1
|
Gupta A, Gonzalez-Chavez Z, Desai SA. Plasmodium falciparum CLAG Paralogs All Traffic to the Host Membrane but Knockouts Have Distinct Phenotypes. Microorganisms 2024; 12:1172. [PMID: 38930554 PMCID: PMC11205492 DOI: 10.3390/microorganisms12061172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 06/04/2024] [Accepted: 06/06/2024] [Indexed: 06/28/2024] Open
Abstract
Malaria parasites increase their host erythrocyte's permeability to obtain essential nutrients from plasma and facilitate intracellular growth. In the human Plasmodium falciparum pathogen, this increase is mediated by the plasmodial surface anion channel (PSAC) and has been linked to CLAG3, a protein integral to the host erythrocyte membrane and encoded by a member of the conserved clag multigene family. Whether paralogs encoded by other clag genes also insert at the host membrane is unknown; their contributions to PSAC formation and other roles served are also unexplored. Here, we generated transfectant lines carrying epitope-tagged versions of each CLAG. Each paralog is colocalized with CLAG3, with concordant trafficking via merozoite rhoptries to the host erythrocyte membrane of newly invaded erythrocytes. Each also exists within infected cells in at least two forms: an alkaline-extractable soluble form and a form integral to the host membrane. Like CLAG3, CLAG2 has a variant region cleaved by extracellular proteases, but CLAG8 and CLAG9 are protease resistant. Paralog knockout lines, generated through CRISPR/Cas9 transfection, exhibited uncompromised growth in PGIM, a modified medium with higher physiological nutrient levels; this finding is in marked contrast to a recently reported CLAG3 knockout parasite. CLAG2 and CLAG8 knockout lines exhibited compensatory increases in the transcription of the remaining clags and associated rhoph genes, yielding increased PSAC-mediated uptake for specific solutes. We also report on the distinct transport properties of these knockout lines. Similar membrane topologies at the host membrane are consistent with each CLAG paralog contributing to PSAC, but other roles require further examination.
Collapse
Affiliation(s)
| | | | - Sanjay A. Desai
- Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MA 20852, USA
| |
Collapse
|
2
|
El-Sayed SAES, Rizk MA, Eldoumani H, Sorour SS, Terkawi MA, AbouLaila M, Igarashi I, Sayed-Ahmed MZ. Identification and Characterization of P0 Protein as a Vaccine Candidate Against Babesia divergens, Blood Parasite of Veterinary and Zoonotic Importance. Front Vet Sci 2022; 8:795906. [PMID: 35071386 PMCID: PMC8776984 DOI: 10.3389/fvets.2021.795906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Accepted: 11/22/2021] [Indexed: 11/18/2022] Open
Abstract
The molecular identification and antigenic characterization of P0 protein in Babesia divergens, a blood parasite of veterinary and zoonotic importance, were carried out in this study for use in developing subunit vaccines against B. divergens infection. Recombinant protein encoding P0 (BdP0) was developed in Escherichia coli, and its antiserum was generated in mice for further molecular characterization. Anti-rBdP0 serum had a specific interaction with the corresponding legitimate B. divergens protein, as confirmed by Western blotting and indirect fluorescent antibody tests. ELISA was used to assess the immunogenicity of BdP0 in a group of 68 bovine field samples, and significant immunological reactivity was found in 19 and 20 positive samples of rBdp0 and B. divergens lysate, respectively. The in vitro growth of B. divergens cultures treated with anti-rBdP0 serum was significantly inhibited (p < 0.05). Furthermore, after 6 h of incubation with 2 mg/ml anti-rBdP0 serum, the ability of pre-incubated free merozoites to invade bovine erythrocytes was reduced by 59.88%. The obtained data suggest the possible use of rBdP0 as diagnostic antigen and may serve as a vaccine candidate against babesiosis caused by B. divergens either in animal or human.
Collapse
Affiliation(s)
- Shimaa Abd El-Salam El-Sayed
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
- Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Mohamed Abdo Rizk
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
- Department of Internal Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
- *Correspondence: Mohamed Abdo Rizk
| | - Haitham Eldoumani
- Department of Anatomy, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
| | - Shimaa Sobhy Sorour
- Department of Parasitology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafr El-Shaikh, Egypt
| | - Mohamad Alaa Terkawi
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
| | - Mahmoud AbouLaila
- Department of Parasitology, Faculty of Veterinary Medicine, Damanhour University, Damanhour, Egypt
| | - Ikuo Igarashi
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Japan
- Ikuo Igarashi
| | - Mohamed Z. Sayed-Ahmed
- Department of Internal Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura, Egypt
- Department of Clinical Pharmacy, College of Pharmacy, Jazan University, Jizan, Saudi Arabia
| |
Collapse
|
3
|
Ito D, Chen JH, Takashima E, Hasegawa T, Otsuki H, Takeo S, Thongkukiatkul A, Han ET, Tsuboi T. Identification of a Novel RAMA/RON3 Rhoptry Protein Complex in Plasmodium falciparum Merozoites. Front Cell Infect Microbiol 2021; 10:605367. [PMID: 33537242 PMCID: PMC7848174 DOI: 10.3389/fcimb.2020.605367] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2020] [Accepted: 11/27/2020] [Indexed: 01/08/2023] Open
Abstract
Malaria causes a half a million deaths annually. The parasite intraerythrocytic lifecycle in the human bloodstream is the major cause of morbidity and mortality. Apical organelles of merozoite stage parasites are involved in the invasion of erythrocytes. A limited number of apical organellar proteins have been identified and characterized for their roles during erythrocyte invasion or subsequent intraerythrocytic parasite development. To expand the repertoire of identified apical organellar proteins we generated a panel of monoclonal antibodies against Plasmodium falciparum schizont-rich parasites and screened the antibodies using immunofluorescence assays. Out of 164 hybridoma lines, 12 clones produced monoclonal antibodies yielding punctate immunofluorescence staining patterns in individual merozoites in late schizonts, suggesting recognition of merozoite apical organelles. Five of the monoclonal antibodies were used to immuno-affinity purify their target antigens and these antigens were identified by liquid chromatography-tandem mass spectrometry (LC-MS/MS). Two known apical organelle protein complexes were identified, the high-molecular mass rhoptry protein complex (PfRhopH1/Clags, PfRhopH2, and PfRhopH3) and the low-molecular mass rhoptry protein complex (rhoptry-associated proteins complex, PfRAP1, and PfRAP2). A novel complex was additionally identified by immunoprecipitation, composed of rhoptry-associated membrane antigen (PfRAMA) and rhoptry neck protein 3 (PfRON3) of P. falciparum. We further identified a region spanning amino acids Q221-E481 within the PfRAMA that may associate with PfRON3 in immature schizonts. Further investigation will be required as to whether PfRAMA and PfRON3 interact directly or indirectly.
Collapse
Affiliation(s)
- Daisuke Ito
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan.,Division of Medical Zoology, Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Jun-Hu Chen
- National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention, Shanghai, China
| | - Eizo Takashima
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Tomoyuki Hasegawa
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| | - Hitoshi Otsuki
- Division of Medical Zoology, Department of Microbiology and Immunology, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Satoru Takeo
- Division of Tropical Diseases and Parasitology, Department of Infectious Diseases, Faculty of Medicine, Kyorin University, Mitaka, Japan
| | | | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Chuncheon, South Korea
| | - Takafumi Tsuboi
- Division of Malaria Research, Proteo-Science Center, Ehime University, Matsuyama, Japan
| |
Collapse
|
4
|
Schureck MA, Darling JE, Merk A, Shao J, Daggupati G, Srinivasan P, Olinares PDB, Rout MP, Chait BT, Wollenberg K, Subramaniam S, Desai SA. Malaria parasites use a soluble RhopH complex for erythrocyte invasion and an integral form for nutrient uptake. eLife 2021; 10:e65282. [PMID: 33393463 PMCID: PMC7840181 DOI: 10.7554/elife.65282] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Accepted: 01/04/2021] [Indexed: 12/12/2022] Open
Abstract
Malaria parasites use the RhopH complex for erythrocyte invasion and channel-mediated nutrient uptake. As the member proteins are unique to Plasmodium spp., how they interact and traffic through subcellular sites to serve these essential functions is unknown. We show that RhopH is synthesized as a soluble complex of CLAG3, RhopH2, and RhopH3 with 1:1:1 stoichiometry. After transfer to a new host cell, the complex crosses a vacuolar membrane surrounding the intracellular parasite and becomes integral to the erythrocyte membrane through a PTEX translocon-dependent process. We present a 2.9 Å single-particle cryo-electron microscopy structure of the trafficking complex, revealing that CLAG3 interacts with the other subunits over large surface areas. This soluble complex is tightly assembled with extensive disulfide bonding and predicted transmembrane helices shielded. We propose a large protein complex stabilized for trafficking but poised for host membrane insertion through large-scale rearrangements, paralleling smaller two-state pore-forming proteins in other organisms.
