance of sporozoite motility and host cell traversal in dermal exit. Importantly, protective immune responses against sporozoites and liver stages of Plasmodium are induced by dendritic cells in the lymph node draining the skin inoculation site. The cellular, molecular, and immunological events that occur in the skin and associated lymph nodes are the topic of this review.
Collapse
|