Collapse
Affiliation(s)
- Marc A Schureck
- Laboratory of Malaria and Vector Research, NIAID, National Institutes of HealthRockvilleUnited States
| | - Joseph E Darling
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesdaUnited States
| | - Alan Merk
- Laboratory of Cell Biology, Center for Cancer Research, National Cancer Institute, National Institutes of HealthBethesdaUnited States
| | - Jinfeng Shao
- Laboratory of Malaria and Vector Research, NIAID, National Institutes of HealthRockvilleUnited States
| | - Geervani Daggupati
- Department of Molecular Microbiology and Immunology, and Johns Hopkins Malaria Institute, Johns Hopkins Bloomberg School of Public HealthBaltimoreUnited States
| | - Prakash Srinivasan
- Department of Molecular Microbiology and Immunology, and Johns Hopkins Malaria Institute, Johns Hopkins Bloomberg School of Public HealthBaltimoreUnited States
| | - Paul Dominic B Olinares
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller UniversityNew YorkUnited States
| | - Michael P Rout
- Laboratory of Cellular and Structural Biology, The Rockefeller UniversityNew YorkUnited States
| | - Brian T Chait
- Laboratory of Mass Spectrometry and Gaseous Ion Chemistry, The Rockefeller UniversityNew YorkUnited States
| | - Kurt Wollenberg
- Office of Cyber Infrastructure & Computational Biology, National Institute of Allergy and Infectious Diseases, National Institutes of HealthBethesdaUnited States
| | - Sriram Subramaniam
- Department of Biochemistry and Molecular Biology, University of British ColumbiaVancouverCanada
| | - Sanjay A Desai
- Laboratory of Malaria and Vector Research, NIAID, National Institutes of HealthRockvilleUnited States
| |
Collapse
|
5
|
Plasmodium falciparum Clag9-Associated PfRhopH Complex Is Involved in Merozoite Binding to Human Erythrocytes. Infect Immun 2020; 88:IAI.00504-19. [PMID: 31712270 DOI: 10.1128/iai.00504-19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2019] [Accepted: 11/01/2019] [Indexed: 02/05/2023] Open
Abstract
Cytoadherence-linked asexual gene 9 (Clag9), a conserved Plasmodium protein expressed during the asexual blood stages, is involved in the cytoadherence of infected red blood cells (RBCs) to the endothelial lining of blood vessels. Here, we show that Plasmodium falciparum Clag9 (PfClag9) is a component of the PfClag9-RhopH complex that is involved in merozoite binding to human erythrocytes. To characterize PfClag9, we expressed four fragments of PfClag9, encompassing the entire protein. Immunostaining analysis using anti-PfClag9 antibodies showed expression and localization of PfClag9 at the apical end of the merozoites. Mass spectrometric analysis of merozoite extracts after immunoprecipitation using anti-PfClag9 antibody identified P. falciparum rhoptry-associated protein 1 (PfRAP1), PfRAP2, PfRAP3, PfRhopH2, and PfRhopH3 as associated proteins. The identified rhoptry proteins were expressed, and their association with PfClag9 domains was assessed by using protein-protein interaction tools. We further showed that PfClag9 binds human RBCs by interacting with the glycophorin A-band 3 receptor-coreceptor complex. In agreement with its cellular localization, PfClag9 was strongly recognized by antibodies generated during natural infection. Mice immunized with the C-terminal domain of PfClag9 were partially protected against a subsequent challenge infection with Plasmodium berghei, further supporting a biological role of PfClag9 during natural infection. Taken together, these results provide direct evidence for the existence of a PfRhopH-Clag9 complex on the Plasmodium merozoite surface that binds to human RBCs.
Collapse
|
6
|
Kochayoo P, Changrob S, Wangriatisak K, Lee SK, Chootong P, Han ET. The persistence of naturally acquired antibodies and memory B cells specific to rhoptry proteins of Plasmodium vivax in patients from areas of low malaria transmission. Malar J 2019; 18:382. [PMID: 31783870 PMCID: PMC6884809 DOI: 10.1186/s12936-019-3009-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Accepted: 11/16/2019] [Indexed: 01/07/2023] Open
Abstract
Background Rhoptries are the large, paired, secretory organelles located at the apical tip of the malaria merozoite that are considered important for parasite invasion processes. Plasmodium vivax rhoptry proteins have been shown to induce humoral immunity during natural infections. Therefore, these proteins may be potential novel vaccine candidates. However, there is a lack of data on the duration of antibody and memory B cell (MBC) responses. Here, the longitudinal analysis of antibody and MBC responses to the P. vivax rhoptry proteins PvRALP1-Ecto and PvRhopH2 were monitored and analysed in individuals to determine their persistence. Methods Thirty-nine samples from P. vivax-infected subjects (age 18–60 years) were recruited to explore the frequency and persistence of antibody and MBC responses against rhoptry proteins (PvRALP1-Ecto and PvRhopH2) using both cross-sectional and longitudinal cohort study designs. Antibody levels were determined by ELISA during clinical malaria, and at 3, 9 and 12 months post-infection. The frequency of MBC sub-sets and presence of rhoptry-specific MBCs in subjects 18 months after treatment were detected by flow cytometry and ELISPOT assay. Results The seroprevalence of antibodies against PvRALP1-Ecto and PvRhopH2 proteins was found to be high during acute infection, with IgG1, IgG2 and IgG3 sub-classes predominant. However, these anti-rhoptry responses were short-lived and significantly decreased at 9 months post-infection. To relate the durability of these antibody responses to MBC persistence at post-infection, 18-month post-infection peripheral blood mononuclear cells (PBMCs) samples were taken to detect rhoptry-specific MBCs and frequency of MBC sub-sets, and correlate with antibody responses. These late post-infection samples revealed that rhoptry-specific MBCs were present in about 70% of total subjects. However, the persistence of specific MBCs was not correlated with antibody responses as the majority of malaria subjects who were positive for PvRALP1-Ecto- or PvRhopH2-specific MBCs were seronegative for the rhoptry antigens. The frequencies of classical MBCs were increased after infection, whereas those of activated and atypical MBCs were decreased, indicating that MBC responses could switch from activated or atypical MBCs to classical MBCs after parasite clearance, and were maintained in blood circulating at post-infection. Conclusion The study showed that rhoptry antigens induced the development and persistence of MBC responses in P. vivax-infected subjects who lived in a region of low malaria transmission, which were not related to the longevity of antibody responses.
Collapse
Affiliation(s)
- Piyawan Kochayoo
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Siriruk Changrob
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Kittikorn Wangriatisak
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand
| | - Seong Kyun Lee
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 200-701, Republic of Korea
| | - Patchanee Chootong
- Department of Clinical Microbiology and Applied Technology, Faculty of Medical Technology, Mahidol University, Bangkok, 10700, Thailand.
| | - Eun-Taek Han
- Department of Medical Environmental Biology and Tropical Medicine, School of Medicine, Kangwon National University, Chuncheon, Gangwon-do, 200-701, Republic of Korea.
| |
Collapse
|
7
|
CLAG3 Self-Associates in Malaria Parasites and Quantitatively Determines Nutrient Uptake Channels at the Host Membrane. mBio 2018; 9:mBio.02293-17. [PMID: 29739907 PMCID: PMC5941077 DOI: 10.1128/mbio.02293-17] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Malaria parasites increase host erythrocyte permeability to ions and nutrients via a broad-selectivity channel known as the plasmodial surface anion channel (PSAC), linked to parasite-encoded CLAG3 and two associated proteins. These proteins lack the multiple transmembrane domains typically present in channel-forming proteins, raising doubts about their precise roles. Using the virulent human Plasmodium falciparum parasite, we report that CLAG3 undergoes self-association and that this protein’s expression determines channel phenotype quantitatively. We overcame epigenetic silencing of clag3 paralogs and engineered parasites that express two CLAG3 isoforms simultaneously. Stoichiometric expression of these isoforms yielded intermediate channel phenotypes, in agreement with observed trafficking of both proteins to the host membrane. Coimmunoprecipitation and surface labeling revealed formation of CLAG3 oligomers. In vitro selections applied to these transfectant lines yielded distinct mutants with correlated changes in channel activity. These findings support involvement of the identified oligomers in PSAC formation and parasite nutrient acquisition. Malaria parasites are globally important pathogens that evade host immunity by replicating within circulating erythrocytes. To facilitate intracellular growth, these parasites increase erythrocyte nutrient uptake through an unusual ion channel. The parasite CLAG3 protein is a key determinant of this channel, but its lack of homology to known ion channels has raised questions about possible mechanisms. Using a new method that allows simultaneous expression of two different CLAG3 proteins, we identify self-association of CLAG3. The two expressed isoforms faithfully traffic to and insert in the host membrane, while remaining associated with two unrelated parasite proteins. Both the channel phenotypes and molecular changes produced upon selections with a highly specific channel inhibitor are consistent with a multiprotein complex that forms the nutrient pore. These studies support direct involvement of the CLAG3 protein in channel formation and are relevant to antimalarial drug discovery projects targeting parasite nutrient acquisition.
Collapse
|
8
|
El-Sayed SAES, Rizk MA, Terkawi MA, Yokoyama N, Igarashi I. Molecular identification and antigenic characterization of Babesia divergens Erythrocyte Binding Protein (BdEBP) as a potential vaccine candidate. Parasitol Int 2017; 66:721-726. [PMID: 28743470 DOI: 10.1016/j.parint.2017.07.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2017] [Revised: 06/07/2017] [Accepted: 07/17/2017] [Indexed: 10/19/2022]
Abstract
Host cell invasion is the only step where Babesia parasites are extracellular, and their survival is menaced during this step. Therefore, interfering with this critical stage is a target for an anti-Babesia intervention strategy. In this regard, recombinant protein encoding Babesia divergens Erythrocyte Binding Protein (BdEBP) was produced in Escherichia coli in the current study, and its antiserum was prepared in mice for further molecular characterization. Western blotting and indirect fluorescent antibody test (IFAT) revealed the specific reaction of the anti-rBdEBP serum with a corresponding authentic protein of B. divergens. Next, bovine RBCs were incubated with a B. divergens lysate, and anti-rBdEBP serum was produced in mice to detect the ability of BdEBP to bind with host cells. Bands corresponding to 29.6-kDa proteins in the protein-bound erythrocyte lysate were detected by specific immune rBdEBP using Western blotting. These results suggest that BdEBP is functional in the merozoite stage and may be involved in attachment to bovine RBCs. A significant inhibition of the in vitro growth of B. divergens culture treated with anti-rBdEBP serum was observed. Moreover, the efficacy of pre-incubated free merozoites to invade bovine erythrocytes was inhibited by 60% after incubation with 2mg/ml of anti-rBdEBP serum for 6h. The obtained data suggest the possible use of rBdEBP as a vaccine candidate against bovine babesiosis.
Collapse
Affiliation(s)
- Shimaa Abd El-Salam El-Sayed
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-Cho, Obihiro, Hokkaido, Japan; Department of Biochemistry and Chemistry of Nutrition, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Mohamed Abdo Rizk
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-Cho, Obihiro, Hokkaido, Japan; Department of Internal Medicine and Infectious Diseases, Faculty of Veterinary Medicine, Mansoura University, Mansoura 35516, Egypt
| | - Mohamad Alaa Terkawi
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-Cho, Obihiro, Hokkaido, Japan
| | - Naoaki Yokoyama
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-Cho, Obihiro, Hokkaido, Japan
| | - Ikuo Igarashi
- National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Inada-Cho, Obihiro, Hokkaido, Japan.
| |
Collapse
|
9
|
Ito D, Schureck MA, Desai SA. An essential dual-function complex mediates erythrocyte invasion and channel-mediated nutrient uptake in malaria parasites. eLife 2017; 6. [PMID: 28221136 PMCID: PMC5349850 DOI: 10.7554/elife.23485] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2016] [Accepted: 02/16/2017] [Indexed: 11/27/2022] Open
Abstract
Malaria parasites evade immune detection by growth and replication within erythrocytes. After erythrocyte invasion, the intracellular pathogen must increase host cell uptake of nutrients from plasma. Here, we report that the parasite-encoded RhopH complex contributes to both invasion and channel-mediated nutrient uptake. As rhoph2 and rhoph3 gene knockouts were not viable in the human P. falciparum pathogen, we used conditional knockdowns to determine that the encoded proteins are essential and to identify their stage-specific functions. We exclude presumed roles for RhopH2 and CLAG3 in erythrocyte invasion but implicate a RhopH3 contribution either through ligand-receptor interactions or subsequent parasite internalization. These proteins then traffic via an export translocon to the host membrane, where they form a nutrient channel. Knockdown of either RhopH2 or RhopH3 disrupts the entire complex, interfering with organellar targeting and subsequent trafficking. Therapies targeting this complex should attack the pathogen at two critical points in its cycle. DOI:http://dx.doi.org/10.7554/eLife.23485.001 The parasites that cause malaria in humans and other animals infect and live inside red blood cells to escape attack by their hosts’ immune systems. Malaria parasites grow and multiply in red blood cells before bursting out and invading new red blood cells. To fuel this growth, the parasite needs access to sugars and other nutrients that are found outside in the bloodstream. Malaria parasites achieve this by inserting some of their own proteins into the membrane of the red blood cell to form an unusual channel that allows the nutrients to enter the cell. A parasite protein called CLAG3 (also known as RhopH1) is involved in formation of the unusual nutrient channel. Unlike most other proteins, malaria parasites make the CLAG3 protein while they are inside one cell and release it later when they invade a new red blood cell. The CLAG3 protein also binds to two other parasite proteins, called RhopH2 and RhopH3, to form a larger protein complex. However, it was not known what roles these other proteins played, or why the complex was made in the preceding red blood cell. Ito et al. have now addressed these unknowns by editing the genes of the parasite that causes the most dangerous form of malaria in people, a parasite called Plasmodium falciparum. These experiments revealed that the parasites could still invade host cells as normal if they lost CLAG3 and RhopH2. This suggests, that contrary to what was expected, CLAG3 and RhopH2 are not needed for the invasion process. Instead, the experiments revealed that RhopH3 serves a major role in invasion, either by helping the parasite to interact with or enter the new red blood cell. After the parasite has invaded the cell, this complex of three proteins is shuttled to the red blood cell’s membrane, where it inserts to help form the nutrient channel. The findings of Ito et al. reveal that one protein complex serves two unrelated but essential roles at different locations and time points in the life cycle of a malaria parasite. Since a parasite will not survive if it cannot enter a host cell and obtain nutrients, interfering with these processes by targeting this protein complex could lead to new therapies against malaria in the future. DOI:http://dx.doi.org/10.7554/eLife.23485.002
Collapse
Affiliation(s)
- Daisuke Ito
- Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, United States
| | - Marc A Schureck
- Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, United States
| | - Sanjay A Desai
- Laboratory of Malaria and Vector Research, NIAID, National Institutes of Health, Rockville, United States
| |
Collapse
|
10
|
A CLAG3 mutation in an amphipathic transmembrane domain alters malaria parasite nutrient channels and confers leupeptin resistance. Infect Immun 2015; 83:2566-74. [PMID: 25870226 DOI: 10.1128/iai.02966-14] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2014] [Accepted: 04/04/2015] [Indexed: 11/20/2022] Open
Abstract
Erythrocytes infected with malaria parasites have increased permeability to ions and nutrients, as mediated by the plasmodial surface anion channel (PSAC) and recently linked to parasite clag3 genes. Although the encoded protein is integral to the host membrane, its precise contribution to solute transport remains unclear because it lacks conventional transmembrane domains and does not have homology to ion channel proteins in other organisms. Here, we identified a probable CLAG3 transmembrane domain adjacent to a variant extracellular motif. Helical-wheel analysis revealed strict segregation of polar and hydrophobic residues to opposite faces of a predicted α-helical transmembrane domain, suggesting that the domain lines a water-filled pore. A single CLAG3 mutation (A1210T) in a leupeptin-resistant PSAC mutant falls within this transmembrane domain and may affect pore structure. Allelic-exchange transfection and site-directed mutagenesis revealed that this mutation alters solute selectivity in the channel. The A1210T mutation also reduces the blocking affinity of PSAC inhibitors that bind on opposite channel faces, consistent with global changes in channel structure. Transfected parasites carrying this mutation survived a leupeptin challenge significantly better than a transfection control did. Thus, the A1210T mutation contributes directly to both altered PSAC activity and leupeptin resistance. These findings reveal the molecular basis of a novel antimalarial drug resistance mechanism, provide a framework for determining the channel's composition and structure, and should guide the development of therapies targeting the PSAC.
Collapse
|
11
|
Zhao X, Chang Z, Tu Z, Yu S, Wei X, Zhou J, Lu H, Jiang N, Chen Q. PfRON3 is an erythrocyte-binding protein and a potential blood-stage vaccine candidate antigen. Malar J 2014; 13:490. [PMID: 25495792 PMCID: PMC4295329 DOI: 10.1186/1475-2875-13-490] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2014] [Accepted: 12/09/2014] [Indexed: 12/20/2022] Open
Abstract
Background Erythrocyte invasion by merozoites is an essential step in Plasmodium falciparum infection and leads to subsequent disease pathology. Proteins both on the merozoite surface and secreted from the apical organelles (micronemes, rhoptries and dense granules) mediate the invasion of erythrocytes; some of the molecules have been regarded as targets in the development of an anti-malaria vaccine. Recently, a subgroup of rhoptry neck proteins (PfRON2, PfRON4 and PfRON5) associated with the microneme protein apical membrane antigen AMA1 has been described as components of the moving junction complex that assists merozoite invasion into erythrocytes. However, unlike PfRON2, PfRON4 and PfRON5, the latest study suggested that PfRON3 might be located in the rhoptry bulb and participates in a novel PfRON complex (PfRON2, 3 and 4), but does not form a complex with AMA1. Additionally, the full-length PfRON3 protein possesses three transmembrane regions at the N-terminus, which is highly conserved among RON3 orthologues in the genus Plasmodium, Toxoplasma gondii and Eimeria tenella. Overall, these findings suggest that PfRON3 may play an important role in merozoite invasion into erythrocytes. Results PfRON3 was primarily expressed during the late trophozoite stage, with a peak in transcription levels at 40 hours post-invasion. The subcellular localization of PfRON3 was confirmed that it is a merozoite rhoptry bulb protein. Additionally, the recombinant form of PfRON3 protein bound to the erythrocyte and was recognized by sera collected from malaria endemic areas in Africa, and anti-PfRON3 antibodies significantly inhibited merozoite invasion into erythrocytes. Methods The expression of PfRON3 was analysed via real-time quantitative PCR, and the recombinant PfRON3 proteins were generated with an Escherichia coli expression system. The subcellular localization of PfRON3 was assessed with immunoelectron microscopy and immunofluorescence assay (IFA). The recognition PfRON3 by malaria immune sera was analysed with an enzyme-linked immunosorbent assay (ELISA). Erythrocyte-binding assays were performed using recombinant PfRON3 proteins and invasion inhibition assays were carried out with PfRON3-specific antibodies. Conclusion This study confirmed that PfRON3 is a rhoptry protein with an erythrocyte-binding property, which is likely associated red blood cell invasion. PfRON3 is a potential vaccine candidate. Electronic supplementary material The online version of this article (doi:10.1186/1475-2875-13-490) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Ning Jiang
- Key Laboratory of Zoonosis, Jilin University, Xi An Da Lu 5333, Changchun 5333, China.
| | | |
Collapse
|
12
|
The conserved clag multigene family of malaria parasites: essential roles in host-pathogen interaction. Drug Resist Updat 2014; 18:47-54. [PMID: 25467627 DOI: 10.1016/j.drup.2014.10.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The clag multigene family is strictly conserved in malaria parasites, but absent from neighboring genera of protozoan parasites. Early research pointed to roles in merozoite invasion and infected cell cytoadherence, but more recent studies have implicated channel-mediated uptake of ions and nutrients from host plasma. Here, we review the current understanding of this gene family, which appears to be central to host-parasite interactions and an important therapeutic target.
Collapse
|
13
|
Rodriguez M, Alhassan A, Ord RL, Cursino-Santos JR, Singh M, Gray J, Lobo CA. Identification and characterization of the RouenBd1987 Babesia divergens Rhopty-Associated Protein 1. PLoS One 2014; 9:e107727. [PMID: 25226276 PMCID: PMC4166668 DOI: 10.1371/journal.pone.0107727] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2014] [Accepted: 08/20/2014] [Indexed: 11/19/2022] Open
Abstract
Human babesiosis is caused by one of several babesial species transmitted by ixodid ticks that have distinct geographical distributions based on the presence of competent animal hosts. The pathology of babesiosis, like malaria, is a consequence of the parasitaemia which develops through the cyclical replication of Babesia parasites in a patient's red blood cells, though symptoms typically are nonspecific. We have identified the gene encoding Rhoptry-Associated Protein -1 (RAP-1) from a human isolate of B. divergens, Rouen1987 and characterized its protein product at the molecular and cellular level. Consistent with other Babesia RAP-1 homologues, BdRAP-1 is expressed as a 46 kDa protein in the parasite rhoptries, suggesting a possible role in red cell invasion. Native BdRAP-1 binds to an unidentified red cell receptor(s) that appears to be non-sialylated and non-proteinacious in nature, but we do not find significant reduction in growth with anti-rRAP1 antibodies in vitro, highlighting the possibility the B. divergens is able to use alternative pathways for invasion, or there is an alternative, complementary, role for BdRAP-1 during the invasion process. As it is the parasite's ability to recognize and then invade host cells which is central to clinical disease, characterising and understanding the role of Babesia-derived proteins involved in these steps are of great interest for the development of an effective prophylaxis.
Collapse
Affiliation(s)
- Marilis Rodriguez
- Department of Blood-Borne Parasites, New York Blood Center, New York, New York, United States of America
| | - Andy Alhassan
- Department of Blood-Borne Parasites, New York Blood Center, New York, New York, United States of America
| | - Rosalynn L. Ord
- Department of Blood-Borne Parasites, New York Blood Center, New York, New York, United States of America
| | - Jeny R. Cursino-Santos
- Department of Blood-Borne Parasites, New York Blood Center, New York, New York, United States of America
| | - Manpreet Singh
- Department of Blood-Borne Parasites, New York Blood Center, New York, New York, United States of America
| | - Jeremy Gray
- University College Dublin School of Biology and Environmental Science, Dublin, Republic of Ireland
| | - Cheryl A. Lobo
- Department of Blood-Borne Parasites, New York Blood Center, New York, New York, United States of America
- * E-mail:
| |
Collapse
|
14
|
High guanidinium permeability reveals dehydration-dependent ion selectivity in the plasmodial surface anion channel. BIOMED RESEARCH INTERNATIONAL 2014; 2014:741024. [PMID: 25243175 PMCID: PMC4160636 DOI: 10.1155/2014/741024] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 07/19/2014] [Accepted: 07/23/2014] [Indexed: 11/17/2022]
Abstract
Malaria parasites grow within vertebrate erythrocytes and increase host cell permeability to access nutrients from plasma. This increase is mediated by the plasmodial surface anion channel (PSAC), an unusual ion channel linked to the conserved clag gene family. Although PSAC recognizes and transports a broad range of uncharged and charged solutes, it must efficiently exclude the small Na+ ion to maintain infected cell osmotic stability. Here, we examine possible mechanisms for this remarkable solute selectivity. We identify guanidinium as an organic cation with high permeability into human erythrocytes infected with Plasmodium falciparum, but negligible uptake by uninfected cells. Transport characteristics and pharmacology indicate that this uptake is specifically mediated by PSAC. The rank order of organic and inorganic cation permeabilities suggests cation dehydration as the rate-limiting step in transport through the channel. The high guanidinium permeability of infected cells also allows rapid and stringent synchronization of parasite cultures, as required for molecular and cellular studies of this pathogen. These studies provide important insights into how nutrients and ions are transported via PSAC, an established target for antimalarial drug development.
Collapse
|
15
|
Nguitragool W, Rayavara K, Desai SA. Proteolysis at a specific extracellular residue implicates integral membrane CLAG3 in malaria parasite nutrient channels. PLoS One 2014; 9:e93759. [PMID: 24699906 PMCID: PMC3974804 DOI: 10.1371/journal.pone.0093759] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2013] [Accepted: 03/09/2014] [Indexed: 11/24/2022] Open
Abstract
The plasmodial surface anion channel mediates uptake of nutrients and other solutes into erythrocytes infected with malaria parasites. The clag3 genes of P. falciparum determine this channel’s activity in human malaria, but how the encoded proteins contribute to transport is unknown. Here, we used proteases to examine the channel’s composition and function. While proteases with distinct specificities all cleaved within an extracellular domain of CLAG3, they produced differing degrees of transport inhibition. Chymotrypsin-induced inhibition depended on parasite genotype, with channels induced by the HB3 parasite affected to a greater extent than those of the Dd2 clone. Inheritance of functional proteolysis in the HB3×Dd2 genetic cross, DNA transfection, and gene silencing experiments all pointed to the clag3 genes, providing independent evidence for a role of these genes. Protease protection assays with a Dd2-specific inhibitor and site-directed mutagenesis revealed that a variant L1115F residue on a CLAG3 extracellular loop contributes to inhibitor binding and accounts for differences in functional proteolysis. These findings indicate that surface-exposed CLAG3 is the relevant pool of this protein for channel function. They also suggest structural models for how exposed CLAG3 domains contribute to pore formation and parasite nutrient uptake.
Collapse
Affiliation(s)
- Wang Nguitragool
- The Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
- * E-mail: (WN); (SAD)
| | - Kempaiah Rayavara
- The Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
| | - Sanjay A. Desai
- The Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, Maryland, United States of America
- * E-mail: (WN); (SAD)
| |
Collapse
|
16
|
Why do malaria parasites increase host erythrocyte permeability? Trends Parasitol 2014; 30:151-9. [PMID: 24507014 DOI: 10.1016/j.pt.2014.01.003] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2013] [Revised: 01/06/2014] [Accepted: 01/09/2014] [Indexed: 11/21/2022]
Abstract
Malaria parasites increase erythrocyte permeability to diverse solutes including anions, some cations, and organic solutes, as characterized with several independent methods. Over the past decade, patch-clamp studies have determined that the permeability results from one or more ion channels on the infected erythrocyte host membrane. However, the biological role(s) served by these channels, if any, remain controversial. Recent studies implicate the plasmodial surface anion channel (PSAC) and a role in parasite nutrient acquisition. A debated alternative role in remodeling host ion composition for the benefit of the parasite appears to be nonessential. Because both channel activity and the associated clag3 genes are strictly conserved in malaria parasites, channel-mediated permeability is an attractive target for development of new therapies.
Collapse
|
17
|
Costa JDN, Zanchi FB, Rodrigues FLDS, Honda ER, Katsuragawa TH, Pereira DB, Taborda RLM, Tada MS, Ferreira RDGM, Pereira-da-Silva LH. Cross-reactive anti-PfCLAG9 antibodies in the sera of asymptomatic parasite carriers of Plasmodium vivax. Mem Inst Oswaldo Cruz 2013; 108:98-105. [PMID: 23440122 PMCID: PMC3974312 DOI: 10.1590/s0074-02762013000100016] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2012] [Accepted: 10/31/2012] [Indexed: 11/22/2022] Open
Abstract
The PfCLAG9 has been extensively studied because their immunogenicity. Thereby, the gene product is important for therapeutics interventions and a potential vaccine candidate. Antibodies against synthetic peptides corresponding to selected sequences of the Plasmodium falciparum antigen PfCLAG9 were found in sera of falciparum malaria patients from Rondônia, in the Brazilian Amazon. Much higher antibody titres were found in semi-immune and immune asymptomatic parasite carriers than in subjects suffering clinical infections, corroborating original findings in Papua Guinea. However, sera of Plasmodium vivax patients from the same Amazon area, in particular from asymptomatic vivax parasite carriers, reacted strongly with the same peptides. Bioinformatic analyses revealed regions of similarity between P. falciparum Pfclag9 and the P. vivax ortholog Pvclag7. Indirect fluorescent microscopy analysis showed that antibodies against PfCLAG9 peptides elicited in BALB/c mice react with human red blood cells (RBCs) infected with both P. falciparum and P. vivax parasites. The patterns of reactivity on the surface of the parasitised RBCs are very similar. The present observations support previous findings that PfCLAG9 may be a target of protective immune responses and raises the possibility that the cross reactive antibodies to PvCLAG7 in mixed infections play a role in regulate the fate of Plasmodium mixed infections.
Collapse
|
18
|
Zheng L, Feng H, Liu D, Pan YY, Cao YM. The expression of malarial invasion-related molecules is affected by two different nitric oxide-based treatments. Folia Parasitol (Praha) 2013; 60:213-7. [PMID: 23951927 DOI: 10.14411/fp.2013.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The host immune response to parasitic infections plays an important role in controlling multiplication of the parasite and reducing clinical symptoms and life-threatening complications. Nitric oxide (NO), an important innate immune factor and classic Th1 immune effector, may play a role in inhibiting plasmodium infection. In this study, we used two different approaches (L-Arginine [precursor of NO] and NOC5 [short-time NO donor]) to prove the roles of NO in malaria infection. We used 6-8 week-old female BALB/c mice infected with the rodent malaria Plasmodium yoelii Landau, Michel et Adam, 1968 - strain 17XL (P.y17XL) as a model. For L-Arg treatment, mice were administered with an oral dose of 1.5 mg/g L-Arg daily for seven consecutive days prior to infection with Py17XL. L-Arg pretreatment resulted in the decrease of the mRNA level of the apical membrane antigen 1 (AMA1) gene, which encodes a protein involved in host invasion. For NOC5 treatment, NOC5 was injected intraperitoneally into the P.y17XL infected mice on day 5 post-infection or incubated in vitro with purified Py17XL schizonts. Both in vivo and in vitro treatments with NOC5 led to down-regulation of the transcript and protein levels of invasion-related molecules (AMA1, merozoites surface protein 1 and Py235). Our results confirmed the protective role of NO in the asexual blood stage of parasitic infection, which may be partially due to reduced expression of parasite invasion molecules.
Collapse
Affiliation(s)
- Li Zheng
- Department of Immunology, College of Basic Medical Sciences, China Medical University, Heping District, Shenyang, China
| | | | | | | | | |
Collapse
|
19
|
Sharma P, Wollenberg K, Sellers M, Zainabadi K, Galinsky K, Moss E, Nguitragool W, Neafsey D, Desai SA. An epigenetic antimalarial resistance mechanism involving parasite genes linked to nutrient uptake. J Biol Chem 2013; 288:19429-40. [PMID: 23720749 DOI: 10.1074/jbc.m113.468371] [Citation(s) in RCA: 64] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Acquired antimalarial drug resistance produces treatment failures and has led to periods of global disease resurgence. In Plasmodium falciparum, resistance is known to arise through genome-level changes such as mutations and gene duplications. We now report an epigenetic resistance mechanism involving genes responsible for the plasmodial surface anion channel, a nutrient channel that also transports ions and antimalarial compounds at the host erythrocyte membrane. Two blasticidin S-resistant lines exhibited markedly reduced expression of clag genes linked to channel activity, but had no genome-level changes. Silencing aborted production of the channel protein and was directly responsible for reduced uptake. Silencing affected clag paralogs on two chromosomes and was mediated by specific histone modifications, allowing a rapidly reversible drug resistance phenotype advantageous to the parasite. These findings implicate a novel epigenetic resistance mechanism that involves reduced host cell uptake and is a worrisome liability for water-soluble antimalarial drugs.
Collapse
Affiliation(s)
- Paresh Sharma
- Laboratory of Malaria and Vector Research, Office of Cyber Infrastructure and Computational Biology, NIAID, National Institutes of Health, Bethesda, Maryland 20852, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Zhang Y, Jiang N, Lu H, Hou N, Piao X, Cai P, Yin J, Wahlgren M, Chen Q. Proteomic analysis of Plasmodium falciparum schizonts reveals heparin-binding merozoite proteins. J Proteome Res 2013; 12:2185-93. [PMID: 23566259 DOI: 10.1021/pr400038j] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
The malaria parasite Plasmodium falciparum utilizes host glycosaminoglycans (GAGs) as receptors for erythrocyte invasion and intravascular sequestration. Heparin and heparan sulfate (HS) are GAGs which can block erythrocyte invasion of the P. falciparum merozoite, albeit the molecular mechanisms remain poorly understood. Characterization of these heparin-binding merozoite proteins and key ligands in the host-parasite interplay will lead to a better understanding of the mechanism of erythrocyte invasion by malaria parasites. Here, schizont-derived proteins that bind heparin were enriched by affinity chromatography, and 6062 peptides from 811 P. falciparum-derived proteins were identified by two-dimensional liquid chromatography-mass spectrometry (LC/LC-MS/MS). The proteins were categorized into 14 functional groups ranging from pathogenesis, protein catabolic process to signal transduction. Proteins with predominant peptide counts were found to mainly originate from the rhoptry organelle of merozoites and the parasitized erythrocyte membrane. The profile of the heparin/HS-binding proteome of P. falciparum suggests they have important functions in the biology of the parasite.
Collapse
Affiliation(s)
- Yan Zhang
- Key Laboratory of Zoonosis, Ministry of Education, Jilin University, Xian Da Lu 5333, Changchun 130062, China
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Ghoneim AM. Trafficking of Plasmodium falciparum chimeric rhoptry protein with Brefeldin A. Folia Parasitol (Praha) 2013; 60:75-8. [DOI: 10.14411/fp.2013.009] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
|
22
|
Kemp LE, Yamamoto M, Soldati-Favre D. Subversion of host cellular functions by the apicomplexan parasites. FEMS Microbiol Rev 2012. [PMID: 23186105 DOI: 10.1111/1574-6976.12013] [Citation(s) in RCA: 74] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Rhoptries are club-shaped secretory organelles located at the anterior pole of species belonging to the phylum of Apicomplexa. Parasites of this phylum are responsible for a huge burden of disease in humans and animals and a loss of economic productivity. Members of this elite group of obligate intracellular parasites include Plasmodium spp. that cause malaria and Cryptosporidium spp. that cause diarrhoeal disease. Although rhoptries are almost ubiquitous throughout the phylum, the relevance and role of the proteins contained within the rhoptries varies. Rhoptry contents separate into two intra-organellar compartments, the neck and the bulb. A number of rhoptry neck proteins are conserved between species and are involved in functions such as host cell invasion. The bulb proteins are less well-conserved and probably evolved for a particular lifestyle. In the majority of species studied to date, rhoptry content is involved in formation and maintenance of the parasitophorous vacuole; however some species live free within the host cytoplasm. In this review, we will summarise the knowledge available regarding rhoptry proteins. Specifically, we will discuss the role of the rhoptry kinases that are used by Toxoplasma gondii and other coccidian parasites to subvert the host cellular functions and prevent parasite death.
Collapse
Affiliation(s)
- Louise E Kemp
- Department of Microbiology and Molecular Medicine, Faculty of Medicine, University of Geneva, Geneva, Switzerland.
| | | | | |
Collapse
|
23
|
Wang B, Lu F, Cheng Y, Li J, Ito D, Sattabongkot J, Tsuboi T, Han ET. Identification and characterization of the Plasmodium falciparum RhopH2 ortholog in Plasmodium vivax. Parasitol Res 2012; 112:585-93. [PMID: 23097184 DOI: 10.1007/s00436-012-3170-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2012] [Accepted: 10/08/2012] [Indexed: 11/28/2022]
Abstract
Plasmodium vivax is one of the most important human malaria species that is geographically widely endemic and potentially affects a larger number of people than its more notorious cousin, Plasmodium falciparum. During invasion of red blood cells, the parasite requires the intervention of high molecular weight complex rhoptry proteins (RhopH) that are also essential for cytoadherence. PfRhopH2, a member of the RhopH multigene family, has been characterized as being crucial during P. falciparum infection. This study describes identifying and characterizing the pfrhoph2 orthologous gene in P. vivax (hereinafter named pvrhoph2). The PvRhopH2 is a 1,369-amino acid polypeptide encoded by PVX_099930 gene, for which orthologous genes have been identified in other Plasmodium species by bioinformatic approaches. Both P. falciparum and P. vivax genes contain nine introns, and there is a high degree of similarity between the deduced amino acid sequences of the two proteins. Moreover, PvRhopH2 contains a signal peptide at its N-terminus and 12 cysteines predominantly in its C-terminal half. PvRhopH2 is localized in one of the apical organelles of the merozoite, the rhoptry, and the localization pattern is similar to that of PfRhopH2 in P. falciparum. The recombinant PvRhopH2 protein is recognized by serum antibodies of patients naturally exposed to P. vivax, suggesting that PvRhopH2 is immunogenic in humans.
Collapse
Affiliation(s)
- Bo Wang
- Department of Medical Environmental Biology and Tropical Medicine, Kangwon National University School of Medicine, Hyoja2-dong, Chuncheon, Gangwon-do 200-701, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Xangsayarath P, Kaewthamasorn M, Yahata K, Nakazawa S, Sattabongkot J, Udomsangpetch R, Kaneko O. Positive diversifying selection on the Plasmodium falciparum surf4.1 gene in Thailand. Trop Med Health 2012; 40:79-89. [PMID: 23264727 PMCID: PMC3521052 DOI: 10.2149/tmh.2012-12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2012] [Accepted: 05/13/2012] [Indexed: 11/18/2022] Open
Abstract
Plasmodium falciparum SURFIN4.1 is a type I transmembrane protein thought to locate on the merozoite surface and to be responsible for a reversible adherence to the erythrocyte before invasion. In this study, we evaluated surf4.1 gene segment encoding extracellular region for polymorphism, the signature of positive selection, the degree of linkage disequilibrium, and temporal change in allele frequency distribution in P. falciparum isolates from Thailand in 1988–89, 2003, and 2005. We found that SURFIN4.1 is highly polymorphic, particularly at the C-terminal side of the variable region located just before a predicted transmembrane region. A signature of positive diversifying selection on the variable region was detected by multiple tests and, to a lesser extent, on conserved N-terminally located cysteine-rich domain by Tajima’s D test. Linkage disequilibrium between sites over a long distance (> 1.5 kb) was detected, and multiple SURFIN4.1 haplotype sequences detected in 1988/89 still circulated in 2003. Few of the single amino acid polymorphism allele frequency distributions were significantly different between the 1988/89 and 2003 groups, suggesting that the frequency distribution of SURFIN4.1 extracellular region remained stable over 14 years.
Collapse
Affiliation(s)
- Phonepadith Xangsayarath
- Department of Protozoology, Institute of Tropical Medicine (NEKKEN) and the Global COE Program, Nagasaki University, Sakamoto, Nagasaki 852-8523, Japan ; National Institute of Public Health, Vientiane, Lao PDR
| | | | | | | | | | | | | |
Collapse
|
25
|
Global kinomic and phospho-proteomic analyses of the human malaria parasite Plasmodium falciparum. Nat Commun 2011; 2:565. [PMID: 22127061 DOI: 10.1038/ncomms1558] [Citation(s) in RCA: 264] [Impact Index Per Article: 18.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2011] [Accepted: 10/19/2011] [Indexed: 12/23/2022] Open
Abstract
The role of protein phosphorylation in the life cycle of malaria parasites is slowly emerging. Here we combine global phospho-proteomic analysis with kinome-wide reverse genetics to assess the importance of protein phosphorylation in Plasmodium falciparum asexual proliferation. We identify 1177 phosphorylation sites on 650 parasite proteins that are involved in a wide range of general cellular activities such as DNA synthesis, transcription and metabolism as well as key parasite processes such as invasion and cyto-adherence. Several parasite protein kinases are themselves phosphorylated on putative regulatory residues, including tyrosines in the activation loop of PfGSK3 and PfCLK3; we show that phosphorylation of PfCLK3 Y526 is essential for full kinase activity. A kinome-wide reverse genetics strategy identified 36 parasite kinases as likely essential for erythrocytic schizogony. These studies not only reveal processes that are regulated by protein phosphorylation, but also define potential anti-malarial drug targets within the parasite kinome.
Collapse
|
26
|
Nguitragool W, Bokhari AA, Pillai AD, Rayavara K, Sharma P, Turpin B, Aravind L, Desai SA. Malaria parasite clag3 genes determine channel-mediated nutrient uptake by infected red blood cells. Cell 2011; 145:665-77. [PMID: 21620134 PMCID: PMC3105333 DOI: 10.1016/j.cell.2011.05.002] [Citation(s) in RCA: 194] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2010] [Revised: 03/21/2011] [Accepted: 05/02/2011] [Indexed: 12/12/2022]
Abstract
Development of malaria parasites within vertebrate erythrocytes requires nutrient uptake at the host cell membrane. The plasmodial surface anion channel (PSAC) mediates this transport and is an antimalarial target, but its molecular basis is unknown. We report a parasite gene family responsible for PSAC activity. We used high-throughput screening for nutrient uptake inhibitors to identify a compound highly specific for channels from the Dd2 line of the human pathogen P. falciparum. Inheritance of this compound's affinity in a Dd2 × HB3 genetic cross maps to a single parasite locus on chromosome 3. DNA transfection and in vitro selections indicate that PSAC-inhibitor interactions are encoded by two clag3 genes previously assumed to function in cytoadherence. These genes are conserved in plasmodia, exhibit expression switching, and encode an integral protein on the host membrane, as predicted by functional studies. This protein increases host cell permeability to diverse solutes.
Collapse
Affiliation(s)
- Wang Nguitragool
- The Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Abdullah A.B. Bokhari
- The Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Ajay D. Pillai
- The Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Kempaiah Rayavara
- The Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Paresh Sharma
- The Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| | - Brad Turpin
- National Instruments, Inc., Austin, TX 78730, USA
| | - L. Aravind
- National Center for Biotechnology Information, National Library of Medicine, National Institutes of Health, Bethesda, MD 20892, USA
| | - Sanjay A. Desai
- The Laboratory of Malaria and Vector Research, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD 20852, USA
| |
Collapse
|
27
|
Moreno-Perez DA, Mongui A, Soler LN, Sanchez-Ladino M, Patarroyo MA. Identifying and characterizing a member of the RhopH1/Clag family in Plasmodium vivax. Gene 2011; 481:17-23. [PMID: 21513780 DOI: 10.1016/j.gene.2011.03.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2011] [Revised: 03/05/2011] [Accepted: 03/28/2011] [Indexed: 10/18/2022]
Abstract
Plasmodium vivax malaria caused is a public health problem that produces very high morbidity worldwide. During invasion of red blood cells the parasite requires the intervention of high molecular weight complex rhoptry proteins that are also essential for cytoadherence. PfClag9, a member of the RhopH multigene family, has been identified as being critical during Plasmodium falciparum infection. This study describes identifying and characterizing the pfclag9 ortholog in P. vivax (hereinafter named pvclag7). The pvclag7 gene is transcribed at the end of the intraerythrocytic cycle and is recognized by sera from humans who have been infected by P. vivax. PvClag7 subcellular localization has been also determined and, similar to what occurs with PfClag9, it co-localize with other proteins from the Rhoptry high molecular weight complex.
Collapse
Affiliation(s)
- Darwin A Moreno-Perez
- Fundación Instituto de Inmunología de Colombia (FIDIC), Carrera 50 No. 26-20, Bogotá, Colombia.
| | | | | | | | | |
Collapse
|
28
|
Crowley VM, Rovira-Graells N, Ribas de Pouplana L, Cortés A. Heterochromatin formation in bistable chromatin domains controls the epigenetic repression of clonally variant Plasmodium falciparum genes linked to erythrocyte invasion. Mol Microbiol 2011; 80:391-406. [PMID: 21306446 DOI: 10.1111/j.1365-2958.2011.07574.x] [Citation(s) in RCA: 59] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Clonally variant gene expression is a common survival strategy used by many pathogens, including the malaria parasite Plasmodium falciparum. Among the genes that show variant expression in this parasite are several members of small gene families linked to erythrocyte invasion, including the clag and eba families. The active or repressed state of these genes is clonally transmitted by epigenetic mechanisms. Here we characterized the promoters of clag3.1, clag3.2 and eba-140, and compared nuclease accessibility and post-translational histone modifications between their active and repressed states. Activity of these promoters in an episomal context is similar between parasite subclones characterized by different patterns of expression of the endogenous genes. Variant expression is controlled by the euchromatic or heterochromatic state of bistable chromatin domains. Repression is mediated by H3K9me3-based heterochromatin, whereas the active state is characterized by H3K9ac. These marks are maintained throughout the asexual blood cycle to transmit the epigenetic memory. Furthermore, eba-140 is organized in two distinct chromatin domains, probably separated by a barrier insulator located within its ORF. The 5' chromatin domain controls expression of the gene, whereas the 3' domain shares the chromatin conformation with the upstream region of the neighbouring phista family gene, which also shows variant expression.
Collapse
Affiliation(s)
- Valerie M Crowley
- Institute for Research in Biomedicine (IRB), 08028 Barcelona, Catalonia, Spain
| | | | | | | |
Collapse
|
29
|
Ito D, Han ET, Takeo S, Thongkukiatkul A, Otsuki H, Torii M, Tsuboi T. Plasmodial ortholog of Toxoplasma gondii rhoptry neck protein 3 is localized to the rhoptry body. Parasitol Int 2011; 60:132-8. [PMID: 21237287 DOI: 10.1016/j.parint.2011.01.001] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2010] [Revised: 12/24/2010] [Accepted: 01/07/2011] [Indexed: 11/24/2022]
Abstract
The proteins in apical organelles of Plasmodium falciparum merozoite play an important role in invasion into erythrocytes. Several rhoptry neck (RON) proteins have been identified in rhoptry proteome of the closely-related apicomplexan parasite, Toxoplasma gondii. Recently, three of P. falciparum proteins orthologous to TgRON proteins, PfRON2, 4 and 5, were found to be located in the rhoptry neck and interact with the micronemal protein apical membrane antigen 1 (PfAMA1) to form a moving junction complex that helps the invasion of merozoite into erythrocyte. However, the other P. falciparum RON proteins have yet to be characterized. Here, we determined that "PFL2505c" (hereafter referred to as pfron3) is the ortholog of the tgron3 in P. falciparum and characterized its protein expression profile, subcellular localization, and complex formation. Protein expression analysis revealed that PfRON3 was expressed primarily in late schizont stage parasites. Immunofluorescence microscopy (IFA) showed that PfRON3 localizes in the apical region of P. falciparum merozoites. Results from immunoelectron microscopy, along with IFA, clarified that PfRON3 localizes in the rhoptry body and not in the rhoptry neck. Even after erythrocyte invasion, PfRON3 was still detectable at the parasite ring stage in the parasitophorous vacuole. Moreover, co-immunoprecipitation studies indicated that PfRON3 interacts with PfRON2 and PfRON4, but not with PfAMA1. These results suggest that PfRON3 partakes in the novel PfRON complex formation (PfRON2, 3, and 4), but not in the moving junction complex (PfRON2, 4, 5, and PfAMA1). The novel PfRON complex, as well as the moving junction complex, might play a fundamental role in erythrocyte invasion by merozoite stage parasites.
Collapse
Affiliation(s)
- Daisuke Ito
- Cell-Free Science and Technology Research Center, Ehime University, Matsuyama, Ehime 790-8577, Japan
| | | | | | | | | | | | | |
Collapse
|
30
|
Curtidor H, Patiño LC, Arévalo-Pinzón G, Patarroyo ME, Patarroyo MA. Identification of the Plasmodium falciparum rhoptry neck protein 5 (PfRON5). Gene 2010; 474:22-8. [PMID: 21185360 DOI: 10.1016/j.gene.2010.12.005] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2010] [Revised: 12/04/2010] [Accepted: 12/14/2010] [Indexed: 10/18/2022]
Abstract
Gathering knowledge about the proteins involved in erythrocyte invasion by Plasmodium merozoites is the starting point for developing new strategies to control malarial disease. Many of these proteins have been studied in Toxoplasma gondii, where some belonging to the Moving Junction complex have been identified. This complex allows a strong interaction between host cell and parasite membranes, required for parasite invasion. In this genus, four rhoptry proteins (RON2, RON4, RON5 and RON8) and one micronemal protein (TgAMA-1) have been found as part of the complex. In Plasmodium falciparum, RON2 and RON4 have been characterized. In the present study, we identify PfRON5, a ~110 kDa protein which is expressed in merozoite and schizont stages of the FCB-2 strain.
Collapse
Affiliation(s)
- Hernando Curtidor
- Fundacion Instituto de Inmunologia de Colombia, Carrera 50 No. 26-20, Bogota, Colombia
| | | | | | | | | |
Collapse
|
31
|
García J, Curtidor H, Vanegas M, Arévalo-Pinzon G, Patarroyo MA, Patarroyo ME. Conserved regions of the Plasmodium falciparum rhoptry-associated protein 3 mediate specific host-pathogen interactions during invasion of red blood cells. Peptides 2010; 31:2165-72. [PMID: 20833215 DOI: 10.1016/j.peptides.2010.09.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/09/2010] [Revised: 09/01/2010] [Accepted: 09/02/2010] [Indexed: 11/25/2022]
Abstract
Invasion of red blood cells (RBCs) by the Plasmodium falciparum malaria merozoite is mediated by parasite surface molecules and proteins contained within apical organelles that are capable of recognizing receptors on the membrane of RBCs. The identification and characterization of these P. falciparum invasion-associated proteins is the first step for unveiling potential new drug and vaccine target molecules to eradicate this deadly disease. Among the exclusive set of malarial vaccine candidates, the members of the rhoptry-associated protein (RAP) family have been associated with the parasite's binding to and invasion of RBCs. Remarkably, the third member of this family (named RAP-3) has been recently detected on the surface of non-infected RBCs exposed to free merozoites, therefore suggesting the participation of this protein during RBC infection. In this study, the sequence of RAP-3 was finely mapped using synthetic peptides in order to identify which are the specific binding regions involved in RAP3-RBC interactions. Two high-activity binding peptides (HABPs) established high affinity interactions with RBC surface molecules of about 27-90 kDa, which were differentially affected by different enzymatic treatments. RAP-1 and RAP-2 HABPs inhibited binding of RAP-3 HABPs to different extents, thus suggesting the recognition of similar binding sites on RBC membrane, as well as ability of RAP-3 HABPs to inhibit P. falciparum infection in vitro. Altogether, these functional analyses of RAP-3 HABPs strongly suggest a potential role for this protein in RBC invasion, and highlight its HABPs as potential targets to develop a fully protective minimal subunit-based malarial vaccine.
Collapse
Affiliation(s)
- Jeison García
- Fundación Instituto de Inmunología de Colombia FIDIC, Carrera 50 No. 26-20, Bogotá, Colombia
| | | | | | | | | | | |
Collapse
|
32
|
Chromatin-mediated epigenetic regulation in the malaria parasite Plasmodium falciparum. EUKARYOTIC CELL 2010; 9:1138-49. [PMID: 20453074 DOI: 10.1128/ec.00036-10] [Citation(s) in RCA: 91] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Malaria is a major public health problem in many developing countries, with the malignant tertian parasite Plasmodium falciparum causing the most malaria-associated mortality. Extensive research, especially with the advancement of genomics and transfection tools, has highlighted the fundamental importance of chromatin-mediated gene regulation in the developmental program of this early-branching eukaryote. The Plasmodium parasite genomes reveal the existence of both canonical and variant histones that make up the nucleosomes, as well as a full collection of conserved enzymes for chromatin remodeling and histone posttranslational modifications (PTMs). Recent studies have identified a wide array of both conserved and novel histone PTMs in P. falciparum, indicating the presence of a complex and divergent "histone code." Genome-wide analysis has begun to decipher the nucleosome landscape and histone modifications associated with the dynamic organization of chromatin structures during the parasite's life cycle. Focused studies on malaria-specific phenomena such as antigenic variation and red cell invasion pathways shed further light on the involvement of epigenetic mechanisms in these processes. Here we review our current understanding of chromatin-mediated gene regulation in malaria parasites, with specific reference to exemplar studies on antigenic variation and host cell invasion.
Collapse
|
33
|
Kuehn A, Simon N, Pradel G. Family members stick together: multi-protein complexes of malaria parasites. Med Microbiol Immunol 2010; 199:209-26. [PMID: 20419315 DOI: 10.1007/s00430-010-0157-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2010] [Indexed: 11/24/2022]
Abstract
Malaria parasites express a broad repertoire of proteins whose expression is tightly regulated depending on the life-cycle stage of the parasite and the environment of target organs in the respective host. Transmission of malaria parasites from the human to the anopheline mosquito is mediated by intraerythrocytic sexual stages, termed gametocytes, which circulate in the peripheral blood and are essential for the spread of the tropical disease. In Plasmodium falciparum, gametocytes express numerous extracellular proteins with adhesive motifs, which might mediate important interactions during transmission. Among these is a family of six secreted proteins with adhesive modules, termed PfCCp proteins, which are highly conserved throughout the apicomplexan clade. In P. falciparum, the proteins are expressed in the parasitophorous vacuole of gametocytes and are subsequently exposed on the surface of macrogametes during parasite reproduction in the mosquito midgut. One characteristic of the family is a co-dependent expression, such that loss of all six proteins occurs if expression of one member is disrupted via gene knockout. The six PfCCp proteins interact by adhesion domain-mediated binding and thus form complexes on the sexual stage surface having adhesive properties. To date, the PfCCp proteins represent the only protein family of the malaria parasite sexual stages that assembles to multimeric complexes, and only a small number of such protein complexes have so far been identified in other life-cycle stages of the parasite.
Collapse
Affiliation(s)
- Andrea Kuehn
- Research Center for Infectious Diseases, University of Würzburg, Josef-Schneider-Strasse 2, Building D15, Würzburg, Germany
| | | | | |
Collapse
|
34
|
Pinzón CG, Curtidor H, García J, Vanegas M, Vizcaíno C, Patarroyo MA, Patarroyo ME. Sequences of the Plasmodium falciparum cytoadherence-linked asexual protein 9 implicated in malaria parasite invasion to erythrocytes. Vaccine 2010; 28:2653-63. [PMID: 20085836 DOI: 10.1016/j.vaccine.2010.01.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2009] [Revised: 12/17/2009] [Accepted: 01/06/2010] [Indexed: 10/20/2022]
Abstract
In this study, we synthesized the complete sequence of the CLAG-9 protein as 67 20-mer-long non-overlapped peptides and assessed their ability to bind to erythrocytes in receptor-ligand assays. Twenty CLAG-9 peptides were found to have specific high-affinity binding ability to erythrocytes (thereby named as HABPs), with nanomolar dissociation constants. CLAG-9 HABPs interacted with different erythrocyte surface receptors having apparent molecular weights of 85, 63 and 34 kDa. CLAG-9 HABPs binding was also affected by pre-treatment of RBCs with enzymes and inhibited erythrocyte invasion in vitro by up to 72% at 200 microM. These results suggest that some protein fragments of CLAG-9 may be part of the molecular machinery used by malaria parasites to invade erythrocytes, hence supporting their study as possible vaccine candidates.
Collapse
|
35
|
García J, Curtidor H, Pinzón CG, Patarroyo MA, Vanegas M, Forero M, Patarroyo ME. Well-Defined Regions of the Plasmodium falciparum Reticulocyte Binding Protein Homologue 4 Mediate Interaction with Red Blood Cell Membrane. J Med Chem 2009; 53:811-21. [DOI: 10.1021/jm901540n] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Jeison García
- Fundación Instituto de Inmunología de Colombia FIDIC, Carrera 50 No. 26-20, Bogotá, Colombia
- Universidad del Rosario, Calle 14 No. 6-25, Bogotá, Colombia
| | - Hernando Curtidor
- Fundación Instituto de Inmunología de Colombia FIDIC, Carrera 50 No. 26-20, Bogotá, Colombia
- Universidad del Rosario, Calle 14 No. 6-25, Bogotá, Colombia
| | - Carlos G. Pinzón
- Fundación Instituto de Inmunología de Colombia FIDIC, Carrera 50 No. 26-20, Bogotá, Colombia
- Universidad del Rosario, Calle 14 No. 6-25, Bogotá, Colombia
| | - Manuel A. Patarroyo
- Fundación Instituto de Inmunología de Colombia FIDIC, Carrera 50 No. 26-20, Bogotá, Colombia
- Universidad del Rosario, Calle 14 No. 6-25, Bogotá, Colombia
| | - Magnolia Vanegas
- Fundación Instituto de Inmunología de Colombia FIDIC, Carrera 50 No. 26-20, Bogotá, Colombia
- Universidad del Rosario, Calle 14 No. 6-25, Bogotá, Colombia
| | - Martha Forero
- Fundación Instituto de Inmunología de Colombia FIDIC, Carrera 50 No. 26-20, Bogotá, Colombia
- Universidad del Rosario, Calle 14 No. 6-25, Bogotá, Colombia
| | - Manuel E. Patarroyo
- Fundación Instituto de Inmunología de Colombia FIDIC, Carrera 50 No. 26-20, Bogotá, Colombia
- Universidad Nacional de Colombia, Carrera 45 No. 26-85, Bogotá, Colombia
| |
Collapse
|
36
|
Characterization and antigenicity of the promising vaccine candidate Plasmodium vivax 34kDa rhoptry antigen (Pv34). Vaccine 2009; 28:415-21. [PMID: 19837093 DOI: 10.1016/j.vaccine.2009.10.034] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2009] [Revised: 09/29/2009] [Accepted: 10/08/2009] [Indexed: 11/22/2022]
|
37
|
Molecular characterizations of three distinctBabesia gibsonirhoptry-associated protein-1s (RAP-1s). Parasitology 2009; 136:1147-60. [DOI: 10.1017/s003118200999045x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
SUMMARYThree cDNAs encoding rhoptry-associated protein 1 (RAP-1) homologues were found in theBabesia gibsoniEST database. Based on similarities to BgRAP-1a, which was identified previously by serological screening of a cDNA merozoite library, the two new genes were designatedBgRAP-1b(33·7%) andBgRAP-1c(57%). Mice antiserum raised against each recombinant protein reacted specifically withB. gibsoniparasites as determined by Western blotting, which showed native molecular sizes of the BgRAP-1a (51 kDa), BgRAP-1b (53 kDa) and BgRAP-1c (47 kDa) consistent with predictable molecular weights. Immunofluoresence using these antibodies revealed localization of all BgRAP-1s within the matrix of merozoites; however, BgRAP-1a appeared to diverge from the other two when it was found secreted into the cytoplasm of infected erythrocytes. Apical localization of all 3 BgRAP-1s during the extracellular stage of the parasite combined with their ability to bind a canine erythrocyte membrane fraction was suggestive of a role for these proteins in erythrocyte attachment. Lastly, the ability of these recombinant proteins to be used as diagnostic reagents was tested by ELISA and the sensitivities of BgRAP-1a and BgRAP-1c were found increased through N-terminal truncation. Taken together, our data suggest divergent roles for the 3 BgRAP-1s in the merozoite stage ofB. gibsoni.
Collapse
|
38
|
Tufet-Bayona M, Janse CJ, Khan SM, Waters AP, Sinden RE, Franke-Fayard B. Localisation and timing of expression of putative Plasmodium berghei rhoptry proteins in merozoites and sporozoites. Mol Biochem Parasitol 2009; 166:22-31. [DOI: 10.1016/j.molbiopara.2009.02.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2008] [Revised: 01/22/2009] [Accepted: 02/17/2009] [Indexed: 02/05/2023]
|
39
|
Fadiel A, Isokpehi RD, Stambouli N, Hamza A, Benammar-Elgaaied A, Scalise TJ. Protozoan parasite aquaporins. Expert Rev Proteomics 2009; 6:199-211. [PMID: 19385945 DOI: 10.1586/epr.09.10] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Protozoan parasites are a major threat to human health with millions of fatalities worldwide, especially in nonindustrialized countries. Currently, there is no cure for many of these parasitic diseases. Consequently, there is an imperative to find treatment targets and develop novel drugs based on the proteins encoded in the genomes of these parasites. Aquaporins, members of membrane proteins discovered and characterized within the past 20 years, are the mechanism through which water is transported through living membranes. The presence of aquaporins explains disease etiology related to water physiology and presents new pharmacogenomic targets. In this article, we review the literature on aquaporins found in Apicomplexan, Kinetoplastida and Microsporidia parasites as potential drug targets. Furthermore, by analyzing protein motion dynamics, we identify impediments that need to be surmounted for developing effective drugs targeting the aquaglyceroporin of Plasmodium falciparum, the causative agent of the most fatal form of human malaria.
Collapse
Affiliation(s)
- Ahmed Fadiel
- Department of OBGYN, New York University School of Medicine, Bellevue Hospital Center, New York, NY 10016, USA.
| | | | | | | | | | | |
Collapse
|
40
|
Cao J, Kaneko O, Thongkukiatkul A, Tachibana M, Otsuki H, Gao Q, Tsuboi T, Torii M. Rhoptry neck protein RON2 forms a complex with microneme protein AMA1 in Plasmodium falciparum merozoites. Parasitol Int 2008; 58:29-35. [PMID: 18952195 DOI: 10.1016/j.parint.2008.09.005] [Citation(s) in RCA: 130] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2008] [Revised: 09/15/2008] [Accepted: 09/18/2008] [Indexed: 11/18/2022]
Abstract
Erythrocyte invasion is an essential step in the establishment of host infection by malaria parasites, and is a major target of intervention strategies that attempt to control the disease. Recent proteome analysis of the closely-related apicomplexan parasite, Toxoplasma gondii, revealed a panel of novel proteins (RONs) located at the neck portion of the rhoptries. Three of these proteins, RON2, RON4, and RON5 have been shown to form a complex with the microneme protein Apical Membrane Protein 1 (AMA1). This complex, termed the Moving Junction complex, localizes at the interface of the parasite and the host cell during the invasion process. Here we characterized a RON2 ortholog in Plasmodium falciparum. PfRON2 transcription peaked at the mature schizont stage and was expressed at the neck portion of the rhoptry in the merozoite. Co-immunoprecipitation of PfRON2, PfRON4 and PfAMA1 indicated that the complex formation is conserved between T. gondii and P. falciparum, suggesting that co-operative function of the rhoptry and microneme proteins is a common mechanism in apicomplexan parasites during host cell invasion. PfRON2 possesses a region displaying homology with the rhoptry body protein PfRhopH1/Clag, a component of the RhopH complex. However, here we present co-immunoprecipitation studies which suggest that PfRON2 is not a component of the RhopH complex and has an independent role. Nucleotide polymorphism analysis suggested that PfRON2 was under diversifying selective pressure. This evidence suggests that RON2 appears to have a fundamental role in host cell invasion by apicomplexan parasites, and is a potential target for malaria intervention strategies.
Collapse
Affiliation(s)
- Jun Cao
- Department of Molecular Parasitology, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan
| | | | | | | | | | | | | | | |
Collapse
|
41
|
Cortés A. Switching Plasmodium falciparum genes on and off for erythrocyte invasion. Trends Parasitol 2008; 24:517-24. [PMID: 18805736 DOI: 10.1016/j.pt.2008.08.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2008] [Revised: 07/23/2008] [Accepted: 08/01/2008] [Indexed: 11/30/2022]
Abstract
Culture-adapted lines of the malaria parasite Plasmodium falciparum use alternative pathways for the invasion of erythrocytes. The expression of parasite ligands that are involved in the different pathways varies among parasite lines. Recently, several studies have attempted to characterize the use of different invasion pathways and the expression of specific invasion ligands in field isolates, opening the way to understand how invasion occurs in natural infections. In this review, these findings are discussed in the context of the most recent data on invasion by culture-adapted parasites to describe the current understanding of how wild parasites invade, how the variant expression of invasion ligands relates to switching between alternative invasion pathways and why so many different pathways are needed.
Collapse
Affiliation(s)
- Alfred Cortés
- ICREA and Institute for Research in Biomedicine, Barcelona Science Park, Barcelona 08028, Catalonia, Spain.
| |
Collapse
|
42
|
Rodriguez LE, Curtidor H, Urquiza M, Cifuentes G, Reyes C, Patarroyo ME. Intimate Molecular Interactions of P. falciparum Merozoite Proteins Involved in Invasion of Red Blood Cells and Their Implications for Vaccine Design. Chem Rev 2008; 108:3656-705. [DOI: 10.1021/cr068407v] [Citation(s) in RCA: 82] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
| | - Hernando Curtidor
- Fundación Instituto de Inmunología de Colombia, Carrera 50 No. 26-00, Bogotá, Colombia
| | - Mauricio Urquiza
- Fundación Instituto de Inmunología de Colombia, Carrera 50 No. 26-00, Bogotá, Colombia
| | - Gladys Cifuentes
- Fundación Instituto de Inmunología de Colombia, Carrera 50 No. 26-00, Bogotá, Colombia
| | - Claudia Reyes
- Fundación Instituto de Inmunología de Colombia, Carrera 50 No. 26-00, Bogotá, Colombia
| | | |
Collapse
|
43
|
Pinzón CG, Curtidor H, Reyes C, Méndez D, Patarroyo ME. Identification of Plasmodium falciparum RhopH3 protein peptides that specifically bind to erythrocytes and inhibit merozoite invasion. Protein Sci 2008; 17:1719-30. [PMID: 18593818 DOI: 10.1110/ps.035923.108] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
The identification of sequences involved in binding to erythrocytes is an important step for understanding the molecular basis of merozoite-erythrocyte interactions that take place during invasion of the Plasmodium falciparum malaria parasite into host cells. Several molecules located in the apical organelles (micronemes, rhoptry, dense granules) of the invasive-stage parasite are essential for erythrocyte recognition, invasion, and establishment of the nascent parasitophorous vacuole. Particularly, it has been demonstrated that rhoptry proteins play an important role in binding to erythrocyte surface receptors, among which is the PfRhopH3 protein, which triggers important immune responses in patients from endemic regions. It has also been reported that anti-RhopH3 antibodies inhibit in vitro invasion of erythrocytes, further supporting its direct involvement in erythrocyte invasion processes. In this study, PfRhopH3 consecutive peptides were synthesized and tested in erythrocyte binding assays for identifying those regions mediating binding to erythrocytes. Fourteen PfRhopH3 peptides presenting high specific binding activity were found, whose bindings were saturable and presented nanomolar dissociation constants. These high-activity binding peptides (HABPs) were characterized by having alpha-helical structural elements, as determined by circular dichroism, and having receptors of a possible sialic acid-dependent and/or glycoprotein-dependent nature, as evidenced in enzyme-treated erythrocyte binding assays and further corroborated by cross-linking assay results. Furthermore, these HABPs inhibited merozoite in vitro invasion of normal erythrocytes at 200 microM by up to 60% and 90%, suggesting that some RhopH3 protein regions are involved in the P. falciparum erythrocyte invasion.
Collapse
|
44
|
Plasmodium yoelii: novel rhoptry proteins identified within the body of merozoite rhoptries in rodent Plasmodium malaria. Exp Parasitol 2008; 120:113-7. [PMID: 18606406 DOI: 10.1016/j.exppara.2008.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2008] [Revised: 05/15/2008] [Accepted: 05/19/2008] [Indexed: 11/22/2022]
Abstract
The biogenesis, organization and function of the rhoptries are not well understood. Antisera were prepared to synthetic peptides prepared as multiple antigenic peptides (MAPs) obtained from a Plasmodium yoelii merozoite rhoptry proteome analysis. The antisera were used in immunofluorescence and immunoelectron microscopy of schizont-infected erythrocytes. Twenty-seven novel rhoptry proteins representing proteases, metabolic enzymes, secreted proteins and hypothetical proteins, were identified in the body of the rhoptries by immunoelectron microscopy. The merozoite rhoptries contain a heterogeneous mixture of proteins that may initiate host cell invasion and establish intracellular parasite development.
Collapse
|
45
|
Sherman IW. References. ADVANCES IN PARASITOLOGY 2008. [DOI: 10.1016/s0065-308x(08)00430-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
46
|
Iriko H, Kaneko O, Otsuki H, Tsuboi T, Su XZ, Tanabe K, Torii M. Diversity and evolution of the rhoph1/clag multigene family of Plasmodium falciparum. Mol Biochem Parasitol 2007; 158:11-21. [PMID: 18155305 DOI: 10.1016/j.molbiopara.2007.11.004] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2007] [Revised: 11/05/2007] [Accepted: 11/06/2007] [Indexed: 11/19/2022]
Abstract
A complex of high-molecular-mass proteins (PfRhopH) of the human malaria parasite Plasmodium falciparum induces host protective immunity and therefore is a candidate for vaccine development. Understanding the level of polymorphism and the evolutionary processes is important for advancements in both vaccine design and knowledge of the evolution of cell invasion in this parasite. In the present study, we sequenced the entire open reading frames of seven genes encoding the proteins of the PfRhopH complex (rhoph2, rhoph3, and five rhoph1/clag gene paralogs). We found that four rhoph1/clag genes (clag2, 3.1, 3.2, and 8) were highly polymorphic. Amino acid substitutions and indels are predominantly clustered around amino acid positions 1000-1200 of these four rhoph1/clag genes. An excess of nonsynonymous substitutions over synonymous substitutions was detected for clag8 and 9, indicating positive selection. The McDonald-Kreitman test with a Plasmodium reichenowi orthologous sequence also supports positive selection on clag8. Based on the ratio of interspecific genetic distance to intraspecific distance, the time to the most recent common ancestor of the clag2 and 8 polymorphisms was estimated to be 1.89 and 0.87 million years ago, respectively, assuming divergence of P. falciparum and P. reichenowi 6 million years ago. In addition to a copy number polymorphism, gene conversion events were detected for the rhoph1/clag genes on chromosome 3, which likely play a role in increasing the diversity of each locus. Our results indicate that a high diversity of the PfRhopH1/Clag multigene family is maintained by diversifying selection forces over a considerably long period.
Collapse
Affiliation(s)
- Hideyuki Iriko
- Department of Molecular Parasitology, Ehime University Graduate School of Medicine, Shitsukawa, Toon, Ehime 791-0295, Japan
| | | | | | | | | | | | | |
Collapse
